1
|
Duan Y, Qi D, Liu Y, Song Y, Wang X, Jiao S, Li H, Gonzalez FJ, Qi Y, Xu Q, Du J, Qu A. Deficiency of peroxisome proliferator-activated receptor α attenuates apoptosis and promotes migration of vascular smooth muscle cells. Biochem Biophys Rep 2021; 27:101091. [PMID: 34381883 PMCID: PMC8339143 DOI: 10.1016/j.bbrep.2021.101091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR) α is widely expressed in the vasculature and has pleiotropic and lipid-lowering independent effects, but its role in the growth and function of vascular smooth muscle cells (VSMCs) during vascular pathophysiology is still unclear. Herein, VSMC-specific PPARα-deficient mice (Ppara ΔSMC) were generated by Cre-LoxP site-specific recombinase technology and VSMCs were isolated from mice aorta. PPARα deficiency attenuated VSMC apoptosis induced by angiotensin (Ang) II and hydrogen peroxide, and increased the migration of Ang II-challenged cells.
Collapse
Key Words
- Ang II, angiotensin II
- Angiotensin II
- EC, endothelial cell
- ECM, extracellular matrix
- ERK, extracellular signal-regulated kinase
- MAPK, mitogen-activated protein kinase
- MCP-1, monocyte chemoattractant protein-1
- PCR, polymerase chain reaction
- PPAR, peroxisome proliferator-activated receptor
- PPARα
- SM22α, smooth muscle 22α
- TGF, tumor growth factor
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- VSMC, vascular smooth muscle cell
- Vascular remodeling
- Vascular smooth muscle cell
Collapse
Affiliation(s)
- Yan Duan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Dan Qi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Ye Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Yanting Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Shiyu Jiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Huihua Li
- Department of Nutrition and Food Hygiene, School of Public Health, Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yongfen Qi
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Qingbo Xu
- School of Cardiovascular Medicine and Sciences, King' s College of London, London, UK
| | - Jie Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China.,Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| |
Collapse
|
2
|
Kim JS, Thomashow MA, Yip NH, Burkart KM, Lo Cascio CM, Shimbo D, Barr RG. Randomization to Omega-3 Fatty Acid Supplementation and Endothelial Function in COPD: The COD-Fish Randomized Controlled Trial. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2021; 8. [PMID: 33150779 DOI: 10.15326/jcopdf.8.1.2020.0132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rationale Studies suggest a pathogenic role of endothelial dysfunction in chronic obstructive lung disease (COPD). Omega-3 (n-3) polyunsaturated fatty acid (PUFA) supplementation improves endothelial function in other diseases but has not been examined in COPD. Objective We hypothesized that n-3 PUFA supplementation would improve systemic endothelial function in COPD. We performed a pilot randomized, placebo-controlled, double-blind, phase 2 superiority trial (NCT00835289). Methods Adults with moderate and severe stable COPD (79% with emphysema on computed tomography [CT]) were randomized to high-dose fish oil capsules or placebo daily for 6 months. The primary endpoint was percentage change in brachial artery flow-mediated dilation (FMD) from baseline to 6 months. Secondary endpoints included peripheral arterial tonometry, endothelial microparticles (EMPs), 6-minute walk distance, respiratory symptoms, and pulmonary function. Results Thirty-three of 40 randomized participants completed all measurements. Change in FMD after 6 months did not differ between the fish oil and placebo arms (-1.1%, 95% CI -5.0-2.9, p=0.59). CD31+ EMPs increased in the fish oil arm (0.9%, 95% CI 0.1-1.7, p=0.04). More participants in the fish oil arm reported at least a 4-point improvement in the St George's Respiratory Questionnaire (SGRQ) compared to placebo (8 versus 1; p=0.01). There were no significant changes in other secondary endpoints. There were 4 serious adverse events determined to be unrelated to the study (3 in the fish oil arm and 1 in the placebo arm). Conclusion Randomization to n-3 PUFAs for 6 months did not change systemic endothelial function in COPD. Changes in EMPs and SGRQ suggest n-3 PUFAs might have biologic and clinical effects that warrant further investigation.
Collapse
Affiliation(s)
- John S Kim
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States.,Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Michael A Thomashow
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States.,Kaiser Permanante San Francisco Medical Center, San Francisco, California, United States
| | - Natalie H Yip
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - Kristin M Burkart
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - Christian M Lo Cascio
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - Daichi Shimbo
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - R Graham Barr
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States.,Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York, United States
| |
Collapse
|
3
|
Selective inhibition of PKR improves vascular inflammation and remodelling in high fructose treated primary vascular smooth muscle cells. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165606. [PMID: 31740405 DOI: 10.1016/j.bbadis.2019.165606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/25/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022]
|
4
|
Pourjafar M, Saidijam M, Mansouri K, Malih S, Ranjbar Nejad T, Shabab N, Najafi R. Cytoprotective effects of endothelin-1 on mesenchymal stem cells: an in vitro study. Clin Exp Pharmacol Physiol 2017; 43:769-76. [PMID: 27161651 DOI: 10.1111/1440-1681.12590] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/02/2016] [Accepted: 05/04/2016] [Indexed: 12/22/2022]
Abstract
Stem cell-based therapies is a promising approach for regenerative therapy in various diseases. Some obstacles remain to be solved before clinical application of the cell therapy is realized, including increasing the survival of transplanted stem cells, reducing loss of transplanted cells, and maintaining adequate vascular supply. Recently, stem cell preconditioning with chemical and pharmacological agents has been shown to increase therapeutic efficacy. The present study investigated the effect of endothelin-1 (ET-1) on survival, angiogenesis, and migration of mesenchymal stem cells (MSCs), in vitro. MSCs were treated with various concentrations of ET-1 and the expression of cyclooxygenase-2 (COX-2), hypoxia-inducible factor-1 (HIF-1), C-X-C chemokine receptor type 4 (CXCR4), C-C chemokine receptor type 2 (CCR2), vascular endothelial growth factor (VEGF), angiopoietin-2 (Ang-2), angiopoietin-4 (Ang-4) and matrix metalloproteinase-2 (MMP-2) were examined. Caspase 3 activity and prostaglandin E2 (PGE2) were determined by ELISA assay. MSCs migration and tube formation potential were assessed using scratch test and three dimensional vessel formation assay. ET-1 enhanced the MSCs viability. In ET-1- treated MSCs, expression of COX-2, HIF-1, CXCR4, CCR2, VEGF, Ang-2, Ang-4 and MMP-2 were increased compared to control groups. Elevation of all these genes were reversed by celecoxib (50 μmol/L), a selective COX-2 inhibitor. PGE2 generation, MSCs migration and tube formation were enhanced by ET-1 conditioning, whereas caspase-3 activity was reduced in these cells, compared to the control group. The results presented here reveal that preconditioning of MSCs with ET-1 has strong cytoprotective effects through activation of survival signalling molecules and trophic factors.
Collapse
Affiliation(s)
- Mona Pourjafar
- Research Centre for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Research Centre for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Kamran Mansouri
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sara Malih
- Research Centre for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Tayebeh Ranjbar Nejad
- Research Centre for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nooshin Shabab
- Research Centre for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Centre for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Endometrium and Endometriosis Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
5
|
Rani K, Aung NY. Docosahexaenoic Acid Inhibits Vascular Smooth Muscle Cell Proliferation Induced by Glucose Variability. Open Biochem J 2017; 11:56-65. [PMID: 28839472 PMCID: PMC5543665 DOI: 10.2174/1874091x01711010056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 02/23/2017] [Accepted: 03/17/2017] [Indexed: 12/02/2022] Open
Abstract
Background: Vascular Smooth Muscle cells (VSMC) enact crucial roles in early vasculogenesis and sustenance of vascular integrity. However, aberrant proliferation of VSMC followed by migration into the blood vessel wall leads to the formation of vascular lesions accounting for the degeneration and remodelling of vascular basement membrane. In diabetes, hyperglycaemia accelerates VSMC proliferation and contributes to the initiation and progression of atherosclerotic lesions. Recently, acute glucose fluctuations have been implicated in the abnormal VSMC proliferation and complications of diabetic atherosclerosis. Docosahexaenoic acid (DHA), a ω-3 polyunsaturated fatty acid (PUFA) has been shown to inhibit proliferation of several cell types implicating several different mechanisms. In the present study, we have investigated the effects of DHA on VSMC proliferation induced by stable and intermittent high glucose levels. Method: Confluent cultures of rat aortic VSMCs were treated with DHA for 24 hrs and then exposed to stable high glucose (25 mmol/L, SHG) or intermittent high glucose (5 mmol/L and 25 mmol/L alternating every 12 hrs, IHG) for 72 hrs. Cell proliferation was examined by the MTT viability assay, while apoptosis process was evaluated by the Hoechst staining, flow cytometry and caspase-3 activity assays. Results: Our data demonstrated that the hyper proliferation induced by stable and intermittent high glucose levels was significantly inhibited by the DHA pre-treatment. DHA significantly increased caspase-3 activity, resulting in enhanced DNA fragmentation and apoptosis. Conclusion: Our results suggest that DHA reduced the high glucose-induced proliferation of VSMC and induced cell apoptosis.
Collapse
Affiliation(s)
| | - Nway Y Aung
- Nanyang Technological University Singapore, Singapore
| |
Collapse
|
6
|
Phytanic acid attenuates insulin-like growth factor-1 activity via nitric oxide-mediated γ-secretase activation in rat aortic smooth muscle cells: possible implications for pathogenesis of infantile Refsum disease. Pediatr Res 2017; 81:531-536. [PMID: 27886192 DOI: 10.1038/pr.2016.258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 10/09/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND Infantile Refsum disease (IRD), a peroxisomal disease with defective phytanic acid oxidation, causes neurological impairment and development delay. Insulin-like growth factor-1 (IGF-1) regulates child development and to understand molecular mechanism(s) of IRD, we examined the effect of phytanic acid (PA) on IGF-1 activity. METHODS Bromodeoxyuridine (BrdU) incorporation was measured in rat aortic smooth muscle cell (SMC) cultures following treatment with fetal bovine serum (FBS), basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF) or IGF-1 in the absence or presence of PA. Gene expression and protein contents of IGF-1 receptor (IGF-1R) and PDGF receptor (PDGFR) were examined using quantitative PCR and western blotting. RESULTS PA inhibited mitogenic activities of FBS, PDGF and IGF-1 with more pronounced effect on IGF-1-induced bromodeoxyuridine (BrdU) incorporation. Palmitic acid or lignoceric acids did not inhibit IGF-1 activity. PA had no effect on PDGFR mRNA/protein levels but markedly increased IGF-1R mRNA levels. PA and nitric oxide (NO) markedly decreased IGF-1R protein. L-NAME, a NO synthase inhibitor and DAPT, a γ-secretase inhibitor, alleviated PA-induced decrease in IGF-1R protein. Both PA and NO donor increased γ-secretase activity which was alleviated by L-NAME. CONCLUSION This study demonstrates that PA attenuates IGF-1 activity possibly through IGF-1R impairment and NO-mediated modulation of γ-secretase activity.
Collapse
|
7
|
Le Guen M, Chaté V, Hininger-Favier I, Laillet B, Morio B, Pieroni G, Schlattner U, Pison C, Dubouchaud H. A 9-wk docosahexaenoic acid-enriched supplementation improves endurance exercise capacity and skeletal muscle mitochondrial function in adult rats. Am J Physiol Endocrinol Metab 2016; 310:E213-24. [PMID: 26646102 DOI: 10.1152/ajpendo.00468.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/04/2015] [Indexed: 01/07/2023]
Abstract
Decline in skeletal muscle mass and function starts during adulthood. Among the causes, modifications of the mitochondrial function could be of major importance. Polyunsaturated fatty (ω-3) acids have been shown to play a role in intracellular functions. We hypothesize that docosahexaenoic acid (DHA) supplementation could improve muscle mitochondrial function that could contribute to limit the early consequences of aging on adult muscle. Twelve-month-old male Wistar rats were fed a low-polyunsaturated fat diet and were given DHA (DHA group) or placebo (control group) for 9 wk. Rats from the DHA group showed a higher endurance capacity (+56%, P < 0.05) compared with control animals. Permeabilized myofibers from soleus muscle showed higher O2 consumptions (P < 0.05) in the DHA group compared with the control group, with glutamate-malate as substrates, both in basal conditions (i.e., state 2) and under maximal conditions (i.e., state 3, using ADP), along with a higher apparent Km for ADP (P < 0.05). Calcium retention capacity of isolated mitochondria was lower in DHA group compared with the control group (P < 0.05). Phospho-AMPK/AMPK ratio and PPARδ mRNA content were higher in the DHA group compared with the control group (P < 0.05). Results showed that DHA enhanced endurance capacity in adult animals, a beneficial effect potentially resulting from improvement in mitochondrial function, as suggested by our results on permeabilized fibers. DHA supplementation could be of potential interest for the muscle function in adults and for fighting the decline in exercise tolerance with age that could imply energy-sensing pathway, as suggested by changes in phospho-AMPK/AMPK ratio.
Collapse
MESH Headings
- 3-Hydroxyacyl CoA Dehydrogenases/drug effects
- 3-Hydroxyacyl CoA Dehydrogenases/metabolism
- Animals
- Blotting, Western
- Calcium/metabolism
- Calorimetry, Indirect
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Cholesterol/metabolism
- Citrate (si)-Synthase/drug effects
- Citrate (si)-Synthase/metabolism
- Dietary Supplements
- Docosahexaenoic Acids/pharmacology
- Electron Transport/drug effects
- Exercise Tolerance/drug effects
- Hydrogen Peroxide/metabolism
- Male
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/metabolism
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Oxygen Consumption/drug effects
- Phospholipids/metabolism
- Physical Conditioning, Animal
- Physical Endurance/drug effects
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Real-Time Polymerase Chain Reaction
- Triglycerides/metabolism
Collapse
Affiliation(s)
- Marie Le Guen
- Université Grenoble Alpes, Laboratoire de Bioénergétique Fondamentale et Appliquée, U1055, Grenoble, France; Institut National de la Santé et de la Recherche Médicale, U1055, Grenoble, France
| | - Valérie Chaté
- Université Grenoble Alpes, Laboratoire de Bioénergétique Fondamentale et Appliquée, U1055, Grenoble, France; Institut National de la Santé et de la Recherche Médicale, U1055, Grenoble, France
| | - Isabelle Hininger-Favier
- Université Grenoble Alpes, Laboratoire de Bioénergétique Fondamentale et Appliquée, U1055, Grenoble, France; Institut National de la Santé et de la Recherche Médicale, U1055, Grenoble, France
| | - Brigitte Laillet
- Institut National de la Recherche Agronomique, Unité de Nutrition Humaine, UMR1019, Clermont-Ferrand, France; and Université d'Auvergne, Unité de Nutrition Humaine, UMR1019, Clermont-Ferrand, France
| | - Béatrice Morio
- Institut National de la Recherche Agronomique, Unité de Nutrition Humaine, UMR1019, Clermont-Ferrand, France; and Université d'Auvergne, Unité de Nutrition Humaine, UMR1019, Clermont-Ferrand, France
| | | | - Uwe Schlattner
- Université Grenoble Alpes, Laboratoire de Bioénergétique Fondamentale et Appliquée, U1055, Grenoble, France; Institut National de la Santé et de la Recherche Médicale, U1055, Grenoble, France
| | - Christophe Pison
- Université Grenoble Alpes, Laboratoire de Bioénergétique Fondamentale et Appliquée, U1055, Grenoble, France; Institut National de la Santé et de la Recherche Médicale, U1055, Grenoble, France
| | - Hervé Dubouchaud
- Université Grenoble Alpes, Laboratoire de Bioénergétique Fondamentale et Appliquée, U1055, Grenoble, France; Institut National de la Santé et de la Recherche Médicale, U1055, Grenoble, France;
| |
Collapse
|
8
|
Słotwiński R, Słotwińska S, Kędziora S, Bałan BJ. Innate immunity signaling pathways: links between immunonutrition and responses to sepsis. Arch Immunol Ther Exp (Warsz) 2011; 59:139-50. [PMID: 21298487 DOI: 10.1007/s00005-011-0117-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Accepted: 07/30/2010] [Indexed: 01/12/2023]
Abstract
Septic infections in patients treated in intensive care units show the highest mortality rates. Despite advances in treatment methods, there is still no therapy available to efficiently reduce the excessive inflammatory response, which can increase the risk of multiple organ failure. One of the ways to discover new, more efficient treatment methods involves regulating the mechanisms of inflammatory response to a massive infection. Toll-like receptors (TLRs) that recognize pathogen-associated molecular patterns play a significant role in innate antibacterial and inflammatory responses. The regulatory impact of immunonutrition on TLR expression in septic patients seems to be a promising research direction. This paper presents the main mechanisms for the innate immune response to lipopolysaccharide, based on the research results for both TLR-dependent and independent signaling pathways. Special emphasis was put on the research results for the TLR-dependent immune response and the anti-bacterial/anti-inflammatory response after applying immunonutrition with increased concentrations of glutamine and unsaturated fatty acids.
Collapse
Affiliation(s)
- Robert Słotwiński
- Department of Surgical Research and Transplantology, Polish Academy of Sciences Medical Research Center, Warsaw, Poland.
| | | | | | | |
Collapse
|
9
|
Effects of short-term infusion of lipid emulsions on pro-inflammatory cytokines and lymphocyte apoptosis in septic and non-septic rats. Br J Nutr 2011; 106:27-32. [DOI: 10.1017/s0007114510005751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Long-term administration of PUFA is known to modulate immune functions and apoptotic pathways depending on the respective amount of n-6 and n-3 fatty acids (FA). Data on short-term effects on apoptotic pathways are rare. Apoptosis of splenic lymphocytes is the hallmark of detrimental sepsis. Therefore, we aimed to compare the immediate effects of parenterally administered n-6-enriched soyabean oil (SO)- and n-3-enriched fish oil (FO)-based lipid emulsions after laparotomy (LAP; sham procedure) and after induction of acute, severe sepsis by caecal ligation and incision. After 390 min of observation time, plasma was analysed for IL-1β, IL-6 and NEFA. Apoptosis in splenic lymphocytes was quantified by Annexin-V expression. After LAP, infusion of both FO and SO did not change cytokine concentrations. Sepsis increased both cytokines. FO but not SO further augmented the rise. After LAP, SO increased NEFA, and both lipid emulsions reduced free arachidonic acid (AA). Sepsis resulted in a dramatic decrease in NEFA and AA. The drop in NEFA and AA was prevented by both SO and FO. In addition, FO resulted in an increased concentration of n-3 FA under both conditions. Infusion of both lipid emulsions induced apoptosis in splenic lymphocytes after LAP. Sepsis-induced apoptosis was not further enhanced by FO or SO. The present study shows that short-term administration of FO as opposed to SO caused pro-inflammatory effects during sepsis. Moreover, short-term administration of both SO and FO suffices to induce apoptosis in splenic lymphocytes. Finally, SO and FO do not further enhance sepsis-induced splenic apoptosis.
Collapse
|
10
|
Abstract
ET (endothelin)-1 was first described as a potent vasoconstrictor. Since then, many other deleterious properties mediated via its two receptors, ETA and ETB, have been described, such as inflammation, fibrosis and hyperplasia. These effects, combined with a wide tissue distribution of the ET system, its up-regulation in pathological situations and a local autocrine/paracrine activity due to a high tissue receptor binding, make the tissue ET system a key local player in end-organ damage. Furthermore, ET-1 interacts in tissues with other systems such as the RAAS (renin-angiotensin-aldosterone system) to exert its effects. In numerous genetically modified animal models, non-specific or organ-targeted ET-1 overexpression causes intense organ damage, especially hypertrophy and fibrosis, in the absence of haemodynamic changes, confirming a local activity of the ET system. ET receptor antagonists have been shown to prevent and sometimes reverse these tissue alterations in an organ-specific manner, leading to long-term benefits and an improvement in survival in different animal models. Potential for such benefits going beyond a pure haemodynamic effect have also been suggested by clinical trial results in which ET receptor antagonism decreased the occurrence of new digital ulcers in patients with systemic sclerosis and delayed the time to clinical worsening in patients with PAH (pulmonary arterial hypertension). The tissue ET system allows therapeutic interventions to provide organ selectivity and beneficial effects in diseases associated with tissue inflammation, hypertrophy or fibrosis.
Collapse
|
11
|
Artwohl M, Lindenmair A, Roden M, Waldhäusl WK, Freudenthaler A, Klosner G, Ilhan A, Luger A, Baumgartner-Parzer SM. Fatty acids induce apoptosis in human smooth muscle cells depending on chain length, saturation, and duration of exposure. Atherosclerosis 2008; 202:351-62. [PMID: 18597758 DOI: 10.1016/j.atherosclerosis.2008.05.030] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 05/04/2008] [Accepted: 05/06/2008] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Plasma free fatty acid (FFA) concentrations are increased in states of insulin resistance. Therefore, this study evaluated apoptosis and underlying mechanisms induced by selected nutritional FFAs, a defined FFA-mix, and human plasma containing high FFA concentrations in human smooth muscle cells (HSMCs). RESEARCH DESIGN AND METHODS HSMCs were incubated (24-72 h) with selected FFAs (100-300 micromol/l), an FFA-mix (palmitic-/stearic-/oleic-/linoleic-/alpha-linolenic acid=2.6/1/3.6/9/1; 300-900 micromol/l), or with high FFA-plasma (600 micromol/l) versus respective control cultures. Apoptosis, caspase activation, and protein expression were determined by DNA-fragmentation assays, flow cytometry, and Western blots, respectively. RESULTS Exposure (24h) of HSMCs to 300 micromol/l stearic-, oleic-, linoleic-, alpha-linolenic-, and arachidonic acid induced apoptosis, correlating (p<0.01) with the FFAs' chain length (r=0.602) and number of FFA double bonds (r=0.956). After 48 h, 100 micromol/l of all tested FFAs - including palmitic acid - were already sufficient to trigger HSMCs' cell death. FFA-exposure resulted in activation of caspases and apoptosis was completely abolished by co-incubation with caspase inhibitors and negatively correlated (p<0.01) with the base-excision repair protein XRCC1 (r=-0.765) and with c-myc's antagonist mad (r=-0.916), whereas positive correlations (p<0.01) were found for protein expression of the proto-oncogene c-myc (r=0.972) and the transcription factor E2F-1 (r=0.971). Exposure of HSMCs to the defined FFA-mix and to plasma samples from individuals with elevated plasma FFAs supported the results obtained by defined FFA stimulation. CONCLUSIONS Since smooth muscle cells surround the macrophage/foam cell/lipid-laden artheromatous core of atherosclerotic lesions with a protective fibrous cap, their FFA-induced HSMC apoptosis could contribute to progression of atherosclerosis by thinning of the fibrous cap and subsequent plaque destabilization.
Collapse
Affiliation(s)
- Michaela Artwohl
- Department of Internal Medicine III, Clinical Division of Endocrinology and Metabolism, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Sweeney B, Puri P, Reen DJ. Induction and modulation of apoptosis in neonatal monocytes by polyunsaturated fatty acids. J Pediatr Surg 2007; 42:620-8. [PMID: 17448756 DOI: 10.1016/j.jpedsurg.2006.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Polyunsaturated fatty acids (PUFAs), known modulators of the immune response, are the source of essential fatty acids in total parenteral nutrition-dependent patients. Critically ill infants on TPN have an increased incidence of sepsis, and lipid emulsions depress various immune functions. Recent studies have demonstrated that PUFAs induce apoptosis in various tissue cells in vitro and ex vivo. The susceptibility of neonatal monocytes, as major early effector cells in the host response to sepsis, to PUFA-mediated apoptosis and the mechanisms associated with PUFA-induced apoptosis were investigated. Both n-3 and n-6 PUFAs induced rapid, dose-dependent cell death in purified monocytes. Polyunsaturated fatty acids induced significant activation of upstream caspases 8 and 9 as well as caspase 3. The PUFA treatment resulted in a 4-fold increase in oxidative stress and a loss of monocyte mitochondrial potential compared with carrier controls (P < .05). The addition of cyclosporin, which blocks the development of mitochondrial transition pores, completely abolished the proapoptotic effects of PUFAs. Although Trolox (Sigma Aldrich) reduced PUFA-induced intracellular oxidative stress in neonatal monocytes, apoptosis was not blocked by this potent antioxidant. The data identify PUFAs as potent inducers of monocyte apoptosis, which can occur independently of the induction of oxidative stress, by using a mitochondrial dependent pathway. The TPN-dependent infant may be particularly sensitive to such PUFA effects, having a relatively poor capacity to both use and clear PUFAs.
Collapse
Affiliation(s)
- Brian Sweeney
- Children's Research Centre, University College Dublin, Our Lady's Hospital for Sick Children, Dublin K1H 8L1, Ireland.
| | | | | |
Collapse
|
13
|
Touyz RM, Schiffrin EL. Peroxisome proliferator-activated receptors in vascular biology-molecular mechanisms and clinical implications. Vascul Pharmacol 2006; 45:19-28. [PMID: 16782410 DOI: 10.1016/j.vph.2005.11.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Revised: 11/01/2005] [Accepted: 11/01/2005] [Indexed: 01/27/2023]
Abstract
Peroxisome proliferator-activated receptors (PPAR)alpha, gamma and beta/delta belong to the nuclear receptor family of ligand-activated transcription factors. PPARs heterodimerize with the retinoid X receptor (RXR) and then act as transcription factors to modulate the function of many target genes. PPARalpha, gamma and beta/delta subtypes have significant differences in their ligand and gene specificities. PPARalpha is activated by polyunsaturated fatty acids and by fibrate drugs (fenofibrate and gemfibrozil) and controls expression of genes involved in lipid metabolism. PPARgamma is activated by fatty acid derivatives, such as hydroxyoctadecadienoic acid (HODEs), prostaglandin derivatives, such as 15-deoxy-Delta12,14-prostaglandin J2, and thiazolidinedione (glitazone) drugs, such as pioglitazone and rosiglitazone. PPARgamma is a key regulator of glucose homeostasis and adipogenesis. PPARbeta/delta ligands include polyunsaturated fatty acids, prostaglandins and synthetic compounds and stimulate fatty acid oxidation. All PPARs are expressed in vascular cells where they exert antiatherogenic, anti-inflammatory and vasculoprotective actions. Activators of PPARalpha (fibrates) and PPARgamma (thiazolidinediones or glitazones) antagonize angiotensin II effects in vivo and in vitro and have cardiovascular antioxidant and anti-inflammatory actions. PPAR agonists slightly reduce blood pressure are cardio-protective and correct vascular structure and endothelial dysfunction in experimental models of hypertension. Because of these beneficial effects, activators of PPARs may have therapeutic potential in the prevention of cardiovascular disease beyond their actions on carbohydrate and lipid metabolism. The present chapter focuses on the role of PPARs in vascular biology and discusses the clinical implications of using PPAR agonists in the management of vascular disease.
Collapse
Affiliation(s)
- Rhian M Touyz
- Kidney Research Centre, Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montreal, Quebec, Canada.
| | | |
Collapse
|
14
|
Dong F, Zhang X, Wold LE, Ren Q, Zhang Z, Ren J. Endothelin-1 enhances oxidative stress, cell proliferation and reduces apoptosis in human umbilical vein endothelial cells: role of ETB receptor, NADPH oxidase and caveolin-1. Br J Pharmacol 2005; 145:323-33. [PMID: 15765100 PMCID: PMC1576147 DOI: 10.1038/sj.bjp.0706193] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2004] [Revised: 12/17/2004] [Accepted: 02/03/2005] [Indexed: 11/08/2022] Open
Abstract
1 Endothelin-1 (ET-1), an endothelium-derived vasoactive peptide, participates in the regulation of endothelial function through mechanisms that are not fully elucidated. This study examined the impact of ET-1 on oxidative stress, apoptosis and cell proliferation in human umbilical vein endothelial cells (HUVEC). HUVECs were challenged for 24 h with ET-1 (10 pM-10 nM) in the absence or presence of the ET(B) receptor antagonist BQ788 (1 microM) or the NADPH oxidase inhibitor apocynin (1 microM). Reactive oxygen species (ROS) were detected using chloromethyl-2',7'-dichlorodihydrofluorescein diacetate. Apoptosis was evaluated with 4',6'-diamidino-2'-phenylindoladihydrochloride staining and by the caspase-3 assay. Cell proliferation was measured by a colorimetric assay. Expression of NADPH oxidase, Akt, pAkt, Bcl-2, Bax, IkappaB, caveolin-1 and eNOS was evaluated by Western blot analysis. 2 ET-1 significantly enhanced ROS generation and cell proliferation following 24-h incubation, both of which were prevented by BQ788 or apocynin, consistent with the ability of ET-1 to directly upregulate NADPH oxidase. ET-1 itself did not affect apoptosis but attenuated homocysteine-induced apoptosis through an ET(B) receptor-mediated mechanism. Western blot analysis indicated that ET-1 alleviated homocysteine (Hcy)-induced apoptosis, likely acting by antagonizing the Hcy-induced decreases in Akt, pAkt, pAkt-to-Akt, Bcl-2-to-Bax ratios and increases in Bax and caveolin-1 expression. Furthermore, ET-1 downregulated expression of caveolin-1 and eNOS, which was attenuated by BQ788 or apocynin. 3 In summary, our results suggest that ET-1 affects oxidative stress, proliferation and apoptosis possibly through ET(B), NADPH oxidase, Akt, Bax and caveolin-1-mediated mechanisms.
Collapse
Affiliation(s)
- Feng Dong
- Division of Pharmaceutical Sciences & Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, WY 82071-3375, U.S.A
| | - Xiaochun Zhang
- Division of Pharmaceutical Sciences & Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, WY 82071-3375, U.S.A
| | - Loren E Wold
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, Grand Forks, ND 58203, U.S.A
| | - Qun Ren
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071-3375, U.S.A
| | - Zhaojie Zhang
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071-3375, U.S.A
| | - Jun Ren
- Division of Pharmaceutical Sciences & Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, WY 82071-3375, U.S.A
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota, Grand Forks, ND 58203, U.S.A
| |
Collapse
|
15
|
Schiffrin EL. Vascular endothelin in hypertension. Vascul Pharmacol 2005; 43:19-29. [PMID: 15955745 DOI: 10.1016/j.vph.2005.03.004] [Citation(s) in RCA: 184] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2004] [Revised: 01/17/2005] [Accepted: 03/01/2005] [Indexed: 12/31/2022]
Abstract
Endothelins are powerful vasoconstrictor peptides that also play numerous other functions in many different organs. Endothelin-1 (ET-1) is the most abundant and important of this family of peptides in blood vessels. Production of ET-1 is increased in the endothelium and the kidney in salt-dependent models of hypertension (e.g.: DOCA-salt rats and Dahl salt-sensitive rats, in salt-loaded SHR-SP, in angiotensin II-infused and in diabetic rats). ET-1 elicits an inflammatory response by increasing oxidant stress in the vascular wall, which induces vascular remodeling and endothelial dysfunction found in the hypertensive models that exhibit an endothelin-mediated component. Endothelin receptor antagonism reduces blood pressure and vascular hypertrophic remodeling present in these hypertensive models. Patients with stage 2 hypertension have enhanced vascular expression of ET-1. Endothelin receptor antagonists lower blood pressure in hypertensive patients. They could become therapeutic agents for prevention of target organ damage in hypertension and in type 2 diabetes, chronic renal failure and congestive heart failure. Side effects of endothelin receptor blockers have prevented up to the present their development for these indications. New endothelin antagonists devoid of these side effects, or alternatively inhibitors of the endothelin converting enzymes that generate ET-1 may in the future become available to block the endothelin system. However, to date endothelin antagonists have been approved only for the treatment of primary pulmonary hypertension, a rapidly fatal condition in which the endothelin system plays an important role and endothelin antagonists exert favorable effects.
Collapse
Affiliation(s)
- Ernesto L Schiffrin
- CIHR Multidisciplinary Research Group on Hypertension, Clinical Research Institute of Montreal, 110 Pine Avenue West, Montreal, Quebec H2W 1R7, Canada.
| |
Collapse
|
16
|
Schiffrin EL. Peroxisome proliferator-activated receptors and cardiovascular remodeling. Am J Physiol Heart Circ Physiol 2005; 288:H1037-43. [DOI: 10.1152/ajpheart.00677.2004] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that heterodimerize with the retinoid X receptor and then modulate the function of many target genes. Three PPARs are known: α, β/δ, and γ. The better known are PPAR-α and PPAR-γ, which may be activated by different synthetic agonists, although the endogenous ligands are unknown. PPAR-α is involved in fatty acid oxidation and expressed in the liver, kidney, and skeletal muscle, whereas PPAR-γ is involved in fat cell differentiation, lipid storage, and insulin sensitivity. However, both have been shown to be present in variable amounts in cardiovascular tissues, including endothelium, smooth muscle cells, macrophages, and the heart. The activators of PPAR-α (fibrates) and PPAR-γ (thiazolidinediones or glitazones) antagonized the actions of angiotensin II in vivo and in vitro and exerted cardiovascular antioxidant and anti-inflammatory effects. PPAR activators lowered blood pressure, induced favorable effects on the heart, and corrected vascular structure and endothelial dysfunction in several rodent models of hypertension. Activators of PPARs may become therapeutic agents useful in the prevention of cardiovascular disease beyond their effects on carbohydrate and lipid metabolism. Some side effects, such as weight gain, as well as documented aggravation of advanced heart failure through fluid retention by glitazones, may, however, limit their therapeutic application in prevention of cardiovascular disease.
Collapse
|
17
|
Abstract
Beneficial effects of omega-3 polyunsaturated fatty acids (n-3 PUFA) have been reported in recent epidemiologic studies and randomized clinical trials in a variety of cardiovascular and autoimmune diseases. Fish and marine oils are the most abundant and convenient sources of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), the two major n-3 fatty acids that serve as substrates for cyclooxygenase and lipoxygenase pathways leading to less potent inflammatory mediators than those produced through the n-6 PUFA substrate, arachidonic acid. N-3 PUFA can also suppress inflammatory and/or immunologic responses through eicosanoid-independent mechanisms. Although the pathophysiology of IgA nephropathy is incompletely understood, it is likely that n-3 PUFA prevents renal disease progression by interfering with a number of effector pathways triggered by mesangial immune-complex deposition. In addition, potential targets of n-3 PUFA relevant to renal disease progression could be similar to those involved in preventing the development and progression of cardiovascular disease by lowering blood pressure, reducing serum lipid levels, decreasing vascular resistance, or preventing thrombosis. In IgA nephropathy, efficacy of n-3 PUFA contained in fish oil supplements has been tested with varying results. The largest randomized clinical trial performed by our collaborative group provided strong evidence that treatment for 2 years with a daily dose of 1.8 g of EPA and 1.2 g of DHA slowed the progression of renal disease in high-risk patients. These benefits persisted after 6.4 years of follow up. With safety, composition, and dosing convenience in mind, we can recommend two products that are available as pharmaceutical-grade fish-oil concentrates, Omacor (Pronova Biocare, Oslo, Norway) and Coromega (European Reference Botanical Laboratories, Carlsbad, CA).
Collapse
Affiliation(s)
- James V Donadio
- Division of Nephrology, the Department of Medicine, and the Department of Laboratory Medicine and Pathology, Mayo Clinic & Mayo Foundation, Rochester, MN, USA.
| | | |
Collapse
|
18
|
Hirafuji M, Machida T, Hamaue N, Minami M. Cardiovascular protective effects of n-3 polyunsaturated fatty acids with special emphasis on docosahexaenoic acid. J Pharmacol Sci 2003; 92:308-16. [PMID: 12939515 DOI: 10.1254/jphs.92.308] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
It is widely accepted that n-3 polyunsaturated fatty acids (PUFAs) rich in fish oils protect against several types of cardiovascular diseases such as myocardial infarction, arrhythmia, atherosclerosis, or hypertension. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) may be the active biological components of these effects. Although the precise cellular and molecular mechanisms underlying the beneficial effects are still uncertain, the protective effects of n-3 PUFAs are attributable to their direct effects on vascular smooth muscle cell (VSMC) functions. These n-3 PUFAs activate K(+)(ATP) channels and inhibit certain types of Ca(2+) channels, probably via at least 2 distinct mechanisms. N-3 PUFAs favorably alter the eicosanoid profile and regulate cytokine-induced expression of inducible nitric oxide synthase and cyclooxygenase-2 via mechanisms involving modulation of signaling transduction events. N-3 PUFAs also modulate VSMC proliferation, migration, and apoptosis. These recent data suggest that modulation of these VSMC functions contribute to the beneficial effects of n-3 PUFAs on various cardiovascular disorders. Furthermore, recent studies strongly suggest that DHA has more potent and beneficial effects than EPA. However, many questions about the cellular and molecular mechanisms still remain to be answered.
Collapse
Affiliation(s)
- Masahiko Hirafuji
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan.
| | | | | | | |
Collapse
|
19
|
Moreau P, Schiffrin EL. Role of endothelins in animal models of hypertension: focus on cardiovascular protection. Can J Physiol Pharmacol 2003; 81:511-21. [PMID: 12839263 DOI: 10.1139/y03-015] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Investigation of the regulation of vascular function by endothelium-derived factors has been a prominent topic of research in the field of hypertension during the last decade. Of the different endothelial factors, endothelins, which play an important role in vasodilatation-vasoconstriction balance, have been the subject of great interest and an impressive number of publications. This peptide, a very potent vasoconstrictor, triggers as well events involved in growth, proliferation, matrix production and local inflammation. In parallel, its role in hypertension has evolved from a simple vasoconstrictor to a central local regulator of vascular homeostasis contributing not only to the elevation of blood pressure, but also to the complications of hypertension. This review summarizes research on endothelins and its receptor antagonists in experimental hypertension, with special emphasis on vascular remodeling and target-organ protection.
Collapse
Affiliation(s)
- Pierre Moreau
- Laboratory of Vascular Pharmacology, Faculty of Pharmacy, Université de Montréal, PO Box 6128, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada.
| | | |
Collapse
|
20
|
Mayer K, Gokorsch S, Fegbeutel C, Hattar K, Rosseau S, Walmrath D, Seeger W, Grimminger F. Parenteral nutrition with fish oil modulates cytokine response in patients with sepsis. Am J Respir Crit Care Med 2003; 167:1321-8. [PMID: 12615625 DOI: 10.1164/rccm.200207-674oc] [Citation(s) in RCA: 168] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Infusion of fish oil-based (n-3) lipids may influence leukocyte function and plasma lipids in critical care patients. Twenty-one patients with sepsis requiring parenteral nutrition were randomized to receive an n-3 lipid emulsion rich in eicosapentaenoic acid and docosahexaenoic acid or a conventional (n-6) lipid emulsion (index fatty acid: arachidonic acid) for 5 days. The impact on plasma-free fatty acids, mononuclear leukocyte cytokine generation, and membrane fatty acid composition was examined. Cytokine synthesis by isolated mononuclear leukocyte was elicited by endotoxin. Before the onset of lipid infusion therapy, plasma-free fatty acid concentrations were greatly increased in septic patients, with arachidonic acid by far surpassing eicosapentaenoic acid and docosahexaenoic acid, a feature maintained during conventional lipid infusion. Within 2 days of fish oil infusion, free n-3 fatty acids increased, and the n-3/n-6 ratio was reversed, with rapid incorporation of n-3 fatty acids into mononuclear leukocyte membranes. Generation of proinflammatory cytokines by mononuclear leukocytes was markedly amplified during n-6 and was suppressed during n-3 lipid application. After termination of lipid administration, free n-3 fatty acid concentrations and mononuclear leukocyte cytokine synthesis returned to preinfusion values. Use of lipid infusions might allow us to combine intravenous alimentation with differential impact on inflammatory events and immunologic functions in patients with sepsis.
Collapse
Affiliation(s)
- Konstantin Mayer
- Medizinische Klinik II, Justus-Liebig-University, Giessen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Yusufi ANK, Cheng J, Thompson MA, Walker HJ, Gray CE, Warner GM, Grande JP. Differential effects of low-dose docosahexaenoic acid and eicosapentaenoic acid on the regulation of mitogenic signaling pathways in mesangial cells. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2003; 141:318-29. [PMID: 12761475 DOI: 10.1016/s0022-2143(03)00005-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although dietary fish oil supplementation has been used to prevent the progression of kidney disease in patients with IgA nephropathy, relatively few studies provide a mechanistic rationale for its use. Using an antithymocyte (ATS) model of mesangial proliferative glomerulonephritis, we recently demonstrated that fish oil inhibits mesangial cell (MC) activation and proliferation, reduces proteinuria, and decreases histologic evidence of glomerular damage. We therefore sought to define potential mechanisms underlying the antiproliferative effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), the predominant omega-3 polyunsaturated fatty acids found in fish oil, in cultured MC. DHA and EPA were administered to MC as bovine serum albumin fatty-acid complexes. Low-dose (10-50 micromol/L) DHA, but not EPA, inhibited basal and epidermal growth factor (EGF)-stimulated [(3)H]-thymidine incorporation in MCs. At higher doses (100 micromol/L), EPA and DHA were equally effective in suppressing basal and EGF-stimulated MC mitogenesis. Low-dose DHA, but not EPA, decreased ERK activation by 30% (P <.01), as assessed with Western-blot analysis using phosphospecific antibodies. JNK activity was increased by low-dose DHA but not by EPA. p38 activity was not significantly altered by DHA or EPA. Cyclin E activity, as assessed with a histone H1 kinase assay, was inhibited by low-dose DHA but not by EPA. DHA increased expression of the cell cycle inhibitor p21 but not p27; EPA had no effect on p21 or p27. We propose that the differential effect of low-dose DHA vs EPA in suppressing MC mitogenesis is related to down-regulation of ERK and cyclin E activity and to induction of p21.
Collapse
Affiliation(s)
- Ahad N K Yusufi
- Renal Pathophysiology Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Endothelin-1 (ET-1) was first characterized as a potent vasoconstrictor and is overexpressed in the vasculature in different models of hypertension, such as deoxycorticosterone acetate-salt rats, Dahl salt-sensitive rats, and stroke-prone spontaneously hypertensive rats. Moreover, patients with moderate to severe hypertension present increased vascular levels of prepro-ET-1 mRNA. In addition to their blood pressure-lowering effects, ET receptor antagonists are able to reduce vascular growth. Recent data suggest the involvement of an inflammatory response in the effects of ET-1, which contributes to vascular remodeling and endothelial dysfunction. Increasing evidence underscores the potential therapeutic benefit of ET receptor antagonists in different hypertension-related complications, not only in essential hypertension, but also in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Marc Iglarz
- Clinical Research Institute of Montreal, 110 Pine Avenue West, Quebec H2W 1R7, Canada
| | | |
Collapse
|
23
|
Eberle J, Fecker LF, Orfanos CE, Geilen CC. Endothelin-1 decreases basic apoptotic rates in human melanoma cell lines. J Invest Dermatol 2002; 119:549-55. [PMID: 12230494 DOI: 10.1046/j.1523-1747.2002.01848.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Normal human melanocytes respond to endothelin-1 with induced proliferation and differentiation. Whereas in cultured melanoma cells the predominant endothelin receptor, ET(B)-R, is consistently downregulated, ET(B)-R upregulation was recently reported for melanoma tumors. Contrary to the pro-survival activity described for endothelin in vascular cells, a proapoptotic activity of endothelin-1 has been reported for melanoma cells, in previous studies. We therefore investigated the role of endothelin for melanoma cells with respect to apoptosis and proliferation. Treatment with 10 nM endothelin-1 was a strong mitogenic signal for normal human melanocytes, which responded with a 4-6-fold increase of thymidine incorporation, whereas the response was only 1.2-fold for SK-Mel-19, the melanoma cell line characterized by the highest ET(B)-R expression, and it was even less in other cell lines. Determination of the apoptotic rates revealed that endothelin-1 significantly reduced basic apoptotic rates to 75% both in SK-Mel-19 and in normal melanocytes. After cell synchronization, an antiapoptotic effect of endothelin-1 was seen in five of seven cell lines investigated. In the cell line Bro, which showed no response and which lacks ET(B)-R expression, responsibility could be restored by overexpression of ET(B)-R after stable transfection, indicating that the effectors of the endothelin-1 signal cascade were active in these cells, and that the antiapoptotic effect of endothelin-1 is mediated in a receptor-specific way. This antiapoptotic activity of endothelin for melanoma cells combined with upregulation of endothelin receptors in the tumor may be a crucial step for melanoma progression.
Collapse
Affiliation(s)
- Jürgen Eberle
- Department of Dermatology, University Medical Center Benjamin Franklin, The Free University of Berlin, Berlin, Germany.
| | | | | | | |
Collapse
|
24
|
Lafuste P, Robert B, Mondon F, Danan JL, Rossi B, Duc-Goiran P, Mignot TM, Nunez EA, Benassayag C, Ferré F. Alpha-fetoprotein gene expression in early and full-term human trophoblast. Placenta 2002; 23:600-12. [PMID: 12361680 DOI: 10.1053/plac.2002.0816] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Alpha-fetoprotein (AFP) is a major serum glycoprotein synthesized during fetal life mainly by the yolk sac and the fetal liver. At term, it reaches high concentrations in the maternal intervillous blood, which is in direct contact with the placental trophoblastic microvillous membrane, and this suggests the placental origin of the AFP at the fetal-maternal interface. We used several experimental approaches to investigate the expression of AFP gene and fetal protein production in early gestation and term placentas. RT-PCR and immunological studies clearly identified AFP messenger RNA and AFP protein in the placental villi from first trimester of pregnancy. The AFP gene was also expressed in highly purified cytotrophoblasts from early placentas, and enzymo-immunoassay showed that AFP protein was synthesized and secreted by early cytotrophoblasts. AFP was also detected in the cytoplasm of these cells by immuno-cytochemistry. However, none of these methods detected any expression of the AFP gene in full-term placental villi or in cultured trophoblasts. These findings demonstrate that both AFP mRNA and protein are present in trophoblastic cells early in pregnancy. The absence of AFP gene expression in term placental villi also suggests, that the AFP at the fetal-maternal interface is attributable to a notable transplacental passage of AFP from fetal blood in late pregnancy.
Collapse
Affiliation(s)
- P Lafuste
- INSERM U. 361, Descartes University, 75014 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Del Bufalo D, Di Castro V, Biroccio A, Varmi M, Salani D, Rosanò L, Trisciuoglio D, Spinella F, Bagnato A. Endothelin-1 protects ovarian carcinoma cells against paclitaxel-induced apoptosis: requirement for Akt activation. Mol Pharmacol 2002; 61:524-32. [PMID: 11854432 DOI: 10.1124/mol.61.3.524] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Endothelin-1 (ET-1) is a powerful mitogenic peptide produced by different tumors. In ovarian carcinoma cells, ET-1 acts as an autocrine growth factor, selectively through ET(A) receptor (ET(A)R), which is predominantly expressed in tumor cells. The aim of this study was to examine whether ET-1 plays a role in the sensitivity of three ovarian carcinoma cell lines (OVCA 433, HEY, and SK-OV-3) to apoptosis induced by two different stimuli. Our results demonstrated that the addition of ET-1 markedly inhibited serum withdrawal and paclitaxel-induced apoptosis in a concentration-dependent manner, as demonstrated by Annexin-V assay, sub-G(1) peak in DNA content histograms, internucleosomal DNA fragmentation, and terminal deoxynucleotidyl transferase-mediated dUTP biotin nick-end labeling method. Pretreatment of the cells with an ET(A)R antagonist, BQ 123, reversed the ET-1-induced protective effect. Paclitaxel-induced apoptosis resulted in the phosphorylation of Bcl-2 that was suppressed by the addition of ET-1. Further analysis of the signaling pathway demonstrated that ET-1 stimulated Akt activation. The phosphatidylinositol 3-kinase (PI3-K) inhibitor wortmannin blocked ET-1-induced Akt phosphorylation. Inhibition of ET-1-stimulated mitogen-activated protein kinase activity did not affect ET-1 protection from paclitaxel-mediated apoptosis. Moreover, BQ 123 blocked the Akt-mediated pathway activated by ET-1, sensitizing ovarian carcinoma cells to paclitaxel treatment. These results establish a novel role for ET-1 in determining protection of ovarian carcinoma cells against paclitaxel-induced apoptosis through Bcl-2-dependent and PI3-K-mediated Akt pathways and suggest that ET-1 and ET(A)R could represent important targets for anticancer therapy.
Collapse
Affiliation(s)
- Donatella Del Bufalo
- Experimental Chemotherapy Laboratories, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Cattaruzza M, Berger MM, Ochs M, Fayyazi A, Füzesi L, Richter J, Hecker M. Deformation-induced endothelin B receptor-mediated smooth muscle cell apoptosis is matrix-dependent. Cell Death Differ 2002; 9:219-26. [PMID: 11840172 DOI: 10.1038/sj.cdd.4400936] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2001] [Revised: 07/17/2001] [Accepted: 07/24/2001] [Indexed: 11/08/2022] Open
Abstract
To maintain normal blood flow, pressure overload in both arteries and veins requires a structural adaptation of the vessel wall (remodelling) that involves smooth muscle cell (SMC) hypertrophy and/or hyperplasia. Due to its potent vasoconstrictor and growth-promoting effects, endothelin-1 (ET-1) is a likely candidate to initiate and/or promote remodelling in blood vessels exposed to a chronic increase in blood pressure. To test this hypothesis, isolated segments of the rabbit carotid artery and jugular vein were perfused at different levels of intraluminal pressure. In both types of segments, pressure overload (160 and 20 mmHg, respectively) resulted in an increase in endothelial prepro-ET-1 and SMC endothelin B receptor (ETB-R) expression. Moreover, in pressurised segments from the carotid artery an ETB-R antagonist-sensitive increase in SMC apoptosis in the media was observed, while in the vein medial SMC started to proliferate. Isolated SMC from these rabbit blood vessels as well as from the aorta and vena cava of the rat, when cultured on a collagen or laminin matrix, uniformly revealed an ETB-R-mediated increase in apoptosis upon exposure to mechanical deformation plus exogenous ET-1 (10 nmol/L). However, when grown on a fibronectin matrix, the cultured SMC did not respond with an increase in apoptosis under otherwise identical experimental conditions. These findings suggest that deformation-induced activation of the endothelin system in the vessel wall not only plays a crucial role in remodelling, but that the structural components of the vessel wall, in particular the cell-matrix interaction, determine how SMC respond phenotypically to these changes in gene expression.
Collapse
MESH Headings
- Animals
- Apoptosis
- Carotid Arteries/metabolism
- Carotid Arteries/ultrastructure
- Caspase 3
- Caspases/metabolism
- Cell Differentiation
- Cell Division
- Chromatin/ultrastructure
- Culture Techniques
- Endothelin Receptor Antagonists
- Endothelin-1/genetics
- Endothelin-1/metabolism
- Extracellular Matrix Proteins/physiology
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/ultrastructure
- Oligopeptides/pharmacology
- Peptides, Cyclic/pharmacology
- Piperidines/pharmacology
- Pressure
- RNA, Messenger/biosynthesis
- Rabbits
- Rats
- Rats, Wistar
- Receptor, Endothelin B
- Receptors, Endothelin/genetics
- Receptors, Endothelin/physiology
- Stress, Mechanical
- Veins/ultrastructure
Collapse
Affiliation(s)
- M Cattaruzza
- Department of Cardiovascular Physiology, University of Goettingen, Humboldtallee 23, 37073 Goettingen, Germany
| | | | | | | | | | | | | |
Collapse
|
27
|
Orlandi A, Marcellini M, Pesce D, Calvani M, Spagnoli LG. Propionyl-L-carnitine reduces intimal hyperplasia after injury in normocholesterolemic rabbit carotid artery by modulating proliferation and caspase 3-dependent apoptosis of vascular smooth muscle cells. Atherosclerosis 2002; 160:81-9. [PMID: 11755925 DOI: 10.1016/s0021-9150(01)00568-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Previously we documented that propionyl-L-carnitine (PLC) reduces the growth of atherosclerotic lesions in cholesterol-fed aged rabbits in association with a decrease of plaque smooth muscle cell (SMC) proliferation and plasma triglycerides. To clarify whether PLC might influence SMC growth through mechanisms other than triglyceride lowering, we investigated the effect of a daily treatment per os with PLC on carotid intimal hyperplasia after ballooning in normocholesterolemic rabbits. PLC did not induce variations of plasma triglyceride and cholesterol. One week later, the number of proliferating SMCs was reduced in PLC as compared with controls. After 3 weeks, morphometric analysis demonstrated a reduced neointimal relative volume and percentage of stenosis but not vessel area in PLC as compared with controls. This associated with an intimal reduced SMC number and an increased apoptotic rate as detected by nick-end labelling (TUNEL) and ligation-mediated polymerase chain reaction (PCR). Western blotting also demonstrated an increase of caspase-3 cleavage in PLC carotids. Antiproliferative and pro-apoptotic effects of PLC were confirmed in vitro on actively proliferating and serum deprived SMCs, respectively. Molecules with an additional cell-specific, pro-apoptotic action may represent a new therapeutic tool in reducing intimal SMC hyperplasia following angioplasty or stenting procedures.
Collapse
Affiliation(s)
- Augusto Orlandi
- Department of Biopathology and Image Diagnostics, Anatomic Pathology Institute, Tor Vergata University of Rome, Via della Ricerca Scientifica, 00133 Rome, Italy
| | | | | | | | | |
Collapse
|
28
|
Filippatos GS, Gangopadhyay N, Lalude O, Parameswaran N, Said SI, Spielman W, Uhal BD. Regulation of apoptosis by vasoactive peptides. Am J Physiol Lung Cell Mol Physiol 2001; 281:L749-61. [PMID: 11557578 DOI: 10.1152/ajplung.2001.281.4.l749] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Although originally discovered because of their ability to affect hemodynamics, vasoactive peptides have been found to function in a variety of capacities including neurotransmission, endocrine functions, and the regulation of cell proliferation. A growing body of evidence describes the ability of vasoactive peptides to regulate cell death by apoptosis in either a positive or negative fashion depending on the peptide and the type of target cell. The available evidence to date is strongest for the peptides endothelin, angiotensin II, vasoactive intestinal peptide, atrial natriuretic peptide, and adrenomedullin. Each of these peptides is discussed, with specific regard to apoptosis, in terms of regulatory activity, target cell specificity, and potential role in pulmonary physiology.
Collapse
Affiliation(s)
- G S Filippatos
- Second Division of Cardiology, Evangelismos General Hospital, GR-11526 Athens, Greece
| | | | | | | | | | | | | |
Collapse
|