1
|
Ma Y, Dong T, Luan F, Yang J, Miao F, Wei P. Interaction of major facilitator superfamily domain containing 2A with the blood-brain barrier. Neural Regen Res 2025; 20:2133-2152. [PMID: 39248155 DOI: 10.4103/nrr.nrr-d-24-00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/08/2024] [Indexed: 09/10/2024] Open
Abstract
The functional and structural integrity of the blood-brain barrier is crucial in maintaining homeostasis in the brain microenvironment; however, the molecular mechanisms underlying the formation and function of the blood-brain barrier remain poorly understood. The major facilitator superfamily domain containing 2A has been identified as a key regulator of blood-brain barrier function. It plays a critical role in promoting and maintaining the formation and functional stability of the blood-brain barrier, in addition to the transport of lipids, such as docosahexaenoic acid, across the blood-brain barrier. Furthermore, an increasing number of studies have suggested that major facilitator superfamily domain containing 2A is involved in the molecular mechanisms of blood-brain barrier dysfunction in a variety of neurological diseases; however, little is known regarding the mechanisms by which major facilitator superfamily domain containing 2A affects the blood-brain barrier. This paper provides a comprehensive and systematic review of the close relationship between major facilitator superfamily domain containing 2A proteins and the blood-brain barrier, including their basic structures and functions, cross-linking between major facilitator superfamily domain containing 2A and the blood-brain barrier, and the in-depth studies on lipid transport and the regulation of blood-brain barrier permeability. This comprehensive systematic review contributes to an in-depth understanding of the important role of major facilitator superfamily domain containing 2A proteins in maintaining the structure and function of the blood-brain barrier and the research progress to date. This will not only help to elucidate the pathogenesis of neurological diseases, improve the accuracy of laboratory diagnosis, and optimize clinical treatment strategies, but it may also play an important role in prognostic monitoring. In addition, the effects of major facilitator superfamily domain containing 2A on blood-brain barrier leakage in various diseases and the research progress on cross-blood-brain barrier drug delivery are summarized. This review may contribute to the development of new approaches for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yilun Ma
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Taiwei Dong
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Fei Luan
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Juanjuan Yang
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi'an, Shaanxi Province, China
| | - Feng Miao
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Peifeng Wei
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi'an, Shaanxi Province, China
| |
Collapse
|
2
|
Massaro M, Quarta S, Calabriso N, Carluccio MA, Scoditti E, Mancuso P, De Caterina R, Madonna R. Omega-3 polyunsaturated fatty acids and pulmonary arterial hypertension: Insights and perspectives. Eur J Clin Invest 2024; 54:e14277. [PMID: 38940236 PMCID: PMC11490397 DOI: 10.1111/eci.14277] [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: 04/27/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and progressive disorder that affects the pulmonary vasculature. Although recent developments in pharmacotherapy have extended the life expectancy of PAH patients, their 5-year survival remains unacceptably low, underscoring the need for multitarget and more comprehensive approaches to managing the disease. This should incorporate not only medical, but also lifestyle interventions, including dietary changes and the use of nutraceutical support. Among these strategies, n-3 polyunsaturated fatty acids (n-3 PUFAs) are emerging as promising agents able to counteract the inflammatory component of PAH. In this narrative review, we aim at analysing the preclinical evidence for the impact of n-3 PUFAs on the pathogenesis and the course of PAH. Although evidence for the role of n-3 PUFAs deficiencies in the development and progression of PAH in humans is limited, preclinical studies suggest that these dietary components may influence several aspects of the pathobiology of PAH. Further clinical research should test the efficacy of n-3 PUFAs on top of approved clinical management. These studies will provide evidence on whether n-3 PUFAs can genuinely serve as a valuable tool to enhance the efficacy of pharmacotherapy in the treatment of PAH.
Collapse
Affiliation(s)
- Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
| | - Stefano Quarta
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
| | - Nadia Calabriso
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
| | | | - Egeria Scoditti
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
| | - Peter Mancuso
- Department of Nutritional Sciences and the Program in Immunology, School of Public Health, University of Michigan, 1415 Washington Hts., Ann Arbor, Michigan 481009
| | | | | |
Collapse
|
3
|
Liu L, Xu J, Huang X, Wang Y, Ma X, Wang X, Liu Y, Ren X, Li J, Wang Y, Zhou S, Yuan L. DHA dietary intervention caused different hippocampal lipid and protein profile in ApoE-/- and C57BL/6J mice. Biomed Pharmacother 2024; 177:117088. [PMID: 38971007 DOI: 10.1016/j.biopha.2024.117088] [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: 05/26/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Changes in protein and lipid levels may occur in the Alzheimer's disease brain, and DHA can have beneficial effects on it. To investigate the impact of DHA dietary intervention on brain protein and lipid profile in ApoE-/- mice and C57 mice. METHOD Three-month-old ApoE-/- mice and C57 mice were randomly divided into two groups respectively, and fed with control diet and DHA-fortified diet for five months. Cortical TC, HDL-C and LDL-C levels and cholesterol metabolism-related protein expression were measured by ELISA or immunohistochemistry methods. Hippocampus were collected for proteomic and lipidomics analysis by LC-MS/MS and differential proteins and lipid metabolites were screened and further analyzed by GO functional annotation and KEGG pathway enrichment analysis. RESULTS DHA intervention decreased cortical TC level in both C57 and ApoE-/- mice (P < 0.05), but caused different change of cortical HDL-C, LDL-C level and LDL-C/HDL-C ratio in C57 and ApoE-/- mice (P < 0.05). Discrepant cortical and hippocampal LDLR, ABCG1, Lox1 and SORT1 protein expression was found between C57 and ApoE-/- mice (P < 0.05), and DHA treatment caused different changes of these proteins in C57 and ApoE-/- mice (P < 0.05). Differential hippocampal proteins and lipids profile were found in C57 and ApoE-/- mice before and after DHA treatment, which were mainly involved in vesicular transport and phospholipid metabolic pathways. CONCLUSION ApoE genetic defect caused abnormal cholesterol metabolism, and affected protein and lipid profile, as well as discrepant response of hippocampal protein and lipids profile in the brain of mice given DHA fortified diet intervention.
Collapse
Affiliation(s)
- Lu Liu
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases
| | - Jingjing Xu
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases
| | - Xiaochen Huang
- School of Public Health, Capital Medical University, Beijing, China
| | - Ying Wang
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Xiaojun Ma
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases
| | - Xixiang Wang
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases
| | - Yu Liu
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases
| | - Xiuwen Ren
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases
| | - Jiahao Li
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases
| | - Yueyong Wang
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases
| | - Shaobo Zhou
- School of Science, Faculty of Engineering and Science, University of Greenwich, Central Avenue, Chatham ME4 4TB, UK.
| | - Linhong Yuan
- School of Public Health, Capital Medical University, Beijing, China; China-British Joint Laboratory of Nutrition Prevention and Control of Chronic Diseases.
| |
Collapse
|
4
|
He S, Mu M, Zhang M, Su X, Jiang Z. Edible CO 2-Responsive Wormlike Micelles with Docosahexaenoic Acid. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:15013-15022. [PMID: 38991116 DOI: 10.1021/acs.langmuir.4c01314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Due to their unique microstructure and modifiable rheological properties, wormlike micelles that respond to environmental stimulation have garnered significant interest in recent years. Among them, CO2-responsive wormlike micelles have the advantages of simple preparation and controllable properties, which have significant development potential in the food chemistry field of thickeners. In this study, CO2-responsive wormlike micelles were prepared using docosahexaenoic acid (DHA), pyridoxamine (PA), and glucosamine (GA); the stimulus-responsive behaviors and mechanisms of the two systems, namely, NaDHA/PA and NaDHA/GA, were investigated using dynamic light scattering (DLS) and cryo-transmission electron microscopy (Cryo-TEM). The nearly unaltered viscosity of the systems confirmed the cyclic reversibility of the CO2 response of the two systems when the two mixed solutions were converted back to aqueous liquids 10 times. The preparation and properties of DHA-based CO2-responsive wormlike micelles are expected to advance fundamental research and establish the theoretical groundwork for their practical application in controllable thickening agents in food chemistry.
Collapse
Affiliation(s)
- Shuai He
- School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, China
- Engineering Laboratory of High Performance Waterborne Polymer Materials of Anhui Province, Anhui University, Hefei 230601, China
| | - Meng Mu
- Postdoctoral Scientific Research Working Station of Shengli Oilfield, SINOPEC, Dongying 257000, China
- Shengli Oilfield Company, SINOPEC, Dongying 257092, China
| | - Mingmin Zhang
- Zhejiang Research Institute of Tianjin University, Shaoxing 312000, China
| | - Xin Su
- Engineering Laboratory of High Performance Waterborne Polymer Materials of Anhui Province, Anhui University, Hefei 230601, China
- Polymer Research Institute, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zequan Jiang
- Shaoxing Shangyu District Cao'e Jiang S&T Innovation Corridor Service Center, Shaoxing 312000, China
- Shaoxing Shangyu District S&T Entrepreneurship Service Center, Shaoxing 312000, China
| |
Collapse
|
5
|
Lyudinina AY, Parshukova OI, Bojko ER. n-3 Polyunsaturated Fatty Acids Are Associated with Stable Nitric Oxide Metabolites in Highly Trained Athletes. Cells 2024; 13:1110. [PMID: 38994963 PMCID: PMC11240318 DOI: 10.3390/cells13131110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND The aim of this study was to investigate the relationships between levels of n-3 essential polyunsaturated fatty acids (n-3 PUFAs) and stable nitric oxide (NO) metabolites in the plasma of athletes. METHODS Highly trained cross-country skiers (males, n = 39) were examined. The fatty acid profile of the total plasma lipids was determined by gas chromatography. The plasma NO level was studied by a colorimetric method via reaction with Griess reagent. RESULTS A widespread deficiency of essential n-3 PUFAs in the plasma of athletes (more than 80% of the subjects) was demonstrated in association with an imbalance in the levels of nitrates (NO3) and nitrites (NO2). A lower value of n-3 linolenic acid in the plasma (0.21 mol/%) was associated with a NO3 level below the normal range (n-3 C18:3 and NO3 Rs = 0.461; p = 0.003). Higher levels of n-3 eicosapentaenoic acid (0.8 mol/%) were associated with a concentration of NO2 above the normal value (n-3 C20:5 and NO2 Rs = 0.449; p = 0.004). CONCLUSION For the first time, the participation of essential n-3 PUFAs in the nitrite-nitrate pathway of NO synthesis in highly trained skiers was demonstrated.
Collapse
Affiliation(s)
- Aleksandra Y Lyudinina
- Institute of Physiology, Komi Science Centre, Ural Branch of the Russian Academy of Sciences, FRC Komi SC UB RAS, 50 Pervomayskaya Str., 167982 Syktyvkar, Russia
| | - Olga I Parshukova
- Institute of Physiology, Komi Science Centre, Ural Branch of the Russian Academy of Sciences, FRC Komi SC UB RAS, 50 Pervomayskaya Str., 167982 Syktyvkar, Russia
| | - Evgeny R Bojko
- Institute of Physiology, Komi Science Centre, Ural Branch of the Russian Academy of Sciences, FRC Komi SC UB RAS, 50 Pervomayskaya Str., 167982 Syktyvkar, Russia
| |
Collapse
|
6
|
Ahiawodzi P, Buzkova P, Lichtenstein A, Matthan N, Ix J, Kizer J, Tracy R, Arnold A, Newman A, Siscovick D, Djousse L, Mukamal K. The Associations of Individual and Subclasses of Nonesterified Fatty Acids With Disability, and Mobility Limitation in Older Adults: The Cardiovascular Health Study. J Gerontol A Biol Sci Med Sci 2023; 78:1155-1163. [PMID: 36156076 PMCID: PMC10329219 DOI: 10.1093/gerona/glac206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND We sought to determine the associations between individual nonesterified fatty acids (NEFAs) and disability and mobility limitation. METHODS We studied 1 734 participants in the Cardiovascular Health Study (CHS), an ongoing population-based cohort study of community-living older American adults. We measured 35 individual NEFA species in fasting serum samples obtained at the 1996-1997 clinic visit. Using yearly assessments of activities of daily living and self-reported mobility, we identified participants with incident disability or mobility limitation during 15 years of follow-up. Cox proportional hazards regression models were used to determine the associations between per SD increment in the individual NEFAs and incident disability and mobility limitations with adjustment for potential confounding factors. RESULTS Higher concentrations of total and a broad range of individual NEFA species were associated with risk of disability and mobility limitation (disability: HR per SD of total NEFA [SD = 174.70] = 1.11, 95% CI = 1.04-1.18, p = .001; mobility limitation: HR per SD of total NEFA = 1.09, 95% CI = 1.02-1.16, p = .01). Among individual saturated NEFAs (SFAs), myristic (14:0) and palmitic (16:0) acids were significantly associated with higher risk of both disability and mobility limitations, but longer-chain FAs were not. Most individual monounsaturated (MUFA), n-6 polyunsaturated fatty acids (PUFAs), and trans FAs were positively significantly associated with higher risks of both disability and mobility limitation. In contrast, most n-3 PUFA species were not associated with disability or mobility limitation. CONCLUSIONS Higher risks of disability and mobility limitation were observed for proinflammatory intermediate-chain SFAs, MUFAs, n-6 PUFAs, and trans FAs. Our findings indicated no significant association for anti-inflammatory n-3 PUFAs.
Collapse
Affiliation(s)
- Peter D Ahiawodzi
- Department of Public Health, Campbell University College of Pharmacy and Health Sciences, Buies Creek, North Carolina, USA
| | - Petra Buzkova
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Nirupa R Matthan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Joachim H Ix
- Divisions of Nephrology-Hypertension, University of California, San Diego, La Jolla, California, USA
| | - Jorge R Kizer
- Cardiology Section, San Francisco VA Health Care System, and Department of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California,USA
| | - Russell P Tracy
- Department of Pathology and Biochemistry, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Alice Arnold
- Department of a Biostatistics, University of Washington, Seattle, Washington, USA
| | - Anne B Newman
- Departments of Epidemiology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David Siscovick
- Division of Research, Evaluation and Policy, The New York Academy of Medicine, New York, New York, USA
| | - Luc Djousse
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kenneth J Mukamal
- Division of General Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Huang S, Taylor CG, Zahradka P. Growth State-Dependent Activation of eNOS in Response to DHA: Involvement of p38 MAPK. Int J Mol Sci 2023; 24:ijms24098346. [PMID: 37176054 PMCID: PMC10179717 DOI: 10.3390/ijms24098346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/19/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Our laboratory previously reported that docosahexaenoic acid (DHA) differentially activates p38 mitogen-activated protein kinase (MAPK) in growing and quiescent human endothelial cells, which represent the dysfunctional and healthy states in vivo, respectively. Since endothelial nitric oxide synthase (eNOS) activity differs between healthy and dysfunctional endothelial cells, and p38 MAPK reportedly regulates both the activity and expression of eNOS, we hypothesized that the beneficial actions of DHA on endothelial cells are due to eNOS activation by p38 MAPK. The contribution of mitogen- and stress-activated protein kinase (MSK), a p38 MAPK substrate, was also investigated. Growing and quiescent EA.hy926 cells, prepared on Matrigel®-coated plates, were incubated with inhibitors of p38MAPK or MSK before adding DHA. eNOS phosphorylation and levels were quantified by Western blotting. Treatment with 20 µM DHA activated eNOS in both growth states whereas 125 µM DHA suppressed eNOS activation in growing cells. Quiescent cells had higher basal levels of eNOS than growing cells, while 125 µM DHA decreased eNOS levels in both growth states. p38 MAPK inhibition enhanced eNOS activation in quiescent cells but suppressed it in growing cells. Interestingly, 125 µM DHA counteracted these effects of p38 MAPK inhibition in both growth states. MSK was required for eNOS activation in both growth states, but it only mediated eNOS activation by DHA in quiescent cells. MSK thus affects eNOS via a pathway independent of p38MAPK. Quiescent cells were also more resistant to the apoptosis-inducing effect of 125 µM DHA compared to growing cells. The growth state-dependent regulation of p38MAPK and eNOS by DHA provides novel insight into the molecular mechanisms by which DHA influences endothelial cell function.
Collapse
Affiliation(s)
- Shiqi Huang
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Carla G Taylor
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Peter Zahradka
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| |
Collapse
|
8
|
Morita S, Sasaki H, Kaneda Y, Rogi T, Izumo T, Nakai M. Effects of Combining Docosahexaenoic Acid and Eicosapentaenoic Acid with Sesame Lignan on Vascular Endothelial Function. J Nutr Sci Vitaminol (Tokyo) 2023; 69:370-376. [PMID: 37940577 DOI: 10.3177/jnsv.69.370] [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] [Indexed: 11/10/2023]
Abstract
Vascular endothelial cells produce vasoactive substances, such as nitric oxide (NO), to regulate vascular relaxation and contraction. Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) enhance NO production in endothelial cells, and sesamin, a sesame lignan contained in sesame seeds, also promotes NO production. This study examined DHA, EPA, and sesamin's combined effects since it was expected that combining them would further enhance NO production in endothelial cells. Using a human umbilical vein endothelial cell (HUVEC), the NO amount secreted in the culture supernatant was analyzed. Sesamin metabolite (SC1) was used in the experiments because it is a major metabolite in human blood after sesamin absorption. When cells were treated with DHA or EPA alone, they increased NO production in a concentration-dependent manner, whereas no change in NO production was observed for SC1. NO production increased when DHA and EPA were treated in combination with SC1, although the low DHA and EPA concentrations showed no difference in NO production. In the concentrations in which the combined effect was observed, SC1 activated eNOS via calcium signaling, whereas DHA and EPA activated eNOS via alterations in the membrane lipid environment. The combined effect of the two pathways was considered to have enhanced the eNOS activity. These results suggested that combining DHA, EPA, and sesamin might improve vascular endothelial function.
Collapse
Affiliation(s)
| | | | | | - Tomohiro Rogi
- Institute for Health Care Science, Suntory Wellness Ltd
| | | | - Masaaki Nakai
- Institute for Health Care Science, Suntory Wellness Ltd
| |
Collapse
|
9
|
Candido VB, Perego SM, Ceroni A, Metzger M, Colquhoun A, Michelini LC. Trained hypertensive rats exhibit decreased transcellular vesicle trafficking, increased tight junctions' density, restored blood-brain barrier permeability and normalized autonomic control of the circulation. Front Physiol 2023; 14:1069485. [PMID: 36909225 PMCID: PMC9997677 DOI: 10.3389/fphys.2023.1069485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction: Chronic hypertension is accompanied by either blood-brain barrier (BBB) leakage and autonomic dysfunction. There is no consensus on the mechanism determining increased BBB permeability within autonomic areas. While some reports suggested tight junction's breakdown, others indicated the involvement of transcytosis rather than paracellular transport changes. Interestingly, exercise training was able to restore both BBB permeability and autonomic control of the circulation. We sought now to clarify the mechanism(s) governing hypertension- and exercise-induced BBB permeability. Methods: Spontaneously hypertensive rats (SHR) and normotensive controls submitted to 4-week aerobic training (T) or sedentary protocol (S) were chronically cannulated for baseline hemodynamic and autonomic recordings and evaluation of BBB permeability. Brains were harvested for measurement of BBB function (FITC-10 kDa leakage), ultrastructural analysis of BBB constituents (transmission electron microscopy) and caveolin-1 expression (immunofluorescence). Results: In SHR-S the increased pressure, augmented sympathetic vasomotor activity, higher sympathetic and lower parasympathetic modulation of the heart and the reduced baroreflex sensitivity were accompanied by robust FITC-10kDa leakage, large increase in transcytotic vesicles number/capillary, but no change in tight junctions' density within the paraventricular nucleus of the hypothalamus, the nucleus of the solitary tract and the rostral ventrolateral medulla. SHR-T exhibited restored BBB permeability and normalized vesicles counting/capillary simultaneously with a normal autonomic modulation of heart and vessels, resting bradycardia and partial pressure reduction. Caveolin-1 expression ratified the counting of transcellular, not other cytoplasmatic vesicles. Additionally, T caused in both groups significant increases in tight junctions' extension/capillary border. Discussion: Data indicate that transcytosis, not the paracellular transport, is the primary mechanism underlying both hypertension- and exercise-induced BBB permeability changes within autonomic areas. The reduced BBB permeability contributes to normalize the autonomic control of the circulation, which suppresses pressure variability and reduces the occurrence of end-organ damage in the trained SHR. Data also disclose that hypertension does not change but exercise training strengthens the resistance of the paracellular pathway in both strains.
Collapse
Affiliation(s)
| | - Sany M Perego
- Department of Physiology and Biophysics, São Paulo, Brazil
| | | | - Martin Metzger
- Department of Physiology and Biophysics, São Paulo, Brazil
| | - Alison Colquhoun
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, São Paulo, Brazil
| | | |
Collapse
|
10
|
Kotlyarov S, Kotlyarova A. The Importance of the Plasma Membrane in Atherogenesis. MEMBRANES 2022; 12:1036. [PMID: 36363591 PMCID: PMC9698587 DOI: 10.3390/membranes12111036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Atherosclerotic cardiovascular diseases are an important medical problem due to their high prevalence, impact on quality of life and prognosis. The pathogenesis of atherosclerosis is an urgent medical and social problem, the solution of which may improve the quality of diagnosis and treatment of patients. Atherosclerosis is a complex chain of events, which proceeds over many years and in which many cells in the bloodstream and the vascular wall are involved. A growing body of evidence suggests that there are complex, closely linked molecular mechanisms that occur in the plasma membranes of cells involved in atherogenesis. Lipid transport, innate immune system receptor function, and hemodynamic regulation are linked to plasma membranes and their biophysical properties. A better understanding of these interrelationships will improve diagnostic quality and treatment efficacy.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
11
|
Kotlyarov S, Kotlyarova A. Clinical significance of polyunsaturated fatty acids in the prevention of cardiovascular diseases. Front Nutr 2022; 9:998291. [PMID: 36276836 PMCID: PMC9582942 DOI: 10.3389/fnut.2022.998291] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular diseases are one of the most important problems of modern medicine. They are associated with a large number of health care visits, hospitalizations and mortality. Prevention of atherosclerosis is one of the most effective strategies and should start as early as possible. Correction of lipid metabolism disorders is associated with definite clinical successes, both in primary prevention and in the prevention of complications of many cardiovascular diseases. A growing body of evidence suggests a multifaceted role for polyunsaturated fatty acids. They demonstrate a variety of functions in inflammation, both participating directly in a number of cellular processes and acting as a precursor for subsequent biosynthesis of lipid mediators. Extensive clinical data also support the importance of polyunsaturated fatty acids, but all questions have not been answered to date, indicating the need for further research.
Collapse
Affiliation(s)
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, Ryazan, Russia
| |
Collapse
|
12
|
Abstract
Several studies have reported a significant association between the metabolic syndrome (MetS) and mortality around the world. Caveolin-1 (CAV-1) has been widely studied in dyslipidaemia, and several studies have indicated that CAV-1 genetic variations may correlate with dietary intake of fatty acids. This study aimed to investigate the interaction of CAV-1 rs3807992 with types of dietary fatty acid in the MetS risk. This cross-sectional study was carried out on 404 overweight and obese females. Dietary intake was obtained from a 147-item FFQ. The CAV-1 genotype was measured using the PCR-restriction fragment length polymorphism method. Anthropometric values and serum levels (TC, LDL, HDL, TAG and FBS) were measured by standard methods. It was observed that the (AA + AG) group had significantly higher BMI, waist circumference and DBP (P = 0·02, P = 0·02, and P = 0·01, respectively) and lower serum LDL, HDL and TC (P < 0·05) than the GG group. It was found that A allele carriers were at higher odds of the MetS (P = 0·01), abdominal obesity (P = 0·06), increased TAG concentration (P = 0·01), elevated blood pressure (BP) (P = 0·01), increased glucose concentration (P = 0·45) and decreased HDL-cholesterol concentration (P = 0·03). Moreover, the interaction of CAV-1 and SFA intake was significant in terms of the MetS (P = 0·03), LDL (P = 0·03) and BP (P = 0·01). Additionally, the (AA + AG) group was significantly related to PUFA intake in terms of the MetS (P = 0·04), TAG (P = 0·02), glucose (P = 0·02) and homoeostasis model assessment insulin resistance (P = 0·01). Higher PUFA consumption might attenuate the CAV-1 rs3807992 associations with the MetS, and individuals with greater genetic predisposition appeared to have a higher risk of the MetS, associated with higher SFA consumption.
Collapse
|
13
|
Parnova RG. Critical Role of Endothelial Lysophosphatidylcholine Transporter Mfsd2a in Maintaining Blood–Brain Barrier Integrity and Delivering Omega 3 PUFA to the Brain. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022030103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
14
|
Wang J, Xu J, Zang G, Zhang T, Wu Q, Zhang H, Chen Y, Wang Y, Qin W, Zhao S, Qin E, Qiu J, Zhang X, Wen L, Wang Y, Wang G. trans-2-Enoyl-CoA Reductase Tecr-Driven Lipid Metabolism in Endothelial Cells Protects against Transcytosis to Maintain Blood-Brain Barrier Homeostasis. RESEARCH 2022; 2022:9839368. [PMID: 35465346 PMCID: PMC9006154 DOI: 10.34133/2022/9839368] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/02/2022] [Indexed: 12/29/2022]
Abstract
The transport and metabolism of lipids in cerebrovascular endothelial cells (ECs) have been hypothesized to regulate blood-brain barrier (BBB) maturation and homeostasis. Long-chain polyunsaturated fatty acids (LCPUFAs) as the important lipids components of cell membranes are essential for the development and function of BBB, but the direct links of lipid metabolism and ECs barrier function remain to be established. Here, we comprehensively characterize the transcriptomic phenotype of developmental cerebrovascular ECs in single-cell resolution and firstly find that trans-2-enoyl-CoA reductase (Tecr), a very-long-chain fatty acid synthesis, is highly expressed during barriergenesis and decreased after BBB maturation. EC-specific knockout of Tecr compromises angiogenesis due to delayed vascular sprouting. Importantly, EC-specific deletion of Tecr loss restrictive quality of vascular permeability from neonatal stages to adulthood, with high levels of transcytosis, but maintains the vascular tight junctions. Moreover, lipidomic analysis shows that the expression of Tecr in ECs is associated with the containing of omega-3 fatty acids, which directly suppresses caveolae vesicles formation. These results reveal a protective role for Tecr in BBB integrity and suggest that Tecr as a novel therapeutic target in the central nervous system (CNS) diseases associated with BBB dysfunction.
Collapse
Affiliation(s)
- Jinxuan Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Jianxiong Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guangchao Zang
- Institute of Life Science, Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China
| | - Tao Zhang
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Qi Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Hongping Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yidan Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Weixi Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Shuang Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Erdai Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Lin Wen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| |
Collapse
|
15
|
Wheeler E, Walsh-Wilcox M, Shah M, Achrekar A, Anderson JR, Walker MK. Interactive Effects of Omega-3 Polyunsaturated Fatty Acids and Secondhand Smoke in Mice and Human Subjects. Cardiovasc Toxicol 2021; 21:115-126. [PMID: 32844369 PMCID: PMC7854812 DOI: 10.1007/s12012-020-09601-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/14/2020] [Indexed: 10/23/2022]
Abstract
Active smoking and secondhand smoke (SHS) exposure increase the risk of cardiovascular morbidity and mortality. Active smoking is associated with reduced levels of omega-3 polyunsaturated fatty acids (n-3 PUFA) and studies show that n-3 PUFA supplementation can improve smoking-induced vascular dysfunction. However, the relationship between n-3 PUFA and SHS exposure has not been studied. Fat-1 transgenic mice, which convert n-6 to n-3 PUFA, were fed diets with n-3 PUFA or without (n-6 PUFA diet), exposed to air or SHS for 4 weeks, and vasoreactivity, antioxidant indices, and omega-3 index (percent eicosapentaenoic + docosahexaenoic acids in RBC) measured. Compared to air-exposed mice, SHS-enhanced aortic constriction in mice fed the n-6 PUFA diet (omega-3 index, 5.9 ± 0.2%; mean ± SE), but not in mice fed the n-3 PUFA diet (omega-3 index, 7.8 ± 0.6%). SHS also significantly induced mRNA expression of cytochrome P4501A1, NADPH:quinone oxidoreductase, heme oxygenase-1, and angiotensinogen in adipose tissue, and increased antioxidant capacity only in mice on the n-6 PUFA diet. Notably, SHS reduced the omega-3 index by 1.0 percentage point (p = 0.003), compared to air-exposed mice irrespective of diet. Additionally, we recruited human nonsmokers (NS) with and without SHS exposure (n = 40) 19-40 years old and measured the omega-3 index and antioxidant capacity. In human subjects SHS exposure was associated with a significantly lower omega-3 index (NS, 4.4 ± 1.1%; NS + SHS, 3.2 ± 1.0%; mean ± SD, p = 0.002) and higher antioxidant capacity (p < 0.001) than unexposed NS. Thus, SHS exposure is associated with lower levels of n-3 PUFA in mice and humans; however, an omega-3 index of ~ 8% in mice has vasoprotective and antioxidant properties.
Collapse
Affiliation(s)
- Emily Wheeler
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, 2703 Frontier Ave NE MSC09 5630, Albuquerque, NM, 87131, USA
- National Jewish Health, Denver, CO, 80206, USA
| | - Mary Walsh-Wilcox
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, 2703 Frontier Ave NE MSC09 5630, Albuquerque, NM, 87131, USA
| | - Meera Shah
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, 2703 Frontier Ave NE MSC09 5630, Albuquerque, NM, 87131, USA
| | - Abinash Achrekar
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Joe R Anderson
- Department of Pharmacy Practice and Administrative Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Mary K Walker
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, 2703 Frontier Ave NE MSC09 5630, Albuquerque, NM, 87131, USA.
| |
Collapse
|
16
|
Yamagata K. Prevention of Endothelial Dysfunction and Cardiovascular Disease by n-3 Fatty Acids-Inhibiting Action on Oxidative Stress and Inflammation. Curr Pharm Des 2021; 26:3652-3666. [PMID: 32242776 DOI: 10.2174/1381612826666200403121952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Prospective cohort studies and randomized controlled trials have shown the protective effect of n-3 fatty acids against cardiovascular disease (CVD). The effect of n-3 fatty acids on vascular endothelial cells indicates their possible role in CVD prevention. OBJECTIVE Here, we describe the effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on endothelial dysfunction-caused by inflammation and oxidative stress-and their role in the development of CVD. METHODS We reviewed epidemiological studies done on n-3 fatty acids in CVD. The effect of DHA and EPA on vascular endothelial cells was examined with regard to changes in various markers, such as arteriosclerosis, inflammation, and oxidative stress, using cell and animal models. RESULTS Epidemiological studies revealed that dietary intake of EPA and DHA was associated with a reduced risk of various CVDs. EPA and DHA inhibited various events involved in arteriosclerosis development by preventing oxidative stress and inflammation associated with endothelial cell damage. In particular, EPA and DHA prevented endothelial cell dysfunction mediated by inflammatory responses and oxidative stress induced by events related to CVD. DHA and EPA also increased eNOS activity and induced nitric oxide production. CONCLUSION The effects of DHA and EPA on vascular endothelial cell damage and dysfunction may involve the induction of nitric oxide, in addition to antioxidant and anti-inflammatory effects. n-3 fatty acids inhibit endothelial dysfunction and prevent arteriosclerosis. Therefore, the intake of n-3 fatty acids may prevent CVDs, like myocardial infarction and stroke.
Collapse
Affiliation(s)
- Kazuo Yamagata
- College of Bioresource Science, Nihon University (UNBS), Kanagawa, Japan
| |
Collapse
|
17
|
Del Pozo MA, Lolo FN, Echarri A. Caveolae: Mechanosensing and mechanotransduction devices linking membrane trafficking to mechanoadaptation. Curr Opin Cell Biol 2020; 68:113-123. [PMID: 33188985 DOI: 10.1016/j.ceb.2020.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023]
Abstract
Mechanical forces (extracellular matrix stiffness, vascular shear stress, and muscle stretching) reaching the plasma membrane (PM) determine cell behavior. Caveolae are PM-invaginated nanodomains with specific lipid and protein composition. Being highly abundant in mechanically challenged tissues (muscles, lungs, vessels, and adipose tissues), they protect cells from mechanical stress damage. Caveolae flatten upon increased PM tension, enabling both force sensing and accommodation, critical for cell mechanoprotection and homeostasis. Thus, caveolae are highly plastic, ranging in complexity from flattened membranes to vacuolar invaginations surrounded by caveolae-rosettes-which also contribute to mechanoprotection. Caveolar components crosstalk with mechanotransduction pathways and recent studies show that they translocate from the PM to the nucleus to convey stress information. Furthermore, caveolae components can regulate membrane traffic from/to the PM to adapt to environmental mechanical forces. The interdependence between lipids and caveolae starts to be understood, and the relevance of caveolae-dependent membrane trafficking linked to mechanoadaption to different physiopathological processes is emerging.
Collapse
Affiliation(s)
- Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| |
Collapse
|
18
|
Modulatory role of dietary polyunsaturated fatty acids in Nrf2-mediated redox homeostasis. Prog Lipid Res 2020; 80:101066. [DOI: 10.1016/j.plipres.2020.101066] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
|
19
|
Russell JS, Griffith TA, Naghipour S, Vider J, Du Toit EF, Patel HH, Peart JN, Headrick JP. Dietary α-Linolenic Acid Counters Cardioprotective Dysfunction in Diabetic Mice: Unconventional PUFA Protection. Nutrients 2020; 12:nu12092679. [PMID: 32887376 PMCID: PMC7551050 DOI: 10.3390/nu12092679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/12/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
Whether dietary omega-3 (n-3) polyunsaturated fatty acid (PUFA) confers cardiac benefit in cardiometabolic disorders is unclear. We test whether dietary -linolenic acid (ALA) enhances myocardial resistance to ischemia-reperfusion (I-R) and responses to ischemic preconditioning (IPC) in type 2 diabetes (T2D); and involvement of conventional PUFA-dependent mechanisms (caveolins/cavins, kinase signaling, mitochondrial function, and inflammation). Eight-week male C57Bl/6 mice received streptozotocin (75 mg/kg) and 21 weeks high-fat/high-carbohydrate feeding. Half received ALA over six weeks. Responses to I-R/IPC were assessed in perfused hearts. Localization and expression of caveolins/cavins, protein kinase B (AKT), and glycogen synthase kinase-3 β (GSK3β); mitochondrial function; and inflammatory mediators were assessed. ALA reduced circulating leptin, without affecting body weight, glycemic dysfunction, or cholesterol. While I-R tolerance was unaltered, paradoxical injury with IPC was reversed to cardioprotection with ALA. However, post-ischemic apoptosis (nucleosome content) appeared unchanged. Benefit was not associated with shifts in localization or expression of caveolins/cavins, p-AKT, p-GSK3β, or mitochondrial function. Despite mixed inflammatory mediator changes, tumor necrosis factor-a (TNF-a) was markedly reduced. Data collectively reveal a novel impact of ALA on cardioprotective dysfunction in T2D mice, unrelated to caveolins/cavins, mitochondrial, or stress kinase modulation. Although evidence suggests inflammatory involvement, the basis of this "un-conventional" protection remains to be identified.
Collapse
Affiliation(s)
- Jake S. Russell
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Tia A. Griffith
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Saba Naghipour
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Jelena Vider
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Eugene F. Du Toit
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Hemal H. Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California, San Diego, CA 92093, USA;
| | - Jason N. Peart
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - John P. Headrick
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
- Correspondence: ; Tel.: +61-7-5552-8292
| |
Collapse
|
20
|
Cao Q, Zhao J, Xing M, Xiao H, Zhang Q, Liang H, Ji A, Song S. Current Research Landscape of Marine-Derived Anti-Atherosclerotic Substances. Mar Drugs 2020; 18:md18090440. [PMID: 32854344 PMCID: PMC7551282 DOI: 10.3390/md18090440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is a chronic disease characterized by lipid accumulation and chronic inflammation of the arterial wall, which is the pathological basis for coronary heart disease, cerebrovascular disease and thromboembolic disease. Currently, there is a lack of low-cost therapeutic agents that effectively slow the progression of atherosclerosis. Therefore, the development of new drugs is urgently needed. The research and development of marine-derived drugs have gained increasing interest from researchers across the world. Many marine organisms provide a rich material basis for the development of atherosclerotic drugs. This review focuses on the latest technological advances in the structures and mechanisms of action of marine-derived anti-atherosclerotic substances and the challenges of the application of these substances including marine polysaccharides, proteins and peptides, polyunsaturated fatty acids and small molecule compounds. Here, we describe the theoretical basis of marine biological resources in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Qi Cao
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Jiarui Zhao
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Maochen Xing
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Han Xiao
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Qian Zhang
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Hao Liang
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Aiguo Ji
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
- Correspondence: (A.J.); (S.S.)
| | - Shuliang Song
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
- Correspondence: (A.J.); (S.S.)
| |
Collapse
|
21
|
Singh H, Thakur S, Sahajpal NS, Singh H, Singh A, Sohal HS, Jain SK. Recent Advances in the Novel Formulation of Docosahexaenoic Acid for Effective Delivery, Associated Challenges and Its Clinical Importance. Curr Drug Deliv 2020; 17:483-504. [DOI: 10.2174/1567201817666200512103402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/03/2020] [Accepted: 02/02/2020] [Indexed: 11/22/2022]
Abstract
Docosahexaenoic Acid (DHA) is an essential polyunsaturated omega-3 fatty acid, and a fundamental structural component of the phospholipid membranes, especially of neural and retinal cells. DHA is found to be critical for the normal development and functioning of neurons and synaptogenesis in the brain, and is required during pre- and post-natal stages of life. DHA has also been observed to exhibit neuroprotective, cardioprotective, and anti-inflammatory properties. However, geographical dietary variations and poor economic conditions lead to insufficient DHA levels resulting in various health deficits like improper brain development, cognitive disorders, and other clinical complications. Thus, to prevent its deficiency-induced derangements, several authorities recommend DHA as a supplement during pregnancy, infancy, and throughout adulthood. In past decades, the soft gelatin capsule was only feasible resolute of DHA, but due to their limitations and invention of new technologies; it led to the development of new dosage forms with improved physicochemical characteristics of DHA. This article will discuss in detail about the role of DHA in brain development, microalgae oil as an emerging source of DHA, clinical- and pharmacological-activities of DHA, issues related to DHA oil, current formulation of DHA along with their application, limitations, and strategies used for improvement and future prospectives.
Collapse
Affiliation(s)
- Harmanpreet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143105, Punjab, India
| | - Shubham Thakur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143105, Punjab, India
| | - Nikhil Shri Sahajpal
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143105, Punjab, India
| | - Harjeet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143105, Punjab, India
| | - Amrinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143105, Punjab, India
| | - Harminder Singh Sohal
- Department of Orthopaedics, Government Medical College, Amritsar 143001, Punjab, India
| | - Subheet Kumar Jain
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143105, Punjab, India
| |
Collapse
|
22
|
Zhao C, Ma J, Wang Z, Li H, Shen H, Li X, Chen G. Mfsd2a Attenuates Blood-Brain Barrier Disruption After Sub-arachnoid Hemorrhage by Inhibiting Caveolae-Mediated Transcellular Transport in Rats. Transl Stroke Res 2020; 11:1012-1027. [PMID: 31907728 DOI: 10.1007/s12975-019-00775-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/09/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023]
Abstract
Blood-brain barrier (BBB) disruption is one of the critical mechanisms of brain injury induced by subarachnoid hemorrhage (SAH). Past studies have often focused on the tight junctions of endothelial cells. However, low transcellular transport levels also play an important role in the normal functioning of the BBB. Major facilitator superfamily domain-containing 2a (Mfsd2a) has been demonstrated to be essential for the maintenance of the normal BBB. Our present study aimed to explore the roles and mechanisms of Mfsd2a in BBB disruption after SAH. In this study, a prechiasmatic cistern single-injection model was used to produce experimental SAH in Sprague-Dawley rats. Specific small-interfering RNA and plasmids were used to downregulate and upregulate the expression of Mfsd2a prior to assessments in our SAH model. Omega-3 fatty acid deficiency diet was used to reduce DHA in rat brain. The expression level of Mfsd2a decreased significantly after SAH and reached its lowest level at 72 h post-SAH, which then gradually recovered. At 72 h after SAH, BBB function was disrupted; upregulation of Mfsd2a reversed this damage, whereas downregulation of Mfsd2a exacerbated this damage. These effects were primarily mediated through transcellular transport, especially for changes in caveolae compared to those of tight junctions. After stopping the supply of omega-3 fatty acids, the effect of Mfsd2a on inhibition of caveolae and protection of the blood-brain barrier was eliminated. Taken together, Mfsd2a inhibits caveolae-based transcellular transport by transporting omega-3 fatty acids to protect the BBB after SAH.
Collapse
Affiliation(s)
- Chongshun Zhao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Junwei Ma
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
23
|
Vu TT, Dieterich P, Vu TT, Deussen A. Docosahexaenoic acid reduces adenosine triphosphate-induced calcium influx via inhibition of store-operated calcium channels and enhances baseline endothelial nitric oxide synthase phosphorylation in human endothelial cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:345-356. [PMID: 31496872 PMCID: PMC6717795 DOI: 10.4196/kjpp.2019.23.5.345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/25/2022]
Abstract
Docosahexaenoic acid (DHA), an omega-3-fatty acid, modulates multiple cellular functions. In this study, we addressed the effects of DHA on human umbilical vein endothelial cell calcium transient and endothelial nitric oxide synthase (eNOS) phosphorylation under control and adenosine triphosphate (ATP, 100 µM) stimulated conditions. Cells were treated for 48 h with DHA concentrations from 3 to 50 µM. Calcium transient was measured using the fluorescent dye Fura-2-AM and eNOS phosphorylation was addressed by western blot. DHA dose-dependently reduced the ATP stimulated Ca2+-transient. This effect was preserved in the presence of BAPTA (10 and 20 µM) which chelated the intracellular calcium, but eliminated after withdrawal of extracellular calcium, application of 2-aminoethoxy-diphenylborane (75 µM) to inhibit store-operated calcium channel or thapsigargin (2 µM) to delete calcium store. In addition, DHA (12 µM) increased ser1177/thr495 phosphorylation of eNOS under baseline conditions but had no significant effect on this ratio under conditions of ATP stimulation. In conclusion, DHA dose-dependently inhibited the ATP-induced calcium transient, probably via store-operated calcium channels. Furthermore, DHA changed eNOS phosphorylation suggesting activation of the enzyme. Hence, DHA may shift the regulation of eNOS away from a Ca2+ activated mode to a preferentially controlled phosphorylation mode.
Collapse
Affiliation(s)
- Thom Thi Vu
- Department of Physiology, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden 01307, Germany
- Department of Basic Sciences in Medicine and Pharmacy, School of Medicine and Pharmacy, Vietnam National University, Hanoi 100000, Vietnam
| | - Peter Dieterich
- Department of Physiology, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden 01307, Germany
| | - Thu Thi Vu
- Faculty of Biology, VNU University of Science, Hanoi 100000, Vietnam
- Dinh Tien Hoang Institute of Medicine, Hanoi 100000, Vietnam
| | - Andreas Deussen
- Department of Physiology, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden 01307, Germany
| |
Collapse
|
24
|
Hughan KS, Wendell SG, Delmastro-Greenwood M, Helbling N, Corey C, Bellavia L, Potti G, Grimes G, Goodpaster B, Kim-Shapiro DB, Shiva S, Freeman BA, Gladwin MT. Conjugated Linoleic Acid Modulates Clinical Responses to Oral Nitrite and Nitrate. Hypertension 2019; 70:634-644. [PMID: 28739973 DOI: 10.1161/hypertensionaha.117.09016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dietary NO3- (nitrate) and NO2- (nitrite) support ˙NO (nitric oxide) generation and downstream vascular signaling responses. These nitrogen oxides also generate secondary nitrosating and nitrating species that react with low molecular weight thiols, heme centers, proteins, and unsaturated fatty acids. To explore the kinetics of NO3-and NO2-metabolism and the impact of dietary lipid on nitrogen oxide metabolism and cardiovascular responses, the stable isotopes Na15NO3 and Na15NO2 were orally administered in the presence or absence of conjugated linoleic acid (cLA). The reduction of 15NO2- to 15NO was indicated by electron paramagnetic resonance spectroscopy detection of hyperfine splitting patterns reflecting 15NO-deoxyhemoglobin complexes. This formation of 15NO also translated to decreased systolic and mean arterial blood pressures and inhibition of platelet function. Upon concurrent administration of cLA, there was a significant increase in plasma cLA nitration products 9- and 12-15NO2-cLA. Coadministration of cLA with 15NO2- also impacted the pharmacokinetics and physiological effects of 15NO2-, with cLA administration suppressing plasma NO3-and NO2-levels, decreasing 15NO-deoxyhemoglobin formation, NO2-inhibition of platelet activation, and the vasodilatory actions of NO2-, while enhancing the formation of 9- and 12-15NO2-cLA. These results indicate that the biochemical reactions and physiological responses to oral 15NO3-and 15NO2-are significantly impacted by dietary constituents, such as unsaturated lipids. This can explain the variable responses to NO3-and NO2-supplementation in clinical trials and reveals dietary strategies for promoting the generation of pleiotropic nitrogen oxide-derived lipid signaling mediators. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT01681836.
Collapse
Affiliation(s)
- Kara S Hughan
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Stacy Gelhaus Wendell
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Meghan Delmastro-Greenwood
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Nicole Helbling
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Catherine Corey
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Landon Bellavia
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Gopal Potti
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - George Grimes
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Bret Goodpaster
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Daniel B Kim-Shapiro
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Sruti Shiva
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Bruce A Freeman
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| | - Mark T Gladwin
- From the Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes (K.S.H.), Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (K.S.H., S.G.W., M.D.-G., N.H., C.C., S.S., B.A.F., M.T.G.), Department of Pharmacology and Chemical Biology (S.G.W., M.D.-G., S.S., B.A.F.), Department of Medicine, Division of Endocrinology (N.H., B.G.), and Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, PA (M.T.G.); Department of Physics, Wake Forest University, Winston Salem, NC (L.B., D.B.K.-S.); and Pharmaceutical Development Section, Department of Pharmacy, Clinical Center, National Institutes of Health, Bethesda, MD (G.P., G.G.)
| |
Collapse
|
25
|
Thota RN, Ferguson JJA, Abbott KA, Dias CB, Garg ML. Science behind the cardio-metabolic benefits of omega-3 polyunsaturated fatty acids: biochemical effects vs. clinical outcomes. Food Funct 2018; 9:3576-3596. [PMID: 29904777 DOI: 10.1039/c8fo00348c] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lower incidence of cardiovascular disease (CVD) in the Greenland Inuit, Northern Canada and Japan has been attributed to their consumption of seafood rich in long chain omega-3 polyunsaturated fatty acids (LCn-3PUFA). While a large majority of pre-clinical and intervention trials have demonstrated heart health benefits of LCn-3PUFA, some studies have shown no effects or even negative effects. LCn-3PUFA have been shown to favourably modulate blood lipid levels, particularly a reduction in circulating levels of triglycerides. High density lipoprotein-cholesterol (HDL-C) levels are elevated following dietary supplementation with LCn-3PUFA. Although LCn-3PUFA have been shown to increase low-density lipoprotein-cholesterol (LDL-C) levels, the increase is primarily in the large-buoyant particles that are less atherogenic than small-dense LDL particles. The anti-inflammatory effects of LCn-3PUFA have been clearly outlined with inhibition of NFkB mediated cytokine production being the main mechanism. In addition, reduction in adhesion molecules (intercellular adhesion molecule, ICAM and vascular cell adhesion molecule 1, VCAM-1) and leukotriene production have also been demonstrated following LCn-3PUFA supplementation. Anti-aggregatory effects of LCn-3PUFA have been a subject of controversy, however, recent studies showing sex-specific effects on platelet aggregation have helped resolve the effects on hyperactive platelets. Improvements in endothelium function, blood flow and blood pressure after LCn-3PUFA supplementation add to the mechanistic explanation on their cardio-protective effects. Modulation of adipose tissue secretions including pro-inflammatory mediators and adipokines by LCn-3PUFA has re-ignited interest in their cardiovascular health benefits. The aim of this narrative review is to filter out the reasons for possible disparity between cohort, mechanistic, pre-clinical and clinical studies. The focus of the article is to provide possible explanation for the observed controversies surrounding heart health benefits of LCn-3PUFA.
Collapse
Affiliation(s)
- Rohith N Thota
- Nutraceuticals Research Program, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.
| | | | | | | | | |
Collapse
|
26
|
Eser Ocak P, Ocak U, Sherchan P, Zhang JH, Tang J. Insights into major facilitator superfamily domain-containing protein-2a (Mfsd2a) in physiology and pathophysiology. What do we know so far? J Neurosci Res 2018; 98:29-41. [PMID: 30345547 DOI: 10.1002/jnr.24327] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/20/2018] [Accepted: 08/28/2018] [Indexed: 01/02/2023]
Abstract
Major facilitator superfamily domain-containing protein-2a (Mfsd2a) which was considered as an orphan transporter has recently gained attention for its regulatory role in the maintenance of proper functioning of the blood-brain barrier. Besides the major role of Mfsd2a in maintaining the barrier function, increasing evidence has emerged with regard to the contributions of Mfsd2a to various biological processes such as transport, cell fusion, cell cycle, inflammation and regeneration, managing tumor growth, functioning of other organs with barrier functions or responses to injury. The purpose of this article is to review the different roles of Mfsd2a and its involvement in the physiological and pathophysiological processes primarily in the central nervous system and throughout the mammalian body under the lights of the current literature.
Collapse
Affiliation(s)
- Pinar Eser Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Umut Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
27
|
Nuno DW, Coppey LJ, Yorek MA, Lamping KG. Dietary fats modify vascular fat composition, eNOS localization within lipid rafts and vascular function in obesity. Physiol Rep 2018; 6:e13820. [PMID: 30105819 PMCID: PMC6090220 DOI: 10.14814/phy2.13820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022] Open
Abstract
We tested whether dietary fatty acids alter membrane composition shifting localization of signaling pathways within caveolae to determine their role in vascular function. Wild type (WT) and caveolin-1-deficient mice (cav-1 KO), required for vascular caveolae formation, were fed low fat (LF), high saturated fat (HF, 60% kcal from lard), or high-fat diet with 50:50 lard and n-3 polyunsaturated fatty acid-enriched menhaden oil (MO). HF and MO increased body weight and fat in WT but had less effect in cav-1 KO. MO increased unsaturated fatty acids and the unsaturation index of aorta from WT and cav-1 KO. In LF WT aorta, endothelial nitric oxide synthase (eNOS) was localized to cav-1-enriched low-density fractions which shifted to actin-enriched high-density fractions with acetylcholine (ACh). HF and MO shifted eNOS to high-density fractions in WT aorta which was not affected by ACh. In cav-1 KO aorta, eNOS was localized in low-density non-caveolar fractions but not shifted by ACh or diet. Inducible NOS and cyclooxygenase 1/2 were not localized in low-density fractions or affected by diet, ACh or genotype. ACh-induced dilation of gracilis arteries from HF WT was similar to dilation in LF but the NOS component was reduced. In WT and cav-1 KO, dilation to ACh was enhanced by MO through increased role for NOS and cyclooxygenase. We conclude that dietary fats affect vascular fatty acid composition and membrane localization of eNOS but the contribution of eNOS and cyclooxygenase in ACh-mediated vascular responses is independent of lipid rafts.
Collapse
Affiliation(s)
- Daniel W. Nuno
- Department of Internal MedicineRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
| | - Lawrence J. Coppey
- Department of Internal MedicineRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
| | - Mark A. Yorek
- Department of Internal MedicineRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
- Iowa City Veterans Affairs Healthcare SystemIowa CityIowa
| | - Kathryn G. Lamping
- Department of Internal MedicineRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
- Iowa City Veterans Affairs Healthcare SystemIowa CityIowa
- Department of PharmacologyRoy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowa
| |
Collapse
|
28
|
Replication of a Gene-Diet Interaction at CD36, NOS3 and PPARG in Response to Omega-3 Fatty Acid Supplements on Blood Lipids: A Double-Blind Randomized Controlled Trial. EBioMedicine 2018; 31:150-156. [PMID: 29703528 PMCID: PMC6013782 DOI: 10.1016/j.ebiom.2018.04.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Modulation of genetic variants on the effect of omega-3 fatty acid supplements on blood lipids is still unclear. METHODS In a double-blind randomized controlled trial, 150 patients with type 2 diabetes (T2D) were randomized into omega-3 fatty acid group (n = 56 for fish oil and 44 for flaxseed oil) and control group (n = 50) for 180 days. All patients were genotyped for genetic variants at CD36 (rs1527483), NOS3 (rs1799983) and PPARG (rs1801282). Linear regression was used to examine the interaction between omega-3 fatty acid intervention and CD36, NOS3 or PPARG variants for blood lipids. FINDINGS Significant interaction with omega-3 fatty acid supplements was observed for CD36 on triglycerides (p-interaction = 0.042) and PPAGR on low-density lipoprotein-cholesterol (p-interaction = 0.02). We also found a significant interaction between change in erythrocyte phospholipid omega-3 fatty acid composition and NOS3 genotype on triglycerides (p-interaction = 0.042), total cholesterol (p-interaction = 0.013) and ratio of total cholesterol to high-density lipoprotein cholesterol (p-interaction = 0.015). The T2D patients of CD36-G allele, PPARG-G allele and NOS3-A allele tended to respond better to omega-3 fatty acids in improving lipid profiles. The interaction results of the omega-3 fatty acid group were mainly attributed to the fish oil supplements. INTERPRETATION This study suggests that T2D patients with different genotypes at CD36, NOS3 and PPARG respond differentially to intervention of omega-3 supplements in blood lipid profiles.
Collapse
|
29
|
Navarini L, Afeltra A, Gallo Afflitto G, Margiotta DPE. Polyunsaturated fatty acids: any role in rheumatoid arthritis? Lipids Health Dis 2017; 16:197. [PMID: 29017507 PMCID: PMC5634864 DOI: 10.1186/s12944-017-0586-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/02/2017] [Indexed: 12/12/2022] Open
Abstract
Background Polyunsaturated fatty acids (PUFAs) are members of the family of fatty acids and are included in the diet. Particularly, western diet is usually low in n-3 PUFAs and high in n-6 PUFAs. PUFAs play a central role in the homeostasis of immune system: n-6 PUFAs have predominantly pro-inflammatory features, while n-3 PUFAs seem to exert anti-inflammatory and pro-resolving properties. Rheumatoid arthritis (RA) is a chronic inflammatory arthritis in which many inflammatory pathways contribute to joint and systemic inflammation, disease activity, and structural damage. Research on PUFAs could represent an important opportunity to better understand the pathogenesis and to improve the management of RA patients. Methods We searched PubMed, Embase, EBSCO-Medline, Cochrane Central Register of Controlled Trials (CENTRAL), CNKI and Wanfang to identify primary research reporting the role of n-3 PUFAs in rheumatoid arthritis both in humans and in animal models up to the end of March 2017. Results Data from animal models allows to hypothesize that n-3 PUFAs supplementation may represent an interesting perspective in future research as much in prevention as in treating RA. In humans, several case-control and prospective cohort studies suggest that a high content of n-3 PUFAs in the diet could have a protective role for incident RA in subjects at risk. Moreover, n-3 PUFAs supplementation has been assessed as a valuable therapeutic option also for patients with RA, particularly in order to improve the pain symptoms, the tender joint count, the duration of morning stiffness and the frequency of NSAIDs assumption. Conclusions n-3 PUFAs supplementation could represent a promising therapeutic option to better control many features of RA. The impact of n-3 PUFAs on radiographic progression and synovial histopathology has not been yet evaluated, as well as their role in early arthritis and the combination with biologics.
Collapse
Affiliation(s)
- Luca Navarini
- Unit of Allergology, Immunology, Rheumatology, Department of Medicine, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128, Rome, Italy.
| | - Antonella Afeltra
- Unit of Allergology, Immunology, Rheumatology, Department of Medicine, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128, Rome, Italy
| | - Gabriele Gallo Afflitto
- Unit of Allergology, Immunology, Rheumatology, Department of Medicine, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128, Rome, Italy
| | - Domenico Paolo Emanuele Margiotta
- Unit of Allergology, Immunology, Rheumatology, Department of Medicine, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128, Rome, Italy
| |
Collapse
|
30
|
Díaz-Coránguez M, Ramos C, Antonetti DA. The inner blood-retinal barrier: Cellular basis and development. Vision Res 2017; 139:123-137. [PMID: 28619516 DOI: 10.1016/j.visres.2017.05.009] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 01/01/2023]
Abstract
The blood-retinal barrier (BRB) regulates transport across retinal capillaries maintaining proper neural homeostasis and protecting the neural tissue from potential blood borne toxicity. Loss of the BRB contributes to the pathophysiology of a number of blinding retinal diseases including diabetic retinopathy. In this review, we address the basis of the BRB, including the molecular mechanisms that regulate flux across the retinal vascular bed. The routes of transcellular and paracellular flux are described as well as alterations in these pathways in response to permeabilizing agents in diabetes. Finally, we provide information on exciting new studies that help to elucidate the process of BRB development or barriergenesis and how understanding this process may lead to new opportunities for barrier restoration in diabetic retinopathy.
Collapse
Affiliation(s)
- Mónica Díaz-Coránguez
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States
| | - Carla Ramos
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
31
|
Blood-Brain Barrier Permeability Is Regulated by Lipid Transport-Dependent Suppression of Caveolae-Mediated Transcytosis. Neuron 2017; 94:581-594.e5. [PMID: 28416077 DOI: 10.1016/j.neuron.2017.03.043] [Citation(s) in RCA: 371] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 03/02/2017] [Accepted: 03/29/2017] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB) provides a constant homeostatic brain environment that is essential for proper neural function. An unusually low rate of vesicular transport (transcytosis) has been identified as one of the two unique properties of CNS endothelial cells, relative to peripheral endothelial cells, that maintain the restrictive quality of the BBB. However, it is not known how this low rate of transcytosis is achieved. Here we provide a mechanism whereby the regulation of CNS endothelial cell lipid composition specifically inhibits the caveolae-mediated transcytotic route readily used in the periphery. An unbiased lipidomic analysis reveals significant differences in endothelial cell lipid signatures from the CNS and periphery, which underlie a suppression of caveolae vesicle formation and trafficking in brain endothelial cells. Furthermore, lipids transported by Mfsd2a establish a unique lipid environment that inhibits caveolae vesicle formation in CNS endothelial cells to suppress transcytosis and ensure BBB integrity.
Collapse
|
32
|
Omega-3 fatty acids and cytochrome P450-derived eicosanoids in cardiovascular diseases: Which actions and interactions modulate hemodynamics? Prostaglandins Other Lipid Mediat 2017; 128-129:34-42. [DOI: 10.1016/j.prostaglandins.2017.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 01/05/2017] [Accepted: 01/18/2017] [Indexed: 12/24/2022]
|
33
|
Auger C, Said A, Nguyen PN, Chabert P, Idris-Khodja N, Schini-Kerth VB. Potential of Food and Natural Products to Promote Endothelial and Vascular Health. J Cardiovasc Pharmacol 2016; 68:11-8. [PMID: 26974893 DOI: 10.1097/fjc.0000000000000382] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endothelial dysfunction is now well established as a pivotal early event in the development of major cardiovascular diseases including hypertension, atherosclerosis, and diabetes. The alteration of the endothelial function is often triggered by an imbalance between the endothelial formation of vasoprotective factors including nitric oxide (NO) and endothelium-dependent hyperpolarization, and an increased level of oxidative stress involving several prooxidant enzymes such as NADPH oxidase and, often also, the appearance of cyclooxygenase-derived vasoconstrictors. Preclinical studies have indicated that polyphenol-rich food and food-derived products such as grape-derived products, black and red berries, green and black teas and cocoa, and omega-3 fatty acids can trigger activating pathways in endothelial cells promoting an increased formation of nitric oxide and endothelium-dependent hyperpolarization. Moreover, intake of such food-derived products has been associated with the prevention and/or the improvement of an established endothelial dysfunction in several experimental models of cardiovascular diseases and in humans with cardiovascular diseases. This review will discuss both experimental and clinical evidences indicating that different types of food and natural products are able to promote endothelial and vascular health, as well as the underlying mechanisms.
Collapse
Affiliation(s)
- Cyril Auger
- UMR CNRS 7213, Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, Université de Strasbourg, Strasbourg, France
| | | | | | | | | | | |
Collapse
|
34
|
Cabrera CP, Ng FL, Warren HR, Barnes MR, Munroe PB, Caulfield MJ. Exploring hypertension genome-wide association studies findings and impact on pathophysiology, pathways, and pharmacogenetics. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:73-90. [DOI: 10.1002/wsbm.1290] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/25/2014] [Accepted: 01/05/2015] [Indexed: 01/11/2023]
Affiliation(s)
- Claudia P Cabrera
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry; Queen Mary University of London; London UK
- NIHR Barts Cardiovascular Biomedical Research Unit; Queen Mary University of London; London UK
| | - Fu Liang Ng
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry; Queen Mary University of London; London UK
| | - Helen R Warren
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry; Queen Mary University of London; London UK
- NIHR Barts Cardiovascular Biomedical Research Unit; Queen Mary University of London; London UK
| | - Michael R Barnes
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry; Queen Mary University of London; London UK
- NIHR Barts Cardiovascular Biomedical Research Unit; Queen Mary University of London; London UK
| | - Patricia B Munroe
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry; Queen Mary University of London; London UK
- NIHR Barts Cardiovascular Biomedical Research Unit; Queen Mary University of London; London UK
| | - Mark J Caulfield
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry; Queen Mary University of London; London UK
- NIHR Barts Cardiovascular Biomedical Research Unit; Queen Mary University of London; London UK
| |
Collapse
|
35
|
Agbor LN, Wiest EF, Rothe M, Schunck WH, Walker MK. Role of CYP1A1 in modulating the vascular and blood pressure benefits of omega-3 polyunsaturated fatty acids. J Pharmacol Exp Ther 2014; 351:688-98. [PMID: 25316121 DOI: 10.1124/jpet.114.219535] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The mechanisms that mediate the cardiovascular protective effects of omega 3 (n-3) polyunsaturated fatty acids (PUFAs) have not been fully elucidated. Cytochrome P450 1A1 efficiently metabolizes n-3 PUFAs to potent vasodilators. Thus, we hypothesized that dietary n-3 PUFAs increase nitric oxide (NO)-dependent blood pressure regulation and vasodilation in a CYP1A1-dependent manner. CYP1A1 wild-type (WT) and knockout (KO) mice were fed an n-3 or n-6 PUFA-enriched diet for 8 weeks and were analyzed for tissue fatty acids and metabolites, NO-dependent blood pressure regulation, NO-dependent vasodilation of acetylcholine (ACh) in mesenteric resistance arterioles, and endothelial NO synthase (eNOS) and phospho-Ser1177-eNOS expression in the aorta. All mice fed the n-3 PUFA diet showed significantly higher levels of n-3 PUFAs and their metabolites, and significantly lower levels of n-6 PUFAs and their metabolites. In addition, KO mice on the n-3 PUFA diet accumulated significantly higher levels of n-3 PUFAs in the aorta and kidney without a parallel increase in the levels of their metabolites. Moreover, KO mice exhibited significantly less NO-dependent regulation of blood pressure on the n-3 PUFA diet and significantly less NO-dependent, ACh-mediated vasodilation in mesenteric arterioles on both diets. Finally, the n-3 PUFA diet significantly increased aortic phospho-Ser1177-eNOS/eNOS ratio in the WT compared with KO mice. These data demonstrate that CYP1A1 contributes to eNOS activation, NO bioavailability, and NO-dependent blood pressure regulation mediated by dietary n-3 PUFAs.
Collapse
Affiliation(s)
- Larry N Agbor
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| | - Elani F Wiest
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| | - Michael Rothe
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| | - Wolf-Hagen Schunck
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| | - Mary K Walker
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| |
Collapse
|
36
|
Zgheel F, Alhosin M, Rashid S, Burban M, Auger C, Schini-Kerth VB. Redox-sensitive induction of Src/PI3-kinase/Akt and MAPKs pathways activate eNOS in response to EPA:DHA 6:1. PLoS One 2014; 9:e105102. [PMID: 25133540 PMCID: PMC4136823 DOI: 10.1371/journal.pone.0105102] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 07/21/2014] [Indexed: 02/07/2023] Open
Abstract
Aims Omega-3 fatty acid products containing eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have vasoprotective effects, in part, by stimulating the endothelial formation of nitric oxide (NO). This study determined the role of the EPA:DHA ratio and amount, and characterized the mechanism leading to endothelial NO synthase (eNOS) activation. Methods and Results EPA:DHA 6∶1 and 9∶1 caused significantly greater endothelium-dependent relaxations in porcine coronary artery rings than EPA:DHA 3∶1, 1∶1, 1∶3, 1∶6, 1∶9, EPA and DHA alone, and EPA:DHA 6∶1 with a reduced EPA + DHA amount, which were inhibited by an eNOS inhibitor. Relaxations to EPA:DHA 6∶1 were insensitive to cyclooxygenase inhibition, and reduced by inhibitors of either oxidative stress, Src kinase, PI3-kinase, p38 MAPK, MEK, or JNK. EPA:DHA 6∶1 induced phosphorylation of Src, Akt, p38 MAPK, ERK, JNK and eNOS; these effects were inhibited by MnTMPyP. EPA:DHA 6∶1 induced the endothelial formation of ROS in coronary artery sections as assessed by dihydroethidium, and of superoxide anions and hydrogen peroxide in cultured endothelial cells as assessed by electron spin resonance with the spin probe CMH, and the Amplex Red based assay, respectively. Conclusion Omega-3 fatty acids cause endothelium-dependent NO-mediated relaxations in coronary artery rings, which are dependent on the EPA:DHA ratio and amount, and involve an intracellular activation of the redox-sensitive PI3-kinase/Akt and MAPKs pathways to activate eNOS.
Collapse
Affiliation(s)
- Faraj Zgheel
- CNRS UMR 7213 Laboratoire de Biophotonique et Pharmacologie, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Mahmoud Alhosin
- CNRS UMR 7213 Laboratoire de Biophotonique et Pharmacologie, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Sherzad Rashid
- CNRS UMR 7213 Laboratoire de Biophotonique et Pharmacologie, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Mélanie Burban
- CNRS UMR 7213 Laboratoire de Biophotonique et Pharmacologie, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Cyril Auger
- CNRS UMR 7213 Laboratoire de Biophotonique et Pharmacologie, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Valérie B. Schini-Kerth
- CNRS UMR 7213 Laboratoire de Biophotonique et Pharmacologie, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
- * E-mail:
| |
Collapse
|
37
|
Hoogeveen EK, Geleijnse JM, Kromhout D, van't Sant P, Gemen EF, Kusters R, Giltay EJ. No effect of n-3 fatty acids supplementation on NT-proBNP after myocardial infarction: The Alpha Omega Trial. Eur J Prev Cardiol 2014; 22:648-55. [DOI: 10.1177/2047487314536694] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 05/01/2014] [Indexed: 11/17/2022]
Affiliation(s)
- Ellen K Hoogeveen
- Department of Internal Medicine and Nephrology, Jeroen Bosch Hospital, Den Bosch, the Netherlands
| | - Johanna M Geleijnse
- Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
| | - Daan Kromhout
- Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
| | - Peter van't Sant
- Department of Clinical Chemistry, Jeroen Bosch Hospital, Den Bosch, the Netherlands
| | - Eugenie F Gemen
- Department of Clinical Chemistry, Jeroen Bosch Hospital, Den Bosch, the Netherlands
| | - Ron Kusters
- Department of Clinical Chemistry, Jeroen Bosch Hospital, Den Bosch, the Netherlands
| | - Erik J Giltay
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
38
|
Yates CM, Calder PC, Ed Rainger G. Pharmacology and therapeutics of omega-3 polyunsaturated fatty acids in chronic inflammatory disease. Pharmacol Ther 2014; 141:272-82. [DOI: 10.1016/j.pharmthera.2013.10.010] [Citation(s) in RCA: 264] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/12/2013] [Indexed: 12/11/2022]
|
39
|
Marchioli R, Levantesi G, Silletta MG, Barlera S, Bernardinangeli M, Carbonieri E, Cosmi F, Franzosi MG, Latini R, Lucci D, Maggioni AP, Moretti L, Nicolosi GL, Porcu M, Rossi MG, Tognoni G, Tavazzi L. Effect of n-3 polyunsaturated fatty acids and rosuvastatin in patients with heart failure: results of the GISSI-HF trial. Expert Rev Cardiovasc Ther 2014; 7:735-48. [DOI: 10.1586/erc.09.70] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Statin-Mediated Low-Density Lipoprotein Lowering in Chronic Congestive Heart Failure. Am J Med Sci 2014; 347:14-22. [DOI: 10.1097/maj.0b013e318273514c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
41
|
Martins MA, Moss MB, Mendes IKS, Águila MB, Mandarim-de-Lacerda CA, Brunini TMC, Mendes-Ribeiro AC. Role of dietary fish oil on nitric oxide synthase activity and oxidative status in mice red blood cells. Food Funct 2014; 5:3208-15. [DOI: 10.1039/c4fo00055b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The consumption of n-3 polyunsaturated fatty acids derived from fish oil is associated with cardiovascular benefits, which may result from the participation of nitric oxide.
Collapse
Affiliation(s)
- Marcela A. Martins
- Department of Pharmacology and Psychobiology
- Biomedical Center
- Institute of Biology
- State University of Rio de Janeiro
- Rio de Janeiro, Brazil
| | - Monique B. Moss
- Discipline of Pharmacology
- Department of Physiology
- Federal University of Rio de Janeiro State
- Rio de Janeiro, Brazil
| | - Iara K. S. Mendes
- Department of Pharmacology and Psychobiology
- Biomedical Center
- Institute of Biology
- State University of Rio de Janeiro
- Rio de Janeiro, Brazil
| | - Márcia B. Águila
- Laboratory of Morphometry
- Metabolism and Cardiovascular Disease
- Biomedical Center
- Institute of Biology
- State University of Rio de Janeiro
| | | | - Tatiana M. C. Brunini
- Department of Pharmacology and Psychobiology
- Biomedical Center
- Institute of Biology
- State University of Rio de Janeiro
- Rio de Janeiro, Brazil
| | - Antônio Cláudio Mendes-Ribeiro
- Department of Pharmacology and Psychobiology
- Biomedical Center
- Institute of Biology
- State University of Rio de Janeiro
- Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Gortan Cappellari G, Losurdo P, Mazzucco S, Panizon E, Jevnicar M, Macaluso L, Fabris B, Barazzoni R, Biolo G, Carretta R, Zanetti M. Treatment with n-3 polyunsaturated fatty acids reverses endothelial dysfunction and oxidative stress in experimental menopause. J Nutr Biochem 2013; 24:371-9. [DOI: 10.1016/j.jnutbio.2012.07.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 07/12/2012] [Accepted: 07/20/2012] [Indexed: 10/27/2022]
|
43
|
Wei W, Chen M, Zhu Y, Wang J, Zhu P, Li Y, Li J. Down-regulation of vascular HMGB1 and RAGE expression by n-3 polyunsaturated fatty acids is accompanied by amelioration of chronic vasculopathy of small bowel allografts. J Nutr Biochem 2012; 23:1333-40. [DOI: 10.1016/j.jnutbio.2011.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Revised: 07/31/2011] [Accepted: 08/10/2011] [Indexed: 02/08/2023]
|
44
|
Agbor LN, Walsh MT, Boberg JR, Walker MK. Elevated blood pressure in cytochrome P4501A1 knockout mice is associated with reduced vasodilation to omega-3 polyunsaturated fatty acids. Toxicol Appl Pharmacol 2012; 264:351-60. [PMID: 22995157 DOI: 10.1016/j.taap.2012.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 09/04/2012] [Accepted: 09/09/2012] [Indexed: 01/19/2023]
Abstract
In vitro cytochrome P4501A1 (CYP1A1) metabolizes omega-3 polyunsaturated fatty acids (n-3 PUFAs); eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), primarily to 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP), respectively. These metabolites have been shown to mediate vasodilation via increases in nitric oxide (NO) and activation of potassium channels. We hypothesized that genetic deletion of CYP1A1 would reduce vasodilatory responses to n-3 PUFAs, but not the metabolites, and increase blood pressure (BP) due to decreases in NO. We assessed BP by radiotelemetry in CYP1A1 wildtype (WT) and knockout (KO) mice±NO synthase (NOS) inhibitor. We also assessed vasodilation to acetylcholine (ACh), EPA, DHA, 17,18-EEQ and 19,20-EDP in aorta and mesenteric arterioles. Further, we assessed vasodilation to an NO donor and to DHA±inhibitors of potassium channels. CYP1A1 KO mice were hypertensive, compared to WT, (mean BP in mmHg, WT 103±1, KO 116±1, n=5/genotype, p<0.05), and exhibited a reduced heart rate (beats per minute, WT 575±5; KO 530±7; p<0.05). However, BP responses to NOS inhibition and vasorelaxation responses to ACh and an NO donor were normal in CYP1A1 KO mice, suggesting that NO bioavailability was not reduced. In contrast, CYP1A1 KO mice exhibited significantly attenuated vasorelaxation responses to EPA and DHA in both the aorta and mesenteric arterioles, but normal vasorelaxation responses to the CYP1A1 metabolites, 17,18-EEQ and 19,20-EDP, and normal responses to potassium channel inhibition. Taken together these data suggest that CYP1A1 metabolizes n-3 PUFAs to vasodilators in vivo and the loss of these vasodilators may lead to increases in BP.
Collapse
Affiliation(s)
- Larry N Agbor
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | | | | | | |
Collapse
|
45
|
Balakumar P, Taneja G. Fish oil and vascular endothelial protection: bench to bedside. Free Radic Biol Med 2012; 53:271-9. [PMID: 22584102 DOI: 10.1016/j.freeradbiomed.2012.05.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Revised: 03/16/2012] [Accepted: 05/03/2012] [Indexed: 02/07/2023]
Abstract
Fish oil is recommended for the management of hypertriglyceridemia and to prevent secondary cardiovascular disorders. Fish oil is a major source of ω-3-polyunsaturated fatty acids (PUFAs) such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). Clinical studies suggest that fish oil not only prevents the incidence of detrimental cardiovascular events, but also lowers the cardiovascular mortality rate. In addition to a classic lipid-lowering action, ω-3-PUFAs in fish oil could regulate blood pressure and enhance vascular integrity and compliance. Additionally, ω-3-PUFAs have the ability to protect vascular endothelial cells by decreasing oxidative stress, halting atherosclerotic events, and preventing vascular inflammatory and adhesion cascades. Intriguingly, recent studies have demonstrated that ω-3-PUFAs improve the function of vascular endothelium by enhancing the generation and bioavailability of endothelium-derived relaxing factor (nitric oxide) through upregulation and activation of endothelial nitric oxide synthase (eNOS). This certainly opens up a new area of research identifying potential mechanisms influencing fish oil-mediated functional regulatory action on vascular endothelium. We address in this review the potential of fish oil to prevent vascular endothelial dysfunction and associated cardiovascular disorders. Moreover, the mechanisms pertaining to fish oil-mediated eNOS activation and nitric oxide generation in improving endothelial function are delineated. We finally suggest the importance of further studies to determine the dose adjustment of fish oil with an optimal ratio of EPA and DHA for achieving consistent cardiovascular protection.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Cardiovascular Pharmacology Division, Department of Pharmacology, Institute of Pharmacy, Rajendra Institute of Technology and Sciences, Sirsa 125 055, India.
| | | |
Collapse
|
46
|
The janus face of lipids in human breast cancer: how polyunsaturated Fatty acids affect tumor cell hallmarks. Int J Breast Cancer 2012; 2012:712536. [PMID: 22811918 PMCID: PMC3395128 DOI: 10.1155/2012/712536] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/06/2012] [Indexed: 12/13/2022] Open
Abstract
For several years, lipids and especially n - 3 and n - 6 polyunsaturated fatty acids (PUFAs) receive much attention in human health. Epidemiological studies tend to correlate a PUFA-rich diet with a reduced incidence of cancer, including breast cancer. However, the molecular and cellular mechanisms supporting the effect of PUFAs in breast cancer cells remain relatively unknown. Here, we review some recent progress in understanding the impact that PUFA may have on breast cancer cell proliferation, apoptosis, migration, and invasion. While most of the results obtained with docosahexaenoic acid and/or eicosapentaenoic acid show a decrease of tumor cell proliferation and/or aggressivity, there is some evidence that other lipids, which accumulate in breast cancer tissues, such as arachidonic acid may have opposite effects. Finally, lipids and especially PUFAs appear as potential adjuvants to conventional cancer therapy.
Collapse
|
47
|
Abstract
(n-3) PUFA are a family of biologically active fatty acids. The simplest member of this family, α-linolenic acid, can be converted to the more biologically active very long-chain (n-3) PUFA EPA and DHA; this process occurs by a series of desaturation and elongation reactions, with stearidonic acid being an intermediate in the pathway. Biological activity of α-linolenic and stearidonic acids most likely relates to their conversion to EPA. The very long-chain (n-3) PUFA have a range of physiological roles that relate to optimal cell membrane structure and optimal cell function and responses. Thus, (n-3) PUFA play a key role in preventing, and perhaps treating, many conditions of poor health and well-being. The multiple actions of (n-3) PUFA appear to involve multiple mechanisms that connect the cell membrane, the cytosol, and the nucleus. For some actions, (n-3) PUFA appear to act via receptors or sensors, so regulating signaling processes that influence patterns of gene expression. Some effects of (n-3) PUFA seem to involve changes in cell membrane fatty acid composition. Changing membrane composition can in turn affect membrane order, formation of lipid rafts, intracellular signaling processes, gene expression, and the production of both lipid and peptide mediators. Under typical Western dietary conditions, human cells tend to have a fairly high content of the (n-6) fatty acid arachidonic acid. Increased oral intake of EPA and DHA modifies the content of arachidonic acid as well as of EPA and DHA. Arachidonic acid is the substrate for eicosanoids involved in physiology and pathophysiology. The eicosanoids produced from EPA frequently have properties that are different from those that are produced from arachidonic acid. EPA and DHA are also substrates for production of resolvins and protectins, which seem to be biologically extremely potent. Increasing the contents of EPA and DHA in membranes modifies the pattern of production of these different lipid mediators.
Collapse
Affiliation(s)
- Philip C Calder
- Institute of Human Nutrition and Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK.
| |
Collapse
|
48
|
A comparison of the changes in cardiac output and systemic vascular resistance during exercise following high-fat meals containing DHA or EPA. Br J Nutr 2012; 108:492-9. [PMID: 22348439 DOI: 10.1017/s0007114511005721] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Long-chain n-3 PUFA can lower blood pressure (BP) but their acute effects on cardiac output, BP and systemic vascular resistance (SVR) in response to dynamic exercise are uncertain. We compared the effects of high-fat meals rich in EPA (20 : 5n-3), DHA (22 : 6n-3) or oleic acid (control) on cardiac output, BP and SVR in response to exercise stress testing. High-fat meals (50 g fat) containing high-oleic sunflower oil enriched with 4·7 g of either EPA or DHA v. control (high-oleic sunflower oil only) were fed to twenty-two healthy males using a randomised cross-over design. Resting measurements of cardiac output, heart rate and BP were made before and hourly over 5 h following the meal. A standardised 12 min exercise test was then conducted with further measurements made during and post-exercise. Blood samples were collected at fasting, 5 h postprandially and immediately post-exercise for the analysis of lipid, glucose and 8-isoprostane-F2α (8-iso-PGF2α). Plasma concentrations of EPA and DHA increased by 0·22 mmol/l 5 h following the EPA and DHA meals, respectively, compared with the control (P < 0·001). Resting cardiac output and 8-iso-PGF2α increased similarly following all meals and there were no significant differences in cardiac output during exercise between the meals. SVR was lower at 5 h and during exercise following the DHA but not EPA meal, compared with the control meal, by 4·9 % (95 % CI 1·3, 8·4; P < 0·01). Meals containing DHA appear to differ from EPA with regard to their effects on cardiovascular haemodynamics during exercise.
Collapse
|
49
|
Kromhout D, Yasuda S, Geleijnse JM, Shimokawa H. Fish oil and omega-3 fatty acids in cardiovascular disease: do they really work? Eur Heart J 2011; 33:436-43. [PMID: 21933782 DOI: 10.1093/eurheartj/ehr362] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Omega-3 fatty acids, which are found abundantly in fish oil, exert pleiotropic cardiometabolic effects with a diverse range of actions. The results of previous studies raised a lot of interest in the role of fish oil and omega-3 fatty acids in primary and secondary prevention of cardiovascular diseases. The present review will focus on the current clinical uses of omega-3 fatty acids and provide an update on their effects. Since recently published trials in patients with coronary artery diseases or post-myocardial infarction did not show an effect of omega-3 fatty acids on major cardiovascular endpoints, this review will examine the limitations of those data and suggest recommendations for the use of omega-3 fatty acids.
Collapse
Affiliation(s)
- Daan Kromhout
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands.
| | | | | | | |
Collapse
|
50
|
Øie E, Ueland T, Dahl CP, Bohov P, Berge C, Yndestad A, Gullestad L, Aukrust P, Berge RK. Fatty acid composition in chronic heart failure: low circulating levels of eicosatetraenoic acid and high levels of vaccenic acid are associated with disease severity and mortality. J Intern Med 2011; 270:263-72. [PMID: 21466599 DOI: 10.1111/j.1365-2796.2011.02384.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Free fatty acids (FFAs) are the major energy sources of the heart, and fatty acids (FAs) are active components of biological membranes. Data indicate that levels of FAs and their composition may influence myocardial function and inflammation. The aim of this study was to investigate whether total levels and composition of FAs and FFAs in plasma are altered in clinical heart failure (HF) and whether any alterations in these parameters are correlated with the severity of HF. SUBJECTS Plasma from 183 patients with stable HF was compared with plasma from 44 healthy control subjects. RESULTS Our main findings are as follows: (i) patients with HF had decreased levels of several lipid parameters and increased levels of FFAs in plasma, compared with controls, which were significantly correlated with clinical disease severity. (ii) Patients with HF also had a decreased proportion in the plasma of several n-3 polyunsaturated FAs, an increased proportion of several monounsaturated FAs, and a decreased proportion of some readily oxidized long-chain saturated FAs. (iii) These changes in FA composition were significantly associated with functional class, impaired cardiac function (i.e., decreased cardiac index and increased plasma N-terminal pro-B-type natriuretic peptide levels) and enhanced systemic inflammation (i.e., increased high-sensitivity C-reactive protein levels). (iv) Low levels of C20:4n-3 (eicosatetraenoic acid) and in particular high levels of C18:1n-7 (vaccenic acid) were significantly associated with total mortality in this HF population. CONCLUSIONS Our data demonstrate that patients with HF are characterized by a certain FA phenotype and may support a link between disturbed FA composition and the progression of HF.
Collapse
Affiliation(s)
- E Øie
- Research Institute for Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|