1
|
Smyth SP, Nixon B, Skerrett-Byrne DA, Burke ND, Bromfield EG. Building an Understanding of Proteostasis in Reproductive Cells: The Impact of Reactive Carbonyl Species on Protein Fate. Antioxid Redox Signal 2024; 41:296-321. [PMID: 38115641 DOI: 10.1089/ars.2023.0314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Significance: Stringent regulation of protein homeostasis pathways, under both physiological and pathological conditions, is necessary for the maintenance of proteome fidelity and optimal cell functioning. However, when challenged by endogenous or exogenous stressors, these proteostasis pathways can become dysregulated with detrimental consequences for protein fate, cell survival, and overall organism health. Most notably, there are numerous somatic pathologies associated with a loss of proteostatic regulation, including neurodegenerative disorders, type 2 diabetes, and some cancers. Recent Advances: Lipid oxidation-derived reactive carbonyl species (RCS), such as 4-hydroxynonenal (4HNE) and malondialdehyde, are relatively underappreciated purveyors of proteostatic dysregulation, which elicit their effects via the nonenzymatic post-translational modification of proteins. Emerging evidence suggests that a subset of germline proteins can serve as substrates for 4HNE modification. Among these, prevalent targets include succinate dehydrogenase, heat shock protein A2 and A-kinase anchor protein 4, all of which are intrinsically associated with fertility. Critical Issues: Despite growing knowledge in this field, the RCS adductomes of spermatozoa and oocytes are yet to be comprehensively investigated. Furthermore, the manner by which RCS-mediated adduction impacts protein fate and drives cellular responses, such as protein aggregation, requires further examination in the germline. Given that RCS-protein adduction has been attributed a role in infertility, there has been sparked research investment into strategies to prevent lipid peroxidation in germ cells. Future Directions: An increased depth of knowledge regarding the mechanisms and substrates of RCS-mediated protein modification in reproductive cells may reveal important targets for the development of novel therapies to improve fertility and pregnancy outcomes for future generations.
Collapse
Affiliation(s)
- Shannon P Smyth
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Bio21 Institute, School of BioSciences, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Brett Nixon
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
| | - David A Skerrett-Byrne
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Nathan D Burke
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Bio21 Institute, School of BioSciences, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth G Bromfield
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Bio21 Institute, School of BioSciences, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
2
|
Agellon LB. Importance of fatty acid binding proteins in cellular function and organismal metabolism. J Cell Mol Med 2024; 28:e17703. [PMID: 36876733 PMCID: PMC10902576 DOI: 10.1111/jcmm.17703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/25/2023] [Accepted: 02/14/2023] [Indexed: 03/07/2023] Open
Abstract
Fatty acid binding proteins (Fabps) are small soluble proteins that are abundant in the cytosol. These proteins are known to bind a myriad of small hydrophobic molecules and have been postulated to serve a variety of roles, yet their precise functions have remained an enigma over half a century of study. Here, we consider recent findings, along with the cumulative findings contributed by many laboratories working on Fabps over the last half century, to synthesize a new outlook for what functions Fabps serve in cells and organisms. Collectively, the findings illustrate that Fabps function as versatile multi-purpose devices serving as sensors, conveyors and modulators to enable cells to detect and handle a specific class of metabolites, and to adjust their metabolic capacity and efficiency.
Collapse
Affiliation(s)
- Luis B. Agellon
- School of Human NutritionMcGill UniversitySte. Anne de BellevueQuebecCanada
| |
Collapse
|
3
|
You H, Wen X, Wang X, Zhu C, Chen H, Bu L, Zhang J, Qu S. Derlin-1 ameliorates nonalcoholic hepatic steatosis by promoting ubiquitylation and degradation of FABP1. Free Radic Biol Med 2023; 207:260-271. [PMID: 37499886 DOI: 10.1016/j.freeradbiomed.2023.07.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND AND AIMS The functions of liver fatty acid binding protein 1 (FABP1) in the regulation of nonalcoholic fatty liver disease (NAFLD) have been previously established. However, how FABP1 expression is dynamically regulated in metabolic disorders is unclear. Previous studies have reported that ubiquitin proteasome-mediated degradation of FABP1 is involved, but the mechanism remains unknown. METHODS Dysregulated expression of hepatic FABP1 and Derlin-1 was observed in NAFLD patients. We performed mice hepatic tissue coimmunoprecipitation based mass spectrum assays. Interaction between Derlin-1 and FABP1, and its impact on FABP1 ubiquitination status was evaluated by coimmunoprecipitation. The role of Derlin-1 in lipid deposition was tested using adenovirus-mediated overexpression in C57BL/6 mice, as well as by Derlin-1 overexpression or knockdown in HepG2 cells. RESULTS As a subunit of the endoplasmic reticulum-associated degradation complex, Derlin-1 was negatively associated with NAFLD patients, interacted with and ubiquitinated FABP1. Derlin-1 suppressed FABP1 levels and inhibited lipid deposition through a FABP1-dependent pathway. Additionally, Trim25, an E3 ubiquitin ligase present in the endoplasmic reticulum, was recruited to promote Derlin-1-related polyubiquitylation of FABP1, thereby creating a ubiquitin-associated network for FABP1 regulation. Derlin-1 overexpression ameliorated hepatic steatosis in both C57BL/6 mice and HepG2 cells, and contributed to attenuated weight gain, lower liver weight, and visceral fat mass. CONCLUSIONS FABP1 was degraded by Derlin-1 through ubiquitin modification. Negative regulation of FABP1 by Derlin-1 overexpression, suppressed lipid metabolism and alleviated lipid deposition in vivo and in vitro. Hence, Derlin-1 activation in hepatocytes may represent a potential therapeutic strategy for NAFLD.
Collapse
Affiliation(s)
- Hui You
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China; Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Xin Wen
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Xingchun Wang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China; Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Cuiling Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Haibing Chen
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Le Bu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China.
| | - Jun Zhang
- Research Center for Translational Medicine at East Hospital, Tongji University School of Medicine, Tongji University, Shanghai, 200092, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200072, PR China.
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China; Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China.
| |
Collapse
|
4
|
Ran H, Li C, Zhang M, Zhong J, Wang H. Neglected PTM in Animal Adipogenesis: E3-mediated Ubiquitination. Gene 2023:147574. [PMID: 37336271 DOI: 10.1016/j.gene.2023.147574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/21/2023]
Abstract
Ubiquitination is a widespread post-transcriptional modification (PTM) that occurs during protein degradation in eukaryotes and participates in almost all physiological and pathological processes, including animal adipogenesis. Ubiquitination is a cascade reaction regulated by the activating enzyme E1, conjugating enzyme E2, and ligase E3. Several recent studies have reported that E3 ligases play important regulatory roles in adipogenesis. However, as a key influencing factor for the recognition and connection between the substrate and ubiquitin during ubiquitination, its regulatory role in adipogenesis has not received adequate attention. In this review, we summarize the E3s' regulation and modification targets in animal adipogenesis, explain the regulatory mechanisms in lipogenic-related pathways, and further analyze the existing positive results to provide research directions of guiding significance for further studies on the regulatory mechanisms of E3s in animal adipogenesis.
Collapse
Affiliation(s)
- Hongbiao Ran
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610041, People's Republic of China
| | - Chunyan Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610041, People's Republic of China
| | - Ming Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610041, People's Republic of China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610041, People's Republic of China
| | - Hui Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610041, People's Republic of China.
| |
Collapse
|
5
|
Zhai C, Lonergan SM, Huff-Lonergan EJ, Johnson LG, Brown K, Prenni JE, N Nair M. Lipid Peroxidation Products Influence Calpain-1 Functionality In Vitro by Covalent Binding. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:7836-7846. [PMID: 37167568 DOI: 10.1021/acs.jafc.3c01225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The objective of the current study was to evaluate the effects of lipid peroxidation products, malondialdehyde (MDA), hexenal, and 4-hydroxynonenal (HNE), on calpain-1 function, and liquid chromatography and tandem mass spectrometry (LC-MS/MS) identification of adducts on calpain-1. Calpain-1 activity slightly increased after incubation with 100 μM MDA but not with 500 and 1000 μM MDA. However, calpain-1 activity was lowered by hexenal and HNE at 100, 500, and 1000 μM. No difference in calpain-1 autolysis was observed between the control and 1000 μM MDA. However, 1000 μM hexenal and HNE treatments slowed the calpain-1 autolysis. Adducts of MDA were detected on glutamine, arginine, lysine, histidine, and asparagine residues via Schiff base formation, while HNE adducts were detected on histidine, lysine, glutamine, and asparagine residues via Michael addition. These results are the first to demonstrate that lipid peroxidation products can impact calpain-1 activity in a concentration-dependent manner and may impact the development of meat tenderness postmortem.
Collapse
Affiliation(s)
- Chaoyu Zhai
- Department of Animal Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Animal Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Steven M Lonergan
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, United States
| | | | - Logan G Johnson
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, United States
| | - Kitty Brown
- Analytical Resources Core-Bioanalysis & Omics, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Jessica E Prenni
- Department of Horticulture and Landscape Architecture, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Mahesh N Nair
- Department of Animal Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| |
Collapse
|
6
|
Yang T, Li Q, Fadoul G, Alraqmany N, Ikonomovic M, Zhang F. Aldo-Keto Reductase 1C15 Characterization and Protection in Ischemic Brain Injury. Antioxidants (Basel) 2023; 12:antiox12040909. [PMID: 37107284 PMCID: PMC10135333 DOI: 10.3390/antiox12040909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Aldo-keto reductase (AKR) 1C15, a member of the AKR superfamily, was recently identified and cloned, and reported to alleviate oxidative stress in endothelial cells in rodent lungs. However, its expression and role in the brain and ischemic brain diseases have not been investigated. AKR1C15 expression was detected with real-time PCR. Mouse ischemic stroke and ischemic preconditioning (IPC) were established with middle cerebral artery occlusion (MCAO) for 1 h or 12 min, respectively. Recombinant AKR1C15 was administered intraperitoneally, and stroke outcome was evaluated with neurobehavioral tests and infarct volumes. Rat primary brain cell cultures were subjected to oxygen-glucose deprivation (OGD) to mimic ischemic injury. Cell survival or in vitro blood-brain barrier (BBB) permeability was measured, and nitric oxide (NO) release was detected. Immunostaining and Western blotting were used to evaluate oxidative-stress-related protein expression. AKR1C15 administration decreased the infarct volume and neurological deficits 2d post-stroke, and its early (1-h) administration after IPC abolished the protection of IPC against stroke. In rat primary brain cell cultures, AKR1C15 was most abundantly expressed in brain microvascular endothelial cells (BMVECs) and microglia. Its expression decreased upon OGD in most cell types except for BMVECs and microglia. In primary neuronal cultures, AKR1C15 treatment prevented OGD-induced cell death accompanied by decreased levels of 4-hydroxynonenal, 8-hydroxy-2'-deoxyguanosine, and heme oxygenase-1. In BMVEC cultures, AKR1C15 treatment protected against OGD-induced cell death and in vitro BBB leakage. In primary microglial cultures, AKR1C15 reduced the release of NO upon proinflammatory stimulation. Our results provide a characterization of the novel antioxidant AKR1C15 and demonstrate its protective role against ischemic injury, both in vivo and in vitro. AKR1C15 may be a promising agent for ischemic stroke treatment.
Collapse
Affiliation(s)
- Tuo Yang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Qianqian Li
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - George Fadoul
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Nour Alraqmany
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Milos Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Feng Zhang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
| |
Collapse
|
7
|
Milkovic L, Zarkovic N, Marusic Z, Zarkovic K, Jaganjac M. The 4-Hydroxynonenal–Protein Adducts and Their Biological Relevance: Are Some Proteins Preferred Targets? Antioxidants (Basel) 2023; 12:antiox12040856. [PMID: 37107229 PMCID: PMC10135105 DOI: 10.3390/antiox12040856] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
It is well known that oxidative stress and lipid peroxidation (LPO) play a role in physiology and pathology. The most studied LPO product with pleiotropic capabilities is 4-hydroxynonenal (4-HNE). It is considered as an important mediator of cellular signaling processes and a second messenger of reactive oxygen species. The effects of 4-HNE are mainly attributed to its adduction with proteins. Whereas the Michael adducts thus formed are preferred in an order of potency of cysteine > histidine > lysine over Schiff base formation, it is not known which proteins are the preferred targets for 4-HNE under what physiological or pathological conditions. In this review, we briefly discuss the methods used to identify 4-HNE–protein adducts, the progress of mass spectrometry in deciphering the specific protein targets, and their biological relevance, focusing on the role of 4-HNE protein adducts in the adaptive response through modulation of the NRF2/KEAP1 pathway and ferroptosis.
Collapse
Affiliation(s)
- Lidija Milkovic
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Neven Zarkovic
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Zlatko Marusic
- Division of Pathology, Clinical Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia
| | - Kamelija Zarkovic
- Division of Pathology, Clinical Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia
| | - Morana Jaganjac
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| |
Collapse
|
8
|
Somin S, Kulasiri D, Samarasinghe S. Alleviating the unwanted effects of oxidative stress on Aβ clearance: a review of related concepts and strategies for the development of computational modelling. Transl Neurodegener 2023; 12:11. [PMID: 36907887 PMCID: PMC10009979 DOI: 10.1186/s40035-023-00344-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Treatment for Alzheimer's disease (AD) can be more effective in the early stages. Although we do not completely understand the aetiology of the early stages of AD, potential pathological factors (amyloid beta [Aβ] and tau) and other co-factors have been identified as causes of AD, which may indicate some of the mechanism at work in the early stages of AD. Today, one of the primary techniques used to help delay or prevent AD in the early stages involves alleviating the unwanted effects of oxidative stress on Aβ clearance. 4-Hydroxynonenal (HNE), a product of lipid peroxidation caused by oxidative stress, plays a key role in the adduction of the degrading proteases. This HNE employs a mechanism which decreases catalytic activity. This process ultimately impairs Aβ clearance. The degradation of HNE-modified proteins helps to alleviate the unwanted effects of oxidative stress. Having a clear understanding of the mechanisms associated with the degradation of the HNE-modified proteins is essential for the development of strategies and for alleviating the unwanted effects of oxidative stress. The strategies which could be employed to decrease the effects of oxidative stress include enhancing antioxidant activity, as well as the use of nanozymes and/or specific inhibitors. One area which shows promise in reducing oxidative stress is protein design. However, more research is needed to improve the effectiveness and accuracy of this technique. This paper discusses the interplay of potential pathological factors and AD. In particular, it focuses on the effect of oxidative stress on the expression of the Aβ-degrading proteases through adduction of the degrading proteases caused by HNE. The paper also elucidates other strategies that can be used to alleviate the unwanted effects of oxidative stress on Aβ clearance. To improve the effectiveness and accuracy of protein design, we explain the application of quantum mechanical/molecular mechanical approach.
Collapse
Affiliation(s)
- Sarawoot Somin
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, 7647, New Zealand.,Department of Wine, Food and Molecular Biosciences, Lincoln University, Christchurch, 7647, New Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, 7647, New Zealand. .,Department of Wine, Food and Molecular Biosciences, Lincoln University, Christchurch, 7647, New Zealand.
| | - Sandhya Samarasinghe
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, 7647, New Zealand
| |
Collapse
|
9
|
Klein L, Phillips D, Kong F, Bowker B, Mohan A. 4-Oxo-2-nonenal (4-ONE)-Induced Degradation of Bovine Skeletal Muscle Proteins. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:12641-12650. [PMID: 36129340 DOI: 10.1021/acs.jafc.2c05550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Lipids are an important component of meat, as they provide desirable sensory characteristics and nutritional benefits. However, lipids are susceptible to degradation through oxidation and produce toxic oxidative byproducts. 4-Oxo-2-nonenal (4-ONE) is an oxidative byproduct that is highly reactive and cytotoxic. In this study, we investigated the influence of 4-ONE-induced protein degradation on fresh and gastric digested bovine skeletal muscle proteins. The results indicated that 4-ONE naturally forms in fresh muscle proteins. We report here for the first time that 4-ONE causes severe degradation of bovine skeletal muscle proteins. An SDS-PAGE gel analysis showed evidence that the skeletal muscle proteins attenuated over the incubation time, as the density of the protein bands faded significantly after 120 h. Additionally, protein and band density analyses showed a significant decrease in protein abundance and band densities throughout the incubation time. This study revealed that the lipid oxidation byproduct, 4-oxo-2-nonenal (4-ONE) is responsible for causing skeletal muscle protein degradation. Future studies should assess the bioprotective role of antioxidants and other food ingredients for their potential to prevent the formation and/or detoxification of 4-ONE in meat.
Collapse
Affiliation(s)
- Loren Klein
- Department of Food Science and Technology, University of Georgia, Athens, Georgia 30602, United States
| | - Dennis Phillips
- Department of Chemistry, University of Georgia, 140 Cedar Street, Athens, Georgia 30602, United States
| | - Fanbin Kong
- Department of Food Science and Technology, University of Georgia, Athens, Georgia 30602, United States
| | - Brian Bowker
- U.S. National Poultry Research Center, USDA-ARS, 950 College Station Rd., Athens, Georgia 30605, United States
| | - Anand Mohan
- Department of Food Science and Technology, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
10
|
|
11
|
Hindle A, Singh SP, Pradeepkiran JA, Bose C, Vijayan M, Kshirsagar S, Sawant NA, Reddy PH. Rlip76: An Unexplored Player in Neurodegeneration and Alzheimer’s Disease? Int J Mol Sci 2022; 23:ijms23116098. [PMID: 35682775 PMCID: PMC9181721 DOI: 10.3390/ijms23116098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and is the most common cause of dementia in older people. AD is associated with the loss of synapses, oxidative stress, mitochondrial structural and functional abnormalities, microRNA deregulation, inflammatory responses, neuronal loss, accumulation of amyloid-beta (Aβ) and phosphorylated tau (p-tau). AD occurs in two forms: early onset, familial AD and late-onset, sporadic AD. Causal factors are still unknown for a vast majority of AD patients. Genetic polymorphisms are proposed to contribute to late-onset AD via age-dependent increases in oxidative stress and mitochondrial abnormalities. Recent research from our lab revealed that reduced levels of Rlip76 induce oxidative stress, mitochondrial dysfunction and synaptic damage, leading to molecular and behavioral phenotypes resembling late-onset AD. Rlip76 is a multifunctional 76 kDa protein encoded by the RALBP1 gene, located on chromosome 18. Rlip is a stress-protective ATPase of the mercapturic acid pathway that couples clathrin-dependent endocytosis with the efflux of glutathione–electrophile conjugates. Rlip is evolutionarily highly conserved across species and is ubiquitously expressed in all tissues, including AD-affected brain regions, the cerebral cortex and hippocampus, where highly active neuronal metabolisms render the cells highly susceptible to intracellular oxidative damage. In the current article, we summarize molecular and cellular features of Rlip and how depleted Rlip may exacerbate oxidative stress, mitochondrial dysfunction and synaptic damage in AD. We also discuss the possible role of Rlip in aspects of learning and memory via axonal growth, dendritic remodeling, and receptor regulation. We conclude with a discussion of the potential for the contribution of genetic polymorphisms in Rlip to AD progression and the potential for Rlip-based therapies.
Collapse
Affiliation(s)
- Ashly Hindle
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (S.P.S.); (J.A.P.); (C.B.); (M.V.); (S.K.); (N.A.S.)
| | - Sharda P. Singh
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (S.P.S.); (J.A.P.); (C.B.); (M.V.); (S.K.); (N.A.S.)
| | - Jangampalli Adi Pradeepkiran
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (S.P.S.); (J.A.P.); (C.B.); (M.V.); (S.K.); (N.A.S.)
| | - Chhanda Bose
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (S.P.S.); (J.A.P.); (C.B.); (M.V.); (S.K.); (N.A.S.)
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (S.P.S.); (J.A.P.); (C.B.); (M.V.); (S.K.); (N.A.S.)
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (S.P.S.); (J.A.P.); (C.B.); (M.V.); (S.K.); (N.A.S.)
| | - Neha A. Sawant
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (S.P.S.); (J.A.P.); (C.B.); (M.V.); (S.K.); (N.A.S.)
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (S.P.S.); (J.A.P.); (C.B.); (M.V.); (S.K.); (N.A.S.)
- Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Correspondence:
| |
Collapse
|
12
|
Akagawa M. Protein carbonylation: molecular mechanisms, biological implications, and analytical approaches. Free Radic Res 2021; 55:307-320. [DOI: 10.1080/10715762.2020.1851027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Mitsugu Akagawa
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Japan
| |
Collapse
|
13
|
Dietary iron overload mitigates atherosclerosis in high-fat diet-fed apolipoprotein E knockout mice: Role of dysregulated hepatic fatty acid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159004. [PMID: 34245925 DOI: 10.1016/j.bbalip.2021.159004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
The atherosclerosis "iron hypothesis" generates a fair amount of debate since it has been proposed. Here, we revisited the "iron hypothesis" by examining whether dietary iron overload would intensify iron deposition in plaques and thus lead to further exacerbation of atherosclerosis in apolipoprotein E knockout (ApoE KO) mice. ApoE KO mice were fed either a normal chow diet (ND) or a high fat diet (HFD) supplemented with or without 2% carbonyl iron (Fe) for 16 weeks. However, contrary to our assumption, dietary iron overloading did not intensify, but rather diminished the atherosclerotic lesion area by 65.3%, which was accompanied by significantly decreased serum total cholesterol, triglyceride and low-density lipoprotein cholesterol contents, together with hepatic lipid accumulation decline, despite the evident existence of aortic iron accumulation and the typical signs of iron overload in ApoE KO mice. Using isobaric tag for absolute quantification (iTRAQ) proteomics approach, hepatic CD36 and fatty acid binding proteins-mediated fatty acid (FA) uptake and trafficking impairment were identified as the key potential pathomechanisms by which iron overload diminishes atherosclerotic lesions. Furthermore, downstream hepatic FA de novo biosynthesis was enhanced and FA oxidation was inhibited to compensate for the FA deficiency triggered by iron overload-impaired fatty acid uptake and trafficking. Our findings suggested that dietary iron overload is not atherogenic in ApoE KO mice, and more research efforts are warranted to revisit the "iron hypothesis" of atherosclerosis.
Collapse
|
14
|
Tsai MY, Yang WC, Lin CF, Wang CM, Liu HY, Lin CS, Lin JW, Lin WL, Lin TC, Fan PS, Hung KH, Lu YW, Chang GR. The Ameliorative Effects of Fucoidan in Thioacetaide-Induced Liver Injury in Mice. Molecules 2021; 26:molecules26071937. [PMID: 33808318 PMCID: PMC8036993 DOI: 10.3390/molecules26071937] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023] Open
Abstract
Liver disorders have been recognized as one major health concern. Fucoidan, a sulfated polysaccharide extracted from the brown seaweed Fucus serratus, has previously been reported as an anti-inflammatory and antioxidant. However, the discovery and validation of its hepatoprotective properties and elucidation of its mechanisms of action are still unknown. The objective of the current study was to investigate the effect and possible modes of action of a treatment of fucoidan against thioacetamide (TAA)-induced liver injury in male C57BL/6 mice by serum biochemical and histological analyses. The mouse model for liver damage was developed by the administration of TAA thrice a week for six weeks. The mice with TAA-induced liver injury were orally administered fucoidan once a day for 42 days. The treated mice showed significantly higher body weights; food intakes; hepatic antioxidative enzymes (catalase, glutathione peroxidase (GPx), and superoxide dismutase (SOD)); and a lower serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and C-reactive protein (CRP) levels. Additionally, a reduced hepatic IL-6 level and a decreased expression of inflammatory-related genes, such as cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) mRNA was observed. These results demonstrated that fucoidan had a hepatoprotective effect on liver injury through the suppression of the inflammatory responses and acting as an antioxidant. In addition, here, we validated the use of fucoidan against liver disorders with supporting molecular data.
Collapse
Affiliation(s)
- Ming-Yang Tsai
- Animal Industry Division, Livestock Research Institute, Council of Agriculture, Executive Yuan, 112 Muchang, Xinhua Dist, Tainan 71246, Taiwan;
- Graduate Institute of Bioresources, National Pingtung University of Science and Technology, 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan
| | - Wei-Cheng Yang
- School of Veterinary Medicine, National Taiwan University, 4 Section, 1 Roosevelt Road, Taipei 10617, Taiwan; (W.-C.Y.); (C.-S.L.)
| | - Chuen-Fu Lin
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Shuefu Road, Neipu, Pingtung 912301, Taiwan;
| | - Chao-Min Wang
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.); (P.-S.F.)
| | - Hsien-Yueh Liu
- Bachelor Degree Program in Animal Healthcare, Hungkuang University, 6 Section, 1018 Taiwan Boulevard, Shalu District, Taichung 433304, Taiwan; (H.-Y.L.); (J.-W.L.); (W.-L.L.)
| | - Chen-Si Lin
- School of Veterinary Medicine, National Taiwan University, 4 Section, 1 Roosevelt Road, Taipei 10617, Taiwan; (W.-C.Y.); (C.-S.L.)
| | - Jen-Wei Lin
- Bachelor Degree Program in Animal Healthcare, Hungkuang University, 6 Section, 1018 Taiwan Boulevard, Shalu District, Taichung 433304, Taiwan; (H.-Y.L.); (J.-W.L.); (W.-L.L.)
| | - Wei-Li Lin
- Bachelor Degree Program in Animal Healthcare, Hungkuang University, 6 Section, 1018 Taiwan Boulevard, Shalu District, Taichung 433304, Taiwan; (H.-Y.L.); (J.-W.L.); (W.-L.L.)
- General Education Center, Chaoyang University of Technology, 168 Jifeng Eastern Road, Taichung 413310, Taiwan
| | - Tzu-Chun Lin
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.); (P.-S.F.)
| | - Pei-Shan Fan
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.); (P.-S.F.)
| | - Kuo-Hsiang Hung
- Graduate Institute of Bioresources, National Pingtung University of Science and Technology, 1 Shuefu Road, Neipu, Pingtung 91201, Taiwan
- Correspondence: (K.-H.H.); (Y.-W.L.); (G.-R.C.)
| | - Yu-Wen Lu
- Department of Chinese Medicine, Show Chwan Memorial Hospital, 1 Section, 542 Chung-Shan Road, Changhua 50008, Taiwan
- Department of Chinese Medicine, Chang Bing Show Chwan Memorial Hospital, 6 Lugong Road, Changhua 50544, Taiwan
- Correspondence: (K.-H.H.); (Y.-W.L.); (G.-R.C.)
| | - Geng-Ruei Chang
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.); (P.-S.F.)
- Correspondence: (K.-H.H.); (Y.-W.L.); (G.-R.C.)
| |
Collapse
|
15
|
Fuloria S, Subramaniyan V, Karupiah S, Kumari U, Sathasivam K, Meenakshi DU, Wu YS, Guad RM, Udupa K, Fuloria NK. A Comprehensive Review on Source, Types, Effects, Nanotechnology, Detection, and Therapeutic Management of Reactive Carbonyl Species Associated with Various Chronic Diseases. Antioxidants (Basel) 2020; 9:E1075. [PMID: 33147856 PMCID: PMC7692604 DOI: 10.3390/antiox9111075] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Continuous oxidation of carbohydrates, lipids, and amino acids generate extremely reactive carbonyl species (RCS). Human body comprises some important RCS namely hexanal, acrolein, 4-hydroxy-2-nonenal, methylglyoxal, malondialdehyde, isolevuglandins, and 4-oxo-2- nonenal etc. These RCS damage important cellular components including proteins, nucleic acids, and lipids, which manifests cytotoxicity, mutagenicity, multitude of adducts and crosslinks that are connected to ageing and various chronic diseases like inflammatory disease, atherosclerosis, cerebral ischemia, diabetes, cancer, neurodegenerative diseases and cardiovascular disease. The constant prevalence of RCS in living cells suggests their importance in signal transduction and gene expression. Extensive knowledge of RCS properties, metabolism and relation with metabolic diseases would assist in development of effective approach to prevent numerous chronic diseases. Treatment approaches for RCS associated diseases involve endogenous RCS metabolizers, carbonyl metabolizing enzyme inducers, and RCS scavengers. Limited bioavailability and bio efficacy of RCS sequesters suggest importance of nanoparticles and nanocarriers. Identification of RCS and screening of compounds ability to sequester RCS employ several bioassays and analytical techniques. Present review describes in-depth study of RCS sources, types, properties, identification techniques, therapeutic approaches, nanocarriers, and their role in various diseases. This study will give an idea for therapeutic development to combat the RCS associated chronic diseases.
Collapse
Affiliation(s)
- Shivkanya Fuloria
- Faculty of Pharmacy, AIMST University, Kedah, Bedong 08100, Malaysia;
| | - Vetriselvan Subramaniyan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur 42610, Malaysia; (V.S.); (Y.S.W.)
| | - Sundram Karupiah
- Faculty of Pharmacy, AIMST University, Kedah, Bedong 08100, Malaysia;
| | - Usha Kumari
- Faculty of Medicine, AIMST University, Kedah, Bedong 08100, Malaysia;
| | | | | | - Yuan Seng Wu
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur 42610, Malaysia; (V.S.); (Y.S.W.)
| | - Rhanye Mac Guad
- Faculty of Medicine and Health Science, University Malaysia Sabah, Kota Kinabalu 88400, Malaysia;
| | - Kaviraja Udupa
- Department of Neurophysiology, NIMHANS, Bangalore 560029, India;
| | | |
Collapse
|
16
|
Alamil H, Galanti L, Heutte N, Van Der Schueren M, Dagher Z, Lechevrel M. Genotoxicity of aldehyde mixtures: profile of exocyclic DNA-adducts as a biomarker of exposure to tobacco smoke. Toxicol Lett 2020; 331:57-64. [PMID: 32442718 DOI: 10.1016/j.toxlet.2020.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 01/14/2023]
Abstract
Electrophilic compounds present in humans, originating from endogenous processes or pollutant exposures, pose a risk to health though their reaction with nucleophilic sites in protein and DNA. Among this chemical class, aldehydes are mainly present in indoor air and they can also be produced by endogenous lipid peroxidation arising from oxidative stress. Known to be very reactive, aldehydes have the ability to form exocyclic adducts to DNA that, for the most if not repaired correctly, are mutagenic and by consequence potential agents involved in carcinogenesis. The aim of this work was to establish profiles of exocyclic DNA adducts induced by aldehyde mixtures, which could ultimately be considered as a genotoxic marker of endogenous and environmental aldehyde exposure. Adducts were quantified by an accurate, sensitive and validated ultra high performance liquid chromatography-electrospray ionization analytical method coupled to mass spectrometry in the tandem mode (UHPLC-ESI-MS/MS). We simultaneously measured nine exocyclic DNA adducts generated during the exposure in vitro of calf thymus DNA to different concentrations of each aldehyde along, as well as, to an equimolar mixture of these aldehydes. This approach has enabled us to establish dose-response relationships that allowed displaying the specific reactivity of aldehydes towards corresponding adducts formation. Profiles of these adducts determined in DNA of current smokers and non-smokers blood samples supported these findings. These first results are encouraging to explore genotoxicity induced by aldehyde mixtures and can furthermore be used as future reference for adductomic approaches.
Collapse
Affiliation(s)
- Héléna Alamil
- Normandie University, UNICAEN, ABTE EA4651, Caen, France; CCC François Baclesse, UNICANCER, Caen, France; L2GE, Microbiology-Tox/Ecotox Team, Faculty of Sciences, Lebanese University, Fanar, Lebanon.
| | | | - Natacha Heutte
- CCC François Baclesse, UNICANCER, Caen, France; Normandie University, UNIROUEN, CETAPS EA3832, Mont Saint Aignan, Cedex, France
| | | | - Zeina Dagher
- L2GE, Microbiology-Tox/Ecotox Team, Faculty of Sciences, Lebanese University, Fanar, Lebanon
| | - Mathilde Lechevrel
- Normandie University, UNICAEN, ABTE EA4651, Caen, France; CCC François Baclesse, UNICANCER, Caen, France.
| |
Collapse
|
17
|
Allegra M, Restivo I, Fucarino A, Pitruzzella A, Vasto S, Livrea MA, Tesoriere L, Attanzio A. Proeryptotic Activity of 4-Hydroxynonenal: A New Potential Physiopathological Role for Lipid Peroxidation Products. Biomolecules 2020; 10:biom10050770. [PMID: 32429353 PMCID: PMC7277761 DOI: 10.3390/biom10050770] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/06/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Eryptosis is a physiological, apoptosis-like death of injured erythrocytes crucial to prevent premature haemolysis and the pathological sequalae generated by cell-free haemoglobin. When dysregulated, the process is associated to several inflammatory-based pathologies. 4-Hydroxy-trans-2-nonenal (HNE) is an endogenous signalling molecule at physiological levels and, at higher concentrations, is involved in the pathogenesis of several inflammatory-based diseases. This work evaluated whether HNE could induce eryptosis in human erythrocytes. Methods: Measurements of phosphatidylserine, cell volume, intracellular oxidants, Ca++, glutathione, ICAM-1, and ceramide were assessed by flow cytometry. Scanning electron microscopy evaluated morphological alterations of erythrocytes. Western blotting assessed caspases. PGE2 was measured by ELISA. Adhesion of erythrocytes on endothelial cells was evaluated by gravity adherence assay. Results: HNE in the concentration range between 10–100 µM induces eryptosis, morphological alterations correlated to caspase-3 activation, and increased Ca++ levels. The process is not mediated by redox-dependent mechanisms; rather, it strongly depends on PGE2 and ceramide. Interestingly, HNE induces significant increase of erythrocytes adhesion to endothelial cells (ECs) that are in turn dysfunctionated as evident by overexpression of ICAM-1. Conclusions: Our results unveil a new physiopathological role for HNE, provide mechanistic details of the HNE-induced eryptosis, and suggest a novel mechanism through which HNE could exert pro-inflammatory effects.
Collapse
Affiliation(s)
- Mario Allegra
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università di Palermo, 90123 Palermo, Italy; (M.A.); (I.R.); (S.V.); (M.A.L.); (A.A.)
| | - Ignazio Restivo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università di Palermo, 90123 Palermo, Italy; (M.A.); (I.R.); (S.V.); (M.A.L.); (A.A.)
| | - Alberto Fucarino
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, 90127 Palermo, Italy; (A.F.); (A.P.)
| | - Alessandro Pitruzzella
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, 90127 Palermo, Italy; (A.F.); (A.P.)
- Consorzio Universitario di Caltanissetta, Università di Palermo, 90127 Palermo, Italy
| | - Sonya Vasto
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università di Palermo, 90123 Palermo, Italy; (M.A.); (I.R.); (S.V.); (M.A.L.); (A.A.)
| | - Maria Antonia Livrea
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università di Palermo, 90123 Palermo, Italy; (M.A.); (I.R.); (S.V.); (M.A.L.); (A.A.)
| | - Luisa Tesoriere
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università di Palermo, 90123 Palermo, Italy; (M.A.); (I.R.); (S.V.); (M.A.L.); (A.A.)
- Correspondence: ; Tel.: +39-091-2389-6824
| | - Alessandro Attanzio
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università di Palermo, 90123 Palermo, Italy; (M.A.); (I.R.); (S.V.); (M.A.L.); (A.A.)
| |
Collapse
|
18
|
Barman S, Srinivasan K. Ameliorative effect of zinc supplementation on compromised small intestinal health in streptozotocin-induced diabetic rats. Chem Biol Interact 2019; 307:37-50. [DOI: 10.1016/j.cbi.2019.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/04/2019] [Accepted: 04/15/2019] [Indexed: 01/29/2023]
|
19
|
Alviz-Amador A, Galindo-Murillo R, Pineda-Alemán R, Pérez-González H, Rodríguez-Cavallo E, Vivas-Reyes R, Méndez-Cuadro D. 4-HNE carbonylation induces local conformational changes on bovine serum albumin and thioredoxin. A molecular dynamics study. J Mol Graph Model 2019; 86:298-307. [DOI: 10.1016/j.jmgm.2018.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 10/22/2018] [Accepted: 11/01/2018] [Indexed: 01/09/2023]
|
20
|
Ali H, Assiri MA, Shearn CT, Fritz KS. Lipid peroxidation derived reactive aldehydes in alcoholic liver disease. CURRENT OPINION IN TOXICOLOGY 2018; 13:110-117. [PMID: 31263795 DOI: 10.1016/j.cotox.2018.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipid peroxidation is a known consequence of oxidative stress and is thought to play a key role in numerous disease pathologies, including alcoholic liver disease (ALD). The overaccumulation of lipid peroxidation products during chronic alcohol consumption results in pathogenic lesions on protein, DNA, and lipids throughout the cell. Molecular adducts due to secondary end products of lipid peroxidation impact a host of biochemical processes, including inflammation, antioxidant defense, and metabolism. The aggregate burden of lipid peroxidation which occurs due to chronic alcohol metabolism, including downstream signaling events, contributes to the development and progression of ALD. In this current opinion we highlight recent studies and approaches relating cellular mechanisms of lipid peroxidation to the pathogenesis of alcoholic liver disease.
Collapse
Affiliation(s)
- Hadi Ali
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Mohammed A Assiri
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Colin T Shearn
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Kristofer S Fritz
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
21
|
Lipid vesicles affect the aggregation of 4-hydroxy-2-nonenal-modified α-synuclein oligomers. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3060-3068. [PMID: 29960040 DOI: 10.1016/j.bbadis.2018.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 11/23/2022]
Abstract
Parkinson's disease (PD) and other synucleinopathies are characterized by accumulation of misfolded aggregates of α-synuclein (α-syn). The normal function of α-syn is still under investigation, but it has been generally linked to synaptic plasticity, neurotransmitter release and the maintenance of the synaptic pool. α-Syn localizes at synaptic terminals where it can bind to synaptic vesicles as well as to other cellular membranes. It has become clear that these interactions have an impact on both α-syn functional role and its propensity to aggregate. In this study, we investigated the aggregation process of α-syn covalently modified with 4-hydroxy-2-nonenal (HNE). HNE is a product of lipid peroxidation and has been implicated in the pathogenesis of different neurodegenerative diseases by modifying the kinetics of soluble toxic oligomers. Although HNE-modified α-syn has been reported to assemble into stable oligomers, we found that slightly acidic conditions promoted further protein aggregation. Lipid vesicles delayed the aggregation process in a concentration-dependent manner, an effect that was observed only when they were added at the beginning of the aggregation process. Co-aggregation of lipid vesicles with HNE-modified α-syn also induced cytotoxic effects on differentiated SHSY-5Y cells. Under conditions in which the aggregation process was delayed cell viability was reduced. By exploring the behavior and potential cytotoxic effects of HNE-α-syn under acidic conditions in relation to protein-lipid interactions our study gives a framework to examine a possible pathway leading from a physiological setting to the pathological outcome of PD.
Collapse
|
22
|
Shah DD, Singh SM, Dzieciatkowska M, Mallela KMG. Biophysical analysis of the effect of chemical modification by 4-oxononenal on the structure, stability, and function of binding immunoglobulin protein (BiP). PLoS One 2017; 12:e0183975. [PMID: 28886061 PMCID: PMC5590874 DOI: 10.1371/journal.pone.0183975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/15/2017] [Indexed: 11/24/2022] Open
Abstract
Binding immunoglobulin protein (BiP) is a molecular chaperone important for the folding of numerous proteins, which include millions of immunoglobulins in human body. It also plays a key role in the unfolded protein response (UPR) in the endoplasmic reticulum. Free radical generation is a common phenomenon that occurs in cells under healthy as well as under stress conditions such as ageing, inflammation, alcohol consumption, and smoking. These free radicals attack the cell membranes and generate highly reactive lipid peroxidation products such as 4-oxononenal (4-ONE). BiP is a key protein that is modified by 4-ONE. In this study, we probed how such chemical modification affects the biophysical properties of BiP. Upon modification, BiP shows significant tertiary structural changes with no changes in its secondary structure. The protein loses its thermodynamic stability, particularly, that of the nucleotide binding domain (NBD) where ATP binds. In terms of function, the modified BiP completely loses its ATPase activity with decreased ATP binding affinity. However, modified BiP retains its immunoglobulin binding function and its chaperone activity of suppressing non-specific protein aggregation. These results indicate that 4-ONE modification can significantly affect the structure-function of key proteins such as BiP involved in cellular pathways, and provide a molecular basis for how chemical modifications can result in the failure of quality control mechanisms inside the cell.
Collapse
Affiliation(s)
- Dinen D Shah
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Surinder M Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Monika Dzieciatkowska
- Biological Mass Spectrometry Facility, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Krishna M G Mallela
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America.,Program in Structural Biology and Biochemistry, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| |
Collapse
|
23
|
Caira S, Iannelli A, Sciarrillo R, Picariello G, Renzone G, Scaloni A, Addeo P. Differential representation of liver proteins in obese human subjects suggests novel biomarkers and promising targets for drug development in obesity. J Enzyme Inhib Med Chem 2017; 32:672-682. [PMID: 28274171 PMCID: PMC6009959 DOI: 10.1080/14756366.2017.1292262] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The proteome of liver biopsies from human obese (O) subjects has been compared to those of nonobese (NO) subjects using two-dimensional gel electrophoresis (2-DE). Differentially represented proteins were identified by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry (MS)-based peptide mass fingerprinting (PMF) and nanoflow-liquid chromatography coupled to electrospray-tandem mass spectrometry (nLC-ESI-MS/MS). Overall, 61 gene products common to all of the liver biopsies were identified within 65 spots, among which 25 ones were differently represented between O and NO subjects. In particular, over-representation of short-chain acyl-CoA dehydrogenase, Δ(3,5)-Δ(2,4)dienoyl-CoA isomerase, acetyl-CoA acetyltransferase, glyoxylate reductase/hydroxypyruvate reductase, fructose-biphosphate aldolase B, peroxiredoxin I, protein DJ-1, catalase, α- and β-hemoglobin subunits, 3-mercaptopyruvate S-transferase, calreticulin, aminoacylase 1, phenazine biosynthesis-like domain-containing protein and a form of fatty acid-binding protein, together with downrepresentation of glutamate dehydrogenase, glutathione S-transferase A1, S-adenosylmethionine synthase 1A and a form of apolipoprotein A-I, was associated with the obesity condition. Some of these metabolic enzymes and antioxidant proteins have already been identified as putative diagnostic markers of liver dysfunction in animal models of steatosis or obesity, suggesting additional investigations on their role in these syndromes. Their differential representation in human liver was suggestive of their consideration as obesity human biomarkers and for the development of novel antiobesity drugs.
Collapse
Affiliation(s)
- Simonetta Caira
- a Proteomics and Mass Spectrometry Laboratory , ISPAAM, National Research Council , Naples , Italy
| | - Antonio Iannelli
- b Département de Chirurgie Digestive , Centre Hospitalier Universitarie de Nice , Nice , France
| | - Rosaria Sciarrillo
- c Dipartimento di Scienze e Tecnologie , Università degli Studi del Sannio , Benevento , Italy
| | | | - Giovanni Renzone
- a Proteomics and Mass Spectrometry Laboratory , ISPAAM, National Research Council , Naples , Italy
| | - Andrea Scaloni
- a Proteomics and Mass Spectrometry Laboratory , ISPAAM, National Research Council , Naples , Italy
| | - Pietro Addeo
- e Service de Chirurgie Hépatique, Pancréatique, Biliaire et Transplantation, Pôle des Pathologies Digestives, Hépatiques et de la Transplantation, Hôpital de Hautepierre , Université de Strasbourg, Hôpitaux Universitaires de Strasbourg , Strasbourg , France
| |
Collapse
|
24
|
Zimmermann L, Moldzio R, Vazdar K, Krewenka C, Pohl EE. Nutrient deprivation in neuroblastoma cells alters 4-hydroxynonenal-induced stress response. Oncotarget 2017; 8:8173-8188. [PMID: 28030790 PMCID: PMC5352392 DOI: 10.18632/oncotarget.14132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/21/2016] [Indexed: 01/29/2023] Open
Abstract
4-hydroxy-2-nonenal (HNE), a toxic lipid peroxidation product, is associated with oxidative damage in cells and involved in various diseases including the initiation and progression of cancer. Cancer cells have a high, adaptable metabolism with a shift from oxidative phosphorylation to glycolysis and rely on high levels of glucose and glutamine as essential nutrients for cell growth. Here we investigated whether the toxic effects of HNE on the mitochondrial membrane potential (MMP) of cancer cells depends on their metabolic state by deprivation of glucose and/or glutamine. The addition of 16 μM HNE to N18TG2 neuroblastoma cells incubated in glucose medium led to a severe reduction of MMP, which was similar to the MMP of cells fed with both glucose and glutamine. In contrast, HNE addition to cells starved in glutamine medium increased their MMP slightly for a prolonged time period and this was accompanied by increased cellular survival. We found that ß-oxidation of HNE did not cause the increased MMP, since the aldehyde dehydrogenase was distinctly more active in cells with glucose medium. However, after blocking fatty acid ß-oxidation in cells starved in glutamine medium with etomoxir, which inhibits carnitine palmitoyltransferase 1, HNE addition induced a strong reduction of MMP similar to cells in glucose medium. Surprisingly, the effect of more toxic 4-oxo-2-nonenal was less pronounced. Our results suggest that in contrast to cells fed with glucose, glutamine-fed cancer cells are capable of ß-oxidizing fatty acids to maintain their MMP to combat the toxic effects of HNE.
Collapse
Affiliation(s)
- Lars Zimmermann
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Rudolf Moldzio
- Institute of Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Katarina Vazdar
- Division of Organic Chemistry and Biochemistry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Christopher Krewenka
- Institute of Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Elena E. Pohl
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
25
|
Zhang H, Forman HJ. Signaling by 4-hydroxy-2-nonenal: Exposure protocols, target selectivity and degradation. Arch Biochem Biophys 2016; 617:145-154. [PMID: 27840096 DOI: 10.1016/j.abb.2016.11.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/20/2016] [Accepted: 11/09/2016] [Indexed: 01/26/2023]
Abstract
4-hydroxy-2-nonenal (HNE), a major non-saturated aldehyde product of lipid peroxidation, has been extensively studied as a signaling messenger. In these studies a wide range of HNE concentrations have been used, ranging from the unstressed plasma concentration to far beyond what would be found in actual pathophysiological condition. In addition, accumulating evidence suggest that signaling protein modification by HNE is specific with only those proteins with cysteine, histidine, and lysine residues located in certain sequence or environments adducted by HNE. HNE-signaling is further regulated through the turnover of HNE-signaling protein adducts through proteolytic process that involve proteasomes, lysosomes and autophagy. This review discusses the HNE concentrations and exposure modes used in signaling studies, the selectivity of the HNE-adduction site, and the turnover of signaling protein adducts.
Collapse
Affiliation(s)
- Hongqiao Zhang
- Andrus Gerontology Center of the Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089-0191, USA.
| | - Henry Jay Forman
- Andrus Gerontology Center of the Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089-0191, USA
| |
Collapse
|
26
|
Martell J, Seo Y, Bak DW, Kingsley SF, Tissenbaum HA, Weerapana E. Global Cysteine-Reactivity Profiling during Impaired Insulin/IGF-1 Signaling in C. elegans Identifies Uncharacterized Mediators of Longevity. Cell Chem Biol 2016; 23:955-66. [PMID: 27499530 DOI: 10.1016/j.chembiol.2016.06.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 06/23/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
In the nematode Caenorhabditis elegans, inactivating mutations in the insulin/IGF-1 receptor, DAF-2, result in a 2-fold increase in lifespan mediated by DAF-16, a FOXO-family transcription factor. Downstream protein activities that directly regulate longevity during impaired insulin/IGF-1 signaling (IIS) are poorly characterized. Here, we use global cysteine-reactivity profiling to identify protein activity changes during impaired IIS. Upon confirming that cysteine reactivity is a good predictor of functionality in C. elegans, we profiled cysteine-reactivity changes between daf-2 and daf-16;daf-2 mutants, and identified 40 proteins that display a >2-fold change. Subsequent RNAi-mediated knockdown studies revealed that lbp-3 and K02D7.1 knockdown caused significant increases in lifespan and dauer formation. The proteins encoded by these two genes, LBP-3 and K02D7.1, are implicated in intracellular fatty acid transport and purine metabolism, respectively. These studies demonstrate that cysteine-reactivity profiling can be complementary to abundance-based transcriptomic and proteomic studies, serving to identify uncharacterized mediators of C. elegans longevity.
Collapse
Affiliation(s)
- Julianne Martell
- Department of Chemistry, Boston College, Chestnut Hill, MA 02467, USA
| | - Yonghak Seo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Daniel W Bak
- Department of Chemistry, Boston College, Chestnut Hill, MA 02467, USA
| | - Samuel F Kingsley
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Heidi A Tissenbaum
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|
27
|
Chacko BK, Wall SB, Kramer PA, Ravi S, Mitchell T, Johnson MS, Wilson L, Barnes S, Landar A, Darley-Usmar VM. Pleiotropic effects of 4-hydroxynonenal on oxidative burst and phagocytosis in neutrophils. Redox Biol 2016; 9:57-66. [PMID: 27393890 PMCID: PMC4939321 DOI: 10.1016/j.redox.2016.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 01/09/2023] Open
Abstract
Metabolic control of cellular function is significant in the context of inflammation-induced metabolic dysregulation in immune cells. Generation of reactive oxygen species (ROS) such as hydrogen peroxide and superoxide are one of the critical events that modulate the immune response in neutrophils. When activated, neutrophil NADPH oxidases consume large quantities of oxygen to rapidly generate ROS, a process that is referred to as the oxidative burst. These ROS are required for the efficient removal of phagocytized cellular debris and pathogens. In chronic inflammatory diseases, neutrophils are exposed to increased levels of oxidants and pro-inflammatory cytokines that can further prime oxidative burst responses and generate lipid oxidation products such as 4-hydroxynonenal (4-HNE). In this study we hypothesized that since 4-HNE can target glycolysis then this could modify the oxidative burst. To address this the oxidative burst was determined in freshly isolated healthy subject neutrophils using 13-phorbol myristate acetate (PMA) and the extracellular flux analyzer. Neutrophils pretreated with 4-HNE exhibited a significant decrease in the oxidative burst response and phagocytosis. Mass spectrometric analysis of alkyne-HNE treated neutrophils followed by click chemistry detected modification of a number of cytoskeletal, metabolic, redox and signaling proteins that are critical for the NADPH oxidase mediated oxidative burst. These modifications were confirmed using a candidate immunoblot approach for critical proteins of the active NADPH oxidase enzyme complex (Nox2 gp91phox subunit and Rac1 of the NADPH oxidase) and glyceraldehyde phosphate dehydrogenase, a critical enzyme in the metabolic regulation of oxidative burst. Taken together, these data suggest that 4-HNE-induces a pleiotropic mechanism to inhibit neutrophil function. These mechanisms may contribute to the immune dysregulation associated with chronic pathological conditions where 4-HNE is generated. Phagocytosis and glycolysis are inhibited in neutrophils by 4-hydroxynonenal. Click chemistry with alkyne-HNE identifies over 100 potential protein targets. Rac1, NOX2 and GAPDH are modified by 4-HNE. The 4-HNE-dependent inhibition of neutrophil function is mediated by a pleiotropic mechanism.
Collapse
Affiliation(s)
- Balu K Chacko
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States
| | - Stephanie B Wall
- Department of Pathology, University of Alabama at Birmingham, United States
| | - Philip A Kramer
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States
| | - Saranya Ravi
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States
| | - Tanecia Mitchell
- Department of Urology, University of Alabama at Birmingham, United States
| | - Michelle S Johnson
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States
| | - Landon Wilson
- Department of Pharmacology and Toxicology, The Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, United States
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, The Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, United States
| | - Aimee Landar
- Department of Pathology, University of Alabama at Birmingham, United States
| | - Victor M Darley-Usmar
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States.
| |
Collapse
|
28
|
Schroeder F, McIntosh AL, Martin GG, Huang H, Landrock D, Chung S, Landrock KK, Dangott LJ, Li S, Kaczocha M, Murphy EJ, Atshaves BP, Kier AB. Fatty Acid Binding Protein-1 (FABP1) and the Human FABP1 T94A Variant: Roles in the Endocannabinoid System and Dyslipidemias. Lipids 2016; 51:655-76. [PMID: 27117865 PMCID: PMC5408584 DOI: 10.1007/s11745-016-4155-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/11/2016] [Indexed: 01/01/2023]
Abstract
The first discovered member of the mammalian FABP family, liver fatty acid binding protein (FABP1, L-FABP), occurs at high cytosolic concentration in liver, intestine, and in the case of humans also in kidney. While the rat FABP1 is well studied, the extent these findings translate to human FABP1 is not clear-especially in view of recent studies showing that endocannabinoids and cannabinoids represent novel rat FABP1 ligands and FABP1 gene ablation impacts the hepatic endocannabinoid system, known to be involved in non-alcoholic fatty liver (NAFLD) development. Although not detectable in brain, FABP1 ablation nevertheless also impacts brain endocannabinoids. Despite overall tertiary structure similarity, human FABP1 differs significantly from rat FABP1 in secondary structure, much larger ligand binding cavity, and affinities/specificities for some ligands. Moreover, while both mouse and human FABP1 mediate ligand induction of peroxisome proliferator activated receptor-α (PPARα), they differ markedly in pattern of genes induced. This is critically important because a highly prevalent human single nucleotide polymorphism (SNP) (26-38 % minor allele frequency and 8.3 ± 1.9 % homozygous) results in a FABP1 T94A substitution that further accentuates these species differences. The human FABP1 T94A variant is associated with altered body mass index (BMI), clinical dyslipidemias (elevated plasma triglycerides and LDL cholesterol), atherothrombotic cerebral infarction, and non-alcoholic fatty liver disease (NAFLD). Resolving human FABP1 and the T94A variant's impact on the endocannabinoid and cannabinoid system is an exciting challenge due to the importance of this system in hepatic lipid accumulation as well as behavior, pain, inflammation, and satiety.
Collapse
Affiliation(s)
- Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA.
| | - Avery L McIntosh
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Huan Huang
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Sarah Chung
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Kerstin K Landrock
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Lawrence J Dangott
- Department of Biochemistry and Biophysics, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| | - Shengrong Li
- Avanti Polar Lipids, 700 Industrial Park Dr., Alabaster, AL, 35007-9105, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Eric J Murphy
- Department of Pharmacology, Physiology, and Therapeutics and Chemistry, University of North Dakota, Grand Forks, ND, 58202-9037, USA
| | - Barbara P Atshaves
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, TVMC, College Station, TX, 77843-4466, USA
| |
Collapse
|
29
|
Furuhashi M, Fuseya T, Murata M, Hoshina K, Ishimura S, Mita T, Watanabe Y, Omori A, Matsumoto M, Sugaya T, Oikawa T, Nishida J, Kokubu N, Tanaka M, Moniwa N, Yoshida H, Sawada N, Shimamoto K, Miura T. Local Production of Fatty Acid-Binding Protein 4 in Epicardial/Perivascular Fat and Macrophages Is Linked to Coronary Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:825-34. [PMID: 27013610 DOI: 10.1161/atvbaha.116.307225] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/14/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Fatty acid-binding protein 4 (FABP4) is expressed in adipocytes and macrophages, and elevated circulating FABP4 level is associated with obesity-mediated metabolic phenotype. We systematically investigated roles of FABP4 in the development of coronary artery atherosclerosis. APPROACH AND RESULTS First, by immunohistochemical analyses, we found that FABP4 was expressed in macrophages within coronary atherosclerotic plaques and epicardial/perivascular fat in autopsy cases and macrophages within thrombi covering ruptured coronary plaques in thrombectomy samples from patients with acute myocardial infarction. Second, we confirmed that FABP4 was secreted from macrophages and adipocytes cultured in vitro. Third, we investigated the effect of exogenous FABP4 on macrophages and human coronary artery-derived smooth muscle cells and endothelial cells in vitro. Treatment of the cells with recombinant FABP4 significantly increased gene expression of inflammatory markers in a dose-dependent manner. Finally, we measured serum FABP4 level in the aortic root (Ao-FABP4) and coronary sinus (CS-FABP4) of 34 patients with suspected or known coronary artery disease. Coronary stenosis score assessed by the modified Gensini score was weakly correlated with CS-FABP4 but was not correlated with Ao-FABP4. A stronger correlation (r=0.59, P<0.01) was observed for the relationship between coronary stenosis score and coronary veno-arterial difference in FABP4 level, (CS-Ao)-FABP4, indicating local production of FABP4 during coronary circulation in the heart. Multivariate analysis indicated that (CS-Ao)-FABP4 was an independent predictor of the severity of coronary stenosis after adjustment of conventional risk factors. CONCLUSIONS FABP4 locally produced by epicardial/perivascular fat and macrophages in vascular plaques contributes to the development of coronary atherosclerosis.
Collapse
Affiliation(s)
- Masato Furuhashi
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.).
| | - Takahiro Fuseya
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Masaki Murata
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Kyoko Hoshina
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Shutaro Ishimura
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Tomohiro Mita
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Yuki Watanabe
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Akina Omori
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Megumi Matsumoto
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Takeshi Sugaya
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Tsuyoshi Oikawa
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Junichi Nishida
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Nobuaki Kokubu
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Marenao Tanaka
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Norihito Moniwa
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Hideaki Yoshida
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Norimasa Sawada
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Kazuaki Shimamoto
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| | - Tetsuji Miura
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine (M.F., T.F., K.H., S.I., T.M., Y.W., A.O., M.M., J.N., N.K., M.T., N.M., H.Y., T.M.) and Molecular and Cellular Pathology (M.M., N.S.), Sapporo Medical University School of Medicine, Sapporo, Japan; Sapporo Medical University, Chuo-ku, Sapporo, Japan (K.S.); Department of Cardiovascular Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan (S.I., T.M.); Department of Nephrology and Hypertension, St. Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki, Kanagawa, Japan (T.S.); and CIMIC Co, Ltd, Yushima, Bunkyo-ku, Tokyo, Japan (T.S., T.O.)
| |
Collapse
|
30
|
Ravi S, Johnson MS, Chacko BK, Kramer PA, Sawada H, Locy ML, Wilson LS, Barnes S, Marques MB, Darley-Usmar VM. Modification of platelet proteins by 4-hydroxynonenal: Potential Mechanisms for inhibition of aggregation and metabolism. Free Radic Biol Med 2016; 91:143-53. [PMID: 26475426 PMCID: PMC4761519 DOI: 10.1016/j.freeradbiomed.2015.10.408] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/06/2015] [Accepted: 10/10/2015] [Indexed: 01/23/2023]
Abstract
Platelet aggregation is an essential response to tissue injury and is associated with activation of pro-oxidant enzymes, such as cyclooxygenase, and is also a highly energetic process. The two central energetic pathways in the cell, glycolysis and mitochondrial oxidative phosphorylation, are susceptible to damage by reactive lipid species. Interestingly, how platelet metabolism is affected by the oxidative stress associated with aggregation is largely unexplored. To address this issue, we examined the response of human platelets to 4-hydroxynonenal (4-HNE), a reactive lipid species which is generated during thrombus formation and during oxidative stress. Elevated plasma 4-HNE has been associated with renal failure, septic shock and cardiopulmonary bypass surgery. In this study, we found that 4-HNE decreased thrombin stimulated platelet aggregation by approximately 60%. The metabolomics analysis demonstrated that underlying our previous observation of a stimulation of platelet energetics by thrombin glycolysis and TCA (Tricarboxylic acid) metabolites were increased. Next, we assessed the effect of both 4-HNE and alkyne HNE (A-HNE) on bioenergetics and targeted metabolomics, and found a stimulatory effect on glycolysis, associated with inhibition of bioenergetic parameters. In the presence of HNE and thrombin glycolysis was further stimulated but the levels of the TCA metabolites were markedly suppressed. Identification of proteins modified by A-HNE followed by click chemistry and mass spectrometry revealed essential targets in platelet activation including proteins involved in metabolism, adhesion, cytoskeletal reorganization, aggregation, vesicular transport, protein folding, antioxidant proteins, and small GTPases. In summary, the biological effects of 4-HNE can be more effectively explained in platelets by the integrated effects of the modification of an electrophile responsive proteome rather than the isolated effects of candidate proteins.
Collapse
Affiliation(s)
- Saranya Ravi
- Department of Pathology; UAB Mitochondrial Medicine Laboratory; Center for Free Radical Biology
| | - Michelle S Johnson
- Department of Pathology; UAB Mitochondrial Medicine Laboratory; Center for Free Radical Biology
| | - Balu K Chacko
- Department of Pathology; UAB Mitochondrial Medicine Laboratory; Center for Free Radical Biology
| | - Philip A Kramer
- Department of Pathology; UAB Mitochondrial Medicine Laboratory; Center for Free Radical Biology
| | - Hirotaka Sawada
- Department of Pathology; UAB Mitochondrial Medicine Laboratory; Center for Free Radical Biology
| | - Morgan L Locy
- Department of Pathology; UAB Mitochondrial Medicine Laboratory; Center for Free Radical Biology
| | | | - Stephen Barnes
- The Targeted Metabolomics and Proteomics Laboratory; Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Victor M Darley-Usmar
- Department of Pathology; UAB Mitochondrial Medicine Laboratory; Center for Free Radical Biology.
| |
Collapse
|
31
|
Schwarzer E, Gallo V, Valente E, Ulliers D, Taglialatela-Scafati O, Arese P, Skorokhod OA. Preferential binding of 4-hydroxynonenal to lysine residues in specific parasite proteins in plakortin-treated Plasmodium falciparum-parasitized red blood cells. Data Brief 2015; 5:893-9. [PMID: 26702418 PMCID: PMC4669491 DOI: 10.1016/j.dib.2015.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 11/30/2022] Open
Abstract
The data show the frequencies by which the amino acid residues lysine, histidine and cysteine of six proteins of the malaria parasite Plasmodium falciparum are post-translationally modified by the lipoperoxydation endproduct 4-hydroxynonenal after challenging the parasitized red blood cell with plakortin. Plakortin is an antimalarial endoperoxide whose molecular anti-parasitic effect is described in Skorokhod et al. (2015) [1]. Plakortin did not elicit hemoglobin leakage from host red blood cells and did not oxidize reduced glutathione.
Collapse
Affiliation(s)
- Evelin Schwarzer
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy
| | - Valentina Gallo
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy
| | - Elena Valente
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy
| | - Daniela Ulliers
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy
| | | | - Paolo Arese
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy
| | - Oleksii A. Skorokhod
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy
- Corresponding author.
| |
Collapse
|
32
|
Shearn CT, Orlicky DJ, Saba LM, Shearn AH, Petersen DR. Increased hepatocellular protein carbonylation in human end-stage alcoholic cirrhosis. Free Radic Biol Med 2015; 89:1144-53. [PMID: 26518673 PMCID: PMC4762037 DOI: 10.1016/j.freeradbiomed.2015.10.420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 10/20/2015] [Accepted: 10/25/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Oxidative stress is a significant contributing factor in the pathogenesis of alcoholic liver disease (ALD). In the murine models of chronic alcohol consumption, induction of oxidative stress results in increased peroxidation of polyunsaturated fatty acids to form highly reactive electrophilic α/β unsaturated aldehydes that post-translationally modify proteins altering activity. Data are presented here suggesting that oxidative stress and the resulting carbonylation of hepatic proteins is an ongoing process involved in alcohol-induced cirrhosis. METHODS Using age-matched pooled hepatic tissue obtained from healthy humans and patients with end stage cirrhotic ALD, overall carbonylation was assessed by immunohistochemistry and LC-MS/MS of streptavidin purified hepatic whole cell extracts treated with biotin hydrazide. Identified carbonylated proteins were further evaluated using bioinformatics analyses. RESULTS Using immunohistochemistry and Western blotting, protein carbonylation was increased in end stage ALD occurring primarily in hepatocytes. Mass spectrometric analysis revealed a total of 1224 carbonylated proteins in normal hepatic and end-stage alcoholic cirrhosis tissue. Of these, 411 were unique to cirrhotic ALD, 261 unique to normal hepatic tissue and 552 common to both groups. Bioinformatic pathway analysis of hepatic carbonylated proteins revealed a propensity of long term EtOH consumption to increase post-translational carbonylation of proteins involved in glutathione homeostatic, glycolytic and cytoskeletal pathways. Western analysis revealed increased expression of GSTA4 and GSTπ in human ALD. Using LC-MS/MS analysis, a nonenaldehyde post-translational modification was identified on Lysine 235 of the cytoskeletal protein vimentin in whole cell extracts prepared from human end stage ALD hepatic tissue. CONCLUSIONS These studies are the first to use LC-MS/MS analysis of carbonylated proteins in human ALD and begin exploring possible mechanistic links with end-stage alcoholic cirrhosis and oxidative stress.
Collapse
Affiliation(s)
- C T Shearn
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver Anschutz Medical Campus, 12850 East Montview Blvd Box C238, Building V20 Room 2131, Aurora, CO 80045, United States.
| | - D J Orlicky
- Department of Pathology, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, United States
| | - L M Saba
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver Anschutz Medical Campus, 12850 East Montview Blvd Box C238, Building V20 Room 2131, Aurora, CO 80045, United States
| | - A H Shearn
- Alpine Achievement Systems, Inc., 9635 Maroon Circle, Suite 120, Englewood, CO 80112, United States
| | - Dennis R Petersen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver Anschutz Medical Campus, 12850 East Montview Blvd Box C238, Building V20 Room 2131, Aurora, CO 80045, United States.
| |
Collapse
|
33
|
Schaur RJ, Siems W, Bresgen N, Eckl PM. 4-Hydroxy-nonenal-A Bioactive Lipid Peroxidation Product. Biomolecules 2015; 5:2247-337. [PMID: 26437435 PMCID: PMC4693237 DOI: 10.3390/biom5042247] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/24/2015] [Accepted: 07/29/2015] [Indexed: 12/23/2022] Open
Abstract
This review on recent research advances of the lipid peroxidation product 4-hydroxy-nonenal (HNE) has four major topics: I. the formation of HNE in various organs and tissues, II. the diverse biochemical reactions with Michael adduct formation as the most prominent one, III. the endogenous targets of HNE, primarily peptides and proteins (here the mechanisms of covalent adduct formation are described and the (patho-) physiological consequences discussed), and IV. the metabolism of HNE leading to a great number of degradation products, some of which are excreted in urine and may serve as non-invasive biomarkers of oxidative stress.
Collapse
Affiliation(s)
- Rudolf J Schaur
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 33a, 8010 Graz, Austria.
| | - Werner Siems
- Institute for Medical Education, KortexMed GmbH, Hindenburgring 12a, 38667 Bad Harzburg, Germany.
| | - Nikolaus Bresgen
- Division of Genetics, Department of Cell Biology, University of Salzburg, Hellbrunnerstasse 34, 5020 Salzburg, Austria.
| | - Peter M Eckl
- Division of Genetics, Department of Cell Biology, University of Salzburg, Hellbrunnerstasse 34, 5020 Salzburg, Austria.
| |
Collapse
|
34
|
Comparative Proteomic Analysis of Carbonylated Proteins from the Striatum and Cortex of Pesticide-Treated Mice. PARKINSONS DISEASE 2015; 2015:812532. [PMID: 26345149 PMCID: PMC4546751 DOI: 10.1155/2015/812532] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/27/2015] [Accepted: 05/30/2015] [Indexed: 01/28/2023]
Abstract
Epidemiological studies indicate exposures to the herbicide paraquat (PQ) and fungicide maneb (MB) are associated with increased risk of Parkinson's disease (PD). Oxidative stress appears to be a premier mechanism that underlies damage to the nigrostriatal dopamine system in PD and pesticide exposure. Enhanced oxidative stress leads to lipid peroxidation and production of reactive aldehydes; therefore, we conducted proteomic analyses to identify carbonylated proteins in the striatum and cortex of pesticide-treated mice in order to elucidate possible mechanisms of toxicity. Male C57BL/6J mice were treated biweekly for 6 weeks with saline, PQ (10 mg/kg), MB (30 mg/kg), or the combination of PQ and MB (PQMB). Treatments resulted in significant behavioral alterations in all treated mice and depleted striatal dopamine in PQMB mice. Distinct differences in 4-hydroxynonenal-modified proteins were observed in the striatum and cortex. Proteomic analyses identified carbonylated proteins and peptides from the cortex and striatum, and pathway analyses revealed significant enrichment in a variety of KEGG pathways. Further analysis showed enrichment in proteins of the actin cytoskeleton in treated samples, but not in saline controls. These data indicate that treatment-related effects on cytoskeletal proteins could alter proper synaptic function, thereby resulting in impaired neuronal function and even neurodegeneration.
Collapse
|
35
|
Martin GG, Atshaves BP, Landrock KK, Landrock D, Schroeder F, Kier AB. Loss of L-FABP, SCP-2/SCP-x, or both induces hepatic lipid accumulation in female mice. Arch Biochem Biophys 2015; 580:41-9. [PMID: 26116377 DOI: 10.1016/j.abb.2015.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 06/08/2015] [Accepted: 06/17/2015] [Indexed: 02/06/2023]
Abstract
Although roles for both sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) and liver fatty acid binding protein (L-FABP) have been proposed in hepatic lipid accumulation, individually ablating these genes has been complicated by concomitant alterations in the other gene product(s). For example, ablating SCP2/SCP-x induces upregulation of L-FABP in female mice. Therefore, the impact of ablating SCP-2/SCP-x (DKO) or L-FABP (LKO) individually or both together (TKO) was examined in female mice. Loss of SCP-2/SCP-x (DKO, TKO) more so than loss of L-FABP alone (LKO) increased hepatic total lipid and total cholesterol content, especially cholesteryl ester. Hepatic accumulation of nonesterified long chain fatty acids (LCFA) and phospholipids occurred only in DKO and TKO mice. Loss of SCP-2/SCP-x (DKO, TKO) increased serum total lipid primarily by increasing triglycerides. Altered hepatic level of proteins involved in cholesterol uptake, efflux, and/or secretion was observed, but did not compensate for the loss of L-FABP, SCP-2/SCP-x or both. However, synergistic responses were not seen with the combinatorial knock out animals-suggesting that inhibiting SCP-2/SCP-x is more correlative with hepatic dysfunction than L-FABP. The DKO- and TKO-induced hepatic accumulation of cholesterol and long chain fatty acids shared significant phenotypic similarities with non-alcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Gregory G Martin
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, United States
| | - Barbara P Atshaves
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States
| | - Kerstin K Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, United States
| | - Danilo Landrock
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, United States
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, United States
| | - Ann B Kier
- Department of Pathobiology, Texas A&M University, College Station, TX 77843-4467, United States.
| |
Collapse
|
36
|
Aldini G, Domingues MR, Spickett CM, Domingues P, Altomare A, Sánchez-Gómez FJ, Oeste CL, Pérez-Sala D. Protein lipoxidation: Detection strategies and challenges. Redox Biol 2015; 5:253-266. [PMID: 26072467 PMCID: PMC4477048 DOI: 10.1016/j.redox.2015.05.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 05/14/2015] [Accepted: 05/19/2015] [Indexed: 12/21/2022] Open
Abstract
Enzymatic and non-enzymatic lipid metabolism can give rise to reactive species that may covalently modify cellular or plasma proteins through a process known as lipoxidation. Under basal conditions, protein lipoxidation can contribute to normal cell homeostasis and participate in signaling or adaptive mechanisms, as exemplified by lipoxidation of Ras proteins or of the cytoskeletal protein vimentin, both of which behave as sensors of electrophilic species. Nevertheless, increased lipoxidation under pathological conditions may lead to deleterious effects on protein structure or aggregation. This can result in impaired degradation and accumulation of abnormally folded proteins contributing to pathophysiology, as may occur in neurodegenerative diseases. Identification of the protein targets of lipoxidation and its functional consequences under pathophysiological situations can unveil the modification patterns associated with the various outcomes, as well as preventive strategies or potential therapeutic targets. Given the wide structural variability of lipid moieties involved in lipoxidation, highly sensitive and specific methods for its detection are required. Derivatization of reactive carbonyl species is instrumental in the detection of adducts retaining carbonyl groups. In addition, use of tagged derivatives of electrophilic lipids enables enrichment of lipoxidized proteins or peptides. Ultimate confirmation of lipoxidation requires high resolution mass spectrometry approaches to unequivocally identify the adduct and the targeted residue. Moreover, rigorous validation of the targets identified and assessment of the functional consequences of these modifications are essential. Here we present an update on methods to approach the complex field of lipoxidation along with validation strategies and functional assays illustrated with well-studied lipoxidation targets.
Collapse
Affiliation(s)
- Giancarlo Aldini
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milan, Italy
| | - M Rosário Domingues
- Mass Spectrometry Centre, QOPNA, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - Corinne M Spickett
- School of Life and Health Sciences, Aston Triangle, Aston University, Birmingham B4 7ET, UK
| | - Pedro Domingues
- Mass Spectrometry Centre, QOPNA, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - Alessandra Altomare
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milan, Italy
| | - Francisco J Sánchez-Gómez
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu, 9, Madrid 28040, Spain
| | - Clara L Oeste
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu, 9, Madrid 28040, Spain
| | - Dolores Pérez-Sala
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu, 9, Madrid 28040, Spain.
| |
Collapse
|
37
|
Role of the lipoperoxidation product 4-hydroxynonenal in the pathogenesis of severe malaria anemia and malaria immunodepression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:638416. [PMID: 25969702 PMCID: PMC4417603 DOI: 10.1155/2015/638416] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 03/31/2015] [Indexed: 12/15/2022]
Abstract
Oxidative stress plays an important role in the pathogenesis of falciparum malaria, a disease still claiming close to 1 million deaths and 200 million new cases per year. Most frequent complications are severe anemia, cerebral malaria, and immunodepression, the latter being constantly present in all forms of malaria. Complications are associated with oxidative stress and lipoperoxidation. Its final product 4-hydroxynonenal (4-HNE), a stable yet very reactive and diffusible molecule, forms covalent conjugates with proteins, DNA, and phospholipids and modulates important cell functions at very low concentrations. Since oxidative stress plays important roles in the pathogenesis of severe malaria, it appears important to explore the role of 4-HNE in two important malaria complications such as malaria anemia and malaria immunodepression where oxidative stress is considered to be involved. In this review we will summarize data about 4-HNE chemistry, its biologically relevant chemical properties, and its role as regulator of physiologic processes and as pathogenic factor. We will review studies documenting the role of 4-HNE in severe malaria with emphasis on malaria anemia and immunodepression. Data from other diseases qualify 4-HNE both as oxidative stress marker and as pathomechanistically important molecule. Further studies are needed to establish 4-HNE as accepted pathogenic factor in severe malaria.
Collapse
|
38
|
Baek JH, Zhang X, Williams MC, Hicks W, Buehler PW, D'Agnillo F. Sodium nitrite potentiates renal oxidative stress and injury in hemoglobin exposed guinea pigs. Toxicology 2015; 333:89-99. [PMID: 25891524 DOI: 10.1016/j.tox.2015.04.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/17/2015] [Accepted: 04/14/2015] [Indexed: 12/18/2022]
Abstract
Methemoglobin-forming drugs, such as sodium nitrite (NaNO2), may exacerbate oxidative toxicity under certain chronic or acute hemolytic settings. In this study, we evaluated markers of renal oxidative stress and injury in guinea pigs exposed to extracellular hemoglobin (Hb) followed by NaNO2 at doses sufficient to simulate clinically relevant acute methemoglobinemia. NaNO2 induced rapid and extensive oxidation of plasma Hb in this model. This was accompanied by increased renal expression of the oxidative response effectors nuclear factor erythroid 2-derived-factor 2 (Nrf-2) and heme oxygenase-1 (HO-1), elevated non-heme iron deposition, lipid peroxidation, interstitial inflammatory cell activation, increased expression of tubular injury markers kidney injury-1 marker (KIM-1) and liver-fatty acid binding protein (L-FABP), podocyte injury, and cell death. Importantly, these indicators of renal oxidative stress and injury were minimal or absent following infusion of Hb or NaNO2 alone. Together, these results suggest that the exposure to NaNO2 in settings associated with increased extracellular Hb may potentiate acute renal toxicity via processes that are independent of NaNO2 induced erythrocyte methemoglobinemia.
Collapse
Affiliation(s)
- Jin Hyen Baek
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Xiaoyuan Zhang
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Matthew C Williams
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Wayne Hicks
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Paul W Buehler
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Felice D'Agnillo
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA.
| |
Collapse
|
39
|
Ronis MJJ, Mercer KE, Gannon B, Engi B, Zimniak P, Shearn CT, Orlicky DJ, Albano E, Badger TM, Petersen DR. Increased 4-hydroxynonenal protein adducts in male GSTA4-4/PPAR-α double knockout mice enhance injury during early stages of alcoholic liver disease. Am J Physiol Gastrointest Liver Physiol 2015; 308:G403-15. [PMID: 25501545 PMCID: PMC4346750 DOI: 10.1152/ajpgi.00154.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
To test the significance of lipid peroxidation in the development of alcoholic liver injury, an ethanol (EtOH) liquid diet was fed to male 129/SvJ mice (wild-type, WT) and glutathione S-transferase A4-4-null (GSTA4-/-) mice for 40 days. GSTA4-/- mice were crossed with peroxisome proliferator-activated receptor-α-null mice (PPAR-α-/-), and the effects of EtOH in the resulting double knockout (dKO) mice were compared with the other strains. EtOH increased lipid peroxidation in all except WT mice (P < 0.05). Increased steatosis and mRNA expression of the inflammatory markers CXCL2, tumor necrosis factor-α (TNF-α), and α-smooth muscle actin (α-SMA) were observed in EtOH GSTA4-/- compared with EtOH WT mice (P < 0.05). EtOH PPAR-α-/- mice had increased steatosis, serum alanine aminotransferase (ALT), and hepatic CD3+ T cell populations and elevated mRNA encoding CD14, CXCL2, TNF-α, IL-6, CD138, transforming growth factor-β, platelet-derived growth factor receptor-β (PDGFR-β), matrix metalloproteinase (MMP)-9, MMP-13, α-SMA, and collagen type 1 compared with EtOH WT mice. EtOH-fed dKO mice displayed elevation of periportal hepatic 4-hydroxynonenal adducts and serum antibodies against malondialdehyde adducts compared with EtOH feeding of GSTA4-/-, PPAR-α-/-, and WT mice (P < 0.05). ALT was higher in EtOH dKO mice compared with all other groups (P < 0.001). EtOH-fed dKO mice displayed elevated mRNAs for TNF-α and CD14, histological evidence of fibrosis, and increased PDGFR, MMP-9, and MMP-13 mRNAs compared with the EtOH GSTA4-/- or EtOH PPAR-α-/- genotype (P < 0.05). These findings demonstrate the central role lipid peroxidation plays in mediating progression of alcohol-induced necroinflammatory liver injury, stellate cell activation, matrix remodeling, and fibrosis.
Collapse
Affiliation(s)
- Martin J. J. Ronis
- 1Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas; ,2Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas; ,4Arkansas Children's Nutrition Center, Little Rock, Arkansas;
| | - Kelly E. Mercer
- 1Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas; ,4Arkansas Children's Nutrition Center, Little Rock, Arkansas;
| | - Brenda Gannon
- 2Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas;
| | - Bridgette Engi
- 3Department of Laboratory Animal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas;
| | - Piotr Zimniak
- 4Arkansas Children's Nutrition Center, Little Rock, Arkansas;
| | - Colin T. Shearn
- 5University of Colorado Anschutz Medical Campus, Aurora, Colorado;
| | - David J. Orlicky
- 5University of Colorado Anschutz Medical Campus, Aurora, Colorado;
| | - Emanuele Albano
- 6Department of Medical Sciences, University A Avogadro of East Piedmonte, Novara, Italy
| | - Thomas M. Badger
- 1Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas; ,4Arkansas Children's Nutrition Center, Little Rock, Arkansas;
| | | |
Collapse
|
40
|
Milkovic L, Cipak Gasparovic A, Zarkovic N. Overview on major lipid peroxidation bioactive factor 4-hydroxynonenal as pluripotent growth-regulating factor. Free Radic Res 2015; 49:850-60. [PMID: 25532703 DOI: 10.3109/10715762.2014.999056] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The reactive aldehyde 4-hydroxynonenal (HNE) is major bioactive marker of lipid peroxidation generated under oxidative stress from polyunsaturated fatty acids. Biomedical significance of HNE was first revealed in pathogenesis of various degenerative and malignant diseases. Thus, HNE was considered for decades only as cytotoxic molecule, "second toxic messenger of free radicals" responsible for numerous undesirable consequences of oxidative stress. However, the increase of knowledge on physiology of redox signaling revealed also desirable, physiological roles of HNE, especially in the field of cellular signaling pathways regulating proliferation, differentiation, and apoptosis. These pluripotent effects of HNE can be explained by its concentration-dependent interactions with the cytokine networks and complex cellular antioxidant systems also showing cell and tissue specificities. Therefore, this paper gives a comprehensive, yet short overview on HNE as pluripotent growth-regulating factor.
Collapse
Affiliation(s)
- L Milkovic
- Laboratory for Oxidative Stress, Rudjer Boskovic Institute , Zagreb , Croatia
| | | | | |
Collapse
|
41
|
Abstract
The interaction between antioxidant glutathione and the free thiol in susceptible cysteine residues of proteins leads to reversible protein S-glutathionylation. This reaction ensures cellular homeostasis control (as a common redox-dependent post-translational modification associated with signal transduction) and intervenes in oxidative stress-related cardiovascular pathology (as initiated by redox imbalance). The purpose of this review is to evaluate the recent knowledge on protein S-glutathionylation in terms of chemistry, broad cellular intervention, specific quantification, and potential for therapeutic exploitation. The data bases searched were Medline and PubMed, from 2009 to 2014 (term: glutathionylation). Protein S-glutathionylation ensures protection of protein thiols against irreversible over-oxidation, operates as a biological redox switch in both cell survival (influencing kinases and protein phosphatases pathways) and cell death (by potentiation of apoptosis), and cross-talks with phosphorylation and with S-nitrosylation. Collectively, protein S-glutathionylation appears as a valuable biomarker for oxidative stress, with potential for translation into novel therapeutic strategies.
Collapse
Affiliation(s)
- Doina Popov
- Institute of Cellular Biology and Pathology "N. Simionescu" of the Romanian Academy , 8, B.P. Hasdeu Street, Bucharest 050568 , Romania
| |
Collapse
|
42
|
Liu JW, Montero M, Bu L, De Leon M. Epidermal fatty acid-binding protein protects nerve growth factor-differentiated PC12 cells from lipotoxic injury. J Neurochem 2014; 132:85-98. [PMID: 25147052 PMCID: PMC4270845 DOI: 10.1111/jnc.12934] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 08/01/2014] [Accepted: 08/13/2014] [Indexed: 12/11/2022]
Abstract
Epidermal fatty acid-binding protein (E-FABP/FABP5/DA11) binds and transport long-chain fatty acids in the cytoplasm and may play a protecting role during neuronal injury. We examined whether E-FABP protects nerve growth factor-differentiated PC12 cells (NGFDPC12 cells) from lipotoxic injury observed after palmitic acid (C16:0; PAM) overload. NGFDPC12 cells cultures treated with PAM/bovine serum albumin at 0.3 mM/0.15 mM show PAM-induced lipotoxicity (PAM-LTx) and apoptosis. The apoptosis was preceded by a cellular accumulation of reactive oxygen species (ROS) and higher levels of E-FABP. Antioxidants MCI-186 and N-acetyl cysteine prevented E-FABP's induction in expression by PAM-LTx, while tert-butyl hydroperoxide increased ROS and E-FABP expression. Non-metabolized methyl ester of PAM, methyl palmitic acid (mPAM), failed to increase cellular ROS, E-FABP gene expression, or trigger apoptosis. Treatment of NGFDPC12 cultures with siE-FABP showed reduced E-FABP levels correlating with higher accumulation of ROS and cell death after exposure to PAM. In contrast, increasing E-FABP cellular levels by pre-loading the cells with recombinant E-FABP diminished the PAM-induced ROS and cell death. Finally, agonists for PPARβ (GW0742) or PPARγ (GW1929) increased E-FABP expression and enhanced the resistance of NGFDPC12 cells to PAM-LTx. We conclude that E-FABP protects NGFDPC12 cells from lipotoxic injury through mechanisms that involve reduction of ROS. Epidermal fatty acid-binding protein (E-FABP) may protect nerve cells from the damaging exposure to high levels of free fatty acids (FA). We show that E-FABP can neutralize the effects of reactive oxygen species (ROS) generated by the high levels of FA in the cell and protect PC12 cells from lipotoxic injuries common in Type 2 diabetes neuropathy. Potentially, E-FABP gene up-regulation may be mediated through the NFkB pathway and future studies are needed to further evaluate this proposition.
Collapse
Affiliation(s)
- Jo-Wen Liu
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California, USA
| | | | | | | |
Collapse
|
43
|
Galligan JJ, Fritz KS, Backos DS, Shearn CT, Smathers RL, Jiang H, MacLean KN, Reigan PR, Petersen DR. Oxidative stress-mediated aldehyde adduction of GRP78 in a mouse model of alcoholic liver disease: functional independence of ATPase activity and chaperone function. Free Radic Biol Med 2014; 73:411-20. [PMID: 24924946 PMCID: PMC4395467 DOI: 10.1016/j.freeradbiomed.2014.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 05/05/2014] [Accepted: 06/02/2014] [Indexed: 01/23/2023]
Abstract
Pathogenesis in alcoholic liver disease (ALD) is complicated and multifactorial but clearly involves oxidative stress and inflammation. Currently, conflicting reports exist regarding the role of endoplasmic reticulum (ER) stress in the etiology of ALD. The glucose-regulated protein 78 (GRP78) is the ER homolog of HSP70 and plays a critical role in the cellular response to ER stress by serving as a chaperone assisting protein folding and by regulating the signaling of the unfolded protein response (UPR). Comprising three functional domains, an ATPase, a peptide-binding, and a lid domain, GRP78 folds nascent polypeptides via the substrate-binding domain. Earlier work has indicated that the ATPase function of GRP78 is intrinsically linked and essential to its chaperone activity. Previous work in our laboratory has indicated that GRP78 and the UPR are not induced in a mouse model of ALD but that GRP78 is adducted by the lipid electrophiles 4-hydroxynonenal (4-HNE) and 4-oxononenal (4-ONE) in vivo. As impairment of GRP78 has the potential to contribute to pathogenesis in ALD, we investigated the functional consequences of aldehyde adduction on GRP78 function. Identification of 4-HNE and 4-ONE target residues in purified human GRP78 revealed a marked propensity for Lys and His adduction within the ATPase domain and a relative paucity of adduct formation within the peptide-binding domain. Consistent with these findings, we observed a concomitant dose-dependent decrease in ATP-binding and ATPase activity without any discernible impairment of chaperone function. Collectively, our data indicate that ATPase activity is not essential for GRP78-mediated chaperone activity and is consistent with the hypothesis that ER stress does not play a primary initiating role in the early stages of ALD.
Collapse
Affiliation(s)
- James J Galligan
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kristofer S Fritz
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Donald S Backos
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Colin T Shearn
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rebecca L Smathers
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hua Jiang
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth N MacLean
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Philip R Reigan
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dennis R Petersen
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
44
|
Shearn CT, Backos DS, Orlicky DJ, Smathers-McCullough RL, Petersen DR. Identification of 5' AMP-activated kinase as a target of reactive aldehydes during chronic ingestion of high concentrations of ethanol. J Biol Chem 2014; 289:15449-62. [PMID: 24722988 DOI: 10.1074/jbc.m113.543942] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The production of reactive aldehydes including 4-hydroxy-2-nonenal (4-HNE) is a key component of the pathogenesis in a spectrum of chronic inflammatory hepatic diseases including alcoholic liver disease (ALD). One consequence of ALD is increased oxidative stress and altered β-oxidation in hepatocytes. A major regulator of β-oxidation is 5' AMP protein kinase (AMPK). In an in vitro cellular model, we identified AMPK as a direct target of 4-HNE adduction resulting in inhibition of both H2O2 and 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR)-induced downstream signaling. By employing biotin hydrazide capture, it was confirmed that 4-HNE treatment of cells resulted in carbonylation of AMPKα/β, which was not observed in untreated cells. Using a murine model of alcoholic liver disease, treatment with high concentrations of ethanol resulted in an increase in phosphorylated as well as carbonylated AMPKα. Despite increased AMPK phosphorylation, there was no significant change in phosphorylation of acetyl CoA carboxylase. Mass spectrometry identified Michael addition adducts of 4-HNE on Cys(130), Cys(174), Cys(227), and Cys(304) on recombinant AMPKα and Cys(225) on recombinant AMPKβ. Molecular modeling analysis of identified 4-HNE adducts on AMPKα suggest that inhibition of AMPK occurs by steric hindrance of the active site pocket and by inhibition of hydrogen peroxide induced oxidation. The observed inhibition of AMPK by 4-HNE provides a novel mechanism for altered β-oxidation in ALD, and these data demonstrate for the first time that AMPK is subject to regulation by reactive aldehydes in vivo.
Collapse
Affiliation(s)
| | | | - David J Orlicky
- Department of Pathology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado 80045 and
| | | | | |
Collapse
|
45
|
Huang H, McIntosh AL, Martin GG, Landrock KK, Landrock D, Gupta S, Atshaves BP, Kier AB, Schroeder F. Structural and functional interaction of fatty acids with human liver fatty acid-binding protein (L-FABP) T94A variant. FEBS J 2014; 281:2266-83. [PMID: 24628888 DOI: 10.1111/febs.12780] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/17/2014] [Accepted: 03/11/2014] [Indexed: 12/13/2022]
Abstract
The human liver fatty acid-binding protein (L-FABP) T94A variant, the most common in the FABP family, has been associated with elevated liver triglyceride levels. How this amino acid substitution elicits these effects is not known. This issue was addressed using human recombinant wild-type (WT) and T94A variant L-FABP proteins as well as cultured primary human hepatocytes expressing the respective proteins (genotyped as TT, TC and CC). The T94A substitution did not alter or only slightly altered L-FABP binding affinities for saturated, monounsaturated or polyunsaturated long chain fatty acids, nor did it change the affinity for intermediates of triglyceride synthesis. Nevertheless, the T94A substitution markedly altered the secondary structural response of L-FABP induced by binding long chain fatty acids or intermediates of triglyceride synthesis. Finally, the T94A substitution markedly decreased the levels of induction of peroxisome proliferator-activated receptor α-regulated proteins such as L-FABP, fatty acid transport protein 5 and peroxisome proliferator-activated receptor α itself meditated by the polyunsaturated fatty acids eicosapentaenoic acid and docosahexaenoic acid in cultured primary human hepatocytes. Thus, although the T94A substitution did not alter the affinity of human L-FABP for long chain fatty acids, it significantly altered human L-FABP structure and stability, as well as the conformational and functional response to these ligands.
Collapse
Affiliation(s)
- Huan Huang
- Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cell death and diseases related to oxidative stress: 4-hydroxynonenal (HNE) in the balance. Cell Death Differ 2013; 20:1615-30. [PMID: 24096871 DOI: 10.1038/cdd.2013.138] [Citation(s) in RCA: 605] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 07/22/2013] [Accepted: 07/29/2013] [Indexed: 11/08/2022] Open
Abstract
During the last three decades, 4-hydroxy-2-nonenal (HNE), a major α,β-unsaturated aldehyde product of n-6 fatty acid oxidation, has been shown to be involved in a great number of pathologies such as metabolic diseases, neurodegenerative diseases and cancers. These multiple pathologies can be explained by the fact that HNE is a potent modulator of numerous cell processes such as oxidative stress signaling, cell proliferation, transformation or cell death. The main objective of this review is to focus on the different aspects of HNE-induced cell death, with a particular emphasis on apoptosis. HNE is a special apoptotic inducer because of its abilities to form protein adducts and to propagate oxidative stress. It can stimulate intrinsic and extrinsic apoptotic pathways and interact with typical actors such as tumor protein 53, JNK, Fas or mitochondrial regulators. At the same time, due to its oxidant status, it can also induce some cellular defense mechanisms against oxidative stress, thus being involved in its own detoxification. These processes in turn limit the apoptotic potential of HNE. These dualities can imbalance cell fate, either toward cell death or toward survival, depending on the cell type, the metabolic state and the ability to detoxify.
Collapse
|
47
|
Colzani M, Aldini G, Carini M. Mass spectrometric approaches for the identification and quantification of reactive carbonyl species protein adducts. J Proteomics 2013; 92:28-50. [DOI: 10.1016/j.jprot.2013.03.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 03/26/2013] [Accepted: 03/27/2013] [Indexed: 01/28/2023]
|
48
|
Increased dietary fat contributes to dysregulation of the LKB1/AMPK pathway and increased damage in a mouse model of early-stage ethanol-mediated steatosis. J Nutr Biochem 2013; 24:1436-45. [PMID: 23465594 DOI: 10.1016/j.jnutbio.2012.12.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 11/30/2012] [Accepted: 12/06/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The objective of the study was to examine the interaction of moderate and high dietary fat and ethanol with respect to formation of steatosis and regulation of the AMP-activated protein kinase (AMPK) pathway in a mouse model of chronic ethanol consumption. METHODS Male C57BL/6J mice were pair-fed a modified Lieber-DeCarli diet composed of either moderate fat [30% fat-derived calories (MF)] or high fat [45% fat-derived calories (HF)] combined with increasing concentrations of ethanol (2%-6%) for 6 weeks. RESULTS Chronic ethanol consumption resulted in significant increases in plasma alanine aminotransferase in MF (1.84-fold) and HF mice (2.33-fold), yet liver triglycerides only increased significantly in the HF model (1.62-fold). Ethanol addition significantly increased plasma adiponectin under conditions of MF but not HF. In combination with MF, the addition of ethanol significantly decreased total and hepatic pThr(172)AMPKα and acetyl CoA Carboxylase (ACC). HF plus ethanol decreased pSer(108)AMPKβ, yet a marked 1.5-fold increase in pThr(172)AMPKα occurred. No change was evident in pSer(79)ACC under conditions of ethanol and HF ingestion. In both models, nuclear levels of sterol response element binding protein 1c and carbohydrate response element binding protein were decreased. Surprisingly, MF plus ethanol significantly elevated protein expression of medium-chain acyl-CoA dehydrogenase (MCAD), long-chain acyl-CoA dehydrogenase (LCAD) and very long chain acyl-CoA dehydrogenase but did not significantly affect mRNA expression of other proteins involved in β-oxidation and fatty acid synthesis. HF plus ethanol significantly reduced mRNA expression of both stearoyl CoA desaturase 1 and fatty acid elongase 5, but did not have an effect on MCAD or LCAD. CONCLUSION These data suggest that, when co-ingested with ethanol, dietary fat differentially contributes to dysregulation of adiponectin-dependent activation of the AMPK pathway in the liver of mice.
Collapse
|
49
|
Smathers RL, Galligan JJ, Shearn CT, Fritz KS, Mercer K, Ronis M, Orlicky DJ, Davidson NO, Petersen DR. Susceptibility of L-FABP-/- mice to oxidative stress in early-stage alcoholic liver. J Lipid Res 2013; 54:1335-45. [PMID: 23359610 DOI: 10.1194/jlr.m034892] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic ethanol consumption is a prominent cause of liver disease worldwide. Dysregulation of an important lipid uptake and trafficking gene, liver-fatty acid binding protein (L-FABP), may contribute to alterations in lipid homeostasis during early-stage alcoholic liver. We have reported the detrimental effects of ethanol on the expression of L-FABP and hypothesize this may deleteriously impact metabolic networks regulating fatty acids. Male wild-type (WT) and L-FABP(-/-) mice were fed a modified Lieber-DeCarli liquid diet for six weeks. To assess the response to chronic ethanol ingestion, standard biochemical indicators for alcoholic liver disease (ALD) and oxidative stress were measured. Ethanol ingestion resulted in attenuation of hepatic triglyceride accumulation and elevation of cholesterol in L-FABP(-/-) mice. Lipidomics analysis validated multiple alterations in hepatic lipids resulting from ethanol treatment. Increased immunohistochemical staining for the reactive aldehydes 4-hydroxynonenal and malondialdehyde were observed in WT mice ingesting ethanol; however, L-FABP(-/-) mice displayed prominent protein adducts in liver sections evaluated from pair-fed and ethanol-fed mice. Likewise, alterations in glutathione, thiobarbituric acid reactive substances (TBARS), 8-isoprostanes, and protein carbonyl content all indicated L-FABP(-/-) mice exhibit high sustained oxidative stress in the liver. These data establish that L-FABP is an indirect antioxidant protein essential for sequestering FFA and that its impairment could contribute to in the pathogenesis of ALD.
Collapse
Affiliation(s)
- Rebecca L Smathers
- Skaggs School of Pharmaceutical Sciences and Pharmacy and University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhao J, Chen J, Zhu H, Xiong YL. Mass spectrometric evidence of malonaldehyde and 4-hydroxynonenal adductions to radical-scavenging soy peptides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:9727-36. [PMID: 22946674 PMCID: PMC3580276 DOI: 10.1021/jf3026277] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Antioxidative peptides in food systems are potential targets of lipid oxidation-generated reactive aldehydes, such as malonaldehyde (MDA) and 4-hydroxynonenal (HNE). In this study, covalent modifications on radical-scavenging peptides prepared from soy protein hydrolysate by MDA and HNE were characterized by liquid chromatography-electrospray ionization-mass spectrometry (LC-ESI-MS/MS). MS/MS analyses detected the formation of Schiff base type adducts of MDA on the side-chain groups of lysine, histidine, arginine, glutamine, and asparagine residues as well as the N-termini of peptides. MDA also formed a fluorescent product with lysine residues. HNE adducted on lysine residues through Schiff base formation and on histidine, arginine, glutamine, and asparagine residues mainly through Michael addition. Despite the extensive MDA modification, peptide cross-linking by this potential mechanism was undetectable.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, USA
| | - Jing Chen
- Department of Biochemistry, University of Kentucky, Lexington, KY 40546, USA
| | - Haining Zhu
- Department of Biochemistry, University of Kentucky, Lexington, KY 40546, USA
| | - Youling L. Xiong
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, USA
| |
Collapse
|