1
|
Jerab D, Blangero F, da Costa PCT, de Brito Alves JL, Kefi R, Jamoussi H, Morio B, Eljaafari A. Beneficial Effects of Omega-3 Fatty Acids on Obesity and Related Metabolic and Chronic Inflammatory Diseases. Nutrients 2025; 17:1253. [PMID: 40219010 PMCID: PMC11990730 DOI: 10.3390/nu17071253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs) are known to help resolve inflammation through generation of anti-inflammatory eicosanoids and specialized pro-resolving mediators, including resolvins, protectins, and maresins. Through binding to the GPR120/FFAR4 receptor, their beneficial effects result from phospholipid membrane remodeling, impairment of inflammatory signaling molecules clustering, subsequent inhibition of NF-κB and inflammasome activation, and a reduction in oxidative stress. Obesity, a chronic inflammatory disease that contributes to metabolic disorders, is alleviated by n-3 PUFAs. In the adipose tissue (AT) of individuals with obesity, n-3 PUFAs counteract hypoxia, inhibit immune cell infiltration and AT inflammation, improve insulin sensitivity, and reduce fat mass. Beyond AT, n-3 PUFAs also alleviate other metabolic disorders such as metabolic-associated steatotic liver disease (MASLD), gut dysbiosis, and/or renal dysfunction. In cardiovascular disease (CVD), they are mainly recommended as a secondary prevention for patients with coronary heart disease risks. This review provides an in-depth analysis of the benefits of n-3 PUFAs in obesity and related metabolic diseases, examining both the mechanistic and clinical aspects. Additionally, it also explores the effects of n-3 PUFAs in obesity-related chronic inflammatory conditions, including inflammatory bowel disease, psoriasis, rheumatoid arthritis, osteoarthritis, and multiple sclerosis, by targeting specific pathophysiological mechanisms. Clinical applications and limitations of n-3 PUFAs are discussed based on findings from human clinical trials.
Collapse
Affiliation(s)
- Donia Jerab
- CarMeN Laboratory, Institut National de Recherche pour l’ Agriculture, l’ Alimentation et l’Environnement, UMR1397, Institut National de la Santé et de la Recherche Médicale, U 1060, Université Claude Bernard Lyon I, 69310 Pierre-Bénite, France (B.M.)
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis 1002, Tunisia;
| | - Ferdinand Blangero
- CarMeN Laboratory, Institut National de Recherche pour l’ Agriculture, l’ Alimentation et l’Environnement, UMR1397, Institut National de la Santé et de la Recherche Médicale, U 1060, Université Claude Bernard Lyon I, 69310 Pierre-Bénite, France (B.M.)
| | - Paulo César Trindade da Costa
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, Brazil (J.L.d.B.A.)
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, Brazil (J.L.d.B.A.)
| | - Rym Kefi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis 1002, Tunisia;
| | - Henda Jamoussi
- Research Unit “Obesity: Etiopathology and Treatment, UR18ES01”, Faculty of Medicine, Tunis El Manar University, Tunis 2092, Tunisia;
| | - Beatrice Morio
- CarMeN Laboratory, Institut National de Recherche pour l’ Agriculture, l’ Alimentation et l’Environnement, UMR1397, Institut National de la Santé et de la Recherche Médicale, U 1060, Université Claude Bernard Lyon I, 69310 Pierre-Bénite, France (B.M.)
| | - Assia Eljaafari
- CarMeN Laboratory, Institut National de Recherche pour l’ Agriculture, l’ Alimentation et l’Environnement, UMR1397, Institut National de la Santé et de la Recherche Médicale, U 1060, Université Claude Bernard Lyon I, 69310 Pierre-Bénite, France (B.M.)
- Department of Clinical Research, Hospices Civils de Lyon, 69002 Lyon, France
| |
Collapse
|
2
|
Hall WL. Long chain n-3 polyunsaturated fatty acid intake across the life span for cardiovascular disease prevention in women. Proc Nutr Soc 2025; 84:98-109. [PMID: 38444046 DOI: 10.1017/s0029665124000181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Cardiovascular diseases (CVDs) are a major health concern for women. Historically there has been a misconception that men are at greater risk because CVD tends to occur earlier in life compared to women. Clinical guidelines for prevention of heart disease are currently the same for both sexes, but accumulating evidence demonstrates that risk profiles diverge. In fact, several CVD risk factors confer an even greater risk in women relative to men, including high blood pressure, obesity, diabetes and raised triglycerides. Furthermore, many female-specific CVD risk factors exist, including early menarche, pregnancy complications, polycystic ovary syndrome, reproductive hormonal treatments and menopause. Little is known about how diet interacts with CVD risk factors at various stages of a woman’s life. Long chain (LC) n-3 polyunsaturated fatty acid (PUFA) intakes are a key dietary factor that may impact risk of CVD throughout the life course differentially in men and women. Oestrogen enhances conversion of the plant n-3 PUFA, alpha-linolenic acid, to LCn-3 PUFA. Increasing the frequency of oily fish consumption or LCn-3 PUFA supplementation may be important for reducing coronary risk during the menopausal transition, during which time oestrogen levels decline and the increase in CVD risk factors is accelerated. Women are under-represented in the evidence base for CVD prevention following LC n-3 PUFA supplementation. Therefore it is not clear whether there are sex differences in response to treatment. Furthermore, there is a lack of evidence on optimal intakes of LC n-3 PUFA across the lifespan for CVD prevention in women.
Collapse
Affiliation(s)
- Wendy Louise Hall
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
3
|
Queathem ED, Stagg D, Nelson A, Chaves AB, Crown SB, Fulghum K, D Avignon DA, Ryder JR, Bolan PJ, Hayir A, Gillingham JR, Jannatpour S, Rome FI, Williams AS, Muoio DM, Ikramuddin S, Hughey CC, Puchalska P, Crawford PA. Ketogenesis protects against MASLD-MASH progression through mechanisms that extend beyond overall fat oxidation rate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618895. [PMID: 39464122 PMCID: PMC11507910 DOI: 10.1101/2024.10.17.618895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The progression of metabolic-dysfunction-associated steatotic liver disease (MASLD) to metabolic-dysfunction-associated steatohepatitis (MASH) involves complex alterations in both liver-autonomous and systemic metabolism that influence the liver's balance of fat accretion and disposal. Here, we quantify the relative contribution of hepatic oxidative pathways to liver injury in MASLD-MASH. Using NMR spectroscopy, UHPLC-MS, and GC-MS, we performed stable-isotope tracing and formal flux modeling to quantify hepatic oxidative fluxes in humans across the spectrum of MASLD-MASH, and in mouse models of impaired ketogenesis. We found in humans with MASH, that liver injury correlated positively with ketogenesis and total fat oxidation, but not with turnover of the tricarboxylic acid cycle. The use of loss-of-function mouse models demonstrated that disruption of mitochondrial HMG-CoA synthase (HMGCS2), the rate-limiting step of ketogenesis, impairs overall hepatic fat oxidation and induces a MASLD-MASH-like phenotype. Disruption of mitochondrial β-hydroxybutyrate dehydrogenase (BDH1), the terminal step of ketogenesis, also impaired fat oxidation, but surprisingly did not exacerbate steatotic liver injury. Taken together, these findings suggest that quantifiable variations in overall hepatic fat oxidation may not be a primary determinant of MASLD-to-MASH progression, but rather, that maintenance of hepatic ketogenesis could serve a protective role through additional mechanisms that extend beyond quantified overall rates of fat oxidation.
Collapse
|
4
|
Sugimoto K, Nishimura M, Ito N, Hosomi R, Fukunaga K, Nishihira J. Effects of Daily Consumption of Scallop Oil Prepared from Internal Organs of Japanese Giant Scallop (Patinopecten yessoensis) on Serum Lipid Composition and Its Safety: A Randomized, Double-blind, Placebo-controlled, Parallel Group Comparison Study. J Oleo Sci 2024; 73:1201-1212. [PMID: 39168625 DOI: 10.5650/jos.ess24105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
Scallop oil (SCO) prepared from the internal organs of the Japanese giant scallop (Patinopecten yessoensis) contains eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and phospholipids (PL). It was previously shown that SCO consumption improves cholesterol and triacylglycerols (TG) contents in mice. The present study demonstrated the effects of daily SCO consumption (1.2 g/day, containing 376 mg of EPA, 63 mg of DHA, and 150 mg of PL) for 12 weeks in human subjects. In this randomized, doubleblind, placebo-controlled, parallel group comparison study, 70 Japanese subjects with serum TG levels ≥120 but < 200 mg/dL were recruited and randomly assigned to the SCO or placebo group. All subjects ingested six capsules per day for 12 weeks. We conducted medical interviews, body composition measurements, vital sign examinations, and blood sampling at weeks 0 (baseline), 4, 8, and 12, and measured peripheral blood flow at weeks 0 and 12. In the case of subjects with higher serum TG levels, SCO consumption decreased the changes in serum TG and malondialdehyde-low density lipoprotein (MDA-LDL) levels compared with the placebo group. Safety assessment revealed no medically significant changes due to continuous SCO consumption. The findings indicate that 1.2 g/day of SCO consumption may be beneficial for reducing serum TG and MDA-LDL levels in persons with higher TG levels.
Collapse
Affiliation(s)
- Koki Sugimoto
- Faculty of Food and Nutritional Sciences, Toyo University
| | - Mie Nishimura
- Department of Medical Management and Informatics, Hokkaido Information University
| | - Naohito Ito
- Department of Medical Management and Informatics, Hokkaido Information University
| | - Ryota Hosomi
- Department of Life Science and Biotechnology, Kansai University
| | - Kenji Fukunaga
- Department of Life Science and Biotechnology, Kansai University
| | - Jun Nishihira
- Department of Medical Management and Informatics, Hokkaido Information University
| |
Collapse
|
5
|
Komiya Y, Sakazaki Y, Goto T, Kawabata F, Suzuki T, Sato Y, Sawano S, Nakamura M, Tatsumi R, Ikeuchi Y, Arihara K, Mizunoya W. Eicosapentaenoic acid increases proportion of type 1 muscle fibers through PPARδ and AMPK pathways in rats. iScience 2024; 27:109816. [PMID: 38779480 PMCID: PMC11108975 DOI: 10.1016/j.isci.2024.109816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/07/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Muscle fiber type composition (% slow-twitch and % fast-twitch fibers) is associated with metabolism, with increased slow-twitch fibers alleviating metabolic disorders. Previously, we reported that dietary fish oil intake induced a muscle fiber-type transition in a slower direction in rats. The aim of this study was to determine the functionality of eicosapentaenoic acid (EPA), a unique fatty acid in fish oil, to skeletal muscle fiber type and metabolism in rats. Here, we showed that dietary EPA promotes whole-body oxidative metabolism and improves muscle function by increasing proportion of slow-twitch type 1 fibers in rats. Transcriptomic and metabolomic analyses revealed that EPA supplementation activated the peroxisome proliferator-activated receptor δ (PPARδ) and AMP-activated protein kinase (AMPK) pathways in L6 myotube cultures, which potentially increasing slow-twitch fiber share. This highlights the role of EPA as an exercise-mimetic dietary component that improves metabolism and muscle function, with potential benefits for health and athletic performance.
Collapse
Affiliation(s)
- Yusuke Komiya
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Yuka Sakazaki
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Tsuyoshi Goto
- Division of Food Science & Biotechnology, Kyoto University, Kyoto, Japan
| | - Fuminori Kawabata
- Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan
| | - Takahiro Suzuki
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yusuke Sato
- Department of Animal Science, School of Agriculture, Tokai University, Kumamoto, Japan
| | - Shoko Sawano
- Department of Food and Life Science, School of Life and Environmental Science, Azabu University, Sagamihara, Japan
| | - Mako Nakamura
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yoshihide Ikeuchi
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Keizo Arihara
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Wataru Mizunoya
- Department of Animal Science and Biotechnology, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| |
Collapse
|
6
|
Su F, Koeberle A. Regulation and targeting of SREBP-1 in hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:673-708. [PMID: 38036934 PMCID: PMC11156753 DOI: 10.1007/s10555-023-10156-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is an increasing burden on global public health and is associated with enhanced lipogenesis, fatty acid uptake, and lipid metabolic reprogramming. De novo lipogenesis is under the control of the transcription factor sterol regulatory element-binding protein 1 (SREBP-1) and essentially contributes to HCC progression. Here, we summarize the current knowledge on the regulation of SREBP-1 isoforms in HCC based on cellular, animal, and clinical data. Specifically, we (i) address the overarching mechanisms for regulating SREBP-1 transcription, proteolytic processing, nuclear stability, and transactivation and (ii) critically discuss their impact on HCC, taking into account (iii) insights from pharmacological approaches. Emphasis is placed on cross-talk with the phosphatidylinositol-3-kinase (PI3K)-protein kinase B (Akt)-mechanistic target of rapamycin (mTOR) axis, AMP-activated protein kinase (AMPK), protein kinase A (PKA), and other kinases that directly phosphorylate SREBP-1; transcription factors, such as liver X receptor (LXR), peroxisome proliferator-activated receptors (PPARs), proliferator-activated receptor γ co-activator 1 (PGC-1), signal transducers and activators of transcription (STATs), and Myc; epigenetic mechanisms; post-translational modifications of SREBP-1; and SREBP-1-regulatory metabolites such as oxysterols and polyunsaturated fatty acids. By carefully scrutinizing the role of SREBP-1 in HCC development, progression, metastasis, and therapy resistance, we shed light on the potential of SREBP-1-targeting strategies in HCC prevention and treatment.
Collapse
Affiliation(s)
- Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
7
|
Pourrajab B, Sharifi-Zahabi E, Soltani S, Shahinfar H, Shidfar F. Comparison of canola oil and olive oil consumption on the serum lipid profile in adults: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 2023; 63:12270-12284. [PMID: 35866510 DOI: 10.1080/10408398.2022.2100314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Several randomized clinical trials have investigated the effects of canola oil (CO) compared to olive oil (OO) on the serum lipid profiles in adults. However, the results of these studies are inconsistent. Thus, this study aimed to assess the comparison of CO and OO consumption on the serum lipid components in adults. METHODS AND RESULTS The following online databases were searched until February 4th, 2022: PubMed/Medline, Scopus, Clarivate Analytics Web of Science, Cochrane Central Register of Controlled Trials, and Google Scholar. The effect sizes were stated as the weighted mean difference (WMD) with 95% confidence intervals (CI). A total of 13 eligible trials were included in this meta-analysis. The results showed that the CO consumption, significantly reduced serum LDL-c (WMD: -6.13 mg/dl, 95%CI: -9.79, -2.46, p = 0.001), TC (WMD: -8.92 mg/dl, 95% CI: -13.52, -4.33, P < 0.001) and LDL-c/HDL-c ratio (WMD: -0.30; 95% CI, -0.53, -0.06, p = 0.01) levels compared to OO. There were no significant changes in the other components of the blood lipids. CONCLUSION The results of this review suggest that CO consumptionhas beneficial effects on LDL-c, TC, and LDL-c/HDL-c ratio compared to OO. Therefore, its replacement with OO can have cardioprotective impacts.
Collapse
Affiliation(s)
- Behnaz Pourrajab
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Sharifi-Zahabi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Shahinfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Torrecilhas JA, Pereira GL, Vito ES, Fiorentini G, Ramirez-Zamudio GD, Fonseca LS, Torres RDNS, Simioni TA, Duarte JM, Machado Neto OR, Curi RA, Chardulo LAL, Baldassini WA, Berchielli TT. Changes in the Lipid Metabolism of the Longissimus thoracis Muscle in Bulls When Using Different Feeding Strategies during the Growing and Finishing Phases. Metabolites 2023; 13:1042. [PMID: 37887367 PMCID: PMC10608670 DOI: 10.3390/metabo13101042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023] Open
Abstract
The objective was to evaluate the supplementation strategy's effect on beef cattle during the growing phase and two systems during the finishing phase. One hundred and twenty young bulls were randomly divided in a 2 × 2 factorial design to receive either mineral (ad libitum) or protein + energy (3 g/kg body weight (BW)/day) during the growing phase and pasture plus concentrate supplementation (20 g/kg BW/day) or feedlot (25:75% corn silage:concentrate) during the finishing phase. Feedlot-fed bulls had meat (Longissimus thoracis-LT) with a higher content of lipids and saturated and monounsaturated fatty acids and a greater upregulation of stearoyl-CoA desaturase and sterol regulatory element-binding protein-1c than animals that fed on pasture (p < 0.05). On the other hand, pasture-fed bulls had meat with a higher content of α-linoleic acid, linolenic acid, and n6 and a greater n6:n3 ratio compared to the feedlot-fed group (p < 0.05). In addition, meat from pasture-fed bulls during the finishing phase had 17.6% more isocitrate dehydrogenase enzyme concentration than the feedlot group (p = 0.02). Mineral-fed and pasture-finished bulls showed down-regulation of peroxisome proliferator-activated receptor gamma (p < 0.05), while the bulls fed protein + energy and finished in the feedlot had higher carnitine palmitoyltransferase 2 expression (p ≤ 0.013). In conclusion, mineral or protein + energy supplementation in the growing does not affect the fatty acid composition of intramuscular fat of LT muscle. In the finishing phase, feeding bulls in the feedlot upregulates the lipogenic genes and consequently improves the intramuscular fat content in the meat.
Collapse
Affiliation(s)
- Juliana Akamine Torrecilhas
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Guilherme Luis Pereira
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Elias San Vito
- Confina Beef Cattle Consulting, Sinop 78555-603, MT, Brazil;
| | - Giovani Fiorentini
- Department of Animal Science, Federal University of Pelotas (UFPEL), Pelotas 96160-000, RS, Brazil;
| | - Germán Darío Ramirez-Zamudio
- College of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga 13635-900, SP, Brazil;
| | - Larissa Simielli Fonseca
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (L.S.F.); (T.A.S.); (J.M.D.); (T.T.B.)
| | - Rodrigo de Nazaré Santos Torres
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Tiago Adriano Simioni
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (L.S.F.); (T.A.S.); (J.M.D.); (T.T.B.)
| | - Juliana Messana Duarte
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (L.S.F.); (T.A.S.); (J.M.D.); (T.T.B.)
| | - Otavio Rodrigues Machado Neto
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Rogério Abdallah Curi
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Luis Artur Loyola Chardulo
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Welder Angelo Baldassini
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Telma Teresinha Berchielli
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (L.S.F.); (T.A.S.); (J.M.D.); (T.T.B.)
| |
Collapse
|
9
|
Schoeler M, Ellero-Simatos S, Birkner T, Mayneris-Perxachs J, Olsson L, Brolin H, Loeber U, Kraft JD, Polizzi A, Martí-Navas M, Puig J, Moschetta A, Montagner A, Gourdy P, Heymes C, Guillou H, Tremaroli V, Fernández-Real JM, Forslund SK, Burcelin R, Caesar R. The interplay between dietary fatty acids and gut microbiota influences host metabolism and hepatic steatosis. Nat Commun 2023; 14:5329. [PMID: 37658064 PMCID: PMC10474162 DOI: 10.1038/s41467-023-41074-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/21/2023] [Indexed: 09/03/2023] Open
Abstract
Dietary lipids can affect metabolic health through gut microbiota-mediated mechanisms, but the influence of lipid-microbiota interaction on liver steatosis is largely unknown. We investigate the impact of dietary lipids on human gut microbiota composition and the effects of microbiota-lipid interactions on steatosis in male mice. In humans, low intake of saturated fatty acids (SFA) is associated with increased microbial diversity independent of fiber intake. In mice, poorly absorbed dietary long-chain SFA, particularly stearic acid, induce a shift in bile acid profile and improved metabolism and steatosis. These benefits are dependent on the gut microbiota, as they are transmitted by microbial transfer. Diets enriched in polyunsaturated fatty acids are protective against steatosis but have minor influence on the microbiota. In summary, we find that diets enriched in poorly absorbed long-chain SFA modulate gut microbiota profiles independent of fiber intake, and this interaction is relevant to improve metabolism and decrease liver steatosis.
Collapse
Affiliation(s)
- Marc Schoeler
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, Toulouse, France
| | - Till Birkner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Lisa Olsson
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Harald Brolin
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Ulrike Loeber
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Jamie D Kraft
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Arnaud Polizzi
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, Toulouse, France
| | - Marian Martí-Navas
- Department of Radiology, Biomedical Research Institute Imaging Research Unit, Diagnostic Imaging Institute, Doctor Josep Trueta University Hospital of Girona, Avinguda de França, s/n, 17007, Girona, Catalonia, Spain
| | - Josep Puig
- Department of Radiology, Biomedical Research Institute Imaging Research Unit, Diagnostic Imaging Institute, Doctor Josep Trueta University Hospital of Girona, Avinguda de França, s/n, 17007, Girona, Catalonia, Spain
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124, Bari, Italy
- Medicina e Chirurgia d'Accettazione E d'Urgenza, Azienda Ospedaliero-Universitaria Policlinico di Bari, 70124, Bari, Italy
- Medicina Sub-Intensiva, Presidio Maxi-Emergenze Fiera del Levante, Azienda Ospedaliero-Universitaria Policlinico di Bari, 70124, Bari, Italy
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1297, Université Paul Sabatier, Université de Toulouse, F-31432, Toulouse, France
| | - Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1297, Université Paul Sabatier, Université de Toulouse, F-31432, Toulouse, France
- Endocrinology-Diabetology-Nutrition Department, Toulouse University Hospital, Toulouse, France
| | - Christophe Heymes
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1297, Université Paul Sabatier, Université de Toulouse, F-31432, Toulouse, France
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, Toulouse, France
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain
| | - Sofia K Forslund
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Remy Burcelin
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1297, Université Paul Sabatier, Université de Toulouse, F-31432, Toulouse, France
| | - Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden.
| |
Collapse
|
10
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic fatty liver disease worldwide, particularly in obese and type 2 diabetic individuals. Currently, there are no therapies for NAFLD that have been approved by the US Food and Drug Administration. Herein, we examine the rationale for using ω3 polyunsaturated fatty acids (PUFAs) in NAFLD therapy. This focus is based on the finding that NAFLD severity is associated with a reduction of hepatic C20-22 ω3 PUFAs. Because C20-22 ω3 PUFAs are pleiotropic regulators of cell function, loss of C20-22 ω3 PUFAs has the potential to significantly impact hepatic function. We describe NAFLD prevalence and pathophysiology as well as current NAFLD therapies. We also present evidence from clinical and preclinical studies that evaluated the capacity of C20-22 ω3 PUFAs to treat NAFLD. Given the clinical and preclinical evidence, dietary C20-22 ω3 PUFA supplementation has the potential to decrease human NAFLD severity by reducing hepatosteatosis and liver injury.
Collapse
Affiliation(s)
- Melinda H Spooner
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA;
| | - Donald B Jump
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA;
| |
Collapse
|
11
|
Kamoshita K, Ishii KA, Tahira Y, Kikuchi A, Abuduwaili H, Tajima-Shirasaki N, Li Q, Takayama H, Matsumoto K, Takamura T. Insulin Suppresses Ubiquitination via the Deubiquitinating Enzyme Ubiquitin-Specific Protease 14, Independent of Proteasome Activity in H4IIEC3 Hepatocytes. J Pharmacol Exp Ther 2023; 385:5-16. [PMID: 36328485 DOI: 10.1124/jpet.122.001088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 08/16/2022] [Accepted: 09/09/2022] [Indexed: 11/06/2022] Open
Abstract
Ubiquitin-proteasome dysfunction contributes to obesity-related metabolic disorders, such as diabetes and fatty liver disease. However, the regulation of ubiquitin-proteasome activity by insulin remains to be elucidated. Here, we show that prolonged insulin stimulation activates proteasome function even though it reduces the ubiquitinated proteins in H4IIEC3 hepatocytes. Looking for a pathway by which insulin inhibits ubiquitination, we found that hepatic expression of ubiquitin-specific protease 14 (USP14) was upregulated in the liver of patients with insulin resistance. Indeed, the USP14-specific inhibitor IU1 canceled the insulin-mediated reduction of ubiquitinated proteins. Furthermore, insulin-induced endoplasmic reticulum (ER) stress, which was canceled by IU1, suggesting that USP14 activity is involved in insulin-induced ER stress. Co-stimulation with insulin and IU1 for 2 hours upregulated the nuclear translocation of the lipogenic transcription factor, sterol regulatory element binding protein-1c (SREBP-1c), upregulated the expression of the lipogenic gene, fatty acid synthase (Fasn), and repressed the gluconeogenic genes. In conclusion, insulin activates proteasome function even though it inhibits protein ubiquitination by activating USP14 in hepatocytes. USP14 activation by insulin inhibits mature SREBP-1c while upregulating ER stress and the expression of genes involved in gluconeogenesis. Further understanding mechanisms underlying the USP14 activation and its pleiotropic effects may lead to therapeutic development for obesity-associated metabolic disorders, such as diabetes and fatty liver disease. SIGNIFICANCE STATEMENT: This study shows that insulin stimulation inhibits ubiquitination by activating USP14, independent of its effect on proteasome activity in hepatocytes. USP14 also downregulates the nuclear translocation of the lipogenic transcription factor SREBP-1c and upregulates the expression of genes involved in gluconeogenesis. Since USP14 is upregulated in the liver of insulin-resistant patients, understanding mechanisms underlying the USP14 activation and its pleiotropic effects will help develop treatments for metabolic disorders such as diabetes and fatty liver.
Collapse
Affiliation(s)
- Kyoko Kamoshita
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kiyo-Aki Ishii
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yumiko Tahira
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Akihiro Kikuchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Halimulati Abuduwaili
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Natsumi Tajima-Shirasaki
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Qifang Li
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroaki Takayama
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kunio Matsumoto
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
12
|
Morrow NM, Mulvihill EE. Complexity in Hepatic Insulin Resistance - Unraveling the Role of Ubiquitin-Specific Protease 14 in Protein Homeostasis of Metabolic Transcription Factors. J Pharmacol Exp Ther 2023; 385:1-4. [PMID: 36927510 DOI: 10.1124/jpet.122.001517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/17/2023] [Indexed: 03/18/2023] Open
Affiliation(s)
- Nadya M Morrow
- Department of Biochemistry, Microbiology and Immunology (N.M.M., E.E.M.) and Centre for Infection, Immunity and Inflammation (E.E.M.), Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Heart Institute, Ottawa, Ontario, Canada (N.M.M., E.E.M.); and Montreal Diabetes Research Group, Montreal, Quebec, Canada (E.E.M.)
| | - Erin E Mulvihill
- Department of Biochemistry, Microbiology and Immunology (N.M.M., E.E.M.) and Centre for Infection, Immunity and Inflammation (E.E.M.), Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Heart Institute, Ottawa, Ontario, Canada (N.M.M., E.E.M.); and Montreal Diabetes Research Group, Montreal, Quebec, Canada (E.E.M.)
| |
Collapse
|
13
|
Li Y, Wu S, Zhao X, Hao S, Li F, Wang Y, Liu B, Zhang D, Wang Y, Zhou H. Key events in cancer: Dysregulation of SREBPs. Front Pharmacol 2023; 14:1130747. [PMID: 36969840 PMCID: PMC10030587 DOI: 10.3389/fphar.2023.1130747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Lipid metabolism reprogramming is an important hallmark of tumor progression. Cancer cells require high levels of lipid synthesis and uptake not only to support their continued replication, invasion, metastasis, and survival but also to participate in the formation of biological membranes and signaling molecules. Sterol regulatory element binding proteins (SREBPs) are core transcription factors that control lipid metabolism and the expression of important genes for lipid synthesis and uptake. A growing number of studies have shown that SREBPs are significantly upregulated in human cancers and serve as intermediaries providing a mechanistic link between lipid metabolism reprogramming and malignancy. Different subcellular localizations, including endoplasmic reticulum, Golgi, and nucleus, play an indispensable role in regulating the cleavage maturation and activity of SREBPs. In this review, we focus on the relationship between aberrant regulation of SREBPs activity in three organelles and tumor progression. Because blocking the regulation of lipid synthesis by SREBPs has gradually become an important part of tumor therapy, this review also summarizes and analyzes several current mainstream strategies.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shiming Hao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Difei Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
- *Correspondence: Yishu Wang, Honglan Zhou,
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Yishu Wang, Honglan Zhou,
| |
Collapse
|
14
|
Composition of Fatty Acids and Localization of SREBP1 and ELOVL2 Genes in Cauda Epididymides of Hu Sheep with Different Fertility. Animals (Basel) 2022; 12:ani12233302. [PMID: 36496823 PMCID: PMC9738327 DOI: 10.3390/ani12233302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
The epididymis is an organ that transports, matures and stores sperm, and has functions such as secretion and absorption. Polyunsaturated fatty acid (PUFA) compositions in sperm membrane were changed during the process of epididymis maturation and influence the male fertility. This study aimed to investigate differences in crude fat and fatty acid content in cauda epididymis between high and low fertility of Hu sheep. One hundred and seventy-nine Hu ram lambs were fed from 56 days to 6 months under the same environment. After the feeding trial, all rams were slaughtered, and the body weight, testicular weight, epididymal weight and sperm density were measured. Pearson correlation analysis showed significantly moderate positive correlation between epididymal weight and sperm density and testicular weight. Eighteen rams were selected and divided into the high fertility group (H, n = 9) and low fertility group (L, n = 9) according to the epididymal weight, sperm density and histomorphology. The crude fat content, fatty acid profile and genes related to fatty acid metabolism were detected. The crude fat content, total fatty acid, total n-3 PUFA and docosahexaenoic acid (C22:6n-3, DHA) content of cauda epididymis in high fertility group was significantly higher than those in low fertility group (p < 0.05). However, the ratio of n-6/n-3 PUFA was significantly lower than that in group L (p < 0.05). Immunohistochemistry results showed that SREBP1 and ELOVL2 were expressed in pseudostratified columnar ciliated epithelium and smooth muscle cells. The mRNA expression of SREBP1 (p = 0.09) and ELOVL2 (p < 0.05) in the high fertility group were increased. In conclusion, the high expression of SREBP1 and ELOVL2 may contribute to high n-3 PUFA content in cauda epididymis of high-fertility Hu sheep.
Collapse
|
15
|
Dong Y, Xie Z, You C, Li M, Li Y, Zhao J, Xie D, Wang S, Li Y. GPR120–ERK1–Srebp1c signaling pathway regulates long-chain polyunsaturated fatty acids biosynthesis in marine teleost Siganus canaliculatus. Comp Biochem Physiol B Biochem Mol Biol 2022; 264:110815. [DOI: 10.1016/j.cbpb.2022.110815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/12/2022] [Accepted: 11/17/2022] [Indexed: 11/21/2022]
|
16
|
Huang Q, Wang X, Liu J, Wang H, Miao Y, Zhang C, Zhang M, Qin C, Qin J, Chen L. Effect of Vitamin A Supplementation on Growth Performance, Lipid Deposition, Antioxidant Ability, and Immunity in Juvenile Chinese Mitten Crab Eriocheir sinensis Fed Diet with Fish Oil Totally Replaced by Palm Oil. AQUACULTURE NUTRITION 2022; 2022:1-19. [DOI: 10.1155/2022/3746245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
This research evaluated the protective effect of vitamin A (VA) on the adverse effect of fish oil (FO) substitution with palm oil (PO) in an economical crab Eriocheir sinensis. Three diets of FO, PO, and
VA as the main lipid sources were fed to crabs, respectively, for 8 weeks. Compared to crabs fed FO diet, crabs fed PO diet showed reduced hemolymph VA concentration, feed utilization efficiency, and growth performance. Besides, crabs fed PO diet showed elevated lipid content in hepatopancreas and body and triglyceride content in hepatopancreas, leading to decreased antioxidant enzyme and immune parameters activities from biochemical analysis, enzymatic determination, and quantitative polymerase chain reaction. In contrast, compared to crabs fed PO only, VA supplementation in PO improved the growth performance and utilization of fatty acids and reduced lipid deposition in the hepatopancreas. In addition, VA supplementation suppressed gene expression related to triglyceride synthesis (dgat1) and positively affected gene expression related to lipid catabolism (cpt1a, cpt1b, cpt2, and caat). Furthermore, VA supplementation upregulated antioxidant genes (CuZnSOD and CAT) through downregulating gene expression of upstream regulator Keap1. Furthermore, VA supplementation upregulated immune genes (Lzm and proPO) expression and reduced proinflammatory genes (LITAF, ADAM17, and IL-16) expression related to Toll2/MyD88/Relish signaling pathway. This study shows the necessity of VA addition in the feed with FO totally replaced by PO because it can relieve PO’s adverse effects and improve the growth of crabs.
Collapse
Affiliation(s)
- Qincheng Huang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaodan Wang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiadai Liu
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Han Wang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yixin Miao
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Cong Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Meiling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Sichuan 641100, China
| | - Jianguang Qin
- College of Science and Engineering, Flinders University, Adelaide, SA 5001, Australia
| | - Liqiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
17
|
Hu T, Zhang H, Du Y, Luo S, Yang X, Zhang H, Feng J, Chen X, Tu X, Wang C, Zhang Y. ELOVL2 restrains cell proliferation, migration, and invasion of prostate cancer via regulation of the tumor suppressor INPP4B. Cell Signal 2022; 96:110373. [DOI: 10.1016/j.cellsig.2022.110373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 12/24/2022]
|
18
|
Quispe R, Alfaddagh A, Kazzi B, Zghyer F, Marvel FA, Blumenthal RS, Sharma G, Martin SS. Controversies in the Use of Omega-3 Fatty Acids to Prevent Atherosclerosis. Curr Atheroscler Rep 2022; 24:571-581. [PMID: 35499805 DOI: 10.1007/s11883-022-01031-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW We discuss current controversies in the clinical use of omega-3 fatty acids (FA), primarily eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), and examine discrepancies between recent trials. Furthermore, we discuss potential side effects reported in these studies and the role of mixed omega-3 FA dietary supplements and concerns about their use. RECENT FINDINGS REDUCE-IT showed that addition of icosapent ethyl, a highly purified form of EPA, can reduce risk of cardiovascular events among statin-treated individuals with high triglycerides. Additional supportive evidence for EPA has come from other trials and meta-analyses of omega-3 FA therapy. In contrast, trials of mixed EPA/DHA products have consistently failed to improve cardiovascular outcomes. Discrepancies in results reported in RCTs could be explained by differences in omega-3 FA products, dosing, study populations, and study designs including the placebo control formulation. Evidence obtained from highly purified forms should not be extrapolated to other mixed formulations, including "over-the-counter" omega-3 supplements. Targeting TG-rich lipoproteins represents a new frontier for mitigating ASCVD risk. Clinical and basic research evidence suggests that the use of omega-3 FA, specifically EPA, appears to slow atherosclerosis by reducing triglyceride-rich lipoproteins and/or inflammation, therefore addressing residual risk of clinical ASCVD.
Collapse
Affiliation(s)
- Renato Quispe
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Abdulhamied Alfaddagh
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Brigitte Kazzi
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Fawzi Zghyer
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Francoise A Marvel
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Roger S Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Garima Sharma
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Seth S Martin
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA.
| |
Collapse
|
19
|
Kizilaslan N, Zekiye Erdem N, Katar M, Gevrek F. The Effects of Probiotics and Omega-3 Fatty Acids in Liver Steatosis Induced in Rats by High-Fructose Corn Syrup. Int J Clin Pract 2022; 2022:7172492. [PMID: 35685520 PMCID: PMC9159191 DOI: 10.1155/2022/7172492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022] Open
Abstract
AIMS This study was designed to reveal the effect of probiotics and omega-3 fatty acids in a fatty liver model in rats induced by high-fructose corn syrup (HFCS). METHODS In the study, 40 male Wistar Albino rats were used, and these rats were divided into five groups. HFCS was added to the drinking water (30% solution) of four groups (Groups 2, 3, 4, and 5) for three weeks, and the animals were fed ad libitum. At the end of three weeks, the rats in Groups 3, 4, and 5 were administered omega-3 fatty acids (400 mg/kg) and probiotics (1.5 × 109 cfu/mL/day) with the gavage method for four weeks. The body weights of rats were weighed and recorded before starting the experiment, at the end of the third week, and before the animals were sacrificed at the last week, all at the same hour. By subtracting the remaining amount of food and water from the daily food and water amount, the amount of food and water consumed was calculated. These values were recorded for seven weeks. At the end of the seven weeks, the rats were sacrificed after blood specimens and tissues were taken. RESULTS Analyzing the changes in the food intake of each group within itself throughout the experiment, it was observed that there was an increase in the food intake in the control group; from the starting week to the last week, the food intake amount of the HFCS group began to decrease particularly after the second week; and it began to decrease after the third week in the groups that were administered probiotics and omega-3 fatty acids. The changes in the sacrifice weights in the HFCS + omega-3 fatty acid, HFCS + probiotic, and HFCS + probiotic + omega-3 fatty acid groups were found to be lower than that in the HFCS group. The maximum levels of glucose, ALT, ALP, serum cholesterol, triglyceride and AST were found to be in the HFCS group. It was determined that the minimum mean steatosis level was in the control group, while the maximum steatosis level was in the HFCS group. CONCLUSIONS As a result, there was a protective effect of probiotic and omega-3 fatty acid.
Collapse
Affiliation(s)
- Nildem Kizilaslan
- Tokat Gaziosmanpasa University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Tokat, Turkey
| | - Nihal Zekiye Erdem
- Istanbul Medipol University, School of Health Sciences, Department of Nutrition and Dietetics, Istanbul, Turkey
| | - Muzaffer Katar
- Tokat Gaziosmanpasa University, Faculty of Medicine, Department of Biochemistry, Tokat, Turkey
| | - Fikret Gevrek
- Tokat Gaziosmanpasa University, Faculty of Medicine, Department of Histology, Tokat, Turkey
| |
Collapse
|
20
|
Vvedenskaya O, Rose TD, Knittelfelder O, Palladini A, Wodke JAH, Schuhmann K, Ackerman JM, Wang Y, Has C, Brosch M, Thangapandi VR, Buch S, Züllig T, Hartler J, Köfeler HC, Röcken C, Coskun Ü, Klipp E, von Schoenfels W, Gross J, Schafmayer C, Hampe J, Pauling JK, Shevchenko A. Nonalcoholic fatty liver disease stratification by liver lipidomics. J Lipid Res 2021; 62:100104. [PMID: 34384788 PMCID: PMC8488246 DOI: 10.1016/j.jlr.2021.100104] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/20/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common metabolic dysfunction leading to hepatic steatosis. However, NAFLD's global impact on the liver lipidome is poorly understood. Using high-resolution shotgun mass spectrometry, we quantified the molar abundance of 316 species from 22 major lipid classes in liver biopsies of 365 patients, including nonsteatotic patients with normal or excessive weight, patients diagnosed with NAFL (nonalcoholic fatty liver) or NASH (nonalcoholic steatohepatitis), and patients bearing common mutations of NAFLD-related protein factors. We confirmed the progressive accumulation of di- and triacylglycerols and cholesteryl esters in the liver of NAFL and NASH patients, while the bulk composition of glycerophospho- and sphingolipids remained unchanged. Further stratification by biclustering analysis identified sphingomyelin species comprising n24:2 fatty acid moieties as membrane lipid markers of NAFLD. Normalized relative abundance of sphingomyelins SM 43:3;2 and SM 43:1;2 containing n24:2 and n24:0 fatty acid moieties, respectively, showed opposite trends during NAFLD progression and distinguished NAFL and NASH lipidomes from the lipidome of nonsteatotic livers. Together with several glycerophospholipids containing a C22:6 fatty acid moiety, these lipids serve as markers of early and advanced stages of NAFL.
Collapse
Affiliation(s)
- Olga Vvedenskaya
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Tim Daniel Rose
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Oskar Knittelfelder
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Alessandra Palladini
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | | | - Kai Schuhmann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Yuting Wang
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Canan Has
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Mario Brosch
- Department of Medicine I, University Hospital Dresden, Technische Universität (TU) Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Veera Raghavan Thangapandi
- Department of Medicine I, University Hospital Dresden, Technische Universität (TU) Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Stephan Buch
- Department of Medicine I, University Hospital Dresden, Technische Universität (TU) Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Thomas Züllig
- Core Facility Mass Spectrometry, Medical University of Graz, Graz, Austria
| | - Jürgen Hartler
- Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria; Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Harald C Köfeler
- Core Facility Mass Spectrometry, Medical University of Graz, Graz, Austria
| | - Christoph Röcken
- Department of Pathology, University Hospital Schleswig Holstein, Kiel, Schleswig-Holstein, Germany
| | - Ünal Coskun
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Hospital Carl Gustav Carus, Technische Universität (TU) Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Department of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus of Technische Universität Dresden, Dresden, Germany
| | - Edda Klipp
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Witigo von Schoenfels
- Department of Visceral and Thoracic Surgery, University Hospital Schleswig-Holstein, Kiel Campus, Christian-Albrechts-University Kiel, Kiel, Germany; Christian Albrechts University in Kiel Center of Clinical Anatomy Kiel, Schleswig-Holstein, Germany
| | - Justus Gross
- Department of General, Visceral, Vascular and Transplant Surgery, Rostock University Medical Center, Rostock, Germany
| | - Clemens Schafmayer
- Department of General, Visceral, Vascular and Transplant Surgery, Rostock University Medical Center, Rostock, Germany
| | - Jochen Hampe
- Department of Medicine I, University Hospital Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Josch Konstantin Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany.
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
21
|
Impact of Amerind ancestry and FADS genetic variation on omega-3 deficiency and cardiometabolic traits in Hispanic populations. Commun Biol 2021; 4:918. [PMID: 34321601 PMCID: PMC8319323 DOI: 10.1038/s42003-021-02431-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/22/2021] [Indexed: 12/31/2022] Open
Abstract
Long chain polyunsaturated fatty acids (LC-PUFAs) have critical signaling roles that regulate dyslipidemia and inflammation. Genetic variation in the FADS gene cluster accounts for a large portion of interindividual differences in circulating and tissue levels of LC-PUFAs, with the genotypes most strongly predictive of low LC-PUFA levels at strikingly higher frequencies in Amerind ancestry populations. In this study, we examined relationships between genetic ancestry and FADS variation in 1102 Hispanic American participants from the Multi-Ethnic Study of Atherosclerosis. We demonstrate strong negative associations between Amerind genetic ancestry and LC-PUFA levels. The FADS rs174537 single nucleotide polymorphism (SNP) accounted for much of the AI ancestry effect on LC-PUFAs, especially for low levels of n-3 LC-PUFAs. Rs174537 was also strongly associated with several metabolic, inflammatory and anthropomorphic traits including circulating triglycerides (TGs) and E-selectin in MESA Hispanics. Our study demonstrates that Amerind ancestry provides a useful and readily available tool to identify individuals most likely to have FADS-related n-3 LC-PUFA deficiencies and associated cardiovascular risk.
Collapse
|
22
|
Bravo-Ruiz I, Medina MÁ, Martínez-Poveda B. From Food to Genes: Transcriptional Regulation of Metabolism by Lipids and Carbohydrates. Nutrients 2021; 13:nu13051513. [PMID: 33946267 PMCID: PMC8145205 DOI: 10.3390/nu13051513] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/28/2021] [Indexed: 12/31/2022] Open
Abstract
Lipids and carbohydrates regulate gene expression by means of molecules that sense these macronutrients and act as transcription factors. The peroxisome proliferator-activated receptor (PPAR), activated by some fatty acids or their derivatives, and the carbohydrate response element binding protein (ChREBP), activated by glucose-derived metabolites, play a key role in metabolic homeostasis, especially in glucose and lipid metabolism. Furthermore, the action of both factors in obesity, diabetes and fatty liver, as well as the pharmacological development in the treatment of these pathologies are indeed of high relevance. In this review we present an overview of the discovery, mechanism of activation and metabolic functions of these nutrient-dependent transcription factors in different tissues contexts, from the nutritional genomics perspective. The possibility of targeting these factors in pharmacological approaches is also discussed. Lipid and carbohydrate-dependent transcription factors are key players in the complex metabolic homeostasis, but these factors also drive an adaptive response to non-physiological situations, such as overeating. Possibly the decisive role of ChREBP and PPAR in metabolic regulation points to them as ideal therapeutic targets, but their pleiotropic functions in different tissues makes it difficult to "hit the mark".
Collapse
Affiliation(s)
- Inés Bravo-Ruiz
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.B.-R.); (M.Á.M.)
| | - Miguel Ángel Medina
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.B.-R.); (M.Á.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), E-29071 Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Málaga, Spain
| | - Beatriz Martínez-Poveda
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.B.-R.); (M.Á.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), E-29071 Málaga, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), E-28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
23
|
Schiano E, Annunziata G, Ciampaglia R, Iannuzzo F, Maisto M, Tenore GC, Novellino E. Bioactive Compounds for the Management of Hypertriglyceridemia: Evidence From Clinical Trials and Putative Action Targets. Front Nutr 2020; 7:586178. [PMID: 33330588 PMCID: PMC7734325 DOI: 10.3389/fnut.2020.586178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/26/2020] [Indexed: 01/22/2023] Open
Abstract
Hypertriglyceridemia refers to the presence of elevated concentrations of triglycerides (TG) in the bloodstream (TG >200 mg/dL). This lipid alteration is known to be associated with an increased risk of atherosclerosis, contributing overall to the onset of atherosclerotic cardiovascular disease (CVD). Guidelines for the management of hypertriglyceridemia are based on both lifestyle intervention and pharmacological treatment, but poor adherence, medication-related costs and side effects can limit the success of these interventions. For this reason, the search for natural alternative approaches to reduce plasma TG levels currently represents a hot research field. This review article summarizes the most relevant clinical trials reporting the TG-reducing effect of different food-derived bioactive compounds. Furthermore, based on the evidence obtained from in vitro studies, we provide a description and classification of putative targets of action through which several bioactive compounds can exert a TG-lowering effect. Future research may lead to investigations of the efficacy of novel nutraceutical formulations consisting in a combination of bioactive compounds which contribute to the management of plasma TG levels through different action targets.
Collapse
Affiliation(s)
| | | | | | - Fortuna Iannuzzo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Maria Maisto
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
24
|
Danesi F, Larsen BD, Di Nunzio M, Nielsen R, de Biase D, Valli V, Mandrup S, Bordoni A. Co-Administration of Propionate or Protocatechuic Acid Does Not Affect DHA-Specific Transcriptional Effects on Lipid Metabolism in Cultured Hepatic Cells. Nutrients 2020; 12:2952. [PMID: 32993128 PMCID: PMC7599819 DOI: 10.3390/nu12102952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/08/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
Long-chain n-3 polyunsaturated fatty acids (n-3 LC-PUFAs) are collectively recognized triglyceride-lowering agents, and their preventive action is likely mediated by changes in gene expression. However, as most studies employ fish oil, which contains a mixture of n-3 LC-PUFAs, the docosahexaenoic acid (DHA)-specific transcriptional effects on lipid metabolism are still unclear. The aim of the present study was to further elucidate the DHA-induced transcriptional effects on lipid metabolism in the liver, and to investigate the effects of co-administration with other bioactive compounds having effects on lipid metabolism. To this purpose, HepG2 cells were treated for 6 or 24 h with DHA, the short-chain fatty acid propionate (PRO), and protocatechuic acid (PCA), the main human metabolite of cyanidin-glucosides. Following supplementation, we mapped the global transcriptional changes. PRO and PCA alone had a very slight effect on the transcriptome; on the contrary, supplementation of DHA highly repressed the steroid and fatty acid biosynthesis pathways, this transcriptional modulation being not affected by co-supplementation. Our results confirm that DHA effect on lipid metabolism are mediated at least in part by modulation of the expression of specific genes. PRO and PCA could contribute to counteracting dyslipidemia through other mechanisms.
Collapse
Affiliation(s)
- Francesca Danesi
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, 47521 Cesena, Italy; (F.D.); (M.D.N.); (V.V.)
- Interdepartmental Center for Agri-food Industrial Research (CIRI Agrifood), University of Bologna, 47521 Cesena, Italy
| | - Bjørk D. Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Syddansk Universitet, 5230 Odense M, Denmark; (B.D.L.); (R.N.); (S.M.)
| | - Mattia Di Nunzio
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, 47521 Cesena, Italy; (F.D.); (M.D.N.); (V.V.)
- Interdepartmental Center for Agri-food Industrial Research (CIRI Agrifood), University of Bologna, 47521 Cesena, Italy
| | - Ronni Nielsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Syddansk Universitet, 5230 Odense M, Denmark; (B.D.L.); (R.N.); (S.M.)
| | - Dario de Biase
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40138 Bologna, Italy;
| | - Veronica Valli
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, 47521 Cesena, Italy; (F.D.); (M.D.N.); (V.V.)
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Syddansk Universitet, 5230 Odense M, Denmark; (B.D.L.); (R.N.); (S.M.)
| | - Alessandra Bordoni
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, 47521 Cesena, Italy; (F.D.); (M.D.N.); (V.V.)
- Interdepartmental Center for Agri-food Industrial Research (CIRI Agrifood), University of Bologna, 47521 Cesena, Italy
| |
Collapse
|
25
|
Li Y, Pang Y, Zhao Z, Xiang X, Mai K, Ai Q. Molecular Characterization, Nutritional and Insulin Regulation of Elovl6 in Rainbow Trout ( Oncorhynchus mykiss). Biomolecules 2020; 10:biom10020264. [PMID: 32050615 PMCID: PMC7072538 DOI: 10.3390/biom10020264] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
Elongation of very long-chain fatty acids protein 6 (Elovl6) is a crucial enzyme in the synthesis of endogenous fatty acids, which participates in the energy balance and metabolic diseases. The main objective of this study was to explore the molecular characterization of Elovl6 and the regulation of elovl6 expression in response to dietary fatty acids and insulin. In the present study, the ORF (open reading frame) of Elovl6 from rainbow trout was cloned and characterized, which showed a high identity (87%) with mammals and other teleost. The results of quantitative PCR showed that the transcriptional levels of elovl6 from rainbow trout that were fed diets containing soybean oil (enriched with 18:2n-6, linoleic acid (LA)) or linseed oil (enriched with 18:3n-3, α-linolenic acid (ALA)) were lower than those in the group that were fed diets containing fish oil (enriched with 20:5n-3, eicosapentaenoic acid (EPA) and 22:6n-3, docosahexaenoic acid (DHA)). Correspondingly, mRNA expression of elovl6 in hepatocytes treated with DHA was dramatically higher than that in LA and ALA groups. The transcriptional expression of elovl6 in hepatocytes treated with insulin was also significantly increased. Moreover, the dual luciferase assay showed the transcription factor CREB1 dramatically up-regulated the promoter activity of elovl6, while FOXO1 significantly down-regulated the elovl6 promoter activity in rainbow trout. The differences in transcriptional expression of crbe1 and foxo1 may contribute to the increase or decrease of elovl6 expression in rainbow trout in response to fatty acids or insulin. These findings revealed the molecular characterization of elovl6 and the regulation of elovl6 expression by CREB1 and FOXO1 in rainbow trout in response to dietary fatty acids or insulin.
Collapse
Affiliation(s)
- Yongnan Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (Y.L.); (Y.P.); (Z.Z.); (X.X.); (K.M.)
| | - Yuning Pang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (Y.L.); (Y.P.); (Z.Z.); (X.X.); (K.M.)
| | - Zengqi Zhao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (Y.L.); (Y.P.); (Z.Z.); (X.X.); (K.M.)
| | - Xiaojun Xiang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (Y.L.); (Y.P.); (Z.Z.); (X.X.); (K.M.)
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (Y.L.); (Y.P.); (Z.Z.); (X.X.); (K.M.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (Y.L.); (Y.P.); (Z.Z.); (X.X.); (K.M.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
- Correspondence: ; Tel.: +86-0532-82031943
| |
Collapse
|
26
|
Amiel A, Tremblay-Franco M, Gautier R, Ducheix S, Montagner A, Polizzi A, Debrauwer L, Guillou H, Bertrand-Michel J, Canlet C. Proton NMR Enables the Absolute Quantification of Aqueous Metabolites and Lipid Classes in Unique Mouse Liver Samples. Metabolites 2019; 10:metabo10010009. [PMID: 31877749 PMCID: PMC7023327 DOI: 10.3390/metabo10010009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatic metabolites provide valuable information on the physiological state of an organism, and thus, they are monitored in many clinical situations. Typically, monitoring requires several analyses for each class of targeted metabolite, which is time consuming. The present study aimed to evaluate a proton nuclear magnetic resonance (1H-NMR) method for obtaining quantitative measurements of aqueous and lipidic metabolites. We optimized the extraction protocol, the standard samples, and the organic solvents for the absolute quantification of lipid species. To validate the method, we analyzed metabolic profiles in livers of mice fed three different diets. We compared our results with values obtained with conventional methods and found strong correlations. The 1H-NMR protocol enabled the absolute quantification of 29 aqueous metabolites and eight lipid classes. Results showed that mice fed a diet enriched in saturated fatty acids had higher levels of triglycerides, cholesterol ester, monounsaturated fatty acids, lactate, 3-hydroxy-butyrate, and alanine and lower levels of glucose, compared to mice fed a control diet. In conclusion, proton NMR provided a rapid overview of the main lipid classes (triglycerides, cholesterol, phospholipids, fatty acids) and the most abundant aqueous metabolites in liver.
Collapse
Affiliation(s)
- Aurélien Amiel
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
| | - Marie Tremblay-Franco
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
| | - Roselyne Gautier
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
| | - Simon Ducheix
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
| | - Alexandra Montagner
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
| | - Arnaud Polizzi
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
| | - Laurent Debrauwer
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
| | - Hervé Guillou
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
| | | | - Cécile Canlet
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
- Correspondence:
| |
Collapse
|
27
|
Skulas-Ray AC, Wilson PWF, Harris WS, Brinton EA, Kris-Etherton PM, Richter CK, Jacobson TA, Engler MB, Miller M, Robinson JG, Blum CB, Rodriguez-Leyva D, de Ferranti SD, Welty FK. Omega-3 Fatty Acids for the Management of Hypertriglyceridemia: A Science Advisory From the American Heart Association. Circulation 2019; 140:e673-e691. [PMID: 31422671 DOI: 10.1161/cir.0000000000000709] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hypertriglyceridemia (triglycerides 200-499 mg/dL) is relatively common in the United States, whereas more severe triglyceride elevations (very high triglycerides, ≥500 mg/dL) are far less frequently observed. Both are becoming increasingly prevalent in the United States and elsewhere, likely driven in large part by growing rates of obesity and diabetes mellitus. In a 2002 American Heart Association scientific statement, the omega-3 fatty acids (n-3 FAs) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were recommended (at a dose of 2-4 g/d) for reducing triglycerides in patients with elevated triglycerides. Since 2002, prescription agents containing EPA+DHA or EPA alone have been approved by the US Food and Drug Administration for treating very high triglycerides; these agents are also widely used for hypertriglyceridemia. The purpose of this advisory is to summarize the lipid and lipoprotein effects resulting from pharmacological doses of n-3 FAs (>3 g/d total EPA+DHA) on the basis of new scientific data and availability of n-3 FA agents. In treatment of very high triglycerides with 4 g/d, EPA+DHA agents reduce triglycerides by ≥30% with concurrent increases in low-density lipoprotein cholesterol, whereas EPA-only did not raise low-density lipoprotein cholesterol in very high triglycerides. When used to treat hypertriglyceridemia, n-3 FAs with EPA+DHA or with EPA-only appear roughly comparable for triglyceride lowering and do not increase low-density lipoprotein cholesterol when used as monotherapy or in combination with a statin. In the largest trials of 4 g/d prescription n-3 FA, non-high-density lipoprotein cholesterol and apolipoprotein B were modestly decreased, indicating reductions in total atherogenic lipoproteins. The use of n-3 FA (4 g/d) for improving atherosclerotic cardiovascular disease risk in patients with hypertriglyceridemia is supported by a 25% reduction in major adverse cardiovascular events in REDUCE-IT (Reduction of Cardiovascular Events With EPA Intervention Trial), a randomized placebo-controlled trial of EPA-only in high-risk patients treated with a statin. The results of a trial of 4 g/d prescription EPA+DHA in hypertriglyceridemia are anticipated in 2020. We conclude that prescription n-3 FAs (EPA+DHA or EPA-only) at a dose of 4 g/d (>3 g/d total EPA+DHA) are an effective and safe option for reducing triglycerides as monotherapy or as an adjunct to other lipid-lowering agents.
Collapse
|
28
|
Tang M, Floyd S, Cai H, Zhang M, Yang R, Dang R. The status of ω-3 PUFAs influence chronic unpredicted mild stress-induced metabolic side effects in rats through INSIG/SREBP pathway. Food Funct 2019; 10:4649-4660. [PMID: 31292598 DOI: 10.1039/c9fo00076c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Metabolic disturbances, including lipid metabolism, bone metabolism, and glycometabolism, are common in depression. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs), which are reported to possess antidepressant effect, have also been shown to regulate metabolism. To further clarify the potential link between ω-3 PUFAs and stress-induced metabolic disturbances, metabolic-related parameters including body weight, visceral fat, fatty acid composition and serum parameters, such as serum lipids, free fatty acid (FFA), glucose (GLU), calcium and phosphorus in rats were measured. Moreover, hepatic insulin induced gene (INSIG)/sterol regulatory element binding protein (SREBP) pathway was also investigated. After 5 weeks of chronic unpredicted mild stress (CUMS) administration, rats were induced to a depressive-like state and exhibited decreased serum high-density lipoprotein (HDL-c), body weight and visceral fat, accompanied by altered C18:2n6c and ω-3/ω-6 PUFAs. Supplement of ω-3 PUFAs showed robust antidepressant effects and has beneficial effects on lipid profile. On the contrary, ω-3 PUFAs deficiency induced the visceral fat accumulation and decreased the serum calcium and phosphorus in stressed rats. Additionally, CUMS significantly increased hepatic expressions of SREBP-cleavage activating protein (SCAP)/SREBP-1 and decreased the expression of INSIG-1. This disturbance of SREBPs system is aggravated by ω-3 PUFAs deficiency and alleviated by ω-3 PUFAs supplementation. This study discloses the novel findings that ω-3 PUFAs deficiency will exacerbate the metabolic disturbances in stressed rats. Furthermore, supplementation of ω-3 PUFAs on individuals with a high risk of depression might be an effective way to prevent metabolic disorders accompanied by depression with the involvement of INSIG/SREBP pathway.
Collapse
Affiliation(s)
- Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China.
| | | | | | | | | | | |
Collapse
|
29
|
Rundblad A, Larsen SV, Myhrstad MC, Ottestad I, Thoresen M, Holven KB, Ulven SM. Differences in peripheral blood mononuclear cell gene expression and triglyceride composition in lipoprotein subclasses in plasma triglyceride responders and non-responders to omega-3 supplementation. GENES AND NUTRITION 2019; 14:10. [PMID: 31057673 PMCID: PMC6485081 DOI: 10.1186/s12263-019-0633-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/20/2019] [Indexed: 01/07/2023]
Abstract
Background Intake of the marine omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) reduces fasting triglyceride (TG) levels and may thereby lower cardiovascular disease risk. However, there are large inter-individual differences in the TG-lowering effect of omega-3 supplementation. Genotype differences partly explain this variation, but gene-environment interactions leading to gene expression differences may also be important. In this study, we aimed to investigate baseline differences and differences in the change in peripheral blood mononuclear cell (PBMC) gene expression and lipoprotein subclass TG levels between TG responders and non-responders to omega-3 fatty acid supplementation. Methods In a previous randomized controlled trial, healthy normotriglyceridemic subjects (n = 35, 71% women) received 1.6 g EPA + DHA/day for 7 weeks. In this exploratory sub-study, we defined TG responders as subjects having a TG reduction beyond the 20% day-to-day variation and non-responders as having a TG change between − 20% and + 20% after omega-3 supplementation. PBMC gene expression was measured using microarray, and lipoprotein subclasses were measured using nuclear magnetic resonance spectroscopy. Results Eight subjects were defined as responders with a median TG reduction of 37%, and 16 subjects were defined as non-responders with a median TG change of 0%. At baseline, responders had higher TG levels in two of four high-density lipoprotein (HDL) subclasses and 909 gene transcripts (p ≤ 0.05) were differentially expressed compared to non-responders. During the intervention, the plasma TG reduction among responders was reflected in TG reductions in four of six different very low-density lipoprotein subclasses and three of four different HDL subclasses. Compared to non-responders, the expression of 454 transcripts was differentially altered in responders (p ≤ 0.05). Pathway analyses revealed that responders had altered signaling pathways related to development and immune function. In addition, two of the top 10 enriched pathways in responders compared to non-responders were related to lysophosphatidic acid signaling. Conclusion TG responders and non-responders to omega-3 supplementation have different lipoprotein subclass and PBMC gene expression profiles at baseline and different lipoprotein subclass and PBMC gene expression responses to omega-3 supplementation. These gene expression differences may partially explain the variability in TG response observed after omega-3 supplementation. Graphical abstract Based on free images from Servier Medical Art (Creative Commons Attribution License) and image from www.colourbox.com.![]() Electronic supplementary material The online version of this article (10.1186/s12263-019-0633-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amanda Rundblad
- 1Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317 Oslo, Norway
| | - Sunniva V Larsen
- 1Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317 Oslo, Norway
| | - Mari C Myhrstad
- 2Department of Nursing and Health Promotion, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, PO Box 4, St Olavs plass, 0130 Oslo, Norway
| | - Inger Ottestad
- 1Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317 Oslo, Norway
| | - Magne Thoresen
- 3Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317 Oslo, Norway
| | - Kirsten B Holven
- 1Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317 Oslo, Norway.,4National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, PO Box 4950, Nydalen, 0424 Oslo, Norway
| | - Stine M Ulven
- 1Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317 Oslo, Norway
| |
Collapse
|
30
|
Kang YP, Yoon JH, Long NP, Koo GB, Noh HJ, Oh SJ, Lee SB, Kim HM, Hong JY, Lee WJ, Lee SJ, Hong SS, Kwon SW, Kim YS. Spheroid-Induced Epithelial-Mesenchymal Transition Provokes Global Alterations of Breast Cancer Lipidome: A Multi-Layered Omics Analysis. Front Oncol 2019; 9:145. [PMID: 30949448 PMCID: PMC6437068 DOI: 10.3389/fonc.2019.00145] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/20/2019] [Indexed: 01/06/2023] Open
Abstract
Metabolic rewiring has been recognized as an important feature to the progression of cancer. However, the essential components and functions of lipid metabolic networks in breast cancer progression are not fully understood. In this study, we investigated the roles of altered lipid metabolism in the malignant phenotype of breast cancer. Using a spheroid-induced epithelial-mesenchymal transition (EMT) model, we conducted multi-layered lipidomic and transcriptomic analysis to comprehensively describe the rewiring of the breast cancer lipidome during the malignant transformation. A tremendous homeostatic disturbance of various complex lipid species including ceramide, sphingomyelin, ether-linked phosphatidylcholines, and ether-linked phosphatidylethanolamine was found in the mesenchymal state of cancer cells. Noticeably, polyunsaturated fatty acids composition in spheroid cells was significantly decreased, accordingly with the gene expression patterns observed in the transcriptomic analysis of associated regulators. For instance, the up-regulation of SCD, ACOX3, and FADS1 and the down-regulation of PTPLB, PECR, and ELOVL2 were found among other lipid metabolic regulators. Significantly, the ratio of C22:6n3 (docosahexaenoic acid, DHA) to C22:5n3 was dramatically reduced in spheroid cells analogously to the down-regulation of ELOVL2. Following mechanistic study confirmed the up-regulation of SCD and down-regulation of PTPLB, PECR, ELOVL2, and ELOVL3 in the spheroid cells. Furthermore, the depletion of ELOVL2 induced metastatic characteristics in breast cancer cells via the SREBPs axis. A subsequent large-scale analysis using 51 breast cancer cell lines demonstrated the reduced expression of ELOVL2 in basal-like phenotypes. Breast cancer patients with low ELOVL2 expression exhibited poor prognoses (HR = 0.76, CI = 0.67–0.86). Collectively, ELOVL2 expression is associated with the malignant phenotypes and appear to be a novel prognostic biomarker in breast cancer. In conclusion, the present study demonstrates that there is a global alteration of the lipid composition during EMT and suggests the down-regulation of ELOVL2 induces lipid metabolism reprogramming in breast cancer and contributes to their malignant phenotypes.
Collapse
Affiliation(s)
- Yun Pyo Kang
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jung-Ho Yoon
- Department of Biochemistry, Ajou University School of Medicine, Suwon, South Korea.,Department of Biomedical Sciences, Graduate School, Ajou University, Suwon, South Korea
| | | | - Gi-Bang Koo
- Department of Biochemistry, Ajou University School of Medicine, Suwon, South Korea.,Department of Biomedical Sciences, Graduate School, Ajou University, Suwon, South Korea
| | - Hyun-Jin Noh
- Department of Biochemistry, Ajou University School of Medicine, Suwon, South Korea.,Department of Biomedical Sciences, Graduate School, Ajou University, Suwon, South Korea
| | - Seung-Jae Oh
- Department of Biochemistry, Ajou University School of Medicine, Suwon, South Korea.,Department of Biomedical Sciences, Graduate School, Ajou University, Suwon, South Korea
| | - Sae Bom Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Hyung Min Kim
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Ji Yeon Hong
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Won Jun Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Seul Ji Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, South Korea
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul, South Korea.,Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Suwon, South Korea.,Department of Biomedical Sciences, Graduate School, Ajou University, Suwon, South Korea
| |
Collapse
|
31
|
Yu X, Huang S, Deng Q, Tang Y, Yao P, Tang H, Dong X. Linseed oil improves hepatic insulin resistance in obese mice through modulating mitochondrial quality control. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
32
|
Dziedzic B, Bewicz-Binkowska D, Zgorzynska E, Stulczewski D, Wieteska L, Kaza B, Walczewska A. DHA upregulates FADS2 expression in primary cortical astrocytes exposed to vitamin A. Physiol Res 2018; 67:663-668. [PMID: 29750879 DOI: 10.33549/physiolres.933708] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The fads2 gene encoding delta6-desaturase, the rate-limiting enzyme of the LCPUFA biosynthesis is expressed in astrocytes. Dietary fatty acids, which cross the blood-brain barrier, may regulate the transcription of lipogenic enzymes through activation of transcription factors such as peroxisome proliferator-activated receptors (PPARs). The PPARs form the transcription complex with retinoid X receptors (RXRs) that are activated by 9-cis retinoic acid, a metabolite of vitamin A (VA). The study examines whether challenge of astrocytes with VA, prior 24-h treatment with palmitic acid (PA), alpha-linolenic acid (ALA) or docosahexaenoic acid (DHA) has the effect on the FADS2 expression. RT-qPCR showed that in astrocytes not challenged with VA, PA increased fads2 gene expression and DHA decreased it. However, in VA-primed astrocytes, PA doubled the FADS2 mRNA levels, while DHA increased fads2 gene expression, oppositely to non-primed cells. Furthermore, similar changes were seen in VA-primed astrocytes with regard to delta6-desaturase protein levels following PA and DHA treatment. ALA did not have any effect on the FADS2 mRNA and protein levels in either VA-primed or non-primed astrocytes. These findings indicate that in the presence of vitamin A, DHA upregulates fads2 gene expression in astrocytes.
Collapse
Affiliation(s)
- B Dziedzic
- Department of Cell-to-Cell Communication, Medical University of Lodz, Lodz, Poland.
| | | | | | | | | | | | | |
Collapse
|
33
|
Scorletti E, Byrne CD. Omega-3 fatty acids and non-alcoholic fatty liver disease: Evidence of efficacy and mechanism of action. Mol Aspects Med 2018; 64:135-146. [PMID: 29544992 DOI: 10.1016/j.mam.2018.03.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/07/2018] [Accepted: 03/09/2018] [Indexed: 02/07/2023]
Abstract
For many years it has been known that high doses of long chain omega-3 fatty acids are beneficial in the treatment of hypertriglyceridaemia. Over the last three decades, there has also been a wealth of in vitro and in vivo data that has accumulated to suggest that long chain omega-3 fatty acid treatment might be beneficial to decrease liver triacylglycerol. Several biological mechanisms have been identified that support this hypothesis; notably, it has been shown that long chain omega-3 fatty acids have a beneficial effect: a) on bioactive metabolites involved in inflammatory pathways, and b) on alteration of nuclear transcription factor activities such as peroxisome proliferator-activated receptors (PPARs), sterol regulatory element-binding protein 1c (SREBP-1c) and carbohydrate-responsive element-binding protein (ChREBP), involved in inflammatory pathways and liver lipid metabolism. Since the pathogenesis of non alcoholic fatty liver disease (NAFLD) begins with the accumulation of liver lipid and progresses with inflammation and then several years later with development of fibrosis; it has been thought in patients with NAFLD omega-3 fatty acid treatment would be beneficial in treating liver lipid and possibly also in ameliorating inflammation. Meta-analyses (of predominantly dietary studies and small trials) have tended to support the assertion that omega-3 fatty acids are beneficial in decreasing liver lipid, but recent randomised controlled trials have produced conflicting data. These trials have suggested that omega-3 fatty acid might be beneficial in decreasing liver triglyceride (docosahexanoic acid also possibly being more effective than eicosapentanoic acid) but not in decreasing other features of steatohepatitis (or liver fibrosis). The purpose of this review is to discuss recent evidence regarding biological mechanisms by which long chain omega-3 fatty acids might act to ameliorate liver disease in NAFLD; to consider the recent evidence from randomised trials in both adults and children with NAFLD; and finally to discuss key 'known unknowns' that need to be considered, before planning future studies that are focussed on testing the effects of omega-3 fatty acid treatment in patients with NAFLD.
Collapse
Affiliation(s)
- Eleonora Scorletti
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| | - Christopher D Byrne
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
34
|
Jump DB, Lytle KA, Depner CM, Tripathy S. Omega-3 polyunsaturated fatty acids as a treatment strategy for nonalcoholic fatty liver disease. Pharmacol Ther 2017; 181:108-125. [PMID: 28723414 DOI: 10.1016/j.pharmthera.2017.07.007] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obese and type 2 diabetic (T2DM) patients have a high prevalence of nonalcoholic fatty liver disease (NAFLD). NAFLD is a continuum of chronic liver diseases ranging from benign hepatosteatosis to nonalcoholic steatohepatitis (NASH), cirrhosis and primary hepatocellular cancer (HCC). Because of its strong association with the obesity epidemic, NAFLD is rapidly becoming a major public health concern worldwide. Surprisingly, there are no FDA approved NAFLD therapies; and current therapies focus on the co-morbidities associated with NAFLD, namely, obesity, hyperglycemia, dyslipidemia, and hypertension. The goal of this review is to provide background on the disease process, discuss human studies and preclinical models that have examined treatment options. We also provide an in-depth rationale for the use of dietary ω3 polyunsaturated fatty acid (ω3 PUFA) supplements as a treatment option for NAFLD. This focus is based on recent studies indicating that NASH patients and preclinical mouse models of NASH have low levels of hepatic C20-22 ω3 PUFA. This decline in hepatic PUFA may account for the major phenotypic features associated with NASH, including steatosis, inflammation and fibrosis. Finally, our discussion will address the strengths and limitations of ω3 PUFA supplements use in NAFLD therapy.
Collapse
Affiliation(s)
- Donald B Jump
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, United States.
| | - Kelli A Lytle
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, United States
| | - Christopher M Depner
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, United States
| | - Sasmita Tripathy
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, United States
| |
Collapse
|
35
|
Tajima-Shirasaki N, Ishii KA, Takayama H, Shirasaki T, Iwama H, Chikamoto K, Saito Y, Iwasaki Y, Teraguchi A, Lan F, Kikuchi A, Takeshita Y, Murao K, Matsugo S, Kaneko S, Misu H, Takamura T. Eicosapentaenoic acid down-regulates expression of the selenoprotein P gene by inhibiting SREBP-1c protein independently of the AMP-activated protein kinase pathway in H4IIEC3 hepatocytes. J Biol Chem 2017; 292:10791-10800. [PMID: 28465347 PMCID: PMC5491766 DOI: 10.1074/jbc.m116.747006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 04/27/2017] [Indexed: 02/05/2023] Open
Abstract
Selenoprotein P (encoded by SELENOP in humans, Selenop in rat), a liver-derived secretory protein, induces resistance to insulin and vascular endothelial growth factor (VEGF) in type 2 diabetes. Suppression of selenoprotein P may provide a novel therapeutic approach to treating type 2 diabetes; however, few drugs inhibiting SELENOP expression in hepatocytes have been identified. The present findings demonstrate that eicosapentaenoic acid (EPA) suppresses SELENOP expression by inactivating sterol regulatory element-binding protein-1c (SREBP-1c, encoded by Srebf1 in rat) in H4IIEC3 hepatocytes. Treatment with EPA caused concentration- and time-dependent reduction in SELENOP promoter activity. EPA activated AMP-activated protein kinase (AMPK); however, the inhibitory effect of EPA on SELENOP promoter activity was not canceled with an AMPK inhibitor compound C and dominant-negative AMPK transfection. Deletion mutant promoter assays and computational analysis of transcription factor-binding sites conserved among the species resulted in identification of a sterol regulatory element (SRE)-like site in the SELENOP promoter. A chromatin immunoprecipitation (ChIP) assay revealed that EPA decreases binding of SREBP-1c to the SELENOP promoter. Knockdown of Srebf1 resulted in a significant down-regulation of Selenop expression. Conversely, SREBP-1c overexpression inhibited the suppressive effect of EPA. These data provide a novel mechanism of action for EPA involving improvement of systemic insulin sensitivity through the regulation of selenoprotein P production independently of the AMPK pathway and suggest an additional approach to developing anti-diabetic drugs.
Collapse
Affiliation(s)
| | - Kiyo-Aki Ishii
- From the Departments of Endocrinology and Metabolism
- System Biology, and
| | - Hiroaki Takayama
- From the Departments of Endocrinology and Metabolism
- System Biology, and
| | - Takayoshi Shirasaki
- System Biology, and
- Advanced Medical Technology, Kanazawa University Graduate School of Health Medicine, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641
| | - Hisakazu Iwama
- the Life Science Research Center, Kagawa University, Ikenobe 1750-1, Miki-cho, Kita-gun, Kagawa 761-0793
| | - Keita Chikamoto
- From the Departments of Endocrinology and Metabolism
- the Division of Natural System, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192
| | - Yoshiro Saito
- the Department of Medical Life Systems, Faculty of Medical and Life Sciences, Systems Life Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394
| | | | | | - Fei Lan
- From the Departments of Endocrinology and Metabolism
| | - Akihiro Kikuchi
- From the Departments of Endocrinology and Metabolism
- System Biology, and
| | | | - Koji Murao
- the Department of Advanced Medicine, Kagawa University, Ikenobe 1750-1, Miki-cho, Kita-gun, Kagawa 761-0793, and
| | - Seiichi Matsugo
- the Division of Natural System, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192
| | | | - Hirofumi Misu
- From the Departments of Endocrinology and Metabolism
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | | |
Collapse
|
36
|
Teixeira PD, Oliveira DM, Chizzotti ML, Chalfun-Junior A, Coelho TC, Gionbelli M, Paiva LV, Carvalho JRR, Ladeira MM. Subspecies and diet affect the expression of genes involved in lipid metabolism and chemical composition of muscle in beef cattle. Meat Sci 2017; 133:110-118. [PMID: 28666109 DOI: 10.1016/j.meatsci.2017.06.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 05/08/2017] [Accepted: 06/16/2017] [Indexed: 01/06/2023]
Abstract
Fourteen Nellore and 14 Angus young bulls with BW of 381±11.8kg were randomly assigned into 2 feeding groups (whole shelled corn without forage (WSC) or corn silage and ground corn (GC)) to evaluate chemical composition and expression of genes involved in lipid metabolism in the longissimus thoracis (LT). We hypothesized that bulls fed the WSC diet have greater amounts of intramuscular fat and Angus have higher expression levels of PPAR and SREBF. Meat from Angus bulls had greater ether extract compared to Nellore (P<0.05). Muscle from bulls fed the WSC diet had greater expression of PPARA (P<0.05) and lower levels of SREBF1 expression (P<0.01). The LT of Nellore fed GC had greater expression of FABP4, ACACA and SCD genes (P<0.01). In conclusion, the greater concentration of starch in the WSC diet did not increase marbling in the beef of bulls fed this diet due to the reduced expression of SREBF1.
Collapse
Affiliation(s)
- Priscilla D Teixeira
- Department of Animal Science, Federal University of Lavras, Lavras, Minas Gerais 37.200-000, Brazil
| | - Dalton M Oliveira
- Department of Animal Science, State University of Mato Grasso do Sul, Aquidauana, Mato Grosso do Sul 79.200-000, Brazil
| | - Mario L Chizzotti
- Department of Animal Science, Federal University of Viçosa, Viçosa, Minas Gerais 37.200-000, Brazil
| | - Antonio Chalfun-Junior
- Department of Biology, Federal University of Lavras, Lavras, Minas Gerais 37.200-000, Brazil
| | - Tamara C Coelho
- Department of Animal Science, Federal University of Lavras, Lavras, Minas Gerais 37.200-000, Brazil
| | - MateusP Gionbelli
- Department of Animal Science, Federal University of Lavras, Lavras, Minas Gerais 37.200-000, Brazil
| | - Luciano V Paiva
- Department of Chemistry, Federal University of Lavras, Lavras, Minas Gerais 37.200-000, Brazil
| | - José Roberto R Carvalho
- Department of Animal Science, Federal University of Lavras, Lavras, Minas Gerais 37.200-000, Brazil
| | - Marcio M Ladeira
- Department of Animal Science, Federal University of Lavras, Lavras, Minas Gerais 37.200-000, Brazil.
| |
Collapse
|
37
|
Yao DW, Luo J, He QY, Li J, Wang H, Shi HB, Xu HF, Wang M, Loor JJ. Characterization of the liver X receptor-dependent regulatory mechanism of goat stearoyl-coenzyme A desaturase 1 gene by linoleic acid. J Dairy Sci 2017; 99:3945-3957. [PMID: 26947306 DOI: 10.3168/jds.2015-10601] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/13/2016] [Indexed: 01/05/2023]
Abstract
Stearoyl-coenzyme A desaturase 1 (SCD1) is a key enzyme in the biosynthesis of palmitoleic and oleic acid. Although the transcriptional regulatory mechanism of SCD1 via polyunsaturated fatty acids (PUFA) has been extensively explored in nonruminants, the existence of such mechanism in ruminant mammary gland remains unknown. In this study, we used goat genomic DNA to clone and sequence a 1,713-bp fragment of the SCD1 5' flanking region. Deletion assays revealed a core region of the promoter located between -415 and -109 bp upstream of the transcription start site, and contained the highly conserved PUFA response region. An intact PUFA response region was required for the basal transcriptional activity of SCD1. Linoleic acid reduced endogenous expression of SCD1 and sterol regulatory element binding factor-1 (SREBF1) in goat mammary epithelial cells. Further analysis indicated that both the sterol response element (SRE) and the nuclear factor Y (NF-Y) binding site in the SCD1 promoter were responsible for the inhibition effect by linoleic acid, whereas the effect was abrogated once NF-Y was deleted. In addition, SRE and NF-Y were partly responsible for the transcriptional activation induced via the liver X receptor agonist T 4506585 (Sigma-Aldrich, St. Louis, MO). When goat mammary epithelial cells were cultured with linoleic acid, addition of T 4506585 markedly increased SCD1 transcription in controls, but had no effect on cells with a deleted SRE promoter. These results demonstrated that linoleic acid can regulate SCD1 expression at the transcriptional level through SRE and NF-Y in a liver X receptor-dependent fashion in the goat mammary gland.
Collapse
Affiliation(s)
- D W Yao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China 712100
| | - J Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China 712100.
| | - Q Y He
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China 712100
| | - J Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, P. R. China 450046
| | - H Wang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China 712100
| | - H B Shi
- College of Animal Sciences, Zhejiang Sci-Tech University, Hangzhou, P. R. China 310058
| | - H F Xu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China 712100
| | - M Wang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P. R. China 712100
| | - J J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
38
|
Rodríguez-Cruz M, Serna DS. Nutrigenomics of ω-3 fatty acids: Regulators of the master transcription factors. Nutrition 2017; 41:90-96. [PMID: 28760435 DOI: 10.1016/j.nut.2017.04.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 03/27/2017] [Accepted: 04/24/2017] [Indexed: 11/19/2022]
Abstract
It is well known that ω-3 long-chain polyunsaturated fatty acids (LC-PUFAs) control some key molecular cell mechanisms, resulting in a beneficial role in inflammatory diseases. Such mechanisms are complex and reflect the diversity of their functions, mainly as modulators of the dynamic properties of membranes, regulators of gene expression, and precursors of active mediators. The aim of this review is to summarize the state of the art of the effects and mechanisms by which ω-3 LC-PUFAs such as eicosapentaenoic acid (EPA, C22:5 ω-3) and docosahexaenoic acid (DHA, C22:6 ω-3) regulate different metabolic processes to maintain homeostasis. Thus, we describe some aspects of these fatty acids-from their structural function in cell membranes to their role as regulators of gene expression, mainly in lipid metabolism. However, further studies are required to elucidate these actions and to have a better understanding in regard to the beneficial effects of ω-3 LC-PUFAs in the pathogenesis of several diseases as well as their functions as nutrients with protective action to avoid or delay development of these diseases. Furthermore, it is necessary to highlight the lack of comprehensive studies including nutritional, biochemical, genetic, and immune aspects to identify specific molecular mechanisms involved in the beneficial effects of consumption of DHA (C22:6 ω-3) and EPA (C22:5 ω-3) and their metabolic derivatives on health promotion.
Collapse
Affiliation(s)
- Maricela Rodríguez-Cruz
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, México City, México.
| | - Donovan Solís Serna
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, México City, México
| |
Collapse
|
39
|
Betancor MB, Ortega A, de la Gándara F, Tocher DR, Mourente G. Lipid metabolism-related gene expression pattern of Atlantic bluefin tuna (Thunnus thynnus L.) larvae fed on live prey. FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:493-516. [PMID: 27815797 PMCID: PMC5374188 DOI: 10.1007/s10695-016-0305-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 10/08/2016] [Indexed: 05/25/2023]
Abstract
The present study is the first to evaluate lipid metabolism in first-feeding Atlantic bluefin tuna (ABT; Thunnus thynnus L.) larvae fed different live prey including enriched rotifers Brachionus plicatilis and Acartia sp. copepod nauplii from 2 days after hatch. Understanding the molecular basis of lipid metabolism and regulation in ABT will provide insights to optimize diet formulations for this high-value species new to aquaculture. To this end, we investigated the effect of dietary lipid on whole larvae lipid class and fatty acid compositions and the expression of key genes involved in lipid metabolism in first feeding ABT larvae fed different live prey. Additionally, the expression of lipid metabolism genes in tissues of adult broodstock ABT was evaluated. Growth and survival data indicated that copepods were the best live prey for first feeding ABT and that differences in growth performance and lipid metabolism observed between larvae from different year classes could be a consequence of broodstock nutrition. In addition, expression patterns of lipid metabolic genes observed in ABT larvae in the trials could reflect differences in lipid class and fatty acid compositions of the live prey. The lipid nutritional requirements, including essential fatty acid requirements of larval ABT during the early feeding stages, are unknown, and the present study represents a first step in addressing these highly relevant issues. However, further studies are required to determine nutritional requirements and understand lipid metabolism during development of ABT larvae and to apply the knowledge to the commercial culture of this iconic species.
Collapse
Affiliation(s)
- Mónica B Betancor
- Institute of Aquaculture, University of Stirling, Stirling, Scotland, FK9 4LA, UK.
| | - Aurelio Ortega
- Planta Experimental de Cultivos Marinos, Instituto Español de Oceanografía (IEO), 30860 Puerto de Mazarrón (Murcia), Madrid, Spain
| | - Fernando de la Gándara
- Planta Experimental de Cultivos Marinos, Instituto Español de Oceanografía (IEO), 30860 Puerto de Mazarrón (Murcia), Madrid, Spain
| | - Douglas R Tocher
- Institute of Aquaculture, University of Stirling, Stirling, Scotland, FK9 4LA, UK
| | - Gabriel Mourente
- Departamento de Biología, Facultad de Ciencias del Mar y Ambientales, Universidad de Cádiz, 11510 Puerto Real, Cádiz, Spain
| |
Collapse
|
40
|
Dossi CG, Cadagan C, San Martín M, Espinosa A, González-Mañán D, Silva D, Mancilla RA, Tapia GS. Effects of rosa mosqueta oil supplementation in lipogenic markers associated with prevention of liver steatosis. Food Funct 2017; 8:832-841. [PMID: 28128380 DOI: 10.1039/c6fo01762b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Rosa mosqueta (RM) oil is rich in α-linolenic acid (ALA) - a precursor of eicosapentaenoic (EPA) and docosahexaenoic acid (DHA), and it has a high antioxidant activity due to its abundant content of tocopherols. Additionally, it has been observed that RM oil administration prevents hepatic steatosis. Thus, the aim of this study was to demonstrate the antilipogenic mechanism related to RM oil administration in a high-fat diet (HFD) fed mice model by evaluating markers associated with the regulation of lipid droplet metabolism (PLIN2, PLIN5 and PPAR-γ), and proteins associated with lipogenesis (FAS and SREBP-1c). C57BL/6J mice were fed either a control diet or a HFD, with and without RM oil supplementation for 12 weeks. The results showed that RM oil supplementation decreases hepatic PLIN2 and PPAR-γ mRNA expression and SREBP-1c, FAS and PLIN2 protein levels, whereas we did not find changes in the level of PLIN5 among the groups. These results suggest that modulation of lipogenic markers could be one of the mechanisms, through which RM oil supplementation prevents the hepatic steatosis induced by HFD consumption in a mice model.
Collapse
Affiliation(s)
- Camila G Dossi
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Cynthia Cadagan
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Marcela San Martín
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Alejandra Espinosa
- Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Daniel González-Mañán
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - David Silva
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Rodrigo A Mancilla
- School of Biochemical Engineering, Faculty of Engineering, Pontifical Catholic University of Valparaiso, Valparaiso, Chile
| | - Gladys S Tapia
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
41
|
Jump DB, Botolin D, Wang Y, Xu J, Christian B. Fatty acids and gene transcription. SCANDINAVIAN JOURNAL OF FOOD & NUTRITION 2016. [DOI: 10.1080/17482970601069318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Donald B. Jump
- Departments of Physiology, Biochemistry and Molecular BiologyMichigan State UniversityMichiganUSA
| | - Daniela Botolin
- Departments of Physiology, Biochemistry and Molecular BiologyMichigan State UniversityMichiganUSA
| | - Yun Wang
- Departments of Physiology, Biochemistry and Molecular BiologyMichigan State UniversityMichiganUSA
| | - Jinghua Xu
- Departments of Physiology, Biochemistry and Molecular BiologyMichigan State UniversityMichiganUSA
| | - Barbara Christian
- Departments of Physiology, Biochemistry and Molecular BiologyMichigan State UniversityMichiganUSA
| |
Collapse
|
42
|
Morton AM, Furtado JD, Lee J, Amerine W, Davidson MH, Sacks FM. The effect of omega-3 carboxylic acids on apolipoprotein CIII−containing lipoproteins in severe hypertriglyceridemia. J Clin Lipidol 2016; 10:1442-1451.e4. [DOI: 10.1016/j.jacl.2016.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/02/2016] [Accepted: 09/05/2016] [Indexed: 10/21/2022]
|
43
|
Libby AE, Bales E, Orlicky DJ, McManaman JL. Perilipin-2 Deletion Impairs Hepatic Lipid Accumulation by Interfering with Sterol Regulatory Element-binding Protein (SREBP) Activation and Altering the Hepatic Lipidome. J Biol Chem 2016; 291:24231-24246. [PMID: 27679530 DOI: 10.1074/jbc.m116.759795] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Indexed: 12/16/2022] Open
Abstract
Perilipin-2 (PLIN2) is a constitutively associated cytoplasmic lipid droplet coat protein that has been implicated in fatty liver formation in non-alcoholic fatty liver disease. Mice with or without whole-body deletion of perilipin-2 (Plin2-null) were fed either Western or control diets for 30 weeks. Perilipin-2 deletion prevents obesity and insulin resistance in Western diet-fed mice and dramatically reduces hepatic triglyceride and cholesterol levels in mice fed Western or control diets. Gene and protein expression studies reveal that PLIN2 deletion suppressed SREBP-1 and SREBP-2 target genes involved in de novo lipogenesis and cholesterol biosynthetic pathways in livers of mice on either diet. GC-MS lipidomics demonstrate that this reduction correlated with profound alterations in the hepatic lipidome with significant reductions in both desaturation and elongation of hepatic neutral lipid species. To examine the possibility that lipidomic actions of PLIN2 deletion contribute to suppression of SREBP activation, we isolated endoplasmic reticulum membrane fractions from long-term Western diet-fed wild type (WT) and Plin2-null mice. Lipidomic analyses reveal that endoplasmic reticulum membranes from Plin2-null mice are markedly enriched in ω-3 and ω-6 long-chain polyunsaturated fatty acids, which others have shown inhibit SREBP activation and de novo lipogenesis. Our results identify PLIN2 as a determinant of global changes in the hepatic lipidome and suggest the hypothesis that these actions contribute to SREBP-regulated de novo lipogenesis involved in non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Andrew E Libby
- From the Integrated Physiology Graduate Program.,Division of Reproductive Sciences, and
| | | | - David J Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - James L McManaman
- From the Integrated Physiology Graduate Program, .,Division of Reproductive Sciences, and
| |
Collapse
|
44
|
Ladeira MM, Schoonmaker JP, Gionbelli MP, Dias JCO, Gionbelli TRS, Carvalho JRR, Teixeira PD. Nutrigenomics and Beef Quality: A Review about Lipogenesis. Int J Mol Sci 2016; 17:ijms17060918. [PMID: 27294923 PMCID: PMC4926451 DOI: 10.3390/ijms17060918] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/06/2016] [Accepted: 05/17/2016] [Indexed: 01/07/2023] Open
Abstract
The objective of the present review is to discuss the results of published studies that show how nutrition affects the expression of genes involved in lipid metabolism and how diet manipulation might change marbling and composition of fat in beef. Several key points in the synthesis of fat in cattle take place at the molecular level, and the association of nutritional factors with the modulation of this metabolism is one of the recent targets of nutrigenomic research. Within this context, special attention has been paid to the study of nuclear receptors associated with fatty acid metabolism. Among the transcription factors involved in lipid metabolism, the peroxisome proliferator-activated receptors (PPARs) and sterol regulatory element-binding proteins (SREBPs) stand out. The mRNA synthesis of these transcription factors is regulated by nutrients, and their metabolic action might be potentiated by diet components and change lipogenesis in muscle. Among the options for dietary manipulation with the objective to modulate lipogenesis, the use of different sources of polyunsaturated fatty acids, starch concentrations, forage ratios and vitamins stand out. Therefore, special care must be exercised in feedlot feed management, mainly when the goal is to produce high marbling beef.
Collapse
Affiliation(s)
- Marcio M Ladeira
- Department of Animal Science, Universidade Federal de Lavras, Lavras 37200-000, Brazil.
| | - Jon P Schoonmaker
- Department of Animal Science, Purdue University, West Lafayette, IN 47906, USA.
| | - Mateus P Gionbelli
- Department of Animal Science, Universidade Federal de Lavras, Lavras 37200-000, Brazil.
| | - Júlio C O Dias
- Department of Animal Science, Universidade Federal de Lavras, Lavras 37200-000, Brazil.
| | | | | | - Priscilla D Teixeira
- Department of Animal Science, Universidade Federal de Lavras, Lavras 37200-000, Brazil.
| |
Collapse
|
45
|
Yuan F, Wang H, Tian Y, Li Q, He L, Li N, Liu Z. Fish oil alleviated high-fat diet-induced non-alcoholic fatty liver disease via regulating hepatic lipids metabolism and metaflammation: a transcriptomic study. Lipids Health Dis 2016; 15:20. [PMID: 26832365 PMCID: PMC4736290 DOI: 10.1186/s12944-016-0190-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/26/2016] [Indexed: 02/06/2023] Open
Abstract
Background Intake of fish oil rich in n-3 polyunsaturated fatty acids (PUFAs) is believed to be beneficial against development of non-alcoholic fatty liver disease (NAFLD). However, the underlying mechanisms remain unclear. This study was to gain further understanding of the potential mechanisms of the protective effects of fish oil against NAFLD. Methods Ten male Sprague–Dawley rats were fed a control diet (CON), a Western style high-fat and high-cholesterol diet (WD), or a WD diet containing fish oil (FOH) for 16 weeks respectively. The development of liver steatosis and fibrosis were verified by histological and biochemical examination. Hepatic transcriptome were extracted for RNA-seq analysis, and particular results were confirmed by real-time polymerase chain reaction (PCR). Results The consumption of fish oil significantly ameliorated WD-induced dyslipidemia, transaminase elevation, hepatic steatosis, inflammatory infiltration, and fibrosis. Hepatic RNA-Seq analysis showed that long-term intake of fish oil restored the expression of circadian clock-related genes per2 and per3, which were reduced in WD fed animals. Fish oil consumption also corrected the expression levels of genes involved in fatty acid and cholesterol metabolism, such as Srebf1, Fasn, Scd1, Insig2, Cd36, Cyp7a1, Abcg5, Abcg8 and Pcsk9. Moreover, the expression levels of pro-inflammation genes Mcp1, Socs2, Sema4a, and Cd44 in the FOH group were lower than that of WD group, implying that fish oil protects the liver against WD-induced hepatic inflammation. Conclusion The present study demonstrates fish oil protects against WD-induced NALFD via improving lipid metabolism and ameliorating hepatic inflammation. Our findings add to the current understanding on the benefits of n-3 PUFAs against NAFLD. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0190-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fahu Yuan
- Wuhan Polytechnic University, School of Biology and Pharmaceutical Engineering, Wuhan, Hubei, 430023, China. .,Jianghan University, School of Medicine, Wuhan, China.
| | - Hualin Wang
- Wuhan Polytechnic University, School of Biology and Pharmaceutical Engineering, Wuhan, Hubei, 430023, China.
| | - Yu Tian
- Wuhan Polytechnic University, School of Biology and Pharmaceutical Engineering, Wuhan, Hubei, 430023, China.
| | - Qi Li
- Wuhan Polytechnic University, School of Biology and Pharmaceutical Engineering, Wuhan, Hubei, 430023, China.
| | - Lei He
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Na Li
- Wuhan Polytechnic University, School of Biology and Pharmaceutical Engineering, Wuhan, Hubei, 430023, China.
| | - Zhiguo Liu
- Wuhan Polytechnic University, School of Biology and Pharmaceutical Engineering, Wuhan, Hubei, 430023, China.
| |
Collapse
|
46
|
Jiménez MJ, Bocos C, Panadero M, Herrera E. Fish oil diet in pregnancy and lactation reduces pup weight and modifies newborn hepatic metabolic adaptations in rats. Eur J Nutr 2015; 56:409-420. [PMID: 26578529 DOI: 10.1007/s00394-015-1091-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/26/2015] [Indexed: 10/22/2022]
Abstract
PURPOSE To determine the effects of a diet containing fish oil (FD) during pregnancy and lactation in rats on the metabolic adaptations made by the offspring during early extrauterine life and to compare it to an olive oil diet (OD). METHODS Rats were mated and randomly allocated to OD or FD containing 10 % of the corresponding oil. During lactation, litters were adjusted to eight pups per dam. Fetuses of 20 days and pups of 0, 1, 10, 20 and 30 days of age were studied. RESULTS Body weight and length were lower in pups of the FD group from birth. The diet, milk, pups' plasma and liver of FD group had higher proportions of n-3 LCPUFA, but the content of arachidonic acid (ARA) was lower. Plasma glucose was higher, but unesterified fatty acids, triacylglycerols (TAG), 3-hydroxybutyrate and liver TAG in 1-day-old pups were lower in the FD group, and differences in some of these variables were also found in pups up to 30 days old. Liver lipoprotein lipase activity and mRNA expression, and the expression of carnitine palmitoyl transferase I, acyl-CoA oxidase and 3-hydroxy 3-methyl glutaryl-CoA synthase increased more at birth in pups of the FD group, but the expression of sterol regulatory element binding protein-1c and Δ6-desaturase mRNA was lower in the FD group. CONCLUSIONS Maternal intake of high n-3 LCPUFA retards postnatal development, which could be the result of impaired ARA synthesis, and affects hepatic metabolic adaptations to extrauterine life.
Collapse
Affiliation(s)
- Maria J Jiménez
- Departamento de Química y Bioquímica, Universidad San Pablo CEU, Ctra. Boadilla del Monte km 5.3, Boadilla del Monte, 28668, Madrid, Spain
| | - Carlos Bocos
- Departamento de Química y Bioquímica, Universidad San Pablo CEU, Ctra. Boadilla del Monte km 5.3, Boadilla del Monte, 28668, Madrid, Spain
| | - Maribel Panadero
- Departamento de Química y Bioquímica, Universidad San Pablo CEU, Ctra. Boadilla del Monte km 5.3, Boadilla del Monte, 28668, Madrid, Spain
| | - Emilio Herrera
- Departamento de Química y Bioquímica, Universidad San Pablo CEU, Ctra. Boadilla del Monte km 5.3, Boadilla del Monte, 28668, Madrid, Spain.
| |
Collapse
|
47
|
Deng X, Dong Q, Bridges D, Raghow R, Park EA, Elam MB. Docosahexaenoic acid inhibits proteolytic processing of sterol regulatory element-binding protein-1c (SREBP-1c) via activation of AMP-activated kinase. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1521-9. [PMID: 26327595 DOI: 10.1016/j.bbalip.2015.08.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 08/06/2015] [Accepted: 08/24/2015] [Indexed: 11/27/2022]
Abstract
In hyperinsulinemic states including obesity and T2DM, overproduction of fatty acid and triglyceride contributes to steatosis of the liver, hyperlipidemia and hepatic insulin resistance. This effect is mediated in part by the transcriptional regulator sterol responsive element binding protein-1c (SREBP-1c), which stimulates the expression of genes involved in hepatic fatty acid and triglyceride synthesis. SREBP-1c is up regulated by insulin both via increased transcription of nascent full-length SREBP-1c and by enhanced proteolytic processing of the endoplasmic reticulum (ER)-bound precursor to yield the transcriptionally active n-terminal form, nSREBP-1c. Polyunsaturated fatty acids of marine origin (n-3 PUFA) prevent induction of SREBP-1c by insulin thereby reducing plasma and hepatic triglycerides. Despite widespread use of n-3 PUFA supplements to reduce triglycerides in clinical practice, the exact mechanisms underlying their hypotriglyceridemic effect remain elusive. Here we demonstrate that the n-3 PUFA docosahexaenoic acid (DHA; 22:5 n-3) reduces nSREBP-1c by inhibiting regulated intramembrane proteolysis (RIP) of the nascent SREBP-1c. We further show that this effect of DHA is mediated both via activation of AMP-activated protein kinase (AMPK) and by inhibition of mechanistic target of rapamycin complex 1 (mTORC1). The inhibitory effect of AMPK on SREBP-1c processing is linked to phosphorylation of serine 365 of SREBP-1c in the rat. We have defined a novel regulatory mechanism by which n-3 PUFA inhibit induction of SREBP-1c by insulin. These findings identify AMPK as an important negative regulator of hepatic lipid synthesis and as a potential therapeutic target for hyperlipidemia in obesity and T2DM.
Collapse
Affiliation(s)
- Xiong Deng
- Department of Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis TN 38104, United States; Department of Pharmacology, 874 Union Avenue, Memphis, TN 38163, United States.
| | - Qingming Dong
- Department of Pharmacology, 874 Union Avenue, Memphis, TN 38163, United States
| | - Dave Bridges
- Department of Physiology, 894 Union Avenue, Memphis TN 38163, United States; Children's Foundation Research Institute, Le Bonheur Children's Hospital, Department of Pediatrics, University of Tennessee Health Science Center, 50 North Dunlap, Memphis TN 38103, United States
| | - Rajendra Raghow
- Department of Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis TN 38104, United States; Department of Pharmacology, 874 Union Avenue, Memphis, TN 38163, United States
| | - Edwards A Park
- Department of Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis TN 38104, United States; Department of Pharmacology, 874 Union Avenue, Memphis, TN 38163, United States
| | - Marshall B Elam
- Department of Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis TN 38104, United States; Department of Pharmacology, 874 Union Avenue, Memphis, TN 38163, United States
| |
Collapse
|
48
|
Regulation of SREBPs by Sphingomyelin in Adipocytes via a Caveolin and Ras-ERK-MAPK-CREB Signaling Pathway. PLoS One 2015; 10:e0133181. [PMID: 26230734 PMCID: PMC4521865 DOI: 10.1371/journal.pone.0133181] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 06/23/2015] [Indexed: 01/07/2023] Open
Abstract
Sterol response element binding protein (SREBP) is a key transcription factor in insulin and glucose metabolism. We previously demonstrated that elevated levels of membrane sphingomyelin (SM) were related to peroxisome proliferator–activated receptor-γ (PPARγ), which is a known target gene of SREBP-1 in adipocytes. However, the role of SM in SREBP expression in adipocytes remains unknown. In human abdominal adipose tissue from obese women with various concentrations of fasting plasma insulin, SREBP-1 proteins decreased in parallel with increases in membrane SM levels. An inverse correlation was found between the membrane SM content and the levels of SREBP-1c/ERK/Ras/PPARγ/CREB proteins. For the first time, we demonstrate the effects of SM and its signaling pathway in 3T3-F442A adipocytes. These cells were enriched or unenriched with SM in a range of concentrations similar to those observed in obese subjects by adding exogenous natural SMs (having different acyl chain lengths) or by inhibiting neutral sphingomyelinase. SM accumulated in caveolae of the plasma membrane within 24 h and then in the intracellular space. SM enrichment decreased SREBP-1 through the inhibition of extracellular signal-regulated protein kinase (ERK) but not JNK or p38 mitogen-activated protein kinase (MAPK). Ras/Raf-1/MEK1/2 and KSR proteins, which are upstream mediators of ERK, were down-regulated, whereas SREBP-2/caveolin and cholesterol were up-regulated. In SM-unmodulated adipocytes treated with DL-1-Phenyl-2-Palmitoylamino-3-morpholino-1-propanol (PPMP), where the ceramide level increased, the expression levels of SREBPs and ERK were modulated in an opposite direction relative to the SM-enriched cells. SM inhibited the insulin-induced expression of SREBP-1. Rosiglitazone, which is an anti-diabetic agent and potent activator of PPARγ, reversed the effects of SM on SREBP-1, PPARγ and CREB. Taken together, these findings provide novel insights indicating that excess membrane SM might be critical for regulating SREBPs in adipocytes via a MAPK-dependent pathway.
Collapse
|
49
|
Tsunoda F, Lamon-Fava S, Asztalos BF, Iyer LK, Richardson K, Schaefer EJ. Effects of oral eicosapentaenoic acid versus docosahexaenoic acid on human peripheral blood mononuclear cell gene expression. Atherosclerosis 2015; 241:400-8. [PMID: 26074314 DOI: 10.1016/j.atherosclerosis.2015.05.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/08/2015] [Accepted: 05/12/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have beneficial effects on inflammation and cardiovascular disease (CVD). Our aim was to assess the effect of a six-week supplementation with either olive oil, EPA, or DHA on gene expression in peripheral blood mononuclear cells (PBMC). METHODS Subjects were sampled at baseline and six weeks after receiving either: olive oil 6.0 g/day (n = 16), EPA 1.8 g/day (n = 16), or DHA 1.8 g/day (n = 18). PBMC were subjected to gene expression analysis by microarray with key findings confirmed by quantitative real-time polymerase chain reaction (Q-PCR). RESULTS Plasma phospholipid EPA increased 3 fold in the EPA group, and DHA increased 63% in the DHA group (both p < 0.01), while no effects were observed in the olive oil group. Microarray analysis indicated that EPA but not DHA or olive oil significantly affected the gene expression in the following pathways: 1) interferon signaling, 2) receptor recognition of bacteria and viruses, 3) G protein signaling, glycolysis and glycolytic shunting, 4) S-adenosyl-l-methionine biosynthesis, and 5) cAMP-mediated signaling including cAMP responsive element protein 1 (CREB1), as well as many other individual genes including hypoxia inducible factor 1, α subunit (HIF1A). The findings for CREB1 and HIF1A were confirmed by Q-PCR analysis. CONCLUSIONS Our data indicate that EPA supplementation was associated with significant effects on gene expression involving the interferon pathway as well as down-regulation of CREB1 and HIF1A, which may relate to its beneficial effect on CVD risk reduction.
Collapse
Affiliation(s)
- Fumiyoshi Tsunoda
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Bela F Asztalos
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Lakshmanan K Iyer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA; Center for Neuroscience Research, Tufts University School of Medicine, Boston, MA, USA
| | - Kris Richardson
- Nutrition and Genomics Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Ernst J Schaefer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA.
| |
Collapse
|
50
|
Shewale SV, Boudyguina E, Zhu X, Shen L, Hutchins PM, Barkley RM, Murphy RC, Parks JS. Botanical oils enriched in n-6 and n-3 FADS2 products are equally effective in preventing atherosclerosis and fatty liver. J Lipid Res 2015; 56:1191-205. [PMID: 25921305 DOI: 10.1194/jlr.m059170] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Indexed: 01/02/2023] Open
Abstract
Echium oil (EO), which is enriched in 18:4 n-3, the immediate product of fatty acid desaturase 2 (FADS2) desaturation of 18:3 n-3, is as atheroprotective as fish oil (FO). The objective of this study was to determine whether botanical oils enriched in the FADS2 products 18:3 n-6 versus 18:4 n-3 are equally atheroprotective. LDL receptor KO mice were fed one of four atherogenic diets containing 0.2% cholesterol and 10% calories as palm oil (PO) plus 10% calories as: 1) PO; 2) borage oil (BO; 18:3 n-6 enriched); 3) EO (18:4 n-3 enriched); or 4) FO for 16 weeks. Mice fed BO, EO, and FO versus PO had significantly lower plasma total and VLDL cholesterol concentrations; hepatic neutral lipid content and inflammation, aortic CE content, aortic root intimal area and macrophage content; and peritoneal macrophage inflammation, CE content, and ex vivo chemotaxis. Atheromas lacked oxidized CEs despite abundant generation of macrophage 12/15 lipooxygenase-derived metabolites. We conclude that botanical oils enriched in 18:3 n-6 and 18:4 n-3 PUFAs beyond the rate-limiting FADS2 enzyme are equally effective in preventing atherosclerosis and hepatosteatosis compared with saturated/monounsaturated fat due to cellular enrichment of ≥20 PUFAs, reduced plasma VLDL, and attenuated macrophage inflammation.
Collapse
Affiliation(s)
- Swapnil V Shewale
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 Physiology/Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Elena Boudyguina
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Xuewei Zhu
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Lulu Shen
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Patrick M Hutchins
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Robert M Barkley
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - John S Parks
- Departments of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|