1
|
Lee S, Kim M, Ahn BJ, Jang Y. Odorant-responsive biological receptors and electronic noses for volatile organic compounds with aldehyde for human health and diseases: A perspective review. JOURNAL OF HAZARDOUS MATERIALS 2023; 455:131555. [PMID: 37156042 DOI: 10.1016/j.jhazmat.2023.131555] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/19/2023] [Accepted: 05/01/2023] [Indexed: 05/10/2023]
Abstract
Volatile organic compounds (VOCs) are gaseous chemicals found in ambient air and exhaled breath. In particular, highly reactive aldehydes are frequently found in polluted air and have been linked to various diseases. Thus, extensive studies have been carried out to elucidate disease-specific aldehydes released from the body to develop potential biomarkers for diagnostic purposes. Mammals possess innate sensory systems, such as receptors and ion channels, to detect these VOCs and maintain physiological homeostasis. Recently, electronic biosensors such as the electronic nose have been developed for disease diagnosis. This review aims to present an overview of natural sensory receptors that can detect reactive aldehydes, as well as electronic noses that have the potential to diagnose certain diseases. In this regard, this review focuses on eight aldehydes that are well-defined as biomarkers in human health and disease. It offers insights into the biological aspects and technological advances in detecting aldehyde-containing VOCs. Therefore, this review will aid in understanding the role of aldehyde-containing VOCs in human health and disease and the technological advances for improved diagnosis.
Collapse
Affiliation(s)
- Solpa Lee
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, South Korea
| | - Minwoo Kim
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, South Korea
| | - Bum Ju Ahn
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, South Korea
| | - Yongwoo Jang
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, South Korea; Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, South Korea.
| |
Collapse
|
2
|
Electrophilic Aldehyde 4-Hydroxy-2-Nonenal Mediated Signaling and Mitochondrial Dysfunction. Biomolecules 2022; 12:biom12111555. [PMID: 36358905 PMCID: PMC9687674 DOI: 10.3390/biom12111555] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 01/21/2023] Open
Abstract
Reactive oxygen species (ROS), a by-product of aerobic life, are highly reactive molecules with unpaired electrons. The excess of ROS leads to oxidative stress, instigating the peroxidation of polyunsaturated fatty acids (PUFA) in the lipid membrane through a free radical chain reaction and the formation of the most bioactive aldehyde, known as 4-hydroxynonenal (4-HNE). 4-HNE functions as a signaling molecule and toxic product and acts mainly by forming covalent adducts with nucleophilic functional groups in proteins, nucleic acids, and lipids. The mitochondria have been implicated as a site for 4-HNE generation and adduction. Several studies clarified how 4-HNE affects the mitochondria's functions, including bioenergetics, calcium homeostasis, and mitochondrial dynamics. Our research group has shown that 4-HNE activates mitochondria apoptosis-inducing factor (AIFM2) translocation and facilitates apoptosis in mice and human heart tissue during anti-cancer treatment. Recently, we demonstrated that a deficiency of SOD2 in the conditional-specific cardiac knockout mouse increases ROS, and subsequent production of 4-HNE inside mitochondria leads to the adduction of several mitochondrial respiratory chain complex proteins. Moreover, we highlighted the physiological functions of HNE and discussed their relevance in human pathophysiology and current discoveries concerning 4-HNE effects on mitochondria.
Collapse
|
3
|
Zhang Y, Qiu Y, Zhang H. Computational Investigation of Structural Basis for Enhanced Binding of Isoflavone Analogues with Mitochondrial Aldehyde Dehydrogenase. ACS OMEGA 2022; 7:8115-8127. [PMID: 35284766 PMCID: PMC8908493 DOI: 10.1021/acsomega.2c00032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/11/2022] [Indexed: 06/14/2023]
Abstract
Isoflavone compounds are potent inhibitors against mitochondrial aldehyde dehydrogenase (ALDH2) for the treatment of alcoholism and drug addiction, and an in-depth understanding of the underlying structural basis helps design new inhibitors for enhanced binding. Here, we investigated the binding poses and strengths of eight isoflavone analogues (including CVT-10216 and daidzin) with ALDH2 via computational methods of molecular docking, molecular dynamics (MD) simulation, molecular mechanics Poisson-Boltzmann surface area (MM-PBSA), steered MD, and umbrella sampling. Neither the Vina scoring of docked and MD-sampled complexes nor the nonbonded protein-inhibitor interaction energy from MD simulations is able to reproduce the relative binding strength of the inhibitors compared to experimental IC50 values. Considering the solvation contribution, MM-PBSA and relatively expensive umbrella sampling yield good performance for the relative binding (free) energies. The isoflavone skeleton prefers to form π-π stacking, π-sulfur, and π-alkyl interactions with planar (Phe and Trp) or sulfur-containing (Cys and Met) residues. The enhanced inhibition of CVT-10216 originates from both end groups of the isoflavone skeleton offering strong van der Waals contacts and from the methylsulfonamide group at the 4' position by hydrogen bonding (HB) with neighboring receptor residues. These results indicate that the hydrophobic binding tunnel of ALDH2 is larger than the isoflavone skeleton in length and thus an extended hydrophobic core is likely a premise for potent inhibitors.
Collapse
|
4
|
Bai Y, Wang M, Yang Y, Liu X, Chen Q, Guo Z. Inhibition of the miR-193b-3p protects against oxidized low-density lipoprotein-induced HUVECs injury by upregulating ALDH2. Cell Biol Int 2021; 46:192-202. [PMID: 34719090 DOI: 10.1002/cbin.11720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 10/14/2021] [Accepted: 10/23/2021] [Indexed: 12/18/2022]
Abstract
Atherosclerosis (AS) is the most dangerous factor for human death, which is a lipid-driven chronic inflammatory disorder of the arteries. Growing evidence has showed that microRNAs play an important role in AS. However, the role of mir-193b-3p in atherosclerosis has been poorly studied to date. Therefore, we focused on the potential role of miR-193b-3p in atherosclerosis. The expressions of miR-193b-3p in the serum of AS patients were detected. We also established an oxidized low density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) apoptosis model in vitro. The mRNA and protein levels of target molecules were detected by RT-qPCR and Western blotting. Apoptosis of HUVECs was determined by Annexin V/PI staining on a flow cytometry. The potential molecular targets of miR-193b-3p were investigated by applying such technologies as dual-luciferase reporter and RIP assay. Our study showed that miR-193b-3p expression level was significantly lower in AS patients than controls. ROC curve analysis showed that the areas under the curve (AUC) of plasma miR-193b-3p was 0.859. We also found that miR-193b-3p was decreased in ox-LDL-induced HUVECs and knockdown of miR-193b-3p suppressed ox-LDL-induced HUVECs injury. By using bioinformatics analysis, aldehyde dehydrogenase (ALDH2) was predicted as a downstream target of miR-193b-3p. The ALDH2 gene is also involved in the development of atherosclerosis. Meanwhile, inhibition of miR-193b-3p and ALDH2 protects ox-LDL-induced HUVECs against endoplasmic-reticulum (ER) stress. In conclusion, inhibition of miR-193b-3p was able to suppress ox-LDL-induced injury in AS through targeting ALDH2 and reducing ER stress.
Collapse
Affiliation(s)
- Yunpeng Bai
- Department of cardiovascular surgery, Chest Hospital, Tianjin University, Tianjin, China.,Department of cardiovascular surgery, Tianjin Chest Hospital, Tianjin Medical University, Tianjin, China
| | - Mei Wang
- Department of Dermatology, Tianjin First Central Hospital, Tianjin, China.,Graduate School of Tianjin Medical University, Tianjin, China
| | - Yin Yang
- Department of cardiovascular surgery, Tianjin Chest Hospital, Tianjin Medical University, Tianjin, China
| | - Xiankun Liu
- Department of cardiovascular surgery, Tianjin Chest Hospital, Tianjin Medical University, Tianjin, China.,Graduate School of Tianjin Medical University, Tianjin, China
| | - Qingliang Chen
- Department of cardiovascular surgery, Tianjin Chest Hospital, Tianjin Medical University, Tianjin, China
| | - Zhigang Guo
- Department of cardiovascular surgery, Tianjin Chest Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
5
|
Endo S, Matsunaga T, Nishinaka T. The Role of AKR1B10 in Physiology and Pathophysiology. Metabolites 2021; 11:332. [PMID: 34063865 PMCID: PMC8224097 DOI: 10.3390/metabo11060332] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
AKR1B10 is a human nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reductase belonging to the aldo-keto reductase (AKR) 1B subfamily. It catalyzes the reduction of aldehydes, some ketones and quinones, and interacts with acetyl-CoA carboxylase and heat shock protein 90α. The enzyme is highly expressed in epithelial cells of the stomach and intestine, but down-regulated in gastrointestinal cancers and inflammatory bowel diseases. In contrast, AKR1B10 expression is low in other tissues, where the enzyme is upregulated in cancers, as well as in non-alcoholic fatty liver disease and several skin diseases. In addition, the enzyme's expression is elevated in cancer cells resistant to clinical anti-cancer drugs. Thus, growing evidence supports AKR1B10 as a potential target for diagnosing and treating these diseases. Herein, we reviewed the literature on the roles of AKR1B10 in a healthy gastrointestinal tract, the development and progression of cancers and acquired chemoresistance, in addition to its gene regulation, functions, and inhibitors.
Collapse
Affiliation(s)
- Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu 502-8585, Japan;
| | - Toru Nishinaka
- Laboratory of Biochemistry, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan;
| |
Collapse
|
6
|
Gianazza E, Brioschi M, Martinez Fernandez A, Casalnuovo F, Altomare A, Aldini G, Banfi C. Lipid Peroxidation in Atherosclerotic Cardiovascular Diseases. Antioxid Redox Signal 2021; 34:49-98. [PMID: 32640910 DOI: 10.1089/ars.2019.7955] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Atherosclerotic cardiovascular diseases (ACVDs) continue to be a primary cause of mortality worldwide in adults aged 35-70 years, occurring more often in countries with lower economic development, and they constitute an ever-growing global burden that has a considerable socioeconomic impact on society. The ACVDs encompass diverse pathologies such as coronary artery disease and heart failure (HF), among others. Recent Advances: It is known that oxidative stress plays a relevant role in ACVDs and some of its effects are mediated by lipid oxidation. In particular, lipid peroxidation (LPO) is a process under which oxidants such as reactive oxygen species attack unsaturated lipids, generating a wide array of oxidation products. These molecules can interact with circulating lipoproteins, to diffuse inside the cell and even to cross biological membranes, modifying target nucleophilic sites within biomolecules such as DNA, lipids, and proteins, and resulting in a plethora of biological effects. Critical Issues: This review summarizes the evidence of the effect of LPO in the development and progression of atherosclerosis-based diseases, HF, and other cardiovascular diseases, highlighting the role of protein adduct formation. Moreover, potential therapeutic strategies targeted at lipoxidation in ACVDs are also discussed. Future Directions: The identification of valid biomarkers for the detection of lipoxidation products and adducts may provide insights into the improvement of the cardiovascular risk stratification of patients and the development of therapeutic strategies against the oxidative effects that can then be applied within a clinical setting.
Collapse
Affiliation(s)
- Erica Gianazza
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| | - Maura Brioschi
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| | | | | | | | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Cristina Banfi
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| |
Collapse
|
7
|
Uman S, Wang LL, Thorn SL, Liu Z, Duncan JS, Sinusas AJ, Burdick JA. Imaging of Injectable Hydrogels Delivered into Myocardium with SPECT/CT. Adv Healthc Mater 2020; 9:e2000294. [PMID: 32543053 PMCID: PMC7482444 DOI: 10.1002/adhm.202000294] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/22/2020] [Indexed: 12/11/2022]
Abstract
Injectable hydrogels are being widely explored for treatment after myocardial infarction (MI) through mechanical bulking or the delivery of therapeutics. Despite this interest, there have been few approaches to image hydrogels upon injection to identify their location, volume, and pattern of delivery, features that are important to understand toward clinical translation. Using a hyaluronic acid (HA) hydrogel as an example, the aim of this study is to introduce radiopacity to hydrogels by encapsulating a clinically used contrast agent (Omnipaque Iohexol, GE Healthcare) for imaging upon placement in the myocardium. Specifically, iohexol is encapsulated into shear-thinning and self-healing hydrogels formed through the mixing of HA-hydrazide and HA-aldehyde. Upon examination of a range of iohexol concentrations, a concentration of 100 mg mL-1 iohexol is deemed optimal based on the greatest contrast, while maintaining hydrogel mechanical properties and acceptable injection forces. In an acute porcine model of MI, hybrid single-photon emission computed tomography/computed tomography (SPECT/CT) perfusion imaging is performed immediately and 3-4 days after hydrogel delivery to assess radiopacity and verify the hydrogel location within the perfusion defect. Hybrid SPECT/CT imaging demonstrates excellent radiopacity of the hydrogel within the perfusion defect immediately after intramyocardial hydrogel injection, demonstrating the feasibility of this method for short-term noninvasive hydrogel monitoring.
Collapse
Affiliation(s)
- Selen Uman
- Department of Bioengineering, University of Pennsylvania, 210 33rd Street, Philadelphia, PA, 19104, USA
| | - Leo L Wang
- Department of Bioengineering, University of Pennsylvania, 210 33rd Street, Philadelphia, PA, 19104, USA
| | - Stephanie L Thorn
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, Yale Translational Research Imaging Center, DANA-3, P.O. Box 208017, New Haven, CT, 06520-8017, USA
| | - Zhao Liu
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, Yale Translational Research Imaging Center, DANA-3, P.O. Box 208017, New Haven, CT, 06520-8017, USA
| | - James S Duncan
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, 06519, USA
| | - Albert J Sinusas
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, Yale Translational Research Imaging Center, DANA-3, P.O. Box 208017, New Haven, CT, 06520-8017, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, 210 33rd Street, Philadelphia, PA, 19104, USA
| |
Collapse
|
8
|
Ribeiro DM, Planchon S, Leclercq CC, Dentinho MTP, Bessa RJB, Santos-Silva J, Paulos K, Jerónimo E, Renaut J, Almeida AM. The effects of improving low dietary protein utilization on the proteome of lamb tissues. J Proteomics 2020; 223:103798. [PMID: 32380293 DOI: 10.1016/j.jprot.2020.103798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/03/2020] [Accepted: 04/25/2020] [Indexed: 02/06/2023]
Abstract
Cistus ladanifer L. is a common shrub endemic to the Mediterranean region with high levels of condensed tannins (CT). CT form complexes with dietary protein resisting microbial degradation in the rumen, which enhances dietary protein utilization in ruminant diets. The objective of this study was to evaluate the utilization of CT in the diet of lambs on the proteomes of muscle, hepatic and adipose tissues. Twenty-four Merino Branco ram lambs were randomly allocated to three treatments (n = 8): C - control (160 g crude protein (CP)) per kg DM, RP - reduced protein (120 g CP/kg DM); and RPCT - reduced protein (120 g CP/kg DM) treated with CT extract. At the end of the trial, lambs were slaughtered and the longissimus lumborum muscle, hepatic and peri-renal adipose tissues sampled. A two-way approach was used for proteomic analysis: 2D-DIGE and nanoLC-MS. In the muscle, C lambs had lower abundance proteins that partake in the glycolysis pathway than the lambs of other treatments. Control lambs had lower abundance of Fe-carrying proteins in the hepatic tissue than RP and RPCT lambs. The latter lambs had highest abundance of hepatic flavin reductase. In the adipose tissue, C lambs had lowest abundance of fatty-acid synthase. SIGNIFICANCE: soybean meal is an expensive feedstuff in which intensive animal production systems heavily rely on. It is a source of protein extensively degraded in the rumen, leading to efficiency losses on dietary protein utilization during digestion. Protection of dietary protein from extensive ruminal degradation throughout the use of plants or extracts rich in CT allow an increase in the digestive utilization of feed proteins. In addition to enhance the protein digestive utilization, dietary CT may induce other beneficial effects in ruminants such as the improvement of the antioxidant status.
Collapse
Affiliation(s)
- D M Ribeiro
- LEAF Linking Landscape, Environment, Agriculture and Food, Instituto Superior de Agronomia, University of Lisbon, Lisboa, Portugal; Luxembourg Institute of Science and Technology (LIST), Green Tech platform, Environmental Research and Innovation Department (ERIN), L-4422 Belvaux, Luxembourg
| | - S Planchon
- Luxembourg Institute of Science and Technology (LIST), Green Tech platform, Environmental Research and Innovation Department (ERIN), L-4422 Belvaux, Luxembourg
| | - C C Leclercq
- Luxembourg Institute of Science and Technology (LIST), Green Tech platform, Environmental Research and Innovation Department (ERIN), L-4422 Belvaux, Luxembourg
| | - M T P Dentinho
- CIISA - Centro Interdisciplinar de Investigação em Sanidade Animal, Faculdade de Medicina Veterinária, Av. Univ. Técnica, Lisboa, Portugal; Instituto Nacional de Investigação Agrária e Veterinária, Pólo Investigação da Fonte Boa (INIAV-Fonte Boa), 2005-048 Santarém, Portugal
| | - R J B Bessa
- CIISA - Centro Interdisciplinar de Investigação em Sanidade Animal, Faculdade de Medicina Veterinária, Av. Univ. Técnica, Lisboa, Portugal
| | - J Santos-Silva
- CIISA - Centro Interdisciplinar de Investigação em Sanidade Animal, Faculdade de Medicina Veterinária, Av. Univ. Técnica, Lisboa, Portugal; Instituto Nacional de Investigação Agrária e Veterinária, Pólo Investigação da Fonte Boa (INIAV-Fonte Boa), 2005-048 Santarém, Portugal
| | - K Paulos
- Instituto Nacional de Investigação Agrária e Veterinária, Pólo Investigação da Fonte Boa (INIAV-Fonte Boa), 2005-048 Santarém, Portugal
| | - E Jerónimo
- Centro de Biotecnologia Agrícola e Agro-Alimentar do Alentejo (CEBAL)/Instituto Politécnico de Beja (IPBeja), 7801-908 Beja, Portugal; MED - Mediterranean Institute for Agriculture, Environment and Development, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal
| | - J Renaut
- Luxembourg Institute of Science and Technology (LIST), Green Tech platform, Environmental Research and Innovation Department (ERIN), L-4422 Belvaux, Luxembourg
| | - A M Almeida
- LEAF Linking Landscape, Environment, Agriculture and Food, Instituto Superior de Agronomia, University of Lisbon, Lisboa, Portugal.
| |
Collapse
|
9
|
Scollo E, Neville DC, Oruna-Concha MJ, Trotin M, Cramer R. UHPLC–MS/MS analysis of cocoa bean proteomes from four different genotypes. Food Chem 2020; 303:125244. [DOI: 10.1016/j.foodchem.2019.125244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/18/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023]
|
10
|
Longo V, Forleo A, Capone S, Scoditti E, Carluccio MA, Siciliano P, Massaro M. In vitro profiling of endothelial volatile organic compounds under resting and pro-inflammatory conditions. Metabolomics 2019; 15:132. [PMID: 31583479 DOI: 10.1007/s11306-019-1602-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The evaluation of volatile organic compounds(VOCs) emitted by human body offers a unique tool to set up new non-invasive devices for early diagnosis and long-lasting monitoring of most human diseases. However, their cellular origin and metabolic fate have not been completely elucidated yet, thus limiting their clinical application. Endothelium acts as an interface between blood and surrounding tissues. As such, it adapts its physiology in response to different environmental modifications thus playing a role in the pathogenesis of many metabolic and inflammatory diseases. OBJECTIVES Since endothelium specifically reshapes its physiologic functions upon environmental changes the objective of this study was to evaluate if and how pro-inflammatory stimuli affect VOC metabolism in endothelial cell in culture. METHODS Gas chromatography with mass spectrometric detection was applied to profile VOCs in the headspace of cultured endothelial cells (EC) in the absence or presence of the pro-inflammatory stimulus lipopolysaccharide (LPS). RESULTS We observed that, under resting conditions, EC affected the amount of 58 VOCs belonging to aldehyde, alkane and ketone families. Among these, LPS significantly altered the amount of 15 VOCs. ROC curves show a perfect performance (AUC = 1) for 10 metabolites including 1-butanol, 3-methyl-1-butanol and 2-ethyl-1-hexanol. DISCUSSION The emission and uptake of the aforementioned VOCs disclose potential unexplored metabolic pathways for EC that deserve to be investigated. Overall, we identified new candidate VOC potentially exploitable, upon experimental confirm in in vivo model of disease, as potential biomarkers of sepsis and pro-inflammatory clinical settings.
Collapse
Affiliation(s)
- V Longo
- National Research Council of Italy, Institute for Microelectronics and Microsystems, Lecce, Italy.
| | - A Forleo
- National Research Council of Italy, Institute for Microelectronics and Microsystems, Lecce, Italy
| | - S Capone
- National Research Council of Italy, Institute for Microelectronics and Microsystems, Lecce, Italy
| | - E Scoditti
- National Research Council of Italy, Institute of Clinical Physiology, Lecce, Italy
| | - M A Carluccio
- National Research Council of Italy, Institute of Clinical Physiology, Lecce, Italy
| | - P Siciliano
- National Research Council of Italy, Institute for Microelectronics and Microsystems, Lecce, Italy
| | - M Massaro
- National Research Council of Italy, Institute of Clinical Physiology, Lecce, Italy.
| |
Collapse
|
11
|
Casin KM, Fallica J, Mackowski N, Veenema RJ, Chan A, St Paul A, Zhu G, Bedja D, Biswal S, Kohr MJ. S-Nitrosoglutathione Reductase Is Essential for Protecting the Female Heart From Ischemia-Reperfusion Injury. Circ Res 2019; 123:1232-1243. [PMID: 30571462 DOI: 10.1161/circresaha.118.313956] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
RATIONALE Protein S-nitros(yl)ation (SNO) has been implicated as an essential mediator of nitric oxide-dependent cardioprotection. Compared with males, female hearts exhibit higher baseline levels of protein SNO and associated with this, reduced susceptibility to myocardial ischemia-reperfusion injury. Female hearts also exhibit enhanced S-nitrosoglutathione reductase (GSNO-R) activity, which would typically favor decreased SNO levels as GSNO-R mediates SNO catabolism. OBJECTIVE Because female hearts exhibit higher SNO levels, we hypothesized that GSNO-R is an essential component of sex-dependent cardioprotection in females. METHODS AND RESULTS Male and female wild-type mouse hearts were subjected to ex vivo ischemia-reperfusion injury with or without GSNO-R inhibition (N6022). Control female hearts exhibited enhanced functional recovery and decreased infarct size versus control males. Interestingly, GSNO-R inhibition reversed this sex disparity, significantly reducing injury in male hearts, and exacerbating injury in females. Similar results were obtained with male and female GSNO-R-/- hearts using ex vivo and in vivo models of ischemia-reperfusion injury. Assessment of SNO levels using SNO-resin assisted capture revealed an increase in total SNO levels with GSNO-R inhibition in males, whereas total SNO levels remained unchanged in females. However, we found that although GSNO-R inhibition significantly increased SNO at the cardioprotective Cys39 residue of nicotinamide adenine dinucleotide (NADH) dehydrogenase subunit 3 in males, SNO-NADH dehydrogenase subunit 3 levels were surprisingly reduced in N6022-treated female hearts. Because GSNO-R also acts as a formaldehyde dehydrogenase, we examined postischemic formaldehyde levels and found that they were nearly 2-fold higher in N6022-treated female hearts compared with nontreated hearts. Importantly, the mitochondrial aldehyde dehydrogenase 2 activator, Alda-1, rescued the phenotype in GSNO-R-/- female hearts, significantly reducing infarct size. CONCLUSIONS These striking findings point to GSNO-R as a critical sex-dependent mediator of myocardial protein SNO and formaldehyde levels and further suggest that different therapeutic strategies may be required to combat ischemic heart disease in males and females.
Collapse
Affiliation(s)
- Kevin M Casin
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Jonathan Fallica
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Nathan Mackowski
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Ryne J Veenema
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Ashley Chan
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Amanda St Paul
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Guangshuo Zhu
- Cardiology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (G.Z., D.B.)
| | - Djahida Bedja
- Cardiology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (G.Z., D.B.)
| | - Shyam Biswal
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| | - Mark J Kohr
- From the Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.C., J.F., N.M., R.J.V., A.C., A.S.P., S.B., M.J.K.)
| |
Collapse
|
12
|
Jin L, Jagatheesan G, Guo L, Nystoriak M, Malovichko M, Lorkiewicz P, Bhatnagar A, Srivastava S, Conklin DJ. Formaldehyde Induces Mesenteric Artery Relaxation via a Sensitive Transient Receptor Potential Ankyrin-1 (TRPA1) and Endothelium-Dependent Mechanism: Potential Role in Postprandial Hyperemia. Front Physiol 2019; 10:277. [PMID: 30984013 PMCID: PMC6448550 DOI: 10.3389/fphys.2019.00277] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/04/2019] [Indexed: 11/30/2022] Open
Abstract
Formaldehyde (FA), the smallest aldehyde, is generated endogenously, and is widespread in the environment in foods, beverages and as a gas phase product of incomplete combustion. The main metabolite of FA, formate, was increased significantly in murine urine (∼3×) after overnight feeding. Because feeding increases mesenteric blood flow, we explored the direct effects of FA in isolated murine superior mesenteric artery (SMA). Over the concentration range of 30–1,200 μM, FA strongly and reversibly relaxed contractions of SMA induced by three different agonists: phenylephrine (PE), thromboxane A2 analog (U46,619) and high potassium (60K, 60 mM K+). Formate (to 1.5 mM) induced a modest relaxation. FA (>1,500 μM) irreversibly depressed vascular function in SMA indicating vasotoxicity. The sensitivity (EC50) but not the efficacy (% relaxation) of FA-induced relaxations was dependent on blood vessel type (SMA << aorta) and contractile agonist (PE, EC50= 52 ± 14 μM; U46,619, EC50= 514 ± 129 μM; 60K, EC50= 1,093 ± 87 μM). The most sensitive component of FA vasorelaxation was within physiological levels (30–150 μM) and was inhibited significantly by: (1) mechanically impaired endothelium; (2) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME); (3) transient receptor potential ankyrin-1 (TRPA1) antagonist (A967079); (4) guanylyl cyclase (GC) inhibitor (ODQ); and, (5) K+ channel inhibitor (BaCl2). A similar mechanism of SMA vasorelaxation was stimulated by the TRPA1 agonist cinnamaldehyde. Positive TRPA1 immunofluorescent staining and gene-specific sequence were present in SMA but not in aorta. These data indicate FA, but not formate, robustly relaxes SMA via a sensitive TRPA1- and endothelium-dependent mechanism that is absent in aorta. Thus, as FA levels increase with feeding, FA likely contributes to the physiological reflex of post-prandial hyperemia via SMA vasodilatation.
Collapse
Affiliation(s)
- L Jin
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, United States
| | - G Jagatheesan
- Envirome Institute, University of Louisville, Louisville, KY, United States.,Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States.,American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, United States
| | - L Guo
- Envirome Institute, University of Louisville, Louisville, KY, United States.,Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States.,American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, United States
| | - M Nystoriak
- Envirome Institute, University of Louisville, Louisville, KY, United States.,Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States.,American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, United States
| | - M Malovichko
- Envirome Institute, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States.,American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, United States
| | - P Lorkiewicz
- Envirome Institute, University of Louisville, Louisville, KY, United States.,Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States.,American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, United States
| | - A Bhatnagar
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, United States.,Envirome Institute, University of Louisville, Louisville, KY, United States.,Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States.,American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, United States
| | - S Srivastava
- Envirome Institute, University of Louisville, Louisville, KY, United States.,Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States.,American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, United States
| | - D J Conklin
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, United States.,Envirome Institute, University of Louisville, Louisville, KY, United States.,Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States.,American Heart Association Tobacco Regulation and Addiction Center, University of Louisville, Louisville, KY, United States
| |
Collapse
|
13
|
Wei S, Zhang L, Bailu Wang, Zhao Y, Dong Q, Pan C, Li C, He D, Yuan Q, Xu F, Chen Y. ALDH2 deficiency inhibits Ox-LDL induced foam cell formation via suppressing CD36 expression. Biochem Biophys Res Commun 2019; 512:41-48. [PMID: 30853183 DOI: 10.1016/j.bbrc.2019.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 02/02/2019] [Indexed: 12/31/2022]
Abstract
Foam cell formation plays an important role in the initiation and progression of atherosclerosis. Aldehyde dehydrogenase 2 (ALDH2), a key enzyme for aldehyde metabolism, is associated with coronary artery disease and affects atherosclerotic plaque vulnerability. However, the role of ALDH2 in foam cell formation remains unclear. Using peritoneal macrophages from ALDH2-deficient and control mice, we found that ALDH2 deficiency suppressed foam cell formation induced by oxidized low-density lipoproteins (ox-LDL) but not acetylated low-density lipoproteins (ac-LDL) ex vivo. After incubation with ox-LDL, ALDH2-deficient macrophages expressed lower levels of CD36 but the expression of other lipid metabolism-related proteins including SRA, LOX-1, ABCA-1, ABCG-1 and ACAT-1 was not changed in ALDH2-/- macrophages. Using CD36 inhibitor, we confirmed that CD36 contributes to the effect of ALDH2 on foam cell formation. PPARγ was downregulated in ox-LDL treated ALDH2-/- macrophages. 4-HNE was increased by ALDH2 deficiency and high concentration of 4-HNE suppressed the expression of PPARγ. These data suggest that ALDH2 plays an important role in foam cell formation via 4-HNE/PPARγ/CD36 pathway.
Collapse
Affiliation(s)
- Shujian Wei
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Luetao Zhang
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Bailu Wang
- Clinical Trial Center, Qilu Hospital, Shandong University, Jinan, China
| | - Yu Zhao
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Qianqian Dong
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Chang Pan
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Chuanbao Li
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Dayu He
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Qiuhuan Yuan
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China.
| | - Yuguo Chen
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China; Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China; Institute of Emergency and Critical Care Medicine, Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China.
| |
Collapse
|
14
|
Han C, Gao L, Zhao L, Sheng Q, Zhang C, An Z, Xia T, Ding Y, Wang J, Bai H, Dou X. Immunohistochemistry Detects Increased Expression of Aldo-Keto Reductase Family 1 Member B10 (AKR1B10) in Early-Stage Hepatocellular Carcinoma. Med Sci Monit 2018; 24:7414-7423. [PMID: 30328412 PMCID: PMC6201704 DOI: 10.12659/msm.910738] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) remains difficult to diagnose at an early stage. Aldo-keto reductase family 1 member B10 (AKR1B10) is an oxidoreductase that is upregulated in some chronic liver diseases. The aim of this study was to use immunohistochemistry to evaluate the expression of AKR1B10 in liver tissue from patients with HCC of different stages. Material/Methods Forty-four patients with a tissue diagnosis of HCC (35 males and 9 females) with 37 control samples of liver tissue containing liver cirrhosis were studied using immunohistochemistry for the expression of AKR1B10. Histological examination determined the grade of HCC; the stage of HCC was determined according to the Barcelona Clinic Liver Cancer (BCLC) staging system. Serum alpha-fetoprotein (AFP) levels were measured and compared between the patients with HCC. Results Immunohistochemistry showed increased expression of AKR1B10 in moderately-differentiated HCC compared with well-differentiated HCC, poorly-differentiated HCC, and liver cirrhosis (P<0.05). Sensitivity and specificity of AKR1B10 expression in HCC were high at a cutoff integral optical density (IOD) value of 89.5. A significant increase in AKR1B10 expression was found in early-stage HCC (P<0.05). Serum AFP levels were increased in patients with poorly-differentiated HCC, were increased in intermediate-stage HCC, and were significantly increased in advanced-stage HCC (P<0.05). Conclusions Immunohistochemistry showed that the expression of AKR1B10 was increased in tumor tissue from patients with early-stage HCC. Further studies are needed to determine the role of AKR1B10 in the early detection of HCC.
Collapse
Affiliation(s)
- Chao Han
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Lanzhu Gao
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland).,Department of Infectious Diseases, Tongliao Infectious Diseases Hospital, Tongliao, Inner Mongolia, China (mainland)
| | - Lianrong Zhao
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Qiuju Sheng
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Chong Zhang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Ziying An
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Tingting Xia
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yang Ding
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Jingyan Wang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Han Bai
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Xiaoguang Dou
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
15
|
Han C, Gao L, Bai H, Dou X. Identification of a role for serum aldo-keto reductase family 1 member B10 in early detection of hepatocellular carcinoma. Oncol Lett 2018; 16:7123-7130. [PMID: 30546447 PMCID: PMC6256343 DOI: 10.3892/ol.2018.9547] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022] Open
Abstract
Despite improved screening programs, the vast majority of patients with hepatocellular carcinoma (HCC) are diagnosed at an advanced stage. A lack of effective diagnosis methods for preclinical HCC has resulted in a low rate of early detection. Aldo-keto reductase family 1 member B10 (AKR1B10) is associated with several cancer types. However, to the best of our knowledge, the diagnostic value of AKR1B10 in early stage HCC is poorly understood. In the current study, the diagnostic performance of serum AKR1B10 in hepatitis B virus/hepatitis C virus (HBV/HCV)-related liver disorders was evaluated and the unique role of AKR1B10 in diagnosing HCC was assessed. Serum AKR1B10 was detected by sandwich ELISA in 84 patients with HBV/HCV-related HCC, 74 patients with liver cirrhosis, 29 patients with chronic hepatitis and 30 healthy controls. Serum AKR1B10 and α-fetoprotein (AFP) levels were analyzed and compared. Elevated levels of serum AKR1B10 were identified in patients with HCC compared with patients with other liver disorders (P<0.05). Compared with advanced and terminal stage HCC, a significant increase in AKR1B10 levels was primarily detected in early and intermediate stage HCC. The sensitivity (81.0%) and specificity (60.9%) for HCC diagnosis with AKR1B10 were high at a cutoff value of 1.51 ng/ml. Conversely, a prominent increase in AFP was observed in advanced and terminal stage HCC. Furthermore, concurrent measurement of serum AKR1B10 and AFP significantly increased sensitivity and negative predictive value for HCC diagnosis. The results presented in the current study strongly indicate AKR1B10 has a unique role as a biomarker for early stage HBV/HCV-related HCC. Compared with AFP alone, a combination of serum AKR1B10 and AFP increased the diagnostic performance in patients with HCC. In summary, the current results identify a unique role of AKR1B10 in HCC diagnosis.
Collapse
Affiliation(s)
- Chao Han
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Lanzhu Gao
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China.,Department of Infectious Diseases, Tongliao Infectious Diseases Hospital, Tongliao, Inner Mongolia Autonomous Region 028000, P.R. China
| | - Han Bai
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Xiaoguang Dou
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| |
Collapse
|
16
|
Chen Y, Zhou CF, Xiao F, Huang HL, Zhang P, Gu HF, Tang XQ. Inhibition of ALDH2 protects PC12 cells against formaldehyde-induced cytotoxicity: involving the protection of hydrogen sulphide. Clin Exp Pharmacol Physiol 2017; 44:595-601. [DOI: 10.1111/1440-1681.12741] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 02/11/2017] [Accepted: 02/23/2017] [Indexed: 01/28/2023]
Affiliation(s)
- Ying Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study; Institute of Pharmacy and Pharmacology; University of South China; Hengyang Hunan China
- Institute of Neuroscience; Medical College; University of South China; Hengyang Hunan China
| | - Cheng-Fang Zhou
- Institute of Neuroscience; Medical College; University of South China; Hengyang Hunan China
| | - Fan Xiao
- Institute of Neuroscience; Medical College; University of South China; Hengyang Hunan China
| | - Hong-Lin Huang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study; Institute of Pharmacy and Pharmacology; University of South China; Hengyang Hunan China
| | - Ping Zhang
- Department of Neurology; Nanhua Affiliated Hospital; University of South China; Hengyang Hunan China
| | - Hong-Feng Gu
- Institute of Neuroscience; Medical College; University of South China; Hengyang Hunan China
| | - Xiao-Qing Tang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study; Institute of Pharmacy and Pharmacology; University of South China; Hengyang Hunan China
- Institute of Neuroscience; Medical College; University of South China; Hengyang Hunan China
| |
Collapse
|
17
|
Liu XZ, Sun X, Shen KP, Jin WJ, Fu ZY, Tao HR, Xu ZX. Aldehyde dehydrogenase 2 overexpression inhibits neuronal apoptosis after spinal cord ischemia/reperfusion injury. Neural Regen Res 2017; 12:1166-1171. [PMID: 28852401 PMCID: PMC5558498 DOI: 10.4103/1673-5374.211198] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is an important factor in inhibiting oxidative stress and has been shown to protect against renal ischemia/reperfusion injury. Therefore, we hypothesized that ALDH2 could reduce spinal cord ischemia/reperfusion injury. Spinal cord ischemia/reperfusion injury was induced in rats using the modified Zivin's method of clamping the abdominal aorta. After successful model establishment, the agonist group was administered a daily consumption of 2.5% alcohol. At 7 days post-surgery, the Basso, Beattie, and Bresnahan score significantly increased in the agonist group compared with the spinal cord ischemia/reperfusion injury group. ALDH2 expression also significantly increased and the number of apoptotic cells significantly decreased in the agonist group than in the spinal cord ischemia/reperfusion injury group. Correlation analysis revealed that ALDH2 expression negatively correlated with the percentage of TUNEL-positive cells (r = −0.485, P < 0.01). In summary, increased ALDH2 expression protected the rat spinal cord against ischemia/reperfusion injury by inhibiting apoptosis.
Collapse
Affiliation(s)
- Xing-Zhen Liu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Sun
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kang-Ping Shen
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Jie Jin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Yi Fu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai-Rong Tao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Xing Xu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Cross-talk between lipid and protein carbonylation in a dynamic cardiomyocyte model of mild nitroxidative stress. Redox Biol 2016; 11:438-455. [PMID: 28086193 PMCID: PMC5226815 DOI: 10.1016/j.redox.2016.12.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen and nitrogen species (ROS/RNS) play an important role in the regulation of cardiac function. Increase in ROS/RNS concentration results in lipid and protein oxidation and is often associated with onset and/or progression of many cardiovascular disorders. However, interplay between lipid and protein modifications has not been simultaneously studied in detail so far. Biomolecule carbonylation is one of the most common biomarkers of oxidative stress. Using a dynamic model of nitroxidative stress we demonstrated rapid changes in biomolecule carbonylation in rat cardiomyocytes. Levels of carbonylated species increased as early as 15min upon treatment with the peroxynitrite donor, 3-morpholinosydnonimine (SIN-1), and decreased to values close to control after 16h. Total (lipids+proteins) vs. protein-specific carbonylation showed different dynamics, with a significant increase in protein-bound carbonyls at later time points. Treatment with SIN-1 in combination with inhibitors of proteasomal and autophagy/lysosomal degradation pathways allowed confirmation of a significant role of the proteasome in the degradation of carbonylated proteins, whereas lipid carbonylation increased in the presence of autophagy/lysosomal inhibitors. Electrophilic aldehydes and ketones formed by lipid peroxidation were identified and relatively quantified using LC-MS/MS. Molecular identity of reactive species was used for data-driven analysis of their protein targets. Combination of different enrichment strategies with LC-MS/MS analysis allowed identification of more than 167 unique proteins with 332 sites modified by electrophilic lipid peroxidation products. Gene ontology analysis of modified proteins demonstrated enrichment of several functional categories including proteins involved in cytoskeleton, extracellular matrix, ion channels and their regulation. Using calcium mobilization assays, the effect of nitroxidative stress on the activity of several ion channels was further confirmed.
Collapse
|
19
|
Li M, Zhang P, Wei HJ, Li MH, Zou W, Li X, Gu HF, Tang XQ. Hydrogen Sulfide Ameliorates Homocysteine-Induced Cognitive Dysfunction by Inhibition of Reactive Aldehydes Involving Upregulation of ALDH2. Int J Neuropsychopharmacol 2016; 20:305-315. [PMID: 27988490 PMCID: PMC5409037 DOI: 10.1093/ijnp/pyw103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/02/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Homocysteine, a risk factor for Alzheimer's disease, induces cognitive dysfunction. Reactive aldehydes play an important role in cognitive dysfunction. Aldehyde-dehydrogenase 2 detoxifies reactive aldehydes. Hydrogen sulfide, a novel neuromodulator, has neuroprotective effects and regulates learning and memory. Our previous work confirmed that the disturbance of hydrogen sulfide synthesis is invovled in homocysteine-induced defects in learning and memory. Therefore, the present work was to explore whether hydrogen sulfide ameliorates homocysteine-generated cognitive dysfunction and to investigate whether its underlying mechanism is related to attenuating accumulation of reactive aldehydes by upregulation of aldehyde-dehydrogenase 2. METHODS The cognitive function of rats was assessed by the Morris water maze test and the novel object recognition test. The levels of malondialdehyde, 4-hydroxynonenal, and glutathione as well as the activity of aldehyde-dehydrogenase 2 were determined by enzyme linked immunosorbent assay; the expression of aldehyde-dehydrogenase 2 was detected by western blot. RESULTS The behavior experiments, Morris water maze test and novel objects recognition test, showed that homocysteine induced deficiency in learning and memory in rats, and this deficiency was reversed by treatment of NaHS (a donor of hydrogen sulfide). We demonstrated that NaHS inhibited homocysteine-induced increases in generations of MDA and 4-HNE in the hippocampus of rats and that hydrogen sulfide reversed homocysteine-induced decreases in the level of glutathione as well as the activity and expression of aldehyde-dehydrogenase 2 in the hippocampus of rats. CONCLUSION Hydrogen sulfide ameliorates homocysteine-induced impairment in cognitive function by decreasing accumulation of reactive aldehydes as a result of upregulations of glutathione and aldehyde-dehydrogenase 2.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, Nanhua Affiliated Hospital (Ms Li, Mr Zhang, Ms Li, Mr Zou, and Dr Tang), and
| | - Ping Zhang
- Department of Neurology, Nanhua Affiliated Hospital (Ms Li, Mr Zhang, Ms Li, Mr Zou, and Dr Tang), and
| | - Hai-jun Wei
- Institute of Neuroscience, Medical College (Mr Wei, Dr Gu, and Dr Tang),,University of South China, Hengyang, Hunan, PR China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Mr Wei and Dr Tang)
| | - Man-Hong Li
- Department of Neurology, Nanhua Affiliated Hospital (Ms Li, Mr Zhang, Ms Li, Mr Zou, and Dr Tang), and
| | - Wei Zou
- Department of Neurology, Nanhua Affiliated Hospital (Ms Li, Mr Zhang, Ms Li, Mr Zou, and Dr Tang), and
| | - Xiang Li
- Department of Anesthesiology, First Affiliated Hospital, University of South China, Hengyang, Hunan, PR China (Mr Li)
| | - Hong-Feng Gu
- Institute of Neuroscience, Medical College (Mr Wei, Dr Gu, and Dr Tang)
| | - Xiao-Qing Tang
- Department of Neurology, Nanhua Affiliated Hospital (Ms Li, Mr Zhang, Ms Li, Mr Zou, and Dr Tang), and,Institute of Neuroscience, Medical College (Mr Wei, Dr Gu, and Dr Tang),,University of South China, Hengyang, Hunan, PR China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Mr Wei and Dr Tang)
| |
Collapse
|
20
|
Conklin DJ. Acute cardiopulmonary toxicity of inhaled aldehydes: role of TRPA1. Ann N Y Acad Sci 2016; 1374:59-67. [PMID: 27152448 DOI: 10.1111/nyas.13055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/26/2016] [Accepted: 03/07/2016] [Indexed: 12/13/2022]
Abstract
Inhalation of high-level volatile aldehydes, as present in smoke from wildfires and in tobacco smoke, is associated with both acute and chronic cardiopulmonary morbidity and mortality, but the underlying mechanisms are unclear. The transient receptor potential ankyrin 1 (TRPA1) protein forms a cation channel (irritant receptor) that mediates tobacco smoke-induced airway and lung injury, yet the role of TRPA1 in the cardiovascular toxicity of aldehyde exposure is unclear. Physiologically, airway-located TRPA1 activation triggers an irritant response (e.g., coughing and "respiratory braking") that alters the rate and depth of breathing to reduce exposure. Acrolein (2-propenal), a volatile, unsaturated aldehyde, activates TRPA1. Acrolein was used as a chemical weapon in World War I and is present at high levels in wildfires and tobacco smoke. Acrolein is thought to contribute to pulmonary and cardiovascular injury caused by tobacco smoke exposure, although the role of TRPA1 in cardiovascular toxicity is unclear. This minireview addresses this gap in our knowledge by exploring literature and recent data indicating a connection between TRPA1 and cardiovascular as well as pulmonary injury due to inhaled aldehydes.
Collapse
Affiliation(s)
- Daniel J Conklin
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
21
|
Faure P, Legendre A, Hanser HI, Andriot I, Artur Y, Guichard E, Coureaud G, Heydel JM. Odorant Metabolism Analysis by an Automated Ex Vivo Headspace Gas-Chromatography Method. Chem Senses 2015; 41:15-23. [DOI: 10.1093/chemse/bjv055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
22
|
Larraufie MH, Yang WS, Jiang E, Thomas AG, Slusher BS, Stockwell BR. Incorporation of metabolically stable ketones into a small molecule probe to increase potency and water solubility. Bioorg Med Chem Lett 2015; 25:4787-4792. [PMID: 26231156 DOI: 10.1016/j.bmcl.2015.07.018] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 12/21/2022]
Abstract
Introducing a reactive carbonyl to a scaffold that does not otherwise have an electrophilic functionality to create a reversible covalent inhibitor is a potentially useful strategy for enhancing compound potency. However, aldehydes are metabolically unstable, which precludes the use of this strategy for compounds to be tested in animal models or in human clinical studies. To overcome this limitation, we designed ketone-based functionalities capable of forming reversible covalent adducts, while displaying high metabolic stability, and imparting improved water solubility to their pendant scaffold. We tested this strategy on the ferroptosis inducer and experimental therapeutic erastin, and observed substantial increases in compound potency. In particular, a new carbonyl erastin analog, termed IKE, displayed improved potency, solubility and metabolic stability, thus representing an ideal candidate for future in vivo cancer therapeutic applications.
Collapse
Affiliation(s)
- Marie-Helene Larraufie
- Department of Biological Sciences, Columbia University, 550 West 120th Street, 1208 Northwest Corner Building, MC 4846, New York, NY 10027, United States
| | - Wan Seok Yang
- Department of Biological Sciences, Columbia University, 550 West 120th Street, 1208 Northwest Corner Building, MC 4846, New York, NY 10027, United States
| | - Elise Jiang
- Department of Chemistry, Columbia University, New York, NY 10027, United States
| | - Ajit G Thomas
- Brain Science Institute, Johns Hopkins Medicine, Baltimore, United States
| | - Barbara S Slusher
- Brain Science Institute, Johns Hopkins Medicine, Baltimore, United States; Department of Neurology, Johns Hopkins Medicine, Baltimore, United States
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, 550 West 120th Street, 1208 Northwest Corner Building, MC 4846, New York, NY 10027, United States; Department of Chemistry, Columbia University, New York, NY 10027, United States; Howard Hughes Medical Institute, Columbia University, New York, NY 10027, United States.
| |
Collapse
|
23
|
Conklin DJ, Guo Y, Jagatheesan G, Kilfoil PJ, Haberzettl P, Hill BG, Baba SP, Guo L, Wetzelberger K, Obal D, Rokosh DG, Prough RA, Prabhu SD, Velayutham M, Zweier JL, Hoetker JD, Riggs DW, Srivastava S, Bolli R, Bhatnagar A. Genetic Deficiency of Glutathione S-Transferase P Increases Myocardial Sensitivity to Ischemia-Reperfusion Injury. Circ Res 2015; 117:437-49. [PMID: 26169370 DOI: 10.1161/circresaha.114.305518] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 07/13/2015] [Indexed: 01/18/2023]
Abstract
RATIONALE Myocardial ischemia-reperfusion (I/R) results in the generation of oxygen-derived free radicals and the accumulation of lipid peroxidation-derived unsaturated aldehydes. However, the contribution of aldehydes to myocardial I/R injury has not been assessed. OBJECTIVE We tested the hypothesis that removal of aldehydes by glutathione S-transferase P (GSTP) diminishes I/R injury. METHODS AND RESULTS In adult male C57BL/6 mouse hearts, Gstp1/2 was the most abundant GST transcript followed by Gsta4 and Gstm4.1, and GSTP activity was a significant fraction of the total GST activity. mGstp1/2 deletion reduced total GST activity, but no compensatory increase in GSTA and GSTM or major antioxidant enzymes was observed. Genetic deficiency of GSTP did not alter cardiac function, but in comparison with hearts from wild-type mice, the hearts isolated from GSTP-null mice were more sensitive to I/R injury. Disruption of the GSTP gene also increased infarct size after coronary occlusion in situ. Ischemia significantly increased acrolein in hearts, and GSTP deficiency induced significant deficits in the metabolism of the unsaturated aldehyde, acrolein, but not in the metabolism of 4-hydroxy-trans-2-nonenal or trans-2-hexanal; on ischemia, the GSTP-null hearts accumulated more acrolein-modified proteins than wild-type hearts. GSTP deficiency did not affect I/R-induced free radical generation, c-Jun N-terminal kinase activation, or depletion of reduced glutathione. Acrolein exposure induced a hyperpolarizing shift in INa, and acrolein-induced cell death was delayed by SN-6, a Na(+)/Ca(++) exchange inhibitor. Cardiomyocytes isolated from GSTP-null hearts were more sensitive than wild-type myocytes to acrolein-induced protein crosslinking and cell death. CONCLUSIONS GSTP protects the heart from I/R injury by facilitating the detoxification of cytotoxic aldehydes, such as acrolein.
Collapse
Affiliation(s)
- Daniel J Conklin
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.).
| | - Yiru Guo
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Ganapathy Jagatheesan
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Peter J Kilfoil
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Petra Haberzettl
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Bradford G Hill
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Shahid P Baba
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Luping Guo
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Karin Wetzelberger
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Detlef Obal
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - D Gregg Rokosh
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Russell A Prough
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Sumanth D Prabhu
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Murugesan Velayutham
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Jay L Zweier
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - J David Hoetker
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Daniel W Riggs
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Sanjay Srivastava
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Roberto Bolli
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| | - Aruni Bhatnagar
- From the Diabetes and Obesity Center (D.J.C., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., K.W., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B.), Institute of Molecular Cardiology (D.J.C., Y.G., P.J.K., P.H., B.G.H., S.P.B., D.O., D.G.R., S.S., R.B., A.B.), Division of Cardiovascular Medicine, Department of Medicine (D.J.C., Y.G., G.J., P.J.K., P.H., B.G.H., S.P.B., L.G., D.O., D.G.R., J.D.H., D.W.R., S.S., R.B., A.B), Department of Anesthesiology and Perioperative Medicine (D.O.), and Department of Biochemistry and Molecular Genetics (P.J.K., R.A.P., A.B.), University of Louisville, KY; Division of Cardiovascular Disease, University of Alabama at Birmingham (S.D.P.); and Center for Biomedical EPR Spectroscopy and Imaging, Davis Heart and Lung Research Institute, and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus (M.V., J.L.Z.)
| |
Collapse
|
24
|
Nakashima Y, Ohsawa I, Nishimaki K, Kumamoto S, Maruyama I, Suzuki Y, Ohta S. Preventive effects of Chlorella on skeletal muscle atrophy in muscle-specific mitochondrial aldehyde dehydrogenase 2 activity-deficient mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:390. [PMID: 25305781 PMCID: PMC4200191 DOI: 10.1186/1472-6882-14-390] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 10/07/2014] [Indexed: 12/24/2022]
Abstract
Background Oxidative stress is involved in age-related muscle atrophy, such as sarcopenia. Since Chlorella, a unicellular green alga, contains various antioxidant substances, we used a mouse model of enhanced oxidative stress to investigate whether Chlorella could prevent muscle atrophy. Methods Aldehyde dehydrogenase 2 (ALDH2) is an anti-oxidative enzyme that detoxifies reactive aldehydes derived from lipid peroxides such as 4-hydroxy-2-nonenal (4-HNE). We therefore used transgenic mice expressing a dominant-negative form of ALDH2 (ALDH2*2 Tg mice) to selectively decrease ALDH2 activity in the muscles. To evaluate the effect of Chlorella, the mice were fed a Chlorella-supplemented diet (CSD) for 6 months. Results ALDH2*2 Tg mice exhibited small body size, muscle atrophy, decreased fat content, osteopenia, and kyphosis, accompanied by increased muscular 4-HNE levels. The CSD helped in recovery of body weight, enhanced oxidative stress, and increased levels of a muscle impairment marker, creatine phosphokinase (CPK) induced by ALDH2*2. Furthermore, histological and histochemical analyses revealed that the consumption of the CSD improved skeletal muscle atrophy and the activity of the mitochondrial cytochrome c oxidase. Conclusions This study suggests that long-term consumption of Chlorella has the potential to prevent age-related muscle atrophy.
Collapse
|
25
|
Chen AH, Zhang P, Yin WL, Wang L, Zou W, Tang XQ. Role of aldehyde dehydrogenase 2 in 1-methy-4-phenylpyridinium ion-induced aldehyde stress and cytotoxicity in PC12 cells. Neurochem Res 2014; 39:1767-75. [PMID: 25005621 DOI: 10.1007/s11064-014-1376-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 06/21/2014] [Accepted: 06/27/2014] [Indexed: 12/15/2022]
Abstract
Aldehyde stress contributes to molecular mechanisms of cell death and the pathogenesis of Parkinson's disease (PD). The neurotoxin 1-Methy-4-Phenylpyridinium Ion (MPP(+)) is commonly used to model PD. Aldehyde dehydrogenase 2 (ALDH2) is an important enzyme detoxifying aldehydes. The aim of this study is to evaluate whether MPP(+)-induced neurotoxicity is involved in aldehyde stress by modulation of ALDH2. Our results demonstrated that treatment of PC12 cells with MPP(+) leads to aldehyde stress by increasing in loads of malondialdehyde and 4-hydroxynonenal, which indicated that MPP(+)-induced aldehyde stress contributes to its cytotoxicity in PC12 cells. We also showed that MPP(+) up-regulates the expression and activity of ALDH2 in PC12 cells and that inhibition of ALDH2 by its specific inhibitor daidzin prevents MPP(+)-induced decrease in cell viability and increases in apoptosis, oxidative stress and aldehyde stress in PC12 cells. These findings suggest that aldehyde stress contributes to MPP(+)-induced toxicity in PC12 cells by upregulation of ALDH2. This study provides a novel insight into the role of ALDH2 in the neurotoxicity of MPP(+).
Collapse
Affiliation(s)
- Ai-Hua Chen
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, 336 E Dongfeng Road, Hengyang, 421001, Hunan, People's Republic of China
| | | | | | | | | | | |
Collapse
|
26
|
Kajita R, Goto T, Lee SH, Oe T. Aldehyde Stress-Mediated Novel Modification of Proteins: Epimerization of the N-Terminal Amino Acid. Chem Res Toxicol 2013; 26:1926-36. [DOI: 10.1021/tx400354d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Ryo Kajita
- Department of Bioanalytical
Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai 980-8578, Japan
| | - Takaaki Goto
- Department of Bioanalytical
Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai 980-8578, Japan
| | - Seon Hwa Lee
- Department of Bioanalytical
Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai 980-8578, Japan
| | - Tomoyuki Oe
- Department of Bioanalytical
Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
27
|
Impaired cardiac SIRT1 activity by carbonyl stress contributes to aging-related ischemic intolerance. PLoS One 2013; 8:e74050. [PMID: 24040162 PMCID: PMC3769351 DOI: 10.1371/journal.pone.0074050] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/25/2013] [Indexed: 01/04/2023] Open
Abstract
Reactive aldehydes can initiate protein oxidative damage which may contribute to heart senescence. Sirtuin 1 (SIRT1) is considered to be a potential interventional target for I/R injury management in the elderly. We hypothesized that aldehyde mediated carbonyl stress increases susceptibility of aged hearts to ischemia/reperfusion (I/R) injury, and elucidate the underlying mechanisms with a focus on SIRT1. Male C57BL/6 young (4-6 mo) and aged (22-24 mo) mice were subjected to myocardial I/R. Cardiac aldehyde dehydrogenase (ALDH2), SIRT1 activity and protein carbonyls were assessed. Our data revealed that aged heart exhibited increased endogenous aldehyde/carbonyl stress due to impaired ALDH2 activity concomitant with blunted SIRT1 activity (P<0.05). Exogenous toxic aldehydes (4-HNE) exposure in isolated cardiomyocyte verified that aldehyde-induced carbonyl modification on SIRT1 impaired SIRT1 activity leading to worse hypoxia/reoxygenation (H/R) injury, which could all be rescued by Alda-1 (ALDH2 activator) (all P<0.05). However, SIRT1 inhibitor blocked the protective effect of Alda-1 on H/R cardiomyocyte. Interestingly, myocardial I/R leads to higher carbonylation but lower activity of SIRT1 in aged hearts than that seen in young hearts (P<0.05). The application of Alda-1 significantly reduced the carbonylation on SIRT1 and markedly improved the tolerance to in vivo I/R injury in aged hearts, but failed to protect Sirt1+/− knockout mice against myocardial I/R injury. This was verified by Alda-1 treatment improved postischemic contractile function recovery in ex vivo perfused aged but not in Sirt1+/− hearts. Thus, aldehyde/carbonyl stress is accelerated in aging heart. These results provide a new insight that impaired cardiac SIRT1 activity by carbonyl stress plays a critical role in the increased susceptibility of aged heart to I/R injury. ALDH2 activation can restore this aging-related myocardial ischemic intolerance.
Collapse
|
28
|
Dunner S, Sevane N, Garcia D, Levéziel H, Williams JL, Mangin B, Valentini A. Genes involved in muscle lipid composition in 15 European Bos taurus breeds. Anim Genet 2013; 44:493-501. [PMID: 23611291 DOI: 10.1111/age.12044] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2013] [Indexed: 11/29/2022]
Abstract
Consumers demand healthy and palatable meat, both factors being affected by fat composition. However, red meat has relatively high concentration of saturated fatty acids and low concentration of the beneficial polyunsaturated fatty acids. To select animals prone to produce particular fat types, it is necessary to identify the genes influencing muscle lipid composition. This paper describes an association study in which a large panel of candidate genes involved in adipogenesis, lipid metabolism and energy homoeostasis was tested for effects on fat composition in 15 European cattle breeds. Sixteen genes were found to have significant effects on different lipid traits, and among these, CFL1 and MYOZ1 were found to have large effects on the ratio of 18:2/18:3, CRI1 on the amount of neutral adrenic acid (22:4 n-6), MMP1 on docosahexaenoic acid (22:6 n-3) and conjugated linoleic acid, PLTP on the ratio of n-6:n-3 and IGF2R on flavour. Several genes - ALDH2, CHRNE, CRHR2, DGAT1, IGFBP3, NEB, SOCS2, SUSP1, TCF12 and FOXO1 - also were found to be associated with both lipid and organoleptic traits although with smaller effect. The results presented here help in understanding the genetic and biochemical background underlying variations in fatty acid composition and flavour in beef.
Collapse
Affiliation(s)
- S Dunner
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Mitochondrial aldehyde dehydrogenase 2 activation and cardioprotection. J Mol Cell Cardiol 2013; 55:58-63. [DOI: 10.1016/j.yjmcc.2012.03.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 03/14/2012] [Accepted: 03/29/2012] [Indexed: 01/10/2023]
|
30
|
Poly(ADP-ribose) polymerase inhibition prevents reactive oxygen species induced inhibition of aldehyde dehydrogenase2 activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:479-86. [PMID: 23159776 DOI: 10.1016/j.bbamcr.2012.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 10/31/2012] [Accepted: 11/07/2012] [Indexed: 01/12/2023]
Abstract
Lipid peroxidation plays a critical role in cardiovascular diseases. Aldehydes are the major end products of lipid peroxidation and can be metabolized into less reactive chemical species by aldehyde dehydrogenase 2 (ALDH2). However, ALDH2 dehydrogenase activity can be affected by many factors including reactive oxygen species. To elucidate how reactive oxygen species inhibit ALDH2 dehydrogenase activity, we stimulated human aortic endothelial cells (HAECs) with oxidized low-density lipoproteins (ox-LDL) and performed a myocardial ischemia-reperfusion model. Ox-LDL treatment and ischemia-reperfusion injury inhibited ALDH2 dehydrogenase activity. Poly(ADP-ribose) polymerase (PARP) was activated by ox-LDL stimulation and ischemia-reperfusion injury and PARP inhibition partly restored ALDH2 dehydrogenase activity in ox-LDL treated HAECs and ischemia-reperfusion rat hearts. SIRT3 was upregulated by ox-LDL stimulation and ischemia-reperfusion injury and downregulated by PARP inhibition. Using siRNA to knock down SIRT3, we demonstrated that SIRT3 mediated deacetylation decreased ALDH2 dehydrogenase activity and PARP inhibition partly restored ALDH2 dehydrogenase activity through preventing SIRT3 expression and subsequently preserving ALDH2 acetylation.
Collapse
|
31
|
Sundaram K, Endo S, Matsunaga T, Tanaka N, Hara A, El-Kabbani O. Structure of the His269Arg mutant of the rat aldose reductase-like protein AKR1B14 complexed with NADPH. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 68:400-3. [PMID: 22505406 PMCID: PMC3325806 DOI: 10.1107/s1744309112008810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 02/27/2012] [Indexed: 11/11/2022]
Abstract
Rat aldose reductase-like protein (AKR1B14) is an orthologue of mouse vas deferens protein (AKR1B7) and plays roles in the detoxification of reactive aldehydes and synthesis of prostaglandin F(2α). Here, the 1.87 Å resolution crystal structure of the His269Arg mutant of AKR1B14 complexed with NADPH is described and shows that the negatively charged 2'-phosphate group of the coenzyme forms an ionic interaction with the positively charged guanidinium group of Arg269 that is also observed in the human aldose reductase (AKR1B1) structure. Previous experiments on the site-directed mutagenesis of His269 to Arg, Phe and Met revealed fourfold, sevenfold and 127-fold increases in the K(m) for NADPH, respectively, which are in agreement with the present molecular-modelling and X-ray crystallographic studies. This is the first tertiary structure of a mutant form of this AKR1B7 orthologue to be reported in order to investigate the structure-function relationship of the nonconserved His269 and its role in coenzyme binding.
Collapse
Affiliation(s)
- Krithika Sundaram
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Daigaku-Nishi, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Daigaku-Nishi, Gifu 501-1196, Japan
| | - Nobutada Tanaka
- School of Pharmaceutical Sciences, Showa University, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Akira Hara
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Daigaku-Nishi, Gifu 501-1196, Japan
| | - Ossama El-Kabbani
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
32
|
He L, Liu B, Dai Z, Zhang HF, Zhang YS, Luo XJ, Ma QL, Peng J. Alpha lipoic acid protects heart against myocardial ischemia-reperfusion injury through a mechanism involving aldehyde dehydrogenase 2 activation. Eur J Pharmacol 2012; 678:32-8. [PMID: 22266491 DOI: 10.1016/j.ejphar.2011.12.042] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 12/22/2011] [Accepted: 12/28/2011] [Indexed: 11/25/2022]
Abstract
Recent studies demonstrate that alpha lipoic acid can prevent nitroglycerin tolerance by restoring aldehyde dehydrogenase 2 (ALDH2) activity and ALDH2-mediated detoxification of aldehydes is thought as an endogenous mechanism against ischemia-reperfusion injury. This study was performed to explore whether the cardioprotective effect of alpha lipoic acid was related to activation of ALDH2 and the underlying mechanisms. In a Langendorff model of ischemia-reperfusion in rats, cardiac function, activities of creatine kinase (CK) and ALDH2, contents of 4-hydroxy-2-nonenal (4-HNE) and malondialdehyde (MDA) were measured. In a cell model of hypoxia-reoxygenation, the apoptosis, ALDH activity, reactive oxygen species level, 4-HNE and MDA contents were examined. In the isolated hearts, ischemia-reperfusion treatment led to cardiac dysfunction accompanied by an increase in 4-HNE and MDA contents. Pretreatment with lipoic acid significantly up-regulated myocardial ALDH2 activity concomitantly with an improvement of cardiac dysfunction and a decrease in 4-HNE and MDA contents, these effects were blocked by the inhibitor of ALDH2. Similarly, in the cultured cardiomyocytes, hypoxia-reoxygenation treatment induced apoptosis accompanied by an increase in the production of reactive oxygen species, 4-HNE and MDA. Administration of lipoic acid significantly up-regulated cellular ALDH2 activity concomitantly with a reduction in apoptosis, production of reactive oxygen species, 4-HNE and MDA, these effects were reversed in the presence of ALDH2 or PKCε inhibitors. Our results suggest that the cardioprotective effects of lipoic acid on ischemia-reperfusion injury are through a mechanism involving ALDH2 activation. The regulatory effect of lipoic acid on ALDH2 activity is dependent on PKCε signaling pathway.
Collapse
Affiliation(s)
- Lan He
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Chippendale TWE, Hu B, El Haj AJ, Smith D. A study of enzymatic activity in cell cultures via the analysis of volatile biomarkers. Analyst 2012; 137:4677-85. [DOI: 10.1039/c2an35815h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
34
|
Kim KJ, Pearl PL, Jensen K, Snead OC, Malaspina P, Jakobs C, Gibson KM. Succinic semialdehyde dehydrogenase: biochemical-molecular-clinical disease mechanisms, redox regulation, and functional significance. Antioxid Redox Signal 2011; 15:691-718. [PMID: 20973619 PMCID: PMC3125545 DOI: 10.1089/ars.2010.3470] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Succinic semialdehyde dehydrogenase (SSADH; aldehyde dehydrogenase 5a1, ALDH5A1; E.C. 1.2.1.24; OMIM 610045, 271980) deficiency is a rare heritable disorder that disrupts the metabolism of the inhibitory neurotransmitter 4-aminobutyric acid (GABA). Identified in conjunction with increased urinary excretion of the GABA analog gamma-hydroxybutyric acid (GHB), numerous patients have been identified worldwide and the autosomal-recessive disorder has been modeled in mice. The phenotype is one of nonprogressive neurological dysfunction in which seizures may be prominently displayed. The murine model is a reasonable phenocopy of the human disorder, yet the severity of the seizure disorder in the mouse exceeds that observed in SSADH-deficient patients. Abnormalities in GABAergic and GHBergic neurotransmission, documented in patients and mice, form a component of disease pathophysiology, although numerous other disturbances (metabolite accumulations, myelin abnormalities, oxidant stress, neurosteroid depletion, altered bioenergetics, etc.) are also likely to be involved in developing the disease phenotype. Most recently, the demonstration of a redox control system in the SSADH protein active site has provided new insights into the regulation of SSADH by the cellular oxidation/reduction potential. The current review summarizes some 30 years of research on this protein and disease, addressing pathological mechanisms in human and mouse at the protein, metabolic, molecular, and whole-animal level.
Collapse
Affiliation(s)
- Kyung-Jin Kim
- Pohang Accelerator Laboratory, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Phillip L. Pearl
- Department of Neurology, Children's National Medical Center, Washington, District of Columbia
| | - Kimmo Jensen
- Synaptic Physiology Laboratory, Department of Physiology and Biophysics, Aarhus University, Aarhus, Denmark
- Center for Psychiatric Research, Aarhus University Hospital, Risskov, Denmark
| | - O. Carter Snead
- Department of Neurology, Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | | | - Cornelis Jakobs
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - K. Michael Gibson
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan
| |
Collapse
|
35
|
Lin Z, Li H, Luo H, Zhang Y, Luo W. Benzylamine and methylamine, substrates of semicarbazide-sensitive amine oxidase, attenuate inflammatory response induced by lipopolysaccharide. Int Immunopharmacol 2011; 11:1079-89. [PMID: 21414430 DOI: 10.1016/j.intimp.2011.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 02/17/2011] [Accepted: 03/01/2011] [Indexed: 02/05/2023]
Abstract
Current evidence indicates that semicarbazide-sensitive amine oxidase (SSAO) substrates possess insulin-mimic effect, which was thought to play an anti-inflammatory role. The purpose of the present study was to determine whether SSAO substrates benzylamine (BZA) and methylamine (MA) attenuate inflammatory response induced by lipopolysaccharide (LPS). BALB/c mice peritoneal macrophages (PMs) that express SSAO and RAW264.7 mouse macrophages that do not express SSAO were used in vitro studies. Experimental mice were given BZA or MA through intraperitoneal injection before LPS challenge. The results showed that BZA or MA treatment significantly reduced LPS-induced pro-inflammatory mediators (nitric oxide, TNF-α) production, the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2, and glucose consumption in murine PMs, but not in RAW264.7 cell line. The metabolites of BZA or MA catalyzed by SSAO, hydrogen peroxide, formaldehyde, and benzaldehyde could also significantly decrease LPS-induced nitric oxide and TNF-α production, iNOS and COX-2 expression, and glucose consumption in vitro. In addition, BZA or MA administration could significantly decrease plasma pro-inflammatory mediators and the expression of iNOS and COX-2 in liver and lung, and could also attenuate LPS-induced transient hyperglycemia and chronic hypoglycemia. These findings indicated that substrates of SSAO might be involved in the anti-inflammatory effects. The metabolites of BZA and MA catalyzed by SSAO might be responsible for the anti-inflammatory effects. Moreover, BZA or MA administration could be useful for normalization of glucose disposal during endotoxemia.
Collapse
Affiliation(s)
- Zhexuan Lin
- Bio-analytical Laboratory, Shantou University Medical College, Shantou, PR China
| | | | | | | | | |
Collapse
|
36
|
Wheat LA, Haberzettl P, Hellmann J, Baba SP, Bertke M, Lee J, McCracken J, O'Toole TE, Bhatnagar A, Conklin DJ. Acrolein inhalation prevents vascular endothelial growth factor-induced mobilization of Flk-1+/Sca-1+ cells in mice. Arterioscler Thromb Vasc Biol 2011; 31:1598-606. [PMID: 21527748 DOI: 10.1161/atvbaha.111.227124] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Acrolein is a toxic chemical present in tobacco, wood, and coal smoke, as well as automobile exhaust. Because exposure to these pollutants is associated with an increase in cardiovascular disease risk, we studied the effects of acrolein on Flk-1(+)/Sca-1(+) cells that are involved in vascular repair. METHODS AND RESULTS In adult male C57BL/6 mice, inhalation of acrolein (1 part per million [ppm], 6 hours/day for 4 days or 5 ppm for 2 or 6 hours) led to the formation of protein-acrolein adducts in the bone marrow and diminished levels of plasma nitric oxide metabolites and circulating Flk-1(+)/Sca-1(+) but not Sca-1(+)-only cells. Acrolein exposure increased the number of apoptotic Flk-1(+)/Sca-1(+) cells in circulation and increased bone marrow-derived cells with endothelial characteristics (DiI-ac-low-density lipoprotein [DiI-acLDL]/UE-lectin and Flk-1(+)/Sca-1(+)) in culture. Deficits in the circulating levels of Flk-1(+)/Sca-1(+) cells were reversed after 7 days of recovery in acrolein-free air. Exposure to acrolein blocked vascular endothelial growth factor (VEGF)/AMD3100-stimulated mobilization of Flk-1(+)/Sca-1(+) but not Sca-1(+)-only cells and prevented VEGF-induced phosphorylation of Akt and endothelial nitric oxide synthase in the aorta. CONCLUSIONS Inhalation of acrolein increases apoptosis and suppresses the circulating levels of Flk-1(+)/Sca-1(+) cells while increasing these cells in the bone marrow and preventing their mobilization by VEGF. Exposure to acrolein-rich pollutants could impair vascular repair capacity.
Collapse
Affiliation(s)
- Laura A Wheat
- Diabetes and Obesity Center, Department of Physiology and Biophysics, University of Louisville, Louisville, KY 40292, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Sundaram K, Dhagat U, Endo S, Chung R, Matsunaga T, Hara A, El-Kabbani O. Structure of rat aldose reductase-like protein AKR1B14 holoenzyme: Probing the role of His269 in coenzyme binding by site-directed mutagenesis. Bioorg Med Chem Lett 2010; 21:801-4. [PMID: 21168333 DOI: 10.1016/j.bmcl.2010.11.086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 11/17/2010] [Accepted: 11/19/2010] [Indexed: 12/12/2022]
Abstract
Rat aldose reductase-like protein (AKR1B14) is the ortholog of mouse vas deferens protein (AKR1B7) playing roles in detoxification of reactive aldehydes and synthesis of prostaglandin F(2α). The crystal structure of the binary complex (AKR1B14-NADPH) was determined at 1.86Å resolution, and showed that the adenine ring and the 2'-phosphate group of the coenzyme formed π-stacking and electrostatic interactions with the imidazole ring and ND1 atom, respectively, of His269, which is not conserved in other aldose reductase-like proteins. The interactions were supported by site-directed mutagenesis of His269 to Arg, Phe and Met, which increased the K(m) for NADPH by 4, 7 and 127-fold, respectively. This is the first report of the tertiary structure of a rodent AKR1B7 ortholog, which describes the role of a novel dual interaction for the non-conserved His269 in coenzyme binding.
Collapse
Affiliation(s)
- Krithika Sundaram
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Everyone encounters various stressors (causes of stress), such as psychological pressure, mental fluctuations, and physical burdens, in their everyday life. It is well accepted that the highest levels of perceived stress correlate with early onset of cardiovascular disease. Conversely, appropriate (mild to moderate) stressors, such as physical activity, have been shown to promote health. This bidirectional dose - response relationship of treatments that are beneficial at low levels but noxious at higher levels is referred to as "hormesis". In the fields of toxicology, pharmacology, radiation biology, and medicine, the significance of the biological effects of low-level exposure to various agents has attracted considerable attention. It is very important to understand how biological systems respond to low levels of stress and their implications within society. Aldehydes, the major endproducts of lipid peroxidation, have been implicated in the pathogenesis of oxidative stress-associated diseases. In addition to the pathogenic effect associated with oxidative stress, sublethal levels of aldehydes interact with signaling systems to upregulate the expression of genes to counteract the stressor challenge and to re-establish homeostasis. The present review article discusses current discoveries regarding the hormetic response to aldehyde and its clinical significance in cardioprotection.
Collapse
Affiliation(s)
- Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
39
|
4-hydroxy-2-nonenal protects against cardiac ischemia-reperfusion injury via the Nrf2-dependent pathway. J Mol Cell Cardiol 2010; 49:576-86. [PMID: 20685357 DOI: 10.1016/j.yjmcc.2010.05.011] [Citation(s) in RCA: 297] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2009] [Revised: 04/24/2010] [Accepted: 05/21/2010] [Indexed: 11/24/2022]
Abstract
Reactive oxygen species (ROS) attack polyunsaturated fatty acids of the membrane and trigger lipid peroxidation, which results in the generation of alpha,beta-unsaturated aldehydes, such as 4-hydroxy-2-nonenal (4-HNE). There is compelling evidence that high concentrations of aldehydes are responsible for much of the damage elicited by cardiac ischemia-reperfusion injury, while sublethal concentrations of aldehydes stimulate stress resistance pathways, to achieve cardioprotection. We investigated the mechanism of cardioprotection mediated by 4-HNE. For cultured cardiomyocytes, 4-HNE was cytotoxic at higher concentrations (>or=20 microM) but had no appreciable cytotoxicity at lower concentrations. Notably, a sublethal concentration (5muM) of 4-HNE primed cardiomyocytes to become resistant to cytotoxic concentrations of 4-HNE. 4-HNE induced nuclear translocation of transcription factor NF-E2-related factor 2 (Nrf2), and enhanced the expression of gamma-glutamylcysteine ligase (GCL) and the core subunit of the Xc(-) high-affinity cystine transporter (xCT), thereby increasing 1.45-fold the intracellular GSH levels. Cardiomyocytes treated with either Nrf2-specific siRNA or the GCL inhibitor l-buthionine sulfoximine (BSO) were less tolerant to 4-HNE. Moreover, the cardioprotective effect of 4-HNE pretreatment against subsequent glucose-free anoxia followed by reoxygenation was completely abolished in these cells. Intravenous administration of 4-HNE (4 mg/kg) activated Nrf2 in the heart and increased the intramyocardial GSH content, and consequently improved the functional recovery of the left ventricle following ischemia-reperfusion in Langendorff-perfused hearts. This cardioprotective effect of 4-HNE was not observed for Nrf2-knockout mice. In summary, 4-HNE activates Nrf2-mediated gene expression and stimulates GSH biosynthesis, thereby conferring on cardiomyocytes protection against ischemia-reperfusion injury.
Collapse
|
40
|
Carbone V, Giglio M, Chung R, Huyton T, Adams J, Maccari R, Ottana R, Hara A, El-Kabbani O. Structure of aldehyde reductase in ternary complex with a 5-arylidene-2,4-thiazolidinedione aldose reductase inhibitor. Eur J Med Chem 2010; 45:1140-5. [DOI: 10.1016/j.ejmech.2009.12.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 12/02/2009] [Accepted: 12/10/2009] [Indexed: 11/24/2022]
|
41
|
MicroRNA group disorganization in aging. Exp Gerontol 2009; 45:269-78. [PMID: 20034554 DOI: 10.1016/j.exger.2009.12.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 12/13/2009] [Accepted: 12/15/2009] [Indexed: 01/01/2023]
Abstract
Among non-coding RNAs, microRNAs may be one of the best known subgroups, due to their unique function of negatively controlling gene expression, by either degrading target messages or binding to their 3'-untranslated region to inhibit translation. Thus gene expression can be repressed through post-transcriptional regulation, implemented as a 'dimmer switch', in contrast to the all-or-none mode of suppression. Work from our laboratory and others shows that during aging, dysregulated expression of microRNAs generally occurs in groups, suggesting that their actions may be functionally coordinated as a 'pack' by common transcriptional regulators; the accumulation of these 'pack' disorganizations may be the underlying culprit contributing to the pathoetiology of many age-dependent disease states. The fact that many microRNAs are coordinated in their expression, due to either the close proximity of their genomic locations or sharing the same transcriptional regulation, suggests that future strategies for correcting age-dependent microRNA disorganization may need to involve a system biology, rather than a reductionist, approach. Therefore, understanding age-dependent changes of microRNA expression in 'packs' may open an entirely new frontier, i.e. how particular groups of non-coding RNAs, functioning together, contribute to mechanisms regulating aging and longevity.
Collapse
|
42
|
|
43
|
Endo J, Sano M, Katayama T, Hishiki T, Shinmura K, Morizane S, Matsuhashi T, Katsumata Y, Zhang Y, Ito H, Nagahata Y, Marchitti S, Nishimaki K, Wolf AM, Nakanishi H, Hattori F, Vasiliou V, Adachi T, Ohsawa I, Taguchi R, Hirabayashi Y, Ohta S, Suematsu M, Ogawa S, Fukuda K. Metabolic Remodeling Induced by Mitochondrial Aldehyde Stress Stimulates Tolerance to Oxidative Stress in the Heart. Circ Res 2009; 105:1118-27. [DOI: 10.1161/circresaha.109.206607] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale
:
Aldehyde accumulation is regarded as a pathognomonic feature of oxidative stress–associated cardiovascular disease.
Objective
:
We investigated how the heart compensates for the accelerated accumulation of aldehydes.
Methods and Results
:
Aldehyde dehydrogenase 2 (ALDH2) has a major role in aldehyde detoxification in the mitochondria, a major source of aldehydes. Transgenic (Tg) mice carrying an
Aldh2
gene with a single nucleotide polymorphism (
Aldh2*2
) were developed. This polymorphism has a dominant-negative effect and the Tg mice exhibited impaired ALDH activity against a broad range of aldehydes. Despite a shift toward the oxidative state in mitochondrial matrices,
Aldh2*2
Tg hearts displayed normal left ventricular function by echocardiography and, because of metabolic remodeling, an unexpected tolerance to oxidative stress induced by ischemia/reperfusion injury. Mitochondrial aldehyde stress stimulated eukaryotic translation initiation factor 2α phosphorylation. Subsequent translational and transcriptional activation of activating transcription factor-4 promoted the expression of enzymes involved in amino acid biosynthesis and transport, ultimately providing precursor amino acids for glutathione biosynthesis. Intracellular glutathione levels were increased 1.37-fold in
Aldh2*2
Tg hearts compared with wild-type controls. Heterozygous knockout of
Atf4
blunted the increase in intracellular glutathione levels in
Aldh2*2
Tg hearts, thereby attenuating the oxidative stress–resistant phenotype. Furthermore, glycolysis and NADPH generation via the pentose phosphate pathway were activated in
Aldh2*2
Tg hearts. (NADPH is required for the recycling of oxidized glutathione.)
Conclusions
:
The findings of the present study indicate that mitochondrial aldehyde stress in the heart induces metabolic remodeling, leading to activation of the glutathione–redox cycle, which confers resistance against acute oxidative stress induced by ischemia/reperfusion.
Collapse
Affiliation(s)
- Jin Endo
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Motoaki Sano
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Takaharu Katayama
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Takako Hishiki
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Ken Shinmura
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Shintaro Morizane
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Tomohiro Matsuhashi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Yoshinori Katsumata
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Yan Zhang
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Hideyuki Ito
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Yoshiko Nagahata
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Satori Marchitti
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Kiyomi Nishimaki
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Alexander Martin Wolf
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Hiroki Nakanishi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Fumiyuki Hattori
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Vasilis Vasiliou
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Takeshi Adachi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Ikuroh Ohsawa
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Ryo Taguchi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Yoshio Hirabayashi
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Shigeo Ohta
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Makoto Suematsu
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Satoshi Ogawa
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| | - Keiichi Fukuda
- From the Department of Regenerative Medicine and Advanced Cardiac Therapeutics (J.E., M. Sano, T.K., S. Morizane, T.M., Y.K., Y.Z., H.I., F.H., K.F.); Cardiology Division (J.E., T.K., T.M., Y.K., S. Ogawa), Department of Internal Medicine; Department of Biochemistry and Integrative Medical Biology (T.H., Y.N., T.A., M. Suematsu); and Division of Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO) (M. Sano),
| |
Collapse
|
44
|
Baba SP, Barski OA, Ahmed Y, O'Toole TE, Conklin DJ, Bhatnagar A, Srivastava S. Reductive metabolism of AGE precursors: a metabolic route for preventing AGE accumulation in cardiovascular tissue. Diabetes 2009; 58:2486-97. [PMID: 19651811 PMCID: PMC2768164 DOI: 10.2337/db09-0375] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To examine the role of aldo-keto reductases (AKRs) in the cardiovascular metabolism of the precursors of advanced glycation end products (AGEs). RESEARCH DESIGN AND METHODS Steady-state kinetic parameters of AKRs with AGE precursors were determined using recombinant proteins expressed in bacteria. Metabolism of methylglyoxal and AGE accumulation were studied in human umbilical vein endothelial cells (HUVECs) and C57 wild-type, akr1b3 (aldose reductase)-null, cardiospecific-akr1b4 (rat aldose reductase), and akr1b8 (FR-1)-transgenic mice. AGE accumulation and atherosclerotic lesions were studied 12 weeks after streptozotocin treatment of C57, akr1b3-null, and apoE- and akr1b3-apoE-null mice. RESULTS Higher levels of AGEs were generated in the cytosol than at the external surface of HUVECs cultured in high glucose, indicating that intracellular metabolism may be an important regulator of AGE accumulation and toxicity. In vitro, AKR 1A and 1B catalyzed the reduction of AGE precursors, whereas AKR1C, AKR6, and AKR7 were relatively ineffective. Highest catalytic efficiency was observed with AKR1B1. Acetol formation in methylglyoxal-treated HUVECs was prevented by the aldose reductase inhibitor sorbinil. Acetol was generated in hearts perfused with methylglyoxal, and its formation was increased in akr1b4- or akr1b8-transgenic mice. Reduction of AGE precursors was diminished in hearts from akr1b3-null mice. Diabetic akr1b3-null mice accumulated more AGEs in the plasma and the heart than wild-type mice, and deletion of akr1b3 increased AGE accumulation and atherosclerotic lesion formation in apoE-null mice. CONCLUSIONS Aldose reductase-catalyzed reduction is an important pathway in the endothelial and cardiac metabolism of AGE precursors, and it prevents AGE accumulation and atherosclerotic lesion formation.
Collapse
Affiliation(s)
- Shahid P. Baba
- From the Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Oleg A. Barski
- From the Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Yonis Ahmed
- From the Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Timothy E. O'Toole
- From the Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Daniel J. Conklin
- From the Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Aruni Bhatnagar
- From the Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Sanjay Srivastava
- From the Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
- Corresponding author: Sanjay Srivastava,
| |
Collapse
|
45
|
Litvinov D, Selvarajan K, Garelnabi M, Brophy L, Parthasarathy S. Anti-atherosclerotic actions of azelaic acid, an end product of linoleic acid peroxidation, in mice. Atherosclerosis 2009; 209:449-54. [PMID: 19880116 DOI: 10.1016/j.atherosclerosis.2009.09.076] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 09/15/2009] [Accepted: 09/29/2009] [Indexed: 01/31/2023]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease associated with the accumulation of oxidized lipids in arterial lesions. Recently we studied the degradation of peroxidized linoleic acid and suggested that oxidation is an essential process that results in the generation of terminal products, namely mono- and dicarboxylic acids that may lack the pro-atherogenic effects of peroxidized lipids. In continuation of that study, we tested the effects of azelaic acid (AzA), one of the end products of linoleic acid peroxidation, on the development of atherosclerosis using low density lipoprotein receptor knockout (LDLr(-/-)) mice. METHODS AND RESULTS LDLr(-/-) mice were fed with a high fat and high cholesterol Western diet (WD group). Another group of animals were fed the same diet with AzA supplementation (WD+AzA group). After 4 months of feeding, mice were sacrificed and atherosclerotic lesions were measured. The results showed that the average lesion area in WD+AzA group was 38% (p<0.001) less as compared to WD group. The athero-protective effect of AzA was not related to changes in plasma lipid content. AzA supplementation decreased the level of CD68 macrophage marker by 34% (p<0.05). CONCLUSIONS The finding that AzA exhibits an anti-atherogenic effect suggests that oxidation of lipid peroxidation-derived aldehydes into carboxylic acids could be an important step in the body's defense against oxidative damage.
Collapse
Affiliation(s)
- Dmitry Litvinov
- Division of Cardiothoracic Surgery, Ohio State University Medical Center, Columbus, OH 43210-1292, USA
| | | | | | | | | |
Collapse
|
46
|
Kinetic studies of AKR1B10, human aldose reductase-like protein: endogenous substrates and inhibition by steroids. Arch Biochem Biophys 2009; 487:1-9. [PMID: 19464995 DOI: 10.1016/j.abb.2009.05.009] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Revised: 05/14/2009] [Accepted: 05/17/2009] [Indexed: 02/02/2023]
Abstract
A human member of the aldo-keto reductase (AKR) superfamily, AKR1B10, was identified as a biomarker of lung cancer, exhibiting high sequence identity with human aldose reductase (AKR1B1). Using recombinant AKR1B10 and AKR1B1, we compared their substrate specificity for biogenic compounds and inhibition by endogenous compounds and found the following unique features of AKR1B10. AKR1B10 efficiently reduced long-chain aliphatic aldehydes including farnesal and geranylgeranial, which are generated from degradation of prenylated proteins and metabolism of farnesol and geranylgeraniol derived from the mevalonate pathway. The enzyme oxidized aliphatic and aromatic alcohols including 20alpha-hydroxysteroids. In addition, AKR1B10 was inhibited by steroid hormones, bile acids and their metabolites, showing IC(50) values of 0.03-25 microM. Kinetic analyses of the alcohol oxidation and inhibition by the steroids and tolrestat, together with the docked model of AKR1B10-inhibitor complex, suggest that the inhibitory steroids and tolrestat bind to overlapping sites within the active site of the enzyme-coenzyme complex. Thus, we propose a novel role of AKR1B10 in controlling isoprenoid homeostasis that is important in cholesterol synthesis and cell proliferation through salvaging isoprenoid alcohols, as well as its metabolic regulation by endogenous steroids.
Collapse
|
47
|
Endo S, Matsunaga T, Mamiya H, Hara A, Kitade Y, Tajima K, El-Kabbani O. Characterization of a rat NADPH-dependent aldo-keto reductase (AKR1B13) induced by oxidative stress. Chem Biol Interact 2008; 178:151-7. [PMID: 18845131 DOI: 10.1016/j.cbi.2008.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 09/11/2008] [Accepted: 09/12/2008] [Indexed: 10/21/2022]
Abstract
A rat aldo-keto reductase (AKR1B13) was identified as a hepatoma-derived protein, exhibiting high sequence identity with mouse fibroblast growth factor (FGF)-induced reductase, AKR1B8. In this study, AKR1B13 was characterized in terms of its enzymatic properties, tissue distribution and regulation. Recombinant AKR1B13 exhibited NADPH-linked reductase activity towards various aldehydes and alpha-dicarbonyl compounds, which include reactive compounds such as methylglyoxal, glyoxal, acrolein, 4-hydroxynonenal and 3-deoxyglucosone. The enzyme exhibited low NADP(+)-linked dehydrogenase activity towards aliphatic and aromatic alcohols, and was inhibited by aldose reductase inhibitors, flavonoids, benzbromarone and hexestrol. Immunochemical and reverse transcription-PCR analyses revealed that the enzyme is expressed in many rat tissues, endothelial cells and fibroblasts. Gene expression in YPEN-1 and NRK cells was up-regulated by treatments with submicromolar concentrations of hydrogen peroxide and 1,4-naphthoquinone, but not with FGF-1, FGF-2, 5alpha-dihydrotestosterone and 17beta-estradiol. These results indicate that AKR1B13 differs from AKR1B8 in tissue distribution and gene regulation, and suggest that it functions as a defense system against oxidative stress in rat tissues.
Collapse
Affiliation(s)
- Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, Japan.
| | | | | | | | | | | | | |
Collapse
|
48
|
Small-interference RNA-mediated knock-down of aldehyde oxidase 1 in 3T3-L1 cells impairs adipogenesis and adiponectin release. FEBS Lett 2008; 582:2965-72. [DOI: 10.1016/j.febslet.2008.07.034] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 07/07/2008] [Accepted: 07/20/2008] [Indexed: 12/14/2022]
|
49
|
Daiyasu H, Watanabe K, Toh H. Recruitment of thioredoxin-like domains into prostaglandin synthases. Biochem Biophys Res Commun 2008; 369:281-6. [DOI: 10.1016/j.bbrc.2008.02.088] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Accepted: 02/13/2008] [Indexed: 10/22/2022]
|
50
|
Jones PB, Brinson RG, Sarma SJ, Elkazaz S. Observation of heavy atom effects in the development of water soluble caged 4-hydroxy-trans-2-nonenal. Org Biomol Chem 2008; 6:4204-11. [DOI: 10.1039/b810954k] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|