1
|
Ma K, Sun L, Jia C, Kui H, Xie J, Zang S, Huang S, Que J, Liu C, Huang J. Potential mechanisms underlying podophyllotoxin-induced cardiotoxicity in male rats: toxicological evidence chain (TEC) concept. Front Pharmacol 2024; 15:1378758. [PMID: 39386032 PMCID: PMC11463157 DOI: 10.3389/fphar.2024.1378758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/31/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Podophyllotoxin (PPT) is a high-content and high-activity compound extracted from the traditional Chinese medicinal plant Dysosma versipellis (DV) which exhibits various biological activities. However, its severe toxicity limits its use. In clinical settings, patients with DV poisoning often experience adverse reactions when taking large doses in a short period. The heart is an important toxic target organ, so it is necessary to conduct 24-h acute cardiac toxicity studies on PPT to understand its underlying toxicity mechanism. Methods Based on the concept of the toxicological evidence chain (TEC), we utilized targeted metabolomic and transcriptomic analyses to reveal the mechanism of the acute cardiotoxicity of PPT. The manifestation of toxicity in Sprague-Dawley rats, including changes in weight and behavior, served as Injury Phenotype Evidence (IPE). To determine Adverse Outcomes Evidence (AOE), the hearts of the rats were evaluated through histopathological examination and by measuring myocardial enzyme and cardiac injury markers levels. Additionally, transcriptome analysis, metabolome analysis, myocardial enzymes, and cardiac injury markers were integrated to obtain Toxic Event Evidence (TEE) using correlation analysis. Results The experiment showed significant epistaxis, hypokinesia, and hunched posture in PPT group rats within 24 h after exposure to 120 mg/kg PPT. It is found that PPT induced cardiac injury in rats within 24 h, as evidenced by increased serum myocardial enzyme levels, elevated concentrations of cardiac injury biomarkers, and altered cardiac cell morphology, all indicating some degree of cardiac toxicity. Transcriptome analysis revealed that primary altered metabolic pathway was arachidonic acid metabolism after PPT exposure. Cyp2e1, Aldob were positively correlated with differential metabolites, while DHA showed positive correlation with differential genes Fmo2 and Timd2, as well as with heart injury markers BNP and Mb. Conclusion This study comprehensively evaluated cardiac toxicity of PPT and initially revealed the mechanism of PPT-induced acute cardiotoxicity, which involved oxidative stress, apoptosis, inflammatory response, and energy metabolism disorder.
Collapse
Affiliation(s)
- Kaiyue Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Sun
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Chunxue Jia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hongqian Kui
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaqi Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shidan Zang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shixin Huang
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jinfeng Que
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chuanxin Liu
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Helal SA, Gerges SH, El-Kadi AOS. Enantioselectivity in some physiological and pathophysiological roles of hydroxyeicosatetraenoic acids. Drug Metab Rev 2024; 56:31-45. [PMID: 38358327 DOI: 10.1080/03602532.2023.2284110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/12/2023] [Indexed: 02/16/2024]
Abstract
The phenomenon of chirality has been shown to greatly impact drug activities and effects. Different enantiomers may exhibit different effects in a certain biological condition or disease state. Cytochrome P450 (CYP) enzymes metabolize arachidonic acid (AA) into a large variety of metabolites with a wide range of activities. Hydroxylation of AA by CYP hydroxylases produces hydroxyeicosatetraenoic acids (HETEs), which are classified into mid-chain (5, 8, 9, 11, 12, and 15-HETE), subterminal (16-, 17-, 18- and 19-HETE) and terminal (20-HETE) HETEs. Except for 20-HETE, these metabolites exist as a racemic mixture of R and S enantiomers in the physiological system. The two enantiomers could have different degrees of activity or sometimes opposing effects. In this review article, we aimed to discuss the role of mid-chain and subterminal HETEs in different organs, importantly the heart and the kidneys. Moreover, we summarized their effects in some conditions such as neutrophil migration, inflammation, angiogenesis, and tumorigenesis, with a focus on the reported enantiospecific effects. We also reported some studies using genetically modified models to investigate the roles of HETEs in different conditions.
Collapse
Affiliation(s)
- Sara A Helal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
3
|
Apostolopoulou A, Tranidou A, Chroni V, Tsakiridis I, Magriplis E, Dagklis T, Chourdakis M. Association of Maternal Diet with Infant Birthweight in Women with Gestational Diabetes Mellitus. Nutrients 2023; 15:4545. [PMID: 37960200 PMCID: PMC10648020 DOI: 10.3390/nu15214545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
OBJECTIVE This study aimed to explore the potential impact of pre-pregnancy and early pregnancy maternal nutrition on the incidence of small-for-gestational-age neonates (SGA) in women with gestational diabetes mellitus (GDM). METHODS A prospective cohort study was conducted between 2020 and 2022 at the 3rd Department of Obstetrics and Gynaecology (School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Greece). Pregnant women from routine care were surveyed about their dietary habits during two distinct periods: six months prior to pregnancy (period A) and from the onset of pregnancy until the oral glucose tolerance test at 24-28 gestational weeks (period B). The intake of various micronutrients and macronutrients was quantified from the questionnaire responses. Logistic regression models, adjusted for potential confounders including age, pre-pregnancy body mass index (BMI), smoking status, physical activity and parity, were used to evaluate the association between nutrient intake and small-for-gestational-age neonate incidence. RESULTS In total, 850 women were screened and of these, 90 (11%) were diagnosed with gestational diabetes mellitus and were included in the study. There were significant associations between the intake of specific nutrients and the occurrence of small-for-gestational-age neonates; higher fat intake compared to non-small for gestationa age during period B (aOR: 1.1, p = 0.005) was associated with an increased risk for small-for-gestational-age neonates, while lower intake of carbohydrates (g) (aOR: 0.95, p = 0.005), fiber intake (aOR: 0.79, p = 0.045), magnesium (aOR: 0.96, p = 0.019), and copper (aOR:0.01, p = 0.018) intake during period B were significantly associated with a decreased risk for small-for-gestational-age neonates. CONCLUSIONS The findings of this study highlight the potential role of maternal nutrition in modulating the risk of small for gestational age neonatesamong women with gestational diabetes mellitus. The results advocate for further research on the assessment and modification of both pre-pregnancy and early pregnancy nutrition for women, especially those at higher risk of gestational diabetes mellitus, to reduce the risk of gestational diabetes mellitus.
Collapse
Affiliation(s)
- Aikaterini Apostolopoulou
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.A.); (A.T.); (V.C.); (M.C.)
| | - Antigoni Tranidou
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.A.); (A.T.); (V.C.); (M.C.)
| | - Violeta Chroni
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.A.); (A.T.); (V.C.); (M.C.)
| | - Ioannis Tsakiridis
- 3rd Department of Obstetrics and Gynecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Emmanuella Magriplis
- Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera odos 75, 118 55 Athens, Greece;
| | - Themistoklis Dagklis
- 3rd Department of Obstetrics and Gynecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Michail Chourdakis
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.A.); (A.T.); (V.C.); (M.C.)
| |
Collapse
|
4
|
He K, Zhou X, Du H, Zhao J, Deng R, Wang J. A review on the relationship between Arachidonic acid 15-Lipoxygenase (ALOX15) and diabetes mellitus. PeerJ 2023; 11:e16239. [PMID: 37849828 PMCID: PMC10578307 DOI: 10.7717/peerj.16239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
Arachidonic acid 15-lipoxygenase (ALOX15), as one of the lipoxygenase family, is mainly responsible for catalyzing the oxidation of various fatty acids to produce a variety of lipid components, contributing to the pathophysiological processes of various immune and inflammatory diseases. Studies have shown that ALOX15 and its related products are widely distributed in human tissues and related to multiple diseases such as liver, cardiovascular, cerebrovascular diseases, diabetes mellitus and other diseases. Diabetes mellitus (DM), the disease studied in this article, is a metabolic disease characterized by a chronic increase in blood glucose levels, which is significantly related to inflammation, oxidative stress, ferroptosis and other mechanisms, and it has a high incidence in the population, accompanied by a variety of complications. Figuring out how ALOX15 is involved in DM is critical to understanding its role in diseases. Therefore, ALOX15 inhibitors or combination therapy containing inhibitors may deliver a novel research direction for the treatment of DM and its complications. This article aims to review the biological effect and the possible function of ALOX15 in the pathogenesis of DM.
Collapse
Affiliation(s)
- Kaiying He
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Xiaochun Zhou
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Hongxuan Du
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Jing Zhao
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Rongrong Deng
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Jianqin Wang
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| |
Collapse
|
5
|
Dong L, Wang H, Chen K, Li Y. Roles of hydroxyeicosatetraenoic acids in diabetes (HETEs and diabetes). Biomed Pharmacother 2022; 156:113981. [DOI: 10.1016/j.biopha.2022.113981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
|
6
|
Gonzalez-Riano C, Santos M, Díaz M, García-Beltran C, Lerin C, Barbas C, Ibáñez L, Sánchez-Infantes D. Birth Weight and Early Postnatal Outcomes: Association with the Cord Blood Lipidome. Nutrients 2022; 14:3760. [PMID: 36145136 PMCID: PMC9505183 DOI: 10.3390/nu14183760] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Being born small or large for gestational age (SGA and LGA, respectively), combined with suboptimal early postnatal outcomes, can entail future metabolic alterations. The exact mechanisms underlying such risks are not fully understood. Lipids are a highly diverse class of molecules that perform multiple structural and metabolic functions. Dysregulation of lipid metabolism underlies the onset and progression of many disorders leading to pathological states. The aim of this pilot study was to investigate the relationships between birth weight, early postnatal outcomes, and cord blood serum lipidomes. We performed a non-targeted lipidomics-based approach to ascertain differences in cord blood lipid species among SGA, LGA, and appropriate-for-GA (AGA) newborns. Moreover, we longitudinally assessed (at birth and at ages of 4 and 12 months) weight and length, body composition (DXA), and clinical parameters. We disclosed distinct cord blood lipidome patterns in SGA, LGA, and AGA newborns; target lipid species distinctly modulated in each SGA, AGA, and LGA individual were associated with parameters related to growth and glucose homeostasis. The distinct lipidome patterns observed in SGA, AGA, and LGA newborns may play a role in adipose tissue remodeling and future metabolic risks. Maternal dietary interventions may potentially provide long-term benefits for the metabolic health of the offspring.
Collapse
Affiliation(s)
- Carolina Gonzalez-Riano
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Marcelo Santos
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Marta Díaz
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina García-Beltran
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carles Lerin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Lourdes Ibáñez
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - David Sánchez-Infantes
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain
| |
Collapse
|
7
|
Xu J, Jin L, Sun Y, Zhang R, Chen X, Zhou R, Gu Y, Hu C. 12(S)-hydroxyeicosatetraenoic acid is significantly increased in diabetic kidney disease and associated with renal function decline. Diabetes Metab Res Rev 2022; 38:e3554. [PMID: 35667014 DOI: 10.1002/dmrr.3554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/09/2022]
Abstract
AIMS 12(S)-hydroxyeicosatetraenoic (12(S)-HETE), an alternate arachidonic acid metabolite, has been recently examined in metabolic disease. However, the role of 12(S)-HETE in diabetic kidney disease (DKD) remains unclear. We studied for the first time the relationship of serum 12(S)-HETE and DKD and renal function parameters in a Chinese population. MATERIALS AND METHODS We recruited 275 subjects who were diagnosed with type 2 diabetes (T2DM) for more than 10 years, including 149 DKD patients and 126 T2DM patients without DKD. Serum 12(S)-HETE was measured using the enzyme-linked immunosorbent assay. RESULTS Serum 12(S)-HETE was significantly higher in DKD patients than controls [384.69 (77.54, 1003.05) pg/ml and 17.77 (8.11, 75.13) pg/ml, respectively, p < 0.0001]. Compared to controls, 12(S)-HETE was significantly increased in both macroalbuminuria and microalbuminuria groups (p < 0.0001). Further, the macroalbuminuria group also had a higher serum 12(S)-HETE level compared to the microalbuminuria group (p = 0.0063). Moreover, serum 12(S)-HETE was positively correlated with the albuminuria level (r = 0.5833, p < 0.0001), serum creatinine (r = 0.2725, p < 0.0001), and was negatively associated with the estimated glomerular filtration rate (r = -0.2085, p = 0.0005). Further, receiver operating characteristic analysis (ROC) revealed that 12(S)-HETE had a good performance of distinguishing DKD from controls (AUC 0.828) with a sensitivity of 0.913 and a specificity of 0.711. CONCLUSION Our findings revealed that serum 12(S)-HETE significantly associated with DKD and disease severity, suggesting that serum 12(S)-HETE may be used as a potential biomarker for the early diagnosis of DKD.
Collapse
Affiliation(s)
- Jie Xu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Li Jin
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yi Sun
- Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xianghui Chen
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ranran Zhou
- Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yunjuan Gu
- Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, China
| |
Collapse
|
8
|
Kotlyarov S. Genetic and Epigenetic Regulation of Lipoxygenase Pathways and Reverse Cholesterol Transport in Atherogenesis. Genes (Basel) 2022; 13:1474. [PMID: 36011386 PMCID: PMC9408222 DOI: 10.3390/genes13081474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is one of the most important medical and social problems of modern society. Atherosclerosis causes a large number of hospitalizations, disability, and mortality. A considerable amount of evidence suggests that inflammation is one of the key links in the pathogenesis of atherosclerosis. Inflammation in the vascular wall has extensive cross-linkages with lipid metabolism, and lipid mediators act as a central link in the regulation of inflammation in the vascular wall. Data on the role of genetics and epigenetic factors in the development of atherosclerosis are of great interest. A growing body of evidence is strengthening the understanding of the significance of gene polymorphism, as well as gene expression dysregulation involved in cross-links between lipid metabolism and the innate immune system. A better understanding of the genetic basis and molecular mechanisms of disease pathogenesis is an important step towards solving the problems of its early diagnosis and treatment.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
9
|
Isse FA, El-Sherbeni AA, El-Kadi AOS. The multifaceted role of cytochrome P450-Derived arachidonic acid metabolites in diabetes and diabetic cardiomyopathy. Drug Metab Rev 2022; 54:141-160. [PMID: 35306928 DOI: 10.1080/03602532.2022.2051045] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding lipid metabolism is a critical key to understanding the pathogenesis of Diabetes Mellitus (DM). It is known that 60-90% of DM patients are obese or used to be obese. The incidence of obesity is rising owing to the modern sedentary lifestyle that leads to insulin resistance and increased levels of free fatty acids, predisposing tissues to utilize more lipids with less glucose uptake. However, the exact mechanism is not yet fully elucidated. Diabetic cardiomyopathy seems to be associated with these alterations in lipid metabolism. Arachidonic acid (AA) is an important fatty acid that is metabolized to several bioactive compounds by cyclooxygenases, lipoxygenases, and the more recently discovered, cytochrome P450 (P450) enzymes. P450 metabolizes AA to either epoxy-AA (EETs) or hydroxy-AA (HETEs). Studies showed that EETs could have cardioprotective effects and beneficial effects in reversing abnormalities in glucose and insulin homeostasis. Conversely, HETEs, most importantly 12-HETE and 20-HETE, were found to interfere with normal glucose and insulin homeostasis and thus, might be involved in diabetic cardiomyopathy. In this review, we highlight the role of P450-derived AA metabolites in the context of DM and diabetic cardiomyopathy and their potential use as a target for developing new treatments for DM and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Fadumo Ahmed Isse
- Departmet of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ahmed A El-Sherbeni
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ayman O S El-Kadi
- Departmet of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
10
|
Qin H, Yu Z, Zhu Z, Lin Y, Xia J, Jia Y. The integrated analyses of metabolomics and transcriptomics in gill of GIFT tilapia in response to long term salinity challenge. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2021.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
11
|
Liang N, Hennebelle M, Gaul S, Johnson CD, Zhang Z, Kirpich IA, McClain CJ, Feldstein AE, Ramsden CE, Taha AY. Feeding mice a diet high in oxidized linoleic acid metabolites does not alter liver oxylipin concentrations. Prostaglandins Leukot Essent Fatty Acids 2021; 172:102316. [PMID: 34403987 PMCID: PMC9157566 DOI: 10.1016/j.plefa.2021.102316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 11/19/2022]
Abstract
The oxidation of dietary linoleic acid (LA) produces oxidized LA metabolites (OXLAMs) known to regulate multiple signaling pathways in vivo. Recently, we reported that feeding OXLAMs to mice resulted in liver inflammation and apoptosis. However, it is not known whether this is due to a direct effect of OXLAMs accumulating in the liver, or to their degradation into bioactive shorter chain molecules (e.g. aldehydes) that can provoke inflammation and related cascades. To address this question, mice were fed a low or high LA diet low in OXLAMs, or a low LA diet supplemented with OXLAMs from heated corn oil (high OXLAM diet). Unesterified oxidized fatty acids (i.e. oxylipins), including OXLAMs, were measured in liver after 8 weeks of dietary intervention using ultra-high pressure liquid chromatography coupled to tandem mass-spectrometry. The high OXLAM diet did not alter liver oxylipin concentrations compared to the low LA diet low in OXLAMs. Significant increases in several omega-6 derived oxylipins and reductions in omega-3 derived oxylipins were observed in the high LA dietary group compared to the low LA group. Our findings suggest that dietary OXLAMs do not accumulate in liver, and likely exert pro-inflammatory and pro-apoptotic effects via downstream secondary metabolites.
Collapse
Affiliation(s)
- Nuanyi Liang
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California Davis, Davis, CA, Unites States
| | - Marie Hennebelle
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California Davis, Davis, CA, Unites States
| | - Susanne Gaul
- Department of Pediatrics, University of California San Diego, La Jolla, CA, Unites States; Klinik und Poliklinik für Kardiologie, University Hospital Leipzig, Leipzig University, Germany
| | - Casey D Johnson
- Department of Pediatrics, University of California San Diego, La Jolla, CA, Unites States
| | - Zhichao Zhang
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California Davis, Davis, CA, Unites States
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY; Hepatobiology and Toxicology Program, University of Louisville, Louisville, KY; Department of Pharmacology and Toxicology and University of Louisville Alcohol Center
| | - Craig J McClain
- Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY; Hepatobiology and Toxicology Program, University of Louisville, Louisville, KY; Department of Pharmacology and Toxicology and University of Louisville Alcohol Center; Veterans Affairs San Diego Healthcare System, San Diego, CA; and Robley Rex Veterans Medical Center, Louisville, KY
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego, La Jolla, CA, Unites States
| | - Christopher E Ramsden
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, Unites States; National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Ameer Y Taha
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California Davis, Davis, CA, Unites States.
| |
Collapse
|
12
|
Pascale JV, Lucchesi PA, Garcia V. Unraveling the Role of 12- and 20- HETE in Cardiac Pathophysiology: G-Protein-Coupled Receptors, Pharmacological Inhibitors, and Transgenic Approaches. J Cardiovasc Pharmacol 2021; 77:707-717. [PMID: 34016841 PMCID: PMC8523029 DOI: 10.1097/fjc.0000000000001013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/03/2021] [Indexed: 12/17/2022]
Abstract
ABSTRACT Arachidonic acid-derived lipid mediators play crucial roles in the development and progression of cardiovascular diseases. Eicosanoid metabolites generated by lipoxygenases and cytochrome P450 enzymes produce several classes of molecules, including the epoxyeicosatrienoic acid (EET) and hydroxyeicosatetraenoic acids (HETE) family of bioactive lipids. In general, the cardioprotective effects of EETs have been documented across a number of cardiac diseases. In contrast, members of the HETE family have been shown to contribute to the pathogenesis of ischemic cardiac disease, maladaptive cardiac hypertrophy, and heart failure. The net effect of 12(S)- and 20-HETE depends upon the relative amounts generated, ratio of HETEs:EETs produced, timing of synthesis, as well as cellular and subcellular mechanisms activated by each respective metabolite. HETEs are synthesized by and affect multiple cell types within the myocardium. Moreover, cytochrome P450-derived and lipoxygenase- derived metabolites have been shown to directly influence cardiac myocyte growth and the regulation of cardiac fibroblasts. The mechanistic data uncovered thus far have employed the use of enzyme inhibitors, HETE antagonists, and the genetic manipulation of lipid-producing enzymes and their respective receptors, all of which influence a complex network of outcomes that complicate data interpretation. This review will summarize and integrate recent findings on the role of 12(S)-/20-HETE in cardiac diseases.
Collapse
Affiliation(s)
| | | | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY
| |
Collapse
|
13
|
Szafran BN, Borazjani A, Seay CN, Carr RL, Lehner R, Kaplan BLF, Ross MK. Effects of Chlorpyrifos on Serine Hydrolase Activities, Lipid Mediators, and Immune Responses in Lungs of Neonatal and Adult Mice. Chem Res Toxicol 2021; 34:1556-1571. [PMID: 33900070 DOI: 10.1021/acs.chemrestox.0c00488] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chlorpyrifos (CPF) is an organophosphate (OP) pesticide that causes acute toxicity by inhibiting acetylcholinesterase (AChE) in the nervous system. However, endocannabinoid (eCB) metabolizing enzymes in brain of neonatal rats are more sensitive than AChE to inhibition by CPF, leading to increased levels of eCBs. Because eCBs are immunomodulatory molecules, we investigated the association between eCB metabolism, lipid mediators, and immune function in adult and neonatal mice exposed to CPF. We focused on lung effects because epidemiologic studies have linked pesticide exposures to respiratory diseases. CPF was hypothesized to disrupt lung eCB metabolism and alter lung immune responses to lipopolysaccharide (LPS), and these effects would be more pronounced in neonatal mice due to an immature immune system. We first assessed the biochemical effects of CPF in adult mice (≥8 weeks old) and neonatal mice after administering CPF (2.5 mg/kg, oral) or vehicle for 7 days. Tissues were harvested 4 h after the last CPF treatment and lung microsomes from both age groups demonstrated CPF-dependent inhibition of carboxylesterases (Ces), a family of xenobiotic and lipid metabolizing enzymes, whereas AChE activity was inhibited in adult lungs only. Activity-based protein profiling (ABPP)-mass spectrometry of lung microsomes identified 31 and 32 individual serine hydrolases in neonatal lung and adult lung, respectively. Of these, Ces1c/Ces1d/Ces1b isoforms were partially inactivated by CPF in neonatal lung, whereas Ces1c/Ces1b and Ces1c/BChE were partially inactivated in adult female and male lungs, respectively, suggesting age- and sex-related differences in their sensitivity to CPF. Monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH) activities in lung were unaffected by CPF. When LPS (1.25 mg/kg, i.p.) was administered following the 7-day CPF dosing period, little to no differences in lung immune responses (cytokines and immunophenotyping) were noted between the CPF and vehicle groups. However, a CPF-dependent increase in the amounts of dendritic cells and certain lipid mediators in female lung following LPS challenge was observed. Experiments in neonatal and adult Ces1d-/- mice yielded similar results as wild type mice (WT) following CPF treatment, except that CPF augmented LPS-induced Tnfa mRNA in adult Ces1d-/- mouse lungs. This effect was associated with decreased expression of Ces1c mRNA in Ces1d-/- mice versus WT mice in the setting of LPS exposure. We conclude that CPF exposure inactivates several Ces isoforms in mouse lung and, during an inflammatory response, increases certain lipid mediators in a female-dependent manner. However, it did not cause widespread altered lung immune effects in response to an LPS challenge.
Collapse
Affiliation(s)
- Brittany N Szafran
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Abdolsamad Borazjani
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Caitlin N Seay
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Russell L Carr
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Richard Lehner
- Departments of Cell Biology and Pediatrics, Group on Molecular & Cell Biology of Lipids, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Barbara L F Kaplan
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Matthew K Ross
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi 39762, United States
| |
Collapse
|
14
|
O'Connell TD, Mason RP, Budoff MJ, Navar AM, Shearer GC. Mechanistic insights into cardiovascular protection for omega-3 fatty acids and their bioactive lipid metabolites. Eur Heart J Suppl 2020; 22:J3-J20. [PMID: 33061864 PMCID: PMC7537803 DOI: 10.1093/eurheartj/suaa115] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Patients with well-controlled low-density lipoprotein cholesterol levels, but persistent high triglycerides, remain at increased risk for cardiovascular events as evidenced by multiple genetic and epidemiologic studies, as well as recent clinical outcome trials. While many trials of low-dose ω3-polyunsaturated fatty acids (ω3-PUFAs), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) have shown mixed results to reduce cardiovascular events, recent trials with high-dose ω3-PUFAs have reignited interest in ω3-PUFAs, particularly EPA, in cardiovascular disease (CVD). REDUCE-IT demonstrated that high-dose EPA (4 g/day icosapent-ethyl) reduced a composite of clinical events by 25% in statin-treated patients with established CVD or diabetes and other cardiovascular risk factors. Outcome trials in similar statin-treated patients using DHA-containing high-dose ω3 formulations have not yet shown the benefits of EPA alone. However, there are data to show that high-dose ω3-PUFAs in patients with acute myocardial infarction had reduced left ventricular remodelling, non-infarct myocardial fibrosis, and systemic inflammation. ω3-polyunsaturated fatty acids, along with their metabolites, such as oxylipins and other lipid mediators, have complex effects on the cardiovascular system. Together they target free fatty acid receptors and peroxisome proliferator-activated receptors in various tissues to modulate inflammation and lipid metabolism. Here, we review these multifactorial mechanisms of ω3-PUFAs in view of recent clinical findings. These findings indicate physico-chemical and biological diversity among ω3-PUFAs that influence tissue distributions as well as disparate effects on membrane organization, rates of lipid oxidation, as well as various receptor-mediated signal transduction pathways and effects on gene expression.
Collapse
Affiliation(s)
- Timothy D O'Connell
- Department of Integrative Biology and Physiology, University of Minnesota, 3-141 CCRB, 2231 6th Street SE, Minneapolis, MN 55414, USA
| | - Richard Preston Mason
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Matthew J Budoff
- Cardiovascular Division, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ann Marie Navar
- Cardiovascular Division, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Gregory C Shearer
- Department of Nutritional Sciences, The Pennsylvania State University, 110 Chandlee Laboratory, University Park, PA 16802, USA
| |
Collapse
|
15
|
Langbøl M, Saruhanian S, Baskaran T, Tiedemann D, Mouhammad ZA, Toft-Kehler AK, Jun B, Vohra R, Bazan NG, Kolko M. Increased Antioxidant Capacity and Pro-Homeostatic Lipid Mediators in Ocular Hypertension-A Human Experimental Model. J Clin Med 2020; 9:jcm9092979. [PMID: 32942740 PMCID: PMC7563216 DOI: 10.3390/jcm9092979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 11/16/2022] Open
Abstract
The main risk factor for primary open-angle glaucoma (POAG) is increased intraocular pressure (IOP). It is of interest that about half of the patients have an IOP within the normal range (normal-tension glaucoma, NTG). Additionally, there is a group of patients with a high IOP but no glaucomatous neurodegeneration (ocular hypertension, OHT). Therefore, risk factors other than IOP are involved in the pathogenesis of glaucoma. Since the retina has a very high oxygen-demand, decreased autoregulation and a fluctuating oxygen supply to the retina have been linked to glaucomatous neurodegeneration. To assess the significance of these mechanisms, we have utilized a human experimental model, in which we stress participants with a fluctuating oxygen supply. Levels of oxidative stress molecules, antioxidants, and lipid mediators were measured in the plasma. Patients with NTG, OHT, and control subjects were found to have similar levels of oxidative stress markers. In contrast, patients with OHT had a higher level of total antioxidant capacity (TAC) and pro-homeostatic lipid mediators. Thus, we suggest that OHT patients manage fluctuating oxygen levels more efficiently and, thus, are less susceptible to glaucomatous neurodegenerations, due to enhanced systemic antioxidant protection.
Collapse
Affiliation(s)
- Mia Langbøl
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (S.S.); (T.B.); (D.T.); (Z.A.M.); (A.K.T.-K.); (R.V.)
- Correspondence: (M.L.); (M.K.); Tel.: +45-30-50-26-62 (M.L.); +45-29-80-76-67 (M.K.)
| | - Sarkis Saruhanian
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (S.S.); (T.B.); (D.T.); (Z.A.M.); (A.K.T.-K.); (R.V.)
| | - Thisayini Baskaran
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (S.S.); (T.B.); (D.T.); (Z.A.M.); (A.K.T.-K.); (R.V.)
| | - Daniel Tiedemann
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (S.S.); (T.B.); (D.T.); (Z.A.M.); (A.K.T.-K.); (R.V.)
| | - Zaynab A. Mouhammad
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (S.S.); (T.B.); (D.T.); (Z.A.M.); (A.K.T.-K.); (R.V.)
| | - Anne Katrine Toft-Kehler
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (S.S.); (T.B.); (D.T.); (Z.A.M.); (A.K.T.-K.); (R.V.)
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA; (B.J.); (N.G.B.)
| | - Rupali Vohra
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (S.S.); (T.B.); (D.T.); (Z.A.M.); (A.K.T.-K.); (R.V.)
- Department of Veterinary and Animal Sciences, University of Copenhagen, 2000 Frederiksberg, Denmark
| | - Nicolas G. Bazan
- Neuroscience Center of Excellence, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA; (B.J.); (N.G.B.)
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (S.S.); (T.B.); (D.T.); (Z.A.M.); (A.K.T.-K.); (R.V.)
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, 2600 Glostrup, Denmark
- Correspondence: (M.L.); (M.K.); Tel.: +45-30-50-26-62 (M.L.); +45-29-80-76-67 (M.K.)
| |
Collapse
|
16
|
Szafran BN, Pinkston R, Perveen Z, Ross MK, Morgan T, Paulsen DB, Penn AL, Kaplan BLF, Noël A. Electronic-Cigarette Vehicles and Flavoring Affect Lung Function and Immune Responses in a Murine Model. Int J Mol Sci 2020; 21:E6022. [PMID: 32825651 PMCID: PMC7504509 DOI: 10.3390/ijms21176022] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
The use of electronic nicotine delivery systems (ENDS), also known as electronic-cigarettes (e-cigs), has raised serious public health concerns, especially in light of the 2019 outbreak of e-cig or vaping product use-associated acute lung injury (EVALI). While these cases have mostly been linked to ENDS that contain vitamin E acetate, there is limited research that has focused on the chronic pulmonary effects of the delivery vehicles (i.e., without nicotine and flavoring). Thus, we investigated lung function and immune responses in a mouse model following exposure to the nearly ubiquitous e-cig delivery vehicles, vegetable glycerin (VG) and propylene glycol (PG), used with a specific 70%/30% ratio, with or without vanilla flavoring. We hypothesized that mice exposed sub-acutely to these e-cig aerosols would exhibit lung inflammation and altered lung function. Adult female C57BL/6 mice (n = 11-12 per group) were exposed to filtered air, 70%/30% VG/PG, or 70%/30% VG/PG with a French vanilla flavoring for 2 h a day for 6 weeks. Prior to sacrifice, lung function was assessed. At sacrifice, broncho-alveolar lavage fluid and lung tissue were collected for lipid mediator analysis, flow cytometry, histopathology, and gene expression analyses. Exposures to VG/PG + vanilla e-cig aerosol increased lung tidal and minute volumes and tissue damping. Immunophenotyping of lung immune cells revealed an increased number of dendritic cells, CD4+ T cells, and CD19+ B cells in the VG/PG-exposed group compared to air, irrespective of the presence of vanilla flavoring. Quantification of bioactive lung lipids demonstrated a >3-fold increase of 2-arachidonoylglycerol (2-AG), an anti-inflammatory mediator, and a 2-fold increase of 12-hydroxyeicosatetraenoic acid (12-HETE), another inflammatory mediator, following VG/PG exposure, with or without vanilla flavoring. This suggests that e-cig aerosol vehicles may affect immunoregulatory molecules. We also found that the two e-cig aerosols dysregulated the expression of lung genes. Ingenuity Pathway Analysis revealed that the gene networks that are dysregulated by the VG/PG e-cig aerosol are associated with metabolism of cellular proteins and lipids. Overall, our findings demonstrate that VG and PG, the main constituents of e-liquid formulations, when aerosolized through an e-cig device, are not harmless to the lungs, since they disrupt immune homeostasis.
Collapse
Affiliation(s)
- Brittany N. Szafran
- Center for Environmental Health Sciences, Department of Basic Sciences, Mississippi State University College of Veterinary Medicine, Mississippi State, MS 39762, USA; (B.N.S.); (M.K.R.); (B.L.F.K.)
| | - Rakeysha Pinkston
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (R.P.); (Z.P.); (A.L.P.)
- Department of Environmental Toxicology, Southern University, Baton Rouge, LA 70803, USA
| | - Zakia Perveen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (R.P.); (Z.P.); (A.L.P.)
| | - Matthew K. Ross
- Center for Environmental Health Sciences, Department of Basic Sciences, Mississippi State University College of Veterinary Medicine, Mississippi State, MS 39762, USA; (B.N.S.); (M.K.R.); (B.L.F.K.)
| | - Timothy Morgan
- Department of Pathobiology and Population Medicine, Mississippi State University College of Veterinary Medicine, Mississippi State, MS 39762, USA;
| | - Daniel B. Paulsen
- Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
| | - Arthur L. Penn
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (R.P.); (Z.P.); (A.L.P.)
| | - Barbara L. F. Kaplan
- Center for Environmental Health Sciences, Department of Basic Sciences, Mississippi State University College of Veterinary Medicine, Mississippi State, MS 39762, USA; (B.N.S.); (M.K.R.); (B.L.F.K.)
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (R.P.); (Z.P.); (A.L.P.)
| |
Collapse
|
17
|
Stamenkovic A, Ganguly R, Aliani M, Ravandi A, Pierce GN. Overcoming the Bitter Taste of Oils Enriched in Fatty Acids to Obtain Their Effects on the Heart in Health and Disease. Nutrients 2019; 11:E1179. [PMID: 31137794 PMCID: PMC6566568 DOI: 10.3390/nu11051179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 01/18/2023] Open
Abstract
Fatty acids come in a variety of structures and, because of this, create a variety of functions for these lipids. Some fatty acids have a role to play in energy metabolism, some help in lipid storage, cell structure, the physical state of the lipid, and even in food stability. Fatty acid metabolism plays a particularly important role in meeting the energy demands of the heart. It is the primary source of myocardial energy in control conditions. Its role changes dramatically in disease states in the heart, but the pathologic role these fatty acids play depends upon the type of cardiovascular disease and the type of fatty acid. However, no matter how good a food is for one's health, its taste will ultimately become a deciding factor in its influence on human health. No food will provide health benefits if it is not ingested. This review discusses the taste characteristics of culinary oils that contain fatty acids and how these fatty acids affect the performance of the heart during healthy and diseased conditions. The contrasting contributions that different fatty acid molecules have in either promoting cardiac pathologies or protecting the heart from cardiovascular disease is also highlighted in this article.
Collapse
Affiliation(s)
- Aleksandra Stamenkovic
- Institute of Cardiovascular Sciences, St Boniface Hospital, Winnipeg, MB R2H2A6, Canada.
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E0W3, Canada.
| | - Riya Ganguly
- Institute of Cardiovascular Sciences, St Boniface Hospital, Winnipeg, MB R2H2A6, Canada.
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E0W3, Canada.
| | - Michel Aliani
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, University of Manitoba, Winnipeg, MB R2H2A6, Canada.
- Department of Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB R2H2A6, Canada.
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, St Boniface Hospital, Winnipeg, MB R2H2A6, Canada.
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E0W3, Canada.
- Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E0W3, Canada.
| | - Grant N Pierce
- Institute of Cardiovascular Sciences, St Boniface Hospital, Winnipeg, MB R2H2A6, Canada.
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E0W3, Canada.
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, University of Manitoba, Winnipeg, MB R2H2A6, Canada.
| |
Collapse
|
18
|
Singh NK, Rao GN. Emerging role of 12/15-Lipoxygenase (ALOX15) in human pathologies. Prog Lipid Res 2019; 73:28-45. [PMID: 30472260 PMCID: PMC6338518 DOI: 10.1016/j.plipres.2018.11.001] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
Abstract
12/15-lipoxygenase (12/15-LOX) is an enzyme, which oxidizes polyunsaturated fatty acids, particularly omega-6 and -3 fatty acids, to generate a number of bioactive lipid metabolites. A large number of studies have revealed the importance of 12/15-LOX role in oxidative and inflammatory responses. The in vitro studies have demonstrated the ability of 12/15-LOX metabolites in the expression of various genes and production of cytokine related to inflammation and resolution of inflammation. The studies with the use of knockout and transgenic animals for 12/15-LOX have further shown its involvement in the pathogenesis of a variety of human diseases, including cardiovascular, renal, neurological and metabolic disorders. This review summarizes our current knowledge on the role of 12/15-LOX in inflammation and various human diseases.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, 71 S. Manassas Street Memphis, Memphis, TN 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, 71 S. Manassas Street Memphis, Memphis, TN 38163, USA.
| |
Collapse
|
19
|
Moon SH, Liu X, Cedars AM, Yang K, Kiebish MA, Joseph SM, Kelley J, Jenkins CM, Gross RW. Heart failure-induced activation of phospholipase iPLA 2γ generates hydroxyeicosatetraenoic acids opening the mitochondrial permeability transition pore. J Biol Chem 2017; 293:115-129. [PMID: 29158256 DOI: 10.1074/jbc.ra117.000405] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/08/2017] [Indexed: 11/06/2022] Open
Abstract
Congestive heart failure typically arises from cardiac myocyte necrosis/apoptosis, associated with the pathological opening of the mitochondrial permeability transition pore (mPTP). mPTP opening decreases the mitochondrial membrane potential leading to the activation of Ca2+-independent phospholipase A2γ (iPLA2γ) and the production of downstream toxic metabolites. However, the array of enzymatic mediators and the exact chemical mechanisms responsible for modulating myocardial mPTP opening remain unclear. Herein, we demonstrate that human heart failure activates specific myocardial mitochondrial phospholipases that increase Ca2+-dependent production of toxic hydroxyeicosatetraenoic acids (HETEs) and attenuate the activity of phospholipases that promote the synthesis of protective epoxyeicosatrienoic acids (EETs). Mechanistically, HETEs activated the Ca2+-induced opening of the mPTP in failing human myocardium, and the highly selective pharmacological blockade of either iPLA2γ or lipoxygenases attenuated mPTP opening in failing hearts. In contrast, pharmacological inhibition of cytochrome P450 epoxygenases opened the myocardial mPTP in human heart mitochondria. Remarkably, the major mitochondrial phospholipase responsible for Ca2+-activated release of arachidonic acid (AA) in mitochondria from non-failing hearts was calcium-dependent phospholipase A2ζ (cPLA2ζ) identified by sequential column chromatographies and activity-based protein profiling. In contrast, iPLA2γ predominated in failing human myocardium. Stable isotope kinetics revealed that in non-failing human hearts, cPLA2ζ metabolically channels arachidonic acid into EETs, whereas in failing hearts, increased iPLA2γ activity channels AA into toxic HETEs. These results mechanistically identify the sequelae of pathological remodeling of human mitochondrial phospholipases in failing myocardium. This remodeling metabolically channels AA into toxic HETEs promoting mPTP opening, which induces necrosis/apoptosis leading to further progression of heart failure.
Collapse
Affiliation(s)
- Sung Ho Moon
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Xinping Liu
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Ari M Cedars
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Kui Yang
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Michael A Kiebish
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Susan M Joseph
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - John Kelley
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Christopher M Jenkins
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Richard W Gross
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110; Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110; Department of Chemistry, Washington University, St. Louis, Missouri 63130.
| |
Collapse
|
20
|
Zhang C, Ma C, Yao H, Zhang L, Yu X, Liu Y, Shen T, Zhang L, Zhang F, Chen X, Zhu D. 12-Lipoxygenase and 12-hydroxyeicosatetraenoic acid regulate hypoxic angiogenesis and survival of pulmonary artery endothelial cells via PI3K/Akt pathway. Am J Physiol Lung Cell Mol Physiol 2017; 314:L606-L616. [PMID: 29074487 DOI: 10.1152/ajplung.00049.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Dysfunction and injury of endothelial cells play critical roles in pulmonary arterial hypertension, including aberrant proliferation, suppressed apoptosis, and excessive angiogenesis. The 12-lipoxygenase and 12-hydroxyeicosatetraenoic acid pathway, which has been considered as a crucial mediator, elevates pulmonary vascular resistance and pulmonary arterial pressure. However, the mechanisms underlying the bioactivity of 12-hydroxyeicosatetraenoic acid in pulmonary vasculature, especially in endothelial cells, are still elusive. Thus we aim to determine the key role of 12-lipoxygenase/12-hydroxyeicosatetraenoic acid in angiogenesis and survival of pulmonary artery endothelial cells and ascertain the signaling pathways participating in the pathological process. Here we establish that hypoxia increases the formation of endogenous 12-hydroxyeicosatetraenoic acid through stimulation of 12-lipoxygenase. Furthermore, we put forward new information that 12-hydroxyeicosatetraenoic acid promotes endothelial cell migration and tube formation, whereas it inhibits the serum deprivation-induced apoptotic responses under hypoxia. Particularly, the regulatory effects of 12-lipoxygenase/12-hydroxyeicosatetraenoic acid on pulmonary artery endothelial cells, at least in part, depend on phosphatidylinositol 3-kinase (PI3K)/Akt signaling activation. Taken together, these results may have significant implications for understanding of pulmonary hypertension and offer a potential therapeutic concept focusing on the 12-lipoxygenase/12-hydroxyeicosatetraenoic acid signaling system.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China
| | - Cui Ma
- Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Hongmin Yao
- Petit Science Center, Department of Biology, College of Arts and Sciences, Georgia State University , Atlanta, Georgia
| | - Lixin Zhang
- Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Xiufeng Yu
- Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Yumei Liu
- Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Tingting Shen
- Department of Pharmacology, Dalian Medical University , Dalian , China
| | - Linlin Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China
| | - Fengying Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China
| | - Xinxin Chen
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China.,Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China.,Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University , Harbin , China
| |
Collapse
|
21
|
Ma K, Xiao A, Park SH, Glenn L, Jackson L, Barot T, Weaver JR, Taylor-Fishwick DA, Luci DK, Maloney DJ, Mirmira RG, Imai Y, Nadler JL. 12-Lipoxygenase Inhibitor Improves Functions of Cytokine-Treated Human Islets and Type 2 Diabetic Islets. J Clin Endocrinol Metab 2017; 102:2789-2797. [PMID: 28609824 PMCID: PMC5546865 DOI: 10.1210/jc.2017-00267] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/03/2017] [Indexed: 12/21/2022]
Abstract
CONTEXT The 12-lipoxygenase (12-LO) pathway produces proinflammatory metabolites, and its activation is implicated in islet inflammation associated with type 1 and type 2 diabetes (T2D). OBJECTIVES We aimed to test the efficacy of ML355, a highly selective, small molecule inhibitor of 12-LO, for the preservation of islet function. DESIGN Human islets from nondiabetic donors were incubated with a mixture of tumor necrosis factor α , interluekin-1β, and interferon-γ to model islet inflammation. Cytokine-treated islets and human islets from T2D donors were incubated in the presence and absence of ML355. SETTING In vitro study. PARTICIPANTS Human islets from organ donors aged >20 years of both sexes and any race were used. T2D status was defined from either medical history or most recent hemoglobin A1c value >6.5%. INTERVENTION Glucose stimulation. MAIN OUTCOME MEASURES Static and dynamic insulin secretion and oxygen consumption rate (OCR). RESULTS ML355 prevented the reduction of insulin secretion and OCR in cytokine-treated human islets and improved both parameters in human islets from T2D donors. CONCLUSIONS ML355 was efficacious in improving human islet function after cytokine treatment and in T2D islets in vitro. The study suggests that the blockade of the 12-LO pathway may serve as a target for both form of diabetes and provides the basis for further study of this small molecule inhibitor in vivo.
Collapse
Affiliation(s)
- Kaiwen Ma
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - An Xiao
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - So Hyun Park
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Lindsey Glenn
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Laura Jackson
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Tatvam Barot
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Jessica R. Weaver
- Department of Microbiology & Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - David A. Taylor-Fishwick
- Department of Microbiology & Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Diane K. Luci
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - David J. Maloney
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Raghavendra G. Mirmira
- Department of Pediatrics, IU Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Departments of Biochemistry and Molecular Biology, Medicine, and Cellular and Integrative Physiology, IU Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Indiana Biosciences Research Institute, Indianapolis, Indiana 46202
| | - Yumi Imai
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Jerry L. Nadler
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
22
|
Brasili E, Filho VC. Metabolomics of cancer cell cultures to assess the effects of dietary phytochemicals. Crit Rev Food Sci Nutr 2017; 57:1328-1339. [DOI: 10.1080/10408398.2014.964799] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Elisa Brasili
- Department of Environmental Biology, “Sapienza” University of Rome, Rome, Italy
| | - Valdir Cechinel Filho
- Programa de Pós-Graduação em Ciências Farmacêuticas e Núcleo de Investigações Químico-Farmacêuticas/CCS, Universidade do Vale do Itajaí, Itajaí, SC, Brazil
| |
Collapse
|
23
|
Charoensin S, Eroglu E, Opelt M, Bischof H, Madreiter-Sokolowski CT, Kirsch A, Depaoli MR, Frank S, Schrammel A, Mayer B, Waldeck-Weiermair M, Graier WF, Malli R. Intact mitochondrial Ca 2+ uniport is essential for agonist-induced activation of endothelial nitric oxide synthase (eNOS). Free Radic Biol Med 2017; 102:248-259. [PMID: 27923677 PMCID: PMC5381715 DOI: 10.1016/j.freeradbiomed.2016.11.049] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 11/14/2016] [Accepted: 11/28/2016] [Indexed: 12/18/2022]
Abstract
Mitochondrial Ca2+ uptake regulates diverse endothelial cell functions and has also been related to nitric oxide (NO•) production. However, it is not entirely clear if the organelles support or counteract NO• biosynthesis by taking up Ca2+. The objective of this study was to verify whether or not mitochondrial Ca2+ uptake influences Ca2+-triggered NO• generation by endothelial NO• synthase (eNOS) in an immortalized endothelial cell line (EA.hy926), respective primary human umbilical vein endothelial cells (HUVECs) and eNOS-RFP (red fluorescent protein) expressing human embryonic kidney (HEK293) cells. We used novel genetically encoded fluorescent NO• probes, the geNOps, and Ca2+ sensors to monitor single cell NO• and Ca2+ dynamics upon cell treatment with ATP, an inositol 1,4,5-trisphosphate (IP3)-generating agonist. Mitochondrial Ca2+ uptake was specifically manipulated by siRNA-mediated knock-down of recently identified key components of the mitochondrial Ca2+ uniporter machinery. In endothelial cells and the eNOS-RFP expressing HEK293 cells we show that reduced mitochondrial Ca2+ uptake upon the knock-down of the mitochondrial calcium uniporter (MCU) protein and the essential MCU regulator (EMRE) yield considerable attenuation of the Ca2+-triggered NO• increase independently of global cytosolic Ca2+ signals. The knock-down of mitochondrial calcium uptake 1 (MICU1), a gatekeeper of the MCU, increased both mitochondrial Ca2+ sequestration and Ca2+-induced NO• signals. The positive correlation between mitochondrial Ca2+ elevation and NO• production was independent of eNOS phosphorylation at serine1177. Our findings emphasize that manipulating mitochondrial Ca2+ uptake may represent a novel strategy to control eNOS-mediated NO• production.
Collapse
Affiliation(s)
- Suphachai Charoensin
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | - Emrah Eroglu
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | - Marissa Opelt
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria
| | - Helmut Bischof
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | | | - Andrijana Kirsch
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | - Maria R Depaoli
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | - Saša Frank
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | - Astrid Schrammel
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria
| | - Bernd Mayer
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria
| | - Markus Waldeck-Weiermair
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | - Wolfgang F Graier
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | - Roland Malli
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria.
| |
Collapse
|
24
|
Moon SH, Mancuso DJ, Sims HF, Liu X, Nguyen AL, Yang K, Guan S, Dilthey BG, Jenkins CM, Weinheimer CJ, Kovacs A, Abendschein D, Gross RW. Cardiac Myocyte-specific Knock-out of Calcium-independent Phospholipase A2γ (iPLA2γ) Decreases Oxidized Fatty Acids during Ischemia/Reperfusion and Reduces Infarct Size. J Biol Chem 2016; 291:19687-700. [PMID: 27453526 DOI: 10.1074/jbc.m116.740597] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Indexed: 12/21/2022] Open
Abstract
Calcium-independent phospholipase A2γ (iPLA2γ) is a mitochondrial enzyme that produces lipid second messengers that facilitate opening of the mitochondrial permeability transition pore (mPTP) and contribute to the production of oxidized fatty acids in myocardium. To specifically identify the roles of iPLA2γ in cardiac myocytes, we generated cardiac myocyte-specific iPLA2γ knock-out (CMiPLA2γKO) mice by removing the exon encoding the active site serine (Ser-477). Hearts of CMiPLA2γKO mice exhibited normal hemodynamic function, glycerophospholipid molecular species composition, and normal rates of mitochondrial respiration and ATP production. In contrast, CMiPLA2γKO mice demonstrated attenuated Ca(2+)-induced mPTP opening that could be rapidly restored by the addition of palmitate and substantially reduced production of oxidized polyunsaturated fatty acids (PUFAs). Furthermore, myocardial ischemia/reperfusion (I/R) in CMiPLA2γKO mice (30 min of ischemia followed by 30 min of reperfusion in vivo) dramatically decreased oxidized fatty acid production in the ischemic border zones. Moreover, CMiPLA2γKO mice subjected to 30 min of ischemia followed by 24 h of reperfusion in vivo developed substantially less cardiac necrosis in the area-at-risk in comparison with their WT littermates. Furthermore, we found that membrane depolarization in murine heart mitochondria was sensitized to Ca(2+) by the presence of oxidized PUFAs. Because mitochondrial membrane depolarization and calcium are known to activate iPLA2γ, these results are consistent with salvage of myocardium after I/R by iPLA2γ loss of function through decreasing mPTP opening, diminishing production of proinflammatory oxidized fatty acids, and attenuating the deleterious effects of abrupt increases in calcium ion on membrane potential during reperfusion.
Collapse
Affiliation(s)
- Sung Ho Moon
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine
| | - David J Mancuso
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine
| | - Harold F Sims
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine
| | - Xinping Liu
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine
| | - Annie L Nguyen
- Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri 63110 and
| | - Kui Yang
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine
| | - Shaoping Guan
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine
| | - Beverly Gibson Dilthey
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine
| | - Christopher M Jenkins
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine
| | - Carla J Weinheimer
- Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri 63110 and
| | - Attila Kovacs
- Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri 63110 and
| | - Dana Abendschein
- Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri 63110 and
| | - Richard W Gross
- From the Division of Bioorganic Chemistry and Molecular Pharmacology, Departments of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri 63110 and Developmental Biology, and the Department of Chemistry, Washington University, Saint Louis, Missouri 63130
| |
Collapse
|
25
|
Imai Y, Dobrian AD, Morris MA, Taylor-Fishwick DA, Nadler JL. Lipids and immunoinflammatory pathways of beta cell destruction. Diabetologia 2016; 59:673-8. [PMID: 26868492 PMCID: PMC4779407 DOI: 10.1007/s00125-016-3890-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 12/30/2015] [Indexed: 12/18/2022]
Abstract
Islet inflammation contributes to beta cell demise in both type 1 and type 2 diabetes. 12-Lipoxygenase (12-LO, gene expressed as ALOX12 in humans and 12-Lo in rodents in this manuscript) produces proinflammatory metabolites such as 12(S)-hydroxyeicosatetraenoic acids through dioxygenation of polyunsaturated fatty acids. 12-LO was first implicated in diabetes when the increase in 12-Lo expression and 12(S)-hydroxyeicosatetraenoic acid was noted in rodent models of diabetes. Subsequently, germline 12-Lo (-/-) was shown to prevent the development of hyperglycemia in mouse models of type 1 diabetes and in high-fat fed mice. More recently, beta cell-specific 12-Lo (-/-) was shown to protect mice against hyperglycaemia after streptozotocin and a high-fat diet. In humans, 12-LO expression is increased in pancreatic islets of autoantibody-positive, type 1 diabetic and type 2 diabetic organ donors. Interestingly, the high expression of ALOX12 is associated with the alteration in first-phase glucose-stimulated insulin secretion in human type 2 diabetic islets. To further clarify the role of islet 12-LO in diabetes and to validate 12-LO as a therapeutic target of diabetes, we have studied selective pharmacological inhibitors for 12-LO. The compounds we have identified show promise: they protect beta cell lines and human islets from apoptosis and preserve insulin secretion when challenged by proinflammatory cytokine mixture. Currently studies are underway to test the compounds in mouse models of diabetes. This review summarises a presentation given at the 'Islet inflammation in type 2 diabetes' symposium at the 2015 annual meeting of the EASD. It is accompanied two other mini-reviews on topics from this symposium (by Simone Baltrusch, DOI: 10.1007/s00125-016-3891-x and Marc Donath, DOI: 10.1007/s00125-016-3873-z ) and a commentary by the Session Chair, Piero Marchetti (DOI: 10.1007/s00125-016-3875-x ).
Collapse
Affiliation(s)
- Yumi Imai
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, 23507, USA.
| | - Anca D Dobrian
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Margaret A Morris
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - David A Taylor-Fishwick
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Jerry L Nadler
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, 23507, USA.
| |
Collapse
|
26
|
Maayah ZH, Althurwi HN, Abdelhamid G, Lesyk G, Jurasz P, El-Kadi AO. CYP1B1 inhibition attenuates doxorubicin-induced cardiotoxicity through a mid-chain HETEs-dependent mechanism. Pharmacol Res 2016; 105:28-43. [DOI: 10.1016/j.phrs.2015.12.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/01/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022]
|
27
|
The role of mid-chain hydroxyeicosatetraenoic acids in the pathogenesis of hypertension and cardiac hypertrophy. Arch Toxicol 2015; 90:119-36. [PMID: 26525395 DOI: 10.1007/s00204-015-1620-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/19/2015] [Indexed: 12/16/2022]
Abstract
The incidence, prevalence, and hospitalization rates associated with cardiovascular diseases (CVDs) are projected to increase substantially in the world. Understanding of the biological and pathophysiological mechanisms of survival can help the researchers to develop new management modalities. Numerous experimental studies have demonstrated that mid-chain HETEs are strongly involved in the pathogenesis of the CVDs. Mid-chain HETEs are biologically active eicosanoids that result from the metabolism of arachidonic acid (AA) by both lipoxygenase and CYP1B1 (lipoxygenase-like reaction). Therefore, identifying the localizations and expressions of the lipoxygenase and CYP1B1 and their associated AA metabolites in the cardiovascular system is of major importance in understanding their pathological roles. Generally, the expression of these enzymes is shown to be induced during several CVDs, including hypertension and cardiac hypertrophy. The induction of these enzymes is associated with the generation of mid-chain HETEs and subsequently causation of cardiovascular events. Of interest, inhibiting the formation of mid-chain HETEs has been reported to confer a protection against different cardiac hypertrophy and hypertension models such as angiotensin II, Goldblatt, spontaneously hypertensive rat and deoxycorticosterone acetate (DOCA)-salt-induced models. Although the exact mechanisms of mid-chain HETEs-mediated cardiovascular dysfunction are not fully understood, the present review proposes several mechanisms which include activating G-protein-coupled receptor, protein kinase C, mitogen-activated protein kinases, and nuclear factor kappa B. This review provides a clear understanding of the role of mid-chain HETEs in the pathogenesis of cardiovascular diseases and their importance as novel targets in the treatment for hypertension and cardiac hypertrophy.
Collapse
|
28
|
Gabbs M, Leng S, Devassy JG, Monirujjaman M, Aukema HM. Advances in Our Understanding of Oxylipins Derived from Dietary PUFAs. Adv Nutr 2015; 6:513-40. [PMID: 26374175 PMCID: PMC4561827 DOI: 10.3945/an.114.007732] [Citation(s) in RCA: 537] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Oxylipins formed from polyunsaturated fatty acids (PUFAs) are the main mediators of PUFA effects in the body. They are formed via cyclooxygenase, lipoxygenase, and cytochrome P450 pathways, resulting in the formation of prostaglandins, thromboxanes, mono-, di-, and tri-hydroxy fatty acids (FAs), epoxy FAs, lipoxins, eoxins, hepoxilins, resolvins, protectins (also called neuroprotectins in the brain), and maresins. In addition to the well-known eicosanoids derived from arachidonic acid, recent developments in lipidomic methodologies have raised awareness of and interest in the large number of oxylipins formed from other PUFAs, including those from the essential FAs and the longer-chain n-3 (ω-3) PUFAs. Oxylipins have essential roles in normal physiology and function, but can also have detrimental effects. Compared with the oxylipins derived from n-3 PUFAs, oxylipins from n-6 PUFAs generally have greater activity and more inflammatory, vasoconstrictory, and proliferative effects, although there are notable exceptions. Because PUFA composition does not necessarily reflect oxylipin composition, comprehensive analysis of the oxylipin profile is necessary to understand the overall physiologic effects of PUFAs mediated through their oxylipins. These analyses should include oxylipins derived from linoleic and α-linolenic acids, because these largely unexplored bioactive oxylipins constitute more than one-half of oxylipins present in tissues. Because collated information on oxylipins formed from different PUFAs is currently unavailable, this review provides a detailed compilation of the main oxylipins formed from PUFAs and describes their functions. Much remains to be elucidated in this emerging field, including the discovery of more oxylipins, and the understanding of the differing biological potencies, kinetics, and isomer-specific activities of these novel PUFA metabolites.
Collapse
Affiliation(s)
| | | | | | | | - Harold M Aukema
- Human Nutritional Sciences, University of Manitoba, Winnipeg, Canada; and Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Canada
| |
Collapse
|
29
|
Evans J, Ko Y, Mata W, Saquib M, Eldridge J, Cohen-Gadol A, Leaver HA, Wang S, Rizzo MT. Arachidonic acid induces brain endothelial cell apoptosis via p38-MAPK and intracellular calcium signaling. Microvasc Res 2014; 98:145-58. [PMID: 24802256 DOI: 10.1016/j.mvr.2014.04.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/23/2014] [Accepted: 04/27/2014] [Indexed: 01/19/2023]
Abstract
Arachidonic acid (AA), a bioactive fatty acid whose levels increase during neuroinflammation, contributes to cerebral vascular damage and dysfunction. However, the mode of injury and underlying signaling mechanisms remain unknown. Challenge of primary human brain endothelial cells (HBECs) with AA activated a stress response resulting in caspase-3 activation, poly(ADP-ribose) polymerase cleavage, and disruption of monolayer integrity. AA also induced loss of mitochondrial membrane potential and cytochrome c release consistent with activation of intrinsic apoptosis. HBEC stimulation with AA resulted in sustained p38-MAPK activation and subsequent phosphorylation of mitogen-activated protein kinase activated protein-2 (MAPKAP-2) kinase and heat shock protein-27 (Hsp27). Conversely, other unsaturated and saturated fatty acids had no effect. Pharmacological and RNA interference-mediated p38α or p38β suppression abrogated AA signaling to caspase-3 and Hsp27, suggesting involvement of both p38 isoforms in AA-induced HBEC apoptosis. Hsp27 silencing also blocked caspase-3 activation. AA stimulated intracellular calcium release, which was attenuated by inositol 1,4,5-trisphosphate (IP3) receptor antagonists. Blockade of intracellular calcium release decreased caspase-3 activation, but had no effect on AA-induced p38-MAPK activation. However, inhibition of p38-MAPK or blockade of intracellular calcium mobilization abrogated AA-induced cytochrome c release. AA-induced caspase-3 activation was abrogated by pharmacological inhibition of lipooxygenases. These findings support a previously unrecognized signaling cooperation between p38-MAPK/MAPKAP-2/Hsp27 and intracellular calcium release in AA-induced HBEC apoptosis and suggest its relevance to neurological disorders associated with vascular inflammation.
Collapse
Affiliation(s)
- Justin Evans
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - YooSeung Ko
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wilmer Mata
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Muhammad Saquib
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel Eldridge
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Aaron Cohen-Gadol
- Goodman Campbell Brain and Spine, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Anne Leaver
- Division of Clinical Neuroscience, Edinburgh University, Edinburgh, UK
| | - Shukun Wang
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maria Teresa Rizzo
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
30
|
Abstract
ALOX12 is a gene encoding arachidonate 12-lipoxygenase (12-LOX), a member of a nonheme lipoxygenase family of dioxygenases. ALOX12 catalyzes the addition of oxygen to arachidonic acid, producing 12-hydroperoxyeicosatetraenoic acid (12-HPETE), which can be reduced to the eicosanoid 12-HETE (12-hydroxyeicosatetraenoic acid). 12-HETE acts in diverse cellular processes, including catecholamine synthesis, vasoconstriction, neuronal function, and inflammation. Consistent with effects on these fundamental mechanisms, allelic variants of ALOX12 are associated with diseases including schizophrenia, atherosclerosis, and cancers, but the mechanisms have not been defined. Toxoplasma gondii is an apicomplexan parasite that causes morbidity and mortality and stimulates an innate and adaptive immune inflammatory reaction. Recently, it has been shown that a gene region known as Toxo1 is critical for susceptibility or resistance to T. gondii infection in rats. An orthologous gene region with ALOX12 centromeric is also present in humans. Here we report that the human ALOX12 gene has susceptibility alleles for human congenital toxoplasmosis (rs6502997 [P, <0.000309], rs312462 [P, <0.028499], rs6502998 [P, <0.029794], and rs434473 [P, <0.038516]). A human monocytic cell line was genetically engineered using lentivirus RNA interference to knock down ALOX12. In ALOX12 knockdown cells, ALOX12 RNA expression decreased and levels of the ALOX12 substrate, arachidonic acid, increased. ALOX12 knockdown attenuated the progression of T. gondii infection and resulted in greater parasite burdens but decreased consequent late cell death of the human monocytic cell line. These findings suggest that ALOX12 influences host responses to T. gondii infection in human cells. ALOX12 has been shown in other studies to be important in numerous diseases. Here we demonstrate the critical role ALOX12 plays in T. gondii infection in humans.
Collapse
|
31
|
12/15-lipoxygenase expressed in non-epithelial cells causes airway epithelial injury in asthma. Sci Rep 2013; 3:1540. [PMID: 23528921 PMCID: PMC3607899 DOI: 10.1038/srep01540] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 03/08/2013] [Indexed: 01/14/2023] Open
Abstract
The mechanisms underlying asthmatic airway epithelial injury are not clear. 12/15-lipoxygenase (an ortholog of human 15-LOX-1), which is induced by IL-13, is associated with mitochondrial degradation in reticulocytes at physiological conditions. In this study, we showed that 12/15-LOX expressed in nonepithelial cells caused epithelial injury in asthma pathogenesis. While 12/15-LOX overexpression or IL-13 administration to naïve mice showed airway epithelial injury, 12/15-LOX knockout/knockdown in allergic mice reduced airway epithelial injury. The constitutive expression of 15-LOX-1 in bronchial epithelia of normal human lungs further indicated that epithelial 15-LOX-1 may not cause epithelial injury. 12/15-LOX expression is increased in various inflammatory cells in allergic mice. Though non-epithelial cells such as macrophages or fibroblasts released 12/15-LOX metabolites upon IL-13 induction, bronchial epithelia didn't release. Further 12-S-HETE, arachidonic acid metabolite of 12/15-LOX leads to epithelial injury. These findings suggested 12/15-LOX expressed in non-epithelial cells such as macrophages and fibroblasts leads to bronchial epithelial injury.
Collapse
|
32
|
Mabalirajan U, Ghosh B. Mitochondrial dysfunction in metabolic syndrome and asthma. J Allergy (Cairo) 2013; 2013:340476. [PMID: 23840225 PMCID: PMC3687506 DOI: 10.1155/2013/340476] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 05/21/2013] [Indexed: 01/15/2023] Open
Abstract
Though severe or refractory asthma merely affects less than 10% of asthma population, it consumes significant health resources and contributes significant morbidity and mortality. Severe asthma does not fell in the routine definition of asthma and requires alternative treatment strategies. It has been observed that asthma severity increases with higher body mass index. The obese-asthmatics, in general, have the features of metabolic syndrome and are progressively causing a significant burden for both developed and developing countries thanks to the westernization of the world. As most of the features of metabolic syndrome seem to be originated from central obesity, the underlying mechanisms for metabolic syndrome could help us to understand the pathobiology of obese-asthma condition. While mitochondrial dysfunction is the common factor for most of the risk factors of metabolic syndrome, such as central obesity, dyslipidemia, hypertension, insulin resistance, and type 2 diabetes, the involvement of mitochondria in obese-asthma pathogenesis seems to be important as mitochondrial dysfunction has recently been shown to be involved in airway epithelial injury and asthma pathogenesis. This review discusses current understanding of the overlapping features between metabolic syndrome and asthma in relation to mitochondrial structural and functional alterations with an aim to uncover mechanisms for obese-asthma.
Collapse
Affiliation(s)
- Ulaganathan Mabalirajan
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | - Balaram Ghosh
- Molecular Immunogenetics Laboratory and Centre of Excellence for Translational Research in Asthma & Lung Disease, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| |
Collapse
|
33
|
Zhang HJ, Sun CH, Kuang HY, Jiang XY, Liu HL, Hua WF, Liu ZJ, Zhou H, Sui H, Qi R. 12S-hydroxyeicosatetraenoic acid levels link to coronary artery disease in Type 2 diabetic patients. J Endocrinol Invest 2013; 36:385-9. [PMID: 23095287 DOI: 10.3275/8654] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND 12(S)-Hydroxyeicosatetraenoic acid (12(S)-HETE) is a metabolite of arachidonic acid. 12(S)-HETE is involved in the pathogenesis of atherosclerosis and diabetes. However, the correlation between 12(S)-HETE and coronary artery disease (CAD) in the diabetic patient is unclear. AIMS The study investigated the relationship between 12(S)-HETE and CAD in Type 2 diabetes (T2D). METHODS Plasma 12(S)- HETE levels were detected by enzyme-linked immunosorbent assay in 103 healthy controls (control), 109 diabetic patients without CAD (diabetic), and 152 diabetic patients with CAD (diabetic-CAD). RESULTS 12(S)-HETE levels were higher in both diabetic and diabetic-CAD groups compared to control and in the diabetic-CAD group compared to the diabetic group. In the multiple linear stepwise regression analysis, 12(S)-HETE levels correlated independently with CAD, systolic blood pressure, and glycated hemoglobin. CONCLUSIONS These results indicate that 12(S)-HETE levels are increased in diabetic patients with CAD, suggesting a role for atherosclerosis in T2D.
Collapse
Affiliation(s)
- H J Zhang
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Moon SH, Jenkins CM, Liu X, Guan S, Mancuso DJ, Gross RW. Activation of mitochondrial calcium-independent phospholipase A2γ (iPLA2γ) by divalent cations mediating arachidonate release and production of downstream eicosanoids. J Biol Chem 2012; 287:14880-95. [PMID: 22389508 DOI: 10.1074/jbc.m111.336776] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Calcium-independent phospholipase A(2)γ (iPLA(2)γ) (PNPLA8) is the predominant phospholipase activity in mammalian mitochondria. However, the chemical mechanisms that regulate its activity are unknown. Here, we utilize iPLA(2)γ gain of function and loss of function genetic models to demonstrate the robust activation of iPLA(2)γ in murine myocardial mitochondria by Ca(2+) or Mg(2+) ions. Calcium ion stimulated the production of 2-arachidonoyl-lysophosphatidylcholine (2-AA-LPC) from 1-palmitoyl-2-[(14)C]arachidonoyl-sn-glycero-3-phosphocholine during incubations with wild-type heart mitochondrial homogenates. Furthermore, incubation of mitochondrial homogenates from transgenic myocardium expressing iPLA(2)γ resulted in 13- and 25-fold increases in the initial rate of radiolabeled 2-AA-LPC and arachidonic acid (AA) production, respectively, in the presence of calcium ion. Mass spectrometric analysis of the products of calcium-activated hydrolysis of endogenous mitochondrial phospholipids in transgenic iPLA(2)γ mitochondria revealed the robust production of AA, 2-AA-LPC, and 2-docosahexaenoyl-LPC that was over 10-fold greater than wild-type mitochondria. The mechanism-based inhibitor (R)-(E)-6-(bromomethylene)-3-(1-naphthalenyl)-2H-tetrahydropyran-2-one (BEL) (iPLA(2)γ selective), but not its enantiomer, (S)-BEL (iPLA(2)β selective) or pyrrolidine (cytosolic PLA(2)α selective), markedly attenuated Ca(2+)-dependent fatty acid release and polyunsaturated LPC production. Moreover, Ca(2+)-induced iPLA(2)γ activation was accompanied by the production of downstream eicosanoid metabolites that were nearly completely ablated by (R)-BEL or by genetic ablation of iPLA(2)γ. Intriguingly, Ca(2+)-induced iPLA(2)γ activation was completely inhibited by long-chain acyl-CoA (IC(50) ∼20 μm) as well as by a nonhydrolyzable acyl-CoA thioether analog. Collectively, these results demonstrate that mitochondrial iPLA(2)γ is activated by divalent cations and inhibited by acyl-CoA modulating the generation of biologically active metabolites that regulate mitochondrial bioenergetic and signaling functions.
Collapse
Affiliation(s)
- Sung Ho Moon
- Department of Medicine, Division of Bioorganic Chemistry and Molecular Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
35
|
Rink C, Khanna S. Significance of brain tissue oxygenation and the arachidonic acid cascade in stroke. Antioxid Redox Signal 2011; 14:1889-903. [PMID: 20673202 PMCID: PMC3078506 DOI: 10.1089/ars.2010.3474] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The significance of the hypoxia component of stroke injury is highlighted by hypermetabolic brain tissue enriched with arachidonic acid (AA), a 22:6n-3 polyunsaturated fatty acid. In an ischemic stroke environment in which cerebral blood flow is arrested, oxygen-starved brain tissue initiates the rapid cleavage of AA from the membrane phospholipid bilayer. Once free, AA undergoes both enzyme-independent and enzyme-mediated oxidative metabolism, resulting in the formation of number of biologically active metabolites which themselves contribute to pathological stroke outcomes. This review is intended to examine two divergent roles of molecular dioxygen in brain tissue as (1) a substrate for life-sustaining homeostatic metabolism of glucose and (2) a substrate for pathogenic metabolism of AA under conditions of stroke. Recent developments in research concerning supplemental oxygen therapy as an intervention to correct the hypoxic component of stroke injury are discussed.
Collapse
Affiliation(s)
- Cameron Rink
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio 43210, USA
| | | |
Collapse
|
36
|
Jäger W, Gruber A, Giessrigl B, Krupitza G, Szekeres T, Sonntag D. Metabolomic Analysis of Resveratrol-Induced Effects in the Human Breast Cancer Cell Lines MCF-7 and MDA-MB-231. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2011; 15:9-14. [DOI: 10.1089/omi.2010.0114] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | | | - Benedikt Giessrigl
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Georg Krupitza
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Thomas Szekeres
- Department of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
37
|
Dobrian AD, Lieb DC, Cole BK, Taylor-Fishwick DA, Chakrabarti SK, Nadler JL. Functional and pathological roles of the 12- and 15-lipoxygenases. Prog Lipid Res 2010; 50:115-31. [PMID: 20970452 DOI: 10.1016/j.plipres.2010.10.005] [Citation(s) in RCA: 246] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 10/13/2010] [Accepted: 10/14/2010] [Indexed: 12/25/2022]
Abstract
The 12/15-lipoxygenase enzymes react with fatty acids producing active lipid metabolites that are involved in a number of significant disease states. The latter include type 1 and type 2 diabetes (and associated complications), cardiovascular disease, hypertension, renal disease, and the neurological conditions Alzheimer's disease and Parkinson's disease. A number of elegant studies over the last thirty years have contributed to unraveling the role that lipoxygenases play in chronic inflammation. The development of animal models with targeted gene deletions has led to a better understanding of the role that lipoxygenases play in various conditions. Selective inhibitors of the different lipoxygenase isoforms are an active area of investigation, and will be both an important research tool and a promising therapeutic target for treating a wide spectrum of human diseases.
Collapse
Affiliation(s)
- Anca D Dobrian
- Eastern Virginia Medical School, Department of Physiological Sciences, Lewis Hall, Room 2027, 700 W. Olney Road, Norfolk, VA 23507, United States.
| | | | | | | | | | | |
Collapse
|
38
|
Comba A, Maestri DM, Berra MA, Garcia CP, Das UN, Eynard AR, Pasqualini ME. Effect of ω-3 and ω-9 fatty acid rich oils on lipoxygenases and cyclooxygenases enzymes and on the growth of a mammary adenocarcinoma model. Lipids Health Dis 2010; 9:112. [PMID: 20932327 PMCID: PMC2959203 DOI: 10.1186/1476-511x-9-112] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 10/08/2010] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Nutritional factors play a major role in cancer initiation and development. Dietary polyunsaturated fatty acids (PUFAs) have the ability to induce modifications in the activity of lipoxygenase (LOX) and cyclooxygenase (COX) enzymes that affect tumour growth. We studied the effect of two diets enriched in 6% Walnut and Peanut oils that are rich in ω-3 and ω9 PUFAs respectively on a murine mammary gland adenocarcinoma as compared with the control (C) that received commercial diet. RESULTS Peanut oil enriched diet induced an increase in membrane arachidonic acid (AA) content and the cyclooxygenase enzyme derived 12-HHT (p < 0.05) and simultaneously showed decrease in 12-LOX, 15-LOX-2, 15-LOX-1 and PGE activities (p < 0.05) that corresponded to higher apoptosis and lower mitosis seen in this group (p < 0.05). Furthermore, Peanut oil group showed lower T-cell infiltration (p < 0.05), number of metastasis (p < 0.05) and tumour volume (p < 0.05) and longer survival rate compared to other groups. CONCLUSIONS The results of the present study showed that Peanut oil-enriched diet protects against mammary cancer development by modulating tumour membrane fatty acids composition and LOX and COX enzyme activities.
Collapse
Affiliation(s)
- Andrea Comba
- Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
A growing body of research supports that members of the vitamin E family are not redundant with respect to their biological function. Palm oil derived from Elaeis guineensis represents the richest source of the lesser characterized vitamin E, alpha-tocotrienol. One of 8 naturally occurring and chemically distinct vitamin E analogs, alpha-tocotrienol possesses unique biological activity that is independent of its potent antioxidant capacity. Current developments in alpha-tocotrienol research demonstrate neuroprotective properties for the lipid-soluble vitamin in brain tissue rich in polyunsaturated fatty acids (PUFAs). Arachidonic acid (AA), one of the most abundant PUFAs of the central nervous system, is highly susceptible to oxidative metabolism under pathologic conditions. Cleaved from the membrane phospholipid bilayer by cytosolic phospholipase A(2), AA is metabolized by both enzymatic and nonenzymatic pathways. A number of neurodegenerative conditions in the human brain are associated with disturbed PUFA metabolism of AA, including acute ischemic stroke. Palm oil-derived alpha-tocotrienol at nanomolar concentrations has been shown to attenuate both enzymatic and nonenzymatic mediators of AA metabolism and neurodegeneration. On a concentration basis, this represents the most potent of all biological functions exhibited by any natural vitamin E molecule. Despite such therapeutic potential, the scientific literature on tocotrienols accounts for roughly 1% of the total literature on vitamin E, thus warranting further investment and investigation.
Collapse
Affiliation(s)
- Chandan K Sen
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio, USA.
| | | | | |
Collapse
|
40
|
Dietrich HH, Abendschein DR, Moon SH, Nayeb-Hashemi N, Mancuso DJ, Jenkins CM, Kaltenbronn KM, Blumer KJ, Turk J, Gross RW. Genetic ablation of calcium-independent phospholipase A(2)beta causes hypercontractility and markedly attenuates endothelium-dependent relaxation to acetylcholine. Am J Physiol Heart Circ Physiol 2010; 298:H2208-20. [PMID: 20382858 DOI: 10.1152/ajpheart.00839.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of phospholipases leads to the release of arachidonic acid and lysophospholipids that play prominent roles in regulating vasomotor tone. To identify the role of calcium-independent phospholipase A(2)beta (iPLA(2)beta) in vasomotor function, we measured vascular responses to phenylephrine (PE) and ACh in mesenteric arterioles from wild-type (WT; iPLA(2)beta(+/+)) mice and those lacking the beta-isoform (iPLA(2)beta(-/-)) both ex vivo and in vivo. Vessels isolated from iPLA(2)beta(-/-) mice demonstrated increased constriction to PE, despite lower basal smooth muscle calcium levels, and decreased vasodilation to ACh compared with iPLA(2)beta(+/+) mice. PE constriction resulted in initial intracellular calcium release with subsequent steady-state constriction that depended on extracellular calcium influx. Endothelial denudation had no effect on vessel tone or PE-induced constriction although the dilation to ACh was significantly reduced in iPLA(2)beta(+/+) vessels. In contrast, vessels from iPLA(2)beta(-/-) constricted by 54% after denudation, indicating smooth muscle hypercontractility. In vivo, blood pressure, resting vessel diameter, and constriction of mesenteric vessels to PE were not different in iPLA(2)beta(-/-) vessels compared with WT mouse vessels. However, relaxation after ACh administration in situ was attenuated, indicating an endothelial inability to induce dilation in response to ACh. In cultured endothelial cells, inhibition of iPLA(2)beta with (S)-(E)-6-(bromomethylene)tetrahydro-3-(1-naphthalenyl)-2H-pyran-2-one (BEL) decreased endothelial nitric oxide synthase phosphorylation and reduced endothelial agonist-induced intracellular calcium release as well as extracellular calcium influx. We conclude that iPLA(2)beta is an important mediator of vascular relaxation and intracellular calcium homeostasis in both smooth muscle and endothelial cells and that ablation of iPLA(2)beta causes agonist-induced smooth muscle hypercontractility and reduced agonist-induced endothelial dilation.
Collapse
Affiliation(s)
- Hans H Dietrich
- Washington Univ. School of Medicine, Dept. of Neurosurgery, 660 South Euclid Ave., Campus Box 8057, St. Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Finocchietto PV, Franco MC, Holod S, Gonzalez AS, Converso DP, Antico Arciuch VG, Serra MP, Poderoso JJ, Carreras MC. Mitochondrial nitric oxide synthase: a masterpiece of metabolic adaptation, cell growth, transformation, and death. Exp Biol Med (Maywood) 2009; 234:1020-8. [PMID: 19546350 DOI: 10.3181/0902-mr-81] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mitochondria are specialized organelles that control energy metabolism and also activate a multiplicity of pathways that modulate cell proliferation and mitochondrial biogenesis or, conversely, promote cell arrest and programmed cell death by a limited number of oxidative or nitrative reactions. Nitric oxide (NO) regulates oxygen uptake by reversible inhibition of cytochrome oxidase and the production of superoxide anion from the mitochondrial electron transfer chain. In this sense, NO produced by mtNOS will set the oxygen uptake level and contribute to oxidation-reduction reaction (redox)-dependent cell signaling. Modulation of translocation and activation of neuronal nitric oxide synthase (mtNOS activity) under different physiologic or pathologic conditions represents an adaptive response properly modulated to adjust mitochondria to different cell challenges.
Collapse
Affiliation(s)
- Paola V Finocchietto
- Laboratory of Oxygen Metabolism, University Hospital, 1120 Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Dedkova EN, Blatter LA. Characteristics and function of cardiac mitochondrial nitric oxide synthase. J Physiol 2009; 587:851-72. [PMID: 19103678 PMCID: PMC2669975 DOI: 10.1113/jphysiol.2008.165423] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2008] [Accepted: 12/15/2008] [Indexed: 12/22/2022] Open
Abstract
We used laser scanning confocal microscopy in combination with the nitric oxide (NO)-sensitive fluorescent dye DAF-2 and the reactive oxygen species (ROS)-sensitive dyes CM-H(2)DCF and MitoSOX Red to characterize NO and ROS production by mitochondrial NO synthase (mtNOS) in permeabilized cat ventricular myocytes. Stimulation of mitochondrial Ca(2+) uptake by exposure to different cytoplasmic Ca(2+) concentrations ([Ca(2+)](i) = 1, 2 and 5 microm) resulted in a dose-dependent increase of NO production by mitochondria when L-arginine, a substrate for mtNOS, was present. Collapsing the mitochondrial membrane potential with the protonophore FCCP or blocking the mitochondrial Ca(2+) uniporter with Ru360 as well as blocking the respiratory chain with rotenone or antimycin A in combination with oligomycin inhibited mitochondrial NO production. In the absence of L-arginine, mitochondrial NO production during stimulation of Ca(2+) uptake was significantly decreased, but accompanied by increase in mitochondrial ROS production. Inhibition of mitochondrial arginase to limit L-arginine availability resulted in 50% inhibition of Ca(2+)-induced ROS production. Both mitochondrial NO and ROS production were blocked by the nNOS inhibitor (4S)-N-(4-amino-5[aminoethyl]aminopentyl)-N'-nitroguanidine and the calmodulin antagonist W-7, while the eNOS inhibitor L-N(5)-(1-iminoethyl)ornithine (L-NIO) or iNOS inhibitor N-(3-aminomethyl)benzylacetamidine, 2HCl (1400W) had no effect. The superoxide dismutase mimetic and peroxynitrite scavenger MnTBAP abolished Ca(2+)-induced ROS generation and increased NO production threefold, suggesting that in the absence of MnTBAP either formation of superoxide radicals suppressed NO production or part of the formed NO was transformed quickly to peroxynitrite. In the absence of L-arginine, mitochondrial Ca(2+) uptake induced opening of the mitochondrial permeability transition pore (PTP), which was blocked by the PTP inhibitor cyclosporin A and MnTBAP, and reversed by L-arginine supplementation. In the presence of the mtNOS cofactor (6R)-5,6,7,8,-tetrahydrobiopterin (BH(4); 100 microm) mitochondrial ROS generation and PTP opening decreased while mitochondrial NO generation slightly increased. These data demonstrate that mitochondrial Ca(2+) uptake activates mtNOS and leads to NO-mediated protection against opening of the mitochondrial PTP, provided sufficient availability of l-arginine and BH(4). In conclusion, our data show the importance of L-arginine and BH(4) for cardioprotection via regulation of mitochondrial oxidative stress and modulation of PTP opening by mtNOS.
Collapse
Affiliation(s)
- Elena N Dedkova
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, IL 60612, USA
| | | |
Collapse
|
43
|
Abstract
Myocardial phospholipids serve as primary reservoirs of arachidonic acid (AA), which is liberated through the rate-determining hydrolytic action of cardiac phospholipases A2 (PLA2s). A predominant PLA2 in myocardium is calcium-independent phospholipase A2beta (iPLA2beta), which, through its calmodulin (CaM) and ATP-binding domains, is regulated by alterations in local cellular Ca2+ concentrations and cardiac bioenergetic status, respectively. Importantly, iPLA2beta has been demonstrated to be activated by ischaemia through elevation of the concentration of myocardial fatty acyl-CoA, which abrogates Ca2+/CaM-mediated inhibition of iPLA2beta. AA released by PLA2-catalysed hydrolysis of phospholipids serves as a precursor for eicosanoids generated by pathways dependent on cyclooxygenases (COX), lipoxygenases (LOX), and cytochromes P450 (CYP). Eicosanoids initiate and propagate diverse signalling cascades, primarily through their interaction with cellular receptors and ion channels. However, during pathologic states such as ischaemia or congestive heart failure, eicosanoids contribute to multiple maladaptive changes including inflammation, alterations of cellular growth programmes, and activation of multiple transcriptional events leading to the deleterious sequelae of these pathologic states. This review summarizes the central roles of myocardial PLA(2)s in eicosanoid signalling in the heart, the major COX, LOX, and CYP pathways of eicosanoid generation in the myocardium, and the effects of important eicosanoids on receptor-, ion channel-, and transcription-mediated processes that facilitate cardiac hypertrophy, mediate ischaemic preconditioning, and precipitate arrhythmogenesis in response to pathologic stimuli.
Collapse
Affiliation(s)
- Christopher M Jenkins
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8020, St Louis, MO 63110, USA
| | | | | |
Collapse
|
44
|
mAtNOS1 induces apoptosis of human mammary adenocarcinoma cells. Life Sci 2008; 82:1077-82. [DOI: 10.1016/j.lfs.2008.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 03/05/2008] [Accepted: 03/17/2008] [Indexed: 12/21/2022]
|
45
|
Parihar MS, Parihar A, Chen Z, Nazarewicz R, Ghafourifar P. mAtNOS1 regulates mitochondrial functions and apoptosis of human neuroblastoma cells. Biochim Biophys Acta Gen Subj 2008; 1780:921-6. [PMID: 18359297 DOI: 10.1016/j.bbagen.2008.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2008] [Revised: 02/19/2008] [Accepted: 02/19/2008] [Indexed: 01/11/2023]
Abstract
mAtNOS1 is a novel gene recently reported in mammalian cells with functions that are not fully understood. The present study generated human neuroblastoma SHSY cells over- and underexpressing mAtNOS1 and shows that mAtNOS1 is involved in regulating mitochondrial nitric oxide, mitochondrial transmembrane potential, protein tyrosine nitration, cytochrome c release, and apoptosis of those cells.
Collapse
Affiliation(s)
- Mordhwaj S Parihar
- Department of Surgery, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | |
Collapse
|
46
|
Parihar MS, Parihar A, Villamena FA, Vaccaro PS, Ghafourifar P. Inactivation of mitochondrial respiratory chain complex I leads mitochondrial nitric oxide synthase to become pro-oxidative. Biochem Biophys Res Commun 2008; 367:761-7. [DOI: 10.1016/j.bbrc.2008.01.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Accepted: 01/04/2008] [Indexed: 11/29/2022]
|
47
|
Ghafourifar P, Parihar MS, Nazarewicz R, Zenebe WJ, Parihar A. Detection assays for determination of mitochondrial nitric oxide synthase activity; advantages and limitations. Methods Enzymol 2008; 440:317-34. [PMID: 18423228 DOI: 10.1016/s0076-6879(07)00821-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nitric oxide (NO) is a reactive radical synthesized by members of the NO synthase (NOS) family, including mitochondrial-specific NOS (mtNOS). Some of the assays used for the determination of cytoplasmic NOS activity have been utilized to detect mtNOS activity. However, it seems that many of those assays need to be adjusted and optimized to detect NO in the unique environment of mitochondria. Additionally, most mtNOS detection assays are designed and optimized for isolated mitochondria and may exert inherent pitfalls and limitations once used in living cells. This chapter describes several assays used commonly for mtNOS detection in isolated mitochondria and in mitochondria of live cells. Those include colorimetric and spectrophotometric methods, Griess reaction, radioassay, and polarographic and chemiluminescence assays. It also describes fluorescent-based assays for the detection of mitochondrial NO in live cells. Advantages and limitations of each assay are discussed.
Collapse
Affiliation(s)
- Pedram Ghafourifar
- Department of Surgery, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | | | | | | |
Collapse
|
48
|
Parihar MS, Nazarewicz RR, Kincaid E, Bringold U, Ghafourifar P. Association of mitochondrial nitric oxide synthase activity with respiratory chain complex I. Biochem Biophys Res Commun 2007; 366:23-8. [PMID: 18036554 DOI: 10.1016/j.bbrc.2007.11.056] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Accepted: 11/11/2007] [Indexed: 11/28/2022]
Abstract
The present study shows that rat liver and brain mitochondrial nitric oxide synthase (mtNOS) are functionally associated with mitochondrial respiratory chain complex I. When complex I is activated, mtNOS exerts high activity and generates nitric oxide, whereas inactivation of complex I leads mtNOS to abandon its NOS activity. Functional association of mtNOS with complex I is potentially important in regulating mtNOS activity and mitochondrial functions.
Collapse
Affiliation(s)
- Mordhwaj S Parihar
- Department of Surgery, Davis Heart and Lung Research Institute, and Institute of Mitochondrial Biology, The Ohio State University, Columbus, OH, USA
| | | | | | | | | |
Collapse
|