1
|
Qian W, Yu H, Zhang C, Zhang H, Fu S, Xia C. Plasma Proteomics Characteristics of Subclinical Vitamin E Deficiency of Dairy Cows During Early Lactation. Front Vet Sci 2021; 8:723898. [PMID: 34957273 PMCID: PMC8703030 DOI: 10.3389/fvets.2021.723898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/15/2021] [Indexed: 11/20/2022] Open
Abstract
Vitamin E (VE) is an essential fat-soluble nutrient for dairy cows. Vitamin E deficiency leads to immune suppression and oxidative stress and increases the susceptibility of cows to reproductive disorders in the early post-partum period. However, studies on plasma proteomics of VE deficiency have not been reported so far. Therefore, the purpose of this study was to understand the changes of blood protein profile in cows with subclinical VE deficiency in the early post-partum period. In this study, plasma protein levels of 14 healthy cows (>4 μg/ml α-tocopherol) and 13 subclinical VE-deficient cows (2–3 μg/ml α-tocopherol) were analyzed by tandem mass tag (TMT). The results showed that there were 26 differentially expressed proteins (DEPs) in the plasma of cows with subclinical VE deficiency compared with healthy controls. Twenty-one kinds of proteins were downregulated, and five kinds were upregulated, among which eight proteins in protein–protein interactions (PPI) network had direct interaction. These proteins are mainly involved in the MAPK signaling pathway, pantothenic acid and coenzyme A (CoA) biosynthesis, PPAR signaling pathway, and glycosylphosphatidylinositol (GPI)-anchor biosynthesis. The top four DEPs in PPI (APOC3, APOC4, SAA4, PHLD) and one important protein (VNN1) by literature review were further verified by ELISA and Western blot. The expression levels of APOC3, VNN1, and SAA4 were significantly lower than those of healthy controls by ELISA. VNN1 was significantly lower than those of healthy controls by Western blot. VNN1 is closely related to dairy cow subclinical VE deficiency and can be a potential biomarker. It lays a foundation for further research on the lack of pathological mechanism and antioxidative stress of VE.
Collapse
Affiliation(s)
- Weidong Qian
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hongyi Yu
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Cuiyu Zhang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hongyou Zhang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shixin Fu
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Cheng Xia
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
2
|
Agarwal S, Yadav A, Chaturvedi RK. Peroxisome proliferator-activated receptors (PPARs) as therapeutic target in neurodegenerative disorders. Biochem Biophys Res Commun 2016; 483:1166-1177. [PMID: 27514452 DOI: 10.1016/j.bbrc.2016.08.043] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/21/2016] [Accepted: 08/07/2016] [Indexed: 01/06/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors and they serve to be a promising therapeutic target for several neurodegenerative disorders, which includes Parkinson disease, Alzheimer's disease, Huntington disease and Amyotrophic Lateral Sclerosis. PPARs play an important role in the downregulation of mitochondrial dysfunction, proteasomal dysfunction, oxidative stress, and neuroinflammation, which are the major causes of the pathogenesis of neurodegenerative disorders. In this review, we discuss about the role of PPARs as therapeutic targets in neurodegenerative disorders. Several experimental approaches suggest potential application of PPAR agonist as well as antagonist in the treatment of neurodegenerative disorders. Several epidemiological studies found that the regular usage of PPAR activating non-steroidal anti-inflammatory drugs is effective in decreasing the progression of neurodegenerative diseases including PD and AD. We also reviewed the neuroprotective effects of PPAR agonists and associated mechanism of action in several neurodegenerative disorders both in vitro as well as in vivo animal models.
Collapse
Affiliation(s)
- Swati Agarwal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India
| | - Anuradha Yadav
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India.
| |
Collapse
|
3
|
Giordano Attianese GMP, Desvergne B. Integrative and systemic approaches for evaluating PPARβ/δ (PPARD) function. NUCLEAR RECEPTOR SIGNALING 2015; 13:e001. [PMID: 25945080 PMCID: PMC4419664 DOI: 10.1621/nrs.13001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/09/2015] [Indexed: 12/13/2022]
Abstract
The peroxisome proliferator-activated receptors (PPARs) are a group of nuclear receptors that function as transcription factors regulating the expression of genes involved in cellular differentiation, development, metabolism and also tumorigenesis. Three PPAR isotypes (α, β/δ and γ) have been identified, among which PPARβ/δ is the most difficult to functionally examine due to its tissue-specific diversity in cell fate determination, energy metabolism and housekeeping activities. PPARβ/δ acts both in a ligand-dependent and -independent manner. The specific type of regulation, activation or repression, is determined by many factors, among which the type of ligand, the presence/absence of PPARβ/δ-interacting corepressor or coactivator complexes and PPARβ/δ protein post-translational modifications play major roles. Recently, new global approaches to the study of nuclear receptors have made it possible to evaluate their molecular activity in a more systemic fashion, rather than deeply digging into a single pathway/function. This systemic approach is ideally suited for studying PPARβ/δ, due to its ubiquitous expression in various organs and its overlapping and tissue-specific transcriptomic signatures. The aim of the present review is to present in detail the diversity of PPARβ/δ function, focusing on the different information gained at the systemic level, and describing the global and unbiased approaches that combine a systems view with molecular understanding.
Collapse
|
4
|
Kim DG, Yoo JC, Kim E, Lee YS, Yarishkin OV, Lee DY, Lee KH, Hong SG, Hwang EM, Park JY. A Novel Cytosolic Isoform of Mitochondrial Trans-2-Enoyl-CoA Reductase Enhances Peroxisome Proliferator-Activated Receptor α Activity. Endocrinol Metab (Seoul) 2014; 29:185-94. [PMID: 25031892 PMCID: PMC4091492 DOI: 10.3803/enm.2014.29.2.185] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/11/2013] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Mitochondrial trans-2-enoyl-CoA reductase (MECR) is involved in mitochondrial synthesis of fatty acids and is highly expressed in mitochondria. MECR is also known as nuclear receptor binding factor-1, which was originally reported with yeast two-hybrid screening as a binding protein of the nuclear hormone receptor peroxisome proliferator-activated receptor α (PPARα). However, MECR and PPARα are localized at different compartment, mitochondria, and the nucleus, respectively. Therefore, the presence of a cytosolic or nuclear isoform of MECR is necessary for functional interaction between MECR and PPARα. METHODS To identify the expression pattern of MECR and the cytosolic form of MECR (cMECR), we performed reverse transcription polymerase chain reaction (RT-PCR) with various tissue samples from Sprague-Dawley rats. To confirm the interaction between cMECR and PPARα, we performed several binding assays such as yeast two-hybrid, coimmunoprecipitation, and bimolecular fluorescence complementation. To observe subcellular localization of these proteins, immunocytochemistry was performed. A luciferase assay was used to measure PPARα activity. RESULTS We provide evidence of an alternatively spliced variant of the rat MECR gene that yields cMECR. The cMECR lacks the N-terminal 76 amino acids of MECR and shows uniform distribution in the cytoplasm and nucleus of HeLa cells. cMECR directly bound PPARα in the nucleus and increased PPARα-dependent luciferase activity in HeLa cells. CONCLUSION We found the cytosolic form of MECR (cMECR) was expressed in the cytosolic and/or nuclear region, directly binds with PPARα, and enhances PPARα activity.
Collapse
Affiliation(s)
- Dong-Gyu Kim
- Department of Physiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Jae Cheal Yoo
- Department of Physiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Eunju Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Korea
| | - Young-Sun Lee
- Department of Physiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Oleg V. Yarishkin
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Korea
| | - Da Yong Lee
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Korea
| | - Kun Ho Lee
- Department of Marine Life Science, Chosun University College of Natural Sciences, Gwangju, Korea
| | - Seong-Geun Hong
- Department of Physiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Korea
| | - Jae-Yong Park
- Department of Physiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju, Korea
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Korea
| |
Collapse
|
5
|
Fruchart JC, Davignon J, Hermans MP, Al-Rubeaan K, Amarenco P, Assmann G, Barter P, Betteridge J, Bruckert E, Cuevas A, Farnier M, Ferrannini E, Fioretto P, Genest J, Ginsberg HN, Gotto AM, Hu D, Kadowaki T, Kodama T, Krempf M, Matsuzawa Y, Núñez-Cortés JM, Monfil CC, Ogawa H, Plutzky J, Rader DJ, Sadikot S, Santos RD, Shlyakhto E, Sritara P, Sy R, Tall A, Tan CE, Tokgözoğlu L, Toth PP, Valensi P, Wanner C, Zambon A, Zhu J, Zimmet P. Residual macrovascular risk in 2013: what have we learned? Cardiovasc Diabetol 2014; 13:26. [PMID: 24460800 PMCID: PMC3922777 DOI: 10.1186/1475-2840-13-26] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/07/2013] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease poses a major challenge for the 21st century, exacerbated by the pandemics of obesity, metabolic syndrome and type 2 diabetes. While best standards of care, including high-dose statins, can ameliorate the risk of vascular complications, patients remain at high risk of cardiovascular events. The Residual Risk Reduction Initiative (R3i) has previously highlighted atherogenic dyslipidaemia, defined as the imbalance between proatherogenic triglyceride-rich apolipoprotein B-containing-lipoproteins and antiatherogenic apolipoprotein A-I-lipoproteins (as in high-density lipoprotein, HDL), as an important modifiable contributor to lipid-related residual cardiovascular risk, especially in insulin-resistant conditions. As part of its mission to improve awareness and clinical management of atherogenic dyslipidaemia, the R3i has identified three key priorities for action: i) to improve recognition of atherogenic dyslipidaemia in patients at high cardiometabolic risk with or without diabetes; ii) to improve implementation and adherence to guideline-based therapies; and iii) to improve therapeutic strategies for managing atherogenic dyslipidaemia. The R3i believes that monitoring of non-HDL cholesterol provides a simple, practical tool for treatment decisions regarding the management of lipid-related residual cardiovascular risk. Addition of a fibrate, niacin (North and South America), omega-3 fatty acids or ezetimibe are all options for combination with a statin to further reduce non-HDL cholesterol, although lacking in hard evidence for cardiovascular outcome benefits. Several emerging treatments may offer promise. These include the next generation peroxisome proliferator-activated receptorα agonists, cholesteryl ester transfer protein inhibitors and monoclonal antibody therapy targeting proprotein convertase subtilisin/kexin type 9. However, long-term outcomes and safety data are clearly needed. In conclusion, the R3i believes that ongoing trials with these novel treatments may help to define the optimal management of atherogenic dyslipidaemia to reduce the clinical and socioeconomic burden of residual cardiovascular risk.
Collapse
Affiliation(s)
- Jean-Charles Fruchart
- R3i Foundation, St. Alban-Anlage 46, Basel, CH 4010, Switzerland
- Fondation Cœur et Artères, Lille, France
| | - Jean Davignon
- Institut de recherches cliniques de Montréal; Centre Hospitalier de l’Université de Montréal and Department of Experimental Medicine, McGill University, Montreal, Canada
| | | | - Khalid Al-Rubeaan
- University Diabetes Center, King Saud University, Riyadh, Saudi Arabia
| | - Pierre Amarenco
- Department of Neurology and Stroke Centre, Bichat University Hospital, Paris, France
| | - Gerd Assmann
- Assmann-Stiftung für Prävention, Münster, Germany
| | - Philip Barter
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | | | - Eric Bruckert
- Department of Endocrinology and Cardiovascular Disease Prevention, Institut of CardioMetabolism and Nutrition (ICAN) Hôpital Pitié-Salpêtrière, Paris, France
| | - Ada Cuevas
- Nutrition Center, Clínica Las Condes, Santiago, Chile
| | | | - Ele Ferrannini
- University of Pisa School of Medicine, and Metabolism Unit of the National Research Council (CNR) Institute of Clinical Physiology, Pisa, Italy
| | - Paola Fioretto
- Department of Medical and Surgical Sciences, University of Padova, Padova, Italy
| | - Jacques Genest
- McGill University and Center for Innovative Medicine, McGill University Health Center/Royal Victoria Hospital, Montreal, Canada
| | - Henry N Ginsberg
- Department of Medicine and Irving Institute for Clinical and Translational Research, Columbia University, New York, USA
| | - Antonio M Gotto
- Weill Cornell Medical College, Cornell University, New York, USA
| | - Dayi Hu
- Heart Institute, People Hospital of Peking University, Beijing, China
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases Unit, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- Department of Systems Biology and Medicine, The University of Tokyo, Tokyo, Japan
| | - Michel Krempf
- Human Nutritional Research Center and Department of Endocrinology, Metabolic Diseases and Nutrition, University Hospital Nantes, Nantes, France
| | | | | | | | - Hisao Ogawa
- Department of Cardiovascular Medicine, Kumamoto University, Kumamoto, Japan
| | - Jorge Plutzky
- Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Smilow Center for Translational Research, Penn Cardiovascular Institute, Philadelphia, PA, USA
| | | | - Raul D Santos
- Unidade Clínica de Lipides InCor-HCFMUSP, Sao Paulo, Brazil
| | - Evgeny Shlyakhto
- Federal Almazov Heart Blood Endocrinology Centre, St Petersburg, Russia
| | | | - Rody Sy
- University of the Philippines-Philippine General Hospital, Manila, The Philippines
| | - Alan Tall
- Specialized Center of Research (SCOR) in Molecular Medicine and Atherosclerosis, Columbia University, College of Physicians & Surgeons, New York, USA
| | | | | | - Peter P Toth
- Sterling Rock Falls Clinic, CGH Medical Center, Sterling and University of Illinois School of Medicine, Peoria, IL, USA
| | - Paul Valensi
- Hôpital Jean Verdier, Department of Endocrinology Diabetology Nutrition, AP-HP, Paris-Nord University, CRNH-IdF, CINFO, Bondy, France
| | | | - Alberto Zambon
- Department of Medical and Surgical Sciences, University of Padova, Padova, Italy
| | - Junren Zhu
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Paul Zimmet
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
6
|
Polyacetylenes from Notopterygium incisum--new selective partial agonists of peroxisome proliferator-activated receptor-gamma. PLoS One 2013; 8:e61755. [PMID: 23630612 PMCID: PMC3632601 DOI: 10.1371/journal.pone.0061755] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 03/12/2013] [Indexed: 12/22/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a key regulator of glucose and lipid metabolism and therefore an important pharmacological target to combat metabolic diseases. Since the currently used full PPARγ agonists display serious side effects, identification of novel ligands, particularly partial agonists, is highly relevant. Searching for new active compounds, we investigated extracts of the underground parts of Notopterygium incisum, a medicinal plant used in traditional Chinese medicine, and observed significant PPARγ activation using a PPARγ-driven luciferase reporter model. Activity-guided fractionation of the dichloromethane extract led to the isolation of six polyacetylenes, which displayed properties of selective partial PPARγ agonists in the luciferase reporter model. Since PPARγ activation by this class of compounds has so far not been reported, we have chosen the prototypical polyacetylene falcarindiol for further investigation. The effect of falcarindiol (10 µM) in the luciferase reporter model was blocked upon co-treatment with the PPARγ antagonist T0070907 (1 µM). Falcarindiol bound to the purified human PPARγ receptor with a Ki of 3.07 µM. In silico docking studies suggested a binding mode within the ligand binding site, where hydrogen bonds to Cys285 and Glu295 are predicted to be formed in addition to extensive hydrophobic interactions. Furthermore, falcarindiol further induced 3T3-L1 preadipocyte differentiation and enhanced the insulin-induced glucose uptake in differentiated 3T3-L1 adipocytes confirming effectiveness in cell models with endogenous PPARγ expression. In conclusion, we identified falcarindiol-type polyacetylenes as a novel class of natural partial PPARγ agonists, having potential to be further explored as pharmaceutical leads or dietary supplements.
Collapse
|
7
|
Sharoni Y, Linnewiel-Hermoni K, Khanin M, Salman H, Veprik A, Danilenko M, Levy J. Carotenoids and apocarotenoids in cellular signaling related to cancer: a review. Mol Nutr Food Res 2011; 56:259-69. [PMID: 22102431 DOI: 10.1002/mnfr.201100311] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 08/22/2011] [Accepted: 09/15/2011] [Indexed: 01/01/2023]
Abstract
The basis for the vivid color of carotenoids and their antioxidant activity is the multiple conjugated double bonds, which are characteristic for these phytonutrients. Moreover, the cleavage of these oxidation-prone double bonds leads to the formation of apocarotenoids. A large number of carbonyl-containing oxidation products are expected to be produced as a result of carotenoid oxidation and these can be further metabolized into the corresponding acids and alcohols. As discussed in this review, many, but not all, of these potential products have been detected and identified in plants as well as in human and animal plasma and tissues. Some of these compounds were found to be biologically active as anticancer agents. In addition to the inhibition of cancer cell proliferation, several carotenoid metabolites were shown to modulate the activity of various transcription systems. These include ligand-activated nuclear receptors, such as the retinoic acid receptor, retinoid X receptor, peroxisome proliferator-activated receptor and estrogen receptor, as well as other transcription systems that have an important role in cancer, such as the electrophile/antioxidant response element pathway and nuclear factor-κB. Therefore, apocarotenoids can be considered as natural compounds with multifunctional, rather than monofunctional, activity and, thus, can be useful in the prevention of cancer and other degenerative diseases.
Collapse
Affiliation(s)
- Yoav Sharoni
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | | | | | | | | | | | |
Collapse
|
8
|
von Lintig J. Colors with functions: elucidating the biochemical and molecular basis of carotenoid metabolism. Annu Rev Nutr 2010; 30:35-56. [PMID: 20415581 DOI: 10.1146/annurev-nutr-080508-141027] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Carotenoids affect a rich variety of physiological functions in nature and are beneficial for human health, serving as antioxidants in lipophilic environments and blue light filters in the macula of human retina. These dietary compounds also serve as precursors of a unique set of apo-carotenoid cleavage products, including retinoids. Although knowledge about retinoid biology has tremendously increased, the metabolism of retinoids' parent precursors remains poorly understood. Recently, molecular players in carotenoid metabolism have been identified and biochemically characterized. Moreover, mutations in their corresponding genes impair carotenoid metabolism and induce various pathologies in animal models. Polymorphisms in these genes alter carotenoid and retinoid homeostasis in humans as well. This review summarizes our current knowledge about the molecular/biochemical basis of carotenoid metabolism and particularly the physiological role of carotenoids in retinoid-dependent physiological processes.
Collapse
Affiliation(s)
- Johannes von Lintig
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106-4965, USA.
| |
Collapse
|
9
|
Supplementation with EPA or fish oil for 11 months lowers circulating lipids, but does not delay the onset of diabetes in UC Davis-type 2 diabetes mellitus rats. Br J Nutr 2010; 104:1628-34. [PMID: 20735878 DOI: 10.1017/s0007114510002655] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
EPA or fish oil supplementation has been suggested as treatments for the prevention of type 2 diabetes mellitus (T2DM) due to their lipid-lowering and potential insulin-sensitising effects. We investigated the effects of supplementation with EPA (1 g/kg body weight per d) or fish oil (3 g/kg body weight per d) on the age of onset of T2DM and circulating glucose, insulin, lipids, leptin and adiponectin in UC Davis (UCD)-T2DM rats. Animals were divided into three groups starting at 1 month of age: control, EPA and fish oil. All the animals were followed until diabetes onset or for up to 12 months of age. Monthly fasting blood samples were collected for the measurement of glucose, lipids, hormones and C-reactive protein (CRP). Neither EPA nor fish oil delayed the onset of T2DM or altered fasting plasma glucose, insulin, CRP, adiponectin or leptin concentrations. The groups did not differ in energy intake or body weight. Fish oil treatment lowered fasting plasma TAG concentrations by 39 (sd 7) % (P < 0.001) and EPA lowered fasting plasma NEFA concentrations by 23 (sd 5) % (P < 0.05) at 4 months of age compared with the control group. EPA and fish oil lowered fasting plasma cholesterol concentrations at 4 months of age by 19 (sd 4) and 22 (sd 4) % compared with the control group, respectively (both P < 0.01). In conclusion, EPA and fish oil supplementation lowers circulating lipid concentrations, but does not delay the onset of T2DM in UCD-T2DM rats.
Collapse
|
10
|
Ruby MA, Goldenson B, Orasanu G, Johnston TP, Plutzky J, Krauss RM. VLDL hydrolysis by LPL activates PPAR-alpha through generation of unbound fatty acids. J Lipid Res 2010; 51:2275-81. [PMID: 20421589 DOI: 10.1194/jlr.m005561] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent evidence suggests that lipoproteins serve as circulating reservoirs of peroxisomal proliferator activated receptor (PPAR) ligands that are accessible through lipolysis. The present study was conducted to determine the biochemical basis of PPAR-alpha activation by lipolysis products and their contribution to PPAR-alpha function in vivo. PPAR-alpha activation was measured in bovine aortic endothelial cells following treatment with human plasma, VLDL lipolysis products, or oleic acid. While plasma failed to activate PPAR-alpha, oleic acid performed similarly to VLDL lipolysis products. Therefore, fatty acids are likely to be the PPAR-alpha ligands generated by VLDL lipolysis. Indeed, unbound fatty acid concentration determined PPAR-alpha activation regardless of fatty acid source, with PPAR-alpha activation occurring only at unbound fatty acid concentrations that are unachievable under physiological conditions without lipase action. In mice, a synthetic lipase inhibitor (poloxamer-407) attenuated fasting-induced changes in expression of PPAR-alpha target genes. Apolipoprotein CIII (apoCIII), an endogenous inhibitor of lipoprotein and hepatic lipase, regulated access to the lipoprotein pool of PPAR-alpha ligands, because addition of exogenous apoCIII inhibited, and removal of endogenous apoCIII potentiated, lipolytic PPAR-alpha activation. These data suggest that the PPAR-alpha response is generated by unbound fatty acids released locally by lipase activity and not by circulating plasma fatty acids.
Collapse
Affiliation(s)
- Maxwell A Ruby
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr Way, Oakland, CA 94609, USA
| | | | | | | | | | | |
Collapse
|
11
|
Shearer GC, Newman JW. Lipoprotein lipase releases esterified oxylipins from very low-density lipoproteins. Prostaglandins Leukot Essent Fatty Acids 2008; 79:215-22. [PMID: 19042114 PMCID: PMC2629508 DOI: 10.1016/j.plefa.2008.09.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 08/06/2008] [Accepted: 09/22/2008] [Indexed: 11/20/2022]
Abstract
We previously demonstrated that defects in lipoprotein metabolism alter the distribution of oxygenated polyunsaturated fatty acids (PUFAs) in lipoprotein particles. If these oxidation products are released by lipoprotein lipase (LpL), then their delivery to peripheral tissues with bulk lipids could influence cellular function. Using 26-week-old normolipidemic and hyperlipidemic Zucker rats, we measured PUFA alcohols, epoxides, diols, ketones, and triols (i.e. oxylipins) in esterified and non-esterified fractions of whole plasma, VLDL, and LpL-generated VLDL-lipolysates. Whole plasma, VLDL, and lipolysate oxylipin profiles were distinct and altered by hyperlipidemia. While >90% of the whole plasma oxylipins were esterified, the fraction of each oxylipin class in the VLDL varied: 46% of alcohols, 30% of epoxides, 19% of diols, <10% of ketones, and <1% triols. Whole plasma was dominated by arachidonate alcohols, while the linoleate alcohols, epoxides, and ketones showed an increased prevalence in VLDL. LpL-mediated VLDL lipolysis of PUFA alcohols, diols and ketones was detected and the relative abundance of oxygenated linoleates was enhanced in the lipolysates, relative to their corresponding VLDL. In summary esterified oxylipins were seen to be LpL substrates with heterogeneous distributions among lipoprotein classes. Moreover, oxylipin distributions are changes within the context of obesity-associated dyslipidemia. These results support the notion that the VLDL-LpL axis may facilitate the delivery of plasma oxylipins to the periphery. The physiological implications of these findings are yet to be elucidated; however, these molecules are plausible indicators of systemic oxidative stress, and could report this status to the peripheral tissues.
Collapse
Affiliation(s)
- Gregory C Shearer
- Department of Veterans Affairs Northern California Health Care System, Mather, CA, USA.
| | | |
Collapse
|
12
|
Peroxisome proliferator-activated receptors and the vascular system: beyond their metabolic effects. ACTA ACUST UNITED AC 2008; 2:227-38. [DOI: 10.1016/j.jash.2007.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 12/28/2007] [Accepted: 12/28/2007] [Indexed: 12/19/2022]
|
13
|
Abstract
Individuals with type 2 diabetes mellitus have increased cardiovascular disease risk compared with those without diabetes. Treatment of the residual risk, other than blood pressure and LDL-cholesterol control, remains important as the rate of diabetes increases worldwide. The accelerated atherosclerosis and cardiovascular disease in diabetes is likely to be multifactorial and therefore several therapeutic approaches can be considered. Results of mechanistic studies done in vitro and in vivo--animals and people--can provide important insights with the potential to improve clinical management decisions and outcomes. In this Review, we focus on three areas in which pathophysiological considerations could be particularly informative--ie, the roles of hyperglycaemia, diabetic dyslipidaemia (other than the control of LDL-cholesterol concentrations), and inflammation (including that in adipose tissue) in the acceleration of vascular injury.
Collapse
Affiliation(s)
- Theodore Mazzone
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | | | | |
Collapse
|
14
|
Ziouzenkova O, Plutzky J. Retinoid metabolism and nuclear receptor responses: New insights into coordinated regulation of the PPAR-RXR complex. FEBS Lett 2007; 582:32-8. [PMID: 18068127 DOI: 10.1016/j.febslet.2007.11.081] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Accepted: 11/28/2007] [Indexed: 12/31/2022]
Abstract
Retinoids, naturally-occurring vitamin A derivatives, regulate metabolism by activating specific nuclear receptors, including the retinoic acid receptor (RAR) and the retinoid X receptor (RXR). RXR, an obligate heterodimeric partner for other nuclear receptors, including peroxisome proliferator-activated receptors (PPARs), helps coordinate energy balance. Recently, many groups have identified new connections between retinoid metabolism and PPAR responses. We found that retinaldehyde (Rald), a molecule that can yield RA through the action of retinaldehyde dehydrogenases (Raldh), is present in fat in vivo and can inhibit PPAR gamma-induced adipogenesis. In vitro, Rald inhibits RXR and PPAR gamma activation. Raldh1-deficient mice have increased Rald levels in fat, higher metabolic rates and body temperatures, and are protected against diet-induced obesity and insulin resistance. Interestingly, one specific asymmetric beta-carotene cleavage product, apo-14'-carotenal, can also inhibit PPAR gamma and PPAR alpha responses. These data highlight how pathways of beta-carotene metabolism and specific retinoid metabolites may have direct distinct metabolic effects.
Collapse
Affiliation(s)
- Ouliana Ziouzenkova
- Department of Human Nutrition, Ohio State University, Columbus, OH 43210, USA
| | | |
Collapse
|
15
|
Abstract
Hypertensive patients are at increased risk for cardiovascular complications. Inhibition of different pathophysiological mechanisms involved in hypertension and hypertension-related target organ damage may revert or prevent the progression of the pathological changes observed and reduce the occurrence of cardiovascular events. One of the new targets that may prevent or regress hypertensive vascular, renal, and perhaps brain changes in hypertension is the activation of nuclear receptors that have metabolic effects but also exert antiinflammatory action, the peroxisome proliferator activator receptor (PPAR) activators alpha and gamma. This review will discuss some of the evidence, both experimental and clinical, that suggests that activation of PPAR alpha and/or gamma in hypertension may exert beneficial cardiovascular protective effects.
Collapse
Affiliation(s)
- Eyal Leibovitz
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
16
|
Newman JW, Kaysen GA, Hammock BD, Shearer GC. Proteinuria increases oxylipid concentrations in VLDL and HDL but not LDL particles in the rat. J Lipid Res 2007; 48:1792-800. [PMID: 17496268 DOI: 10.1194/jlr.m700146-jlr200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously established that proteinuria alters the apolipoprotein content of lipoproteins. This study was conducted to establish whether proteinuria also alters the concentrations of oxidized lipids within lipoprotein density fractions. To this end, we induced passive Heymann nephritis in Sprague Dawley rats and measured an array of alkaline-stable oxylipids in VLDL, LDL, and HDL particles. Proteinuria increased the total oxylipid amounts in the HDL and VLDL fractions. More importantly, these levels were increased when expressed per unit lipoprotein protein, indicating that the oxidized lipid load per particle was increased. Epoxides and diols increased approximately 2-fold in HDL and approximately 5-fold in VLDL, whereas LDL showed approximately 2-fold decreases. The hydroxyeicosatetraenoic acids and hydroxyoctadecadienoic acids (HODEs) increased >4-fold in HDL and >20-fold in VLDL, whereas LDL showed approximately 2-fold decreases in the HODEs. Therefore, nephrotic syndrome alters the lipoprotein oxylipid composition independently of an increase in total lipoprotein levels. These proteinuria-induced changes may be associated with the cardiovascular risk of lipoprotein oxidation.
Collapse
Affiliation(s)
- John W Newman
- Western Human Nutrition Research Center, United States Department of Agriculture, USA.
| | | | | | | |
Collapse
|
17
|
Yakubu MA, Nsaif RH, Oyekan AO. peroxisome proliferator-activated receptor alpha activation-mediated regulation of endothelin-1 production via nitric oxide and protein kinase C signaling pathways in piglet cerebral microvascular endothelial cell culture. J Pharmacol Exp Ther 2006; 320:774-81. [PMID: 17105827 PMCID: PMC4502983 DOI: 10.1124/jpet.106.104992] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Elevated endothelin (ET)-1 has been implicated in cerebrovascular complications following brain trauma characterized by dysregulation of endothelial nitric oxide synthase (eNOS), protein kinase C (PKC), and cerebral function. Recently, vascular expression of PPARalpha has been observed and suggested to improve vascular dysfunction. We speculate that activation of PPARalpha in cerebral microvessels can improve cerebral dysfunction following trauma, and we tested the hypothesis that activation of cerebral endothelial peroxisome proliferator-activated receptor (PPAR)alpha will attenuate ET-1 production via a mechanism involving nitric oxide (NO) and PKC. Phorbol 12-myristate 13-acetate (PMA) (1 microM), bradykinin (BK, 1 microM), angiotensin II (AII, 1 microM), or hemoglobin (Hem, 10 microM) increased ET-1 levels by 24-, 11.4-, 3.6-, or 1.3-fold increasing ET-1 levels from 0.36 +/- 0.08 to 8.6 +/- 0.8, 4.1 +/- 0.7, 1.30 +/- 0.1, or 0.47 +/- 0.03 fmol/microg protein (p < 0.05), respectively. Clofibrate (10 microM) reduced basal ET-1 from 0.36 +/- 0.08 (control) to 0.03 +/- 0.01 and blunted vasoactive agent-induced increase to 0.12 +/- 0.07 (PMA), 0.6 +/- 0.04 (BK), 0.25 +/- 0.03 (AII), or 0.12 +/- 0.03 (Hem) fM/microg protein (p < 0.05). L-arginine methyl ester (100 microM) inhibited clofibrate-induced reduction in basal ET-1 production. Clofibrate increased PPARalpha expression, accompanied by increased NO production and eNOS expression. PKC inhibition by calphostin C (10 microM) blocked these effects, whereas activation by PMA reduced basal PPARalpha expression. Thus, PPARalpha activation attenuated ET-1 production by agents that mediate brain injury through mechanisms that probably result from PPARalpha-induced increase in eNOS expression/NO production and complex PKC signaling pathways. Therefore, PPARalpha activators can be appropriate therapeutic agents to alleviate cerebrovascular dysfunction following cerebral vasospasm.
Collapse
Affiliation(s)
- Momoh A Yakubu
- Vascular Biology Unit, Center for Cardiovascular Diseases, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX 77004, USA.
| | | | | |
Collapse
|
18
|
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
|