1
|
Martínez-Beamonte R, Barranquero C, Gascón S, Mariño J, Arnal C, Estopañán G, Rodriguez-Yoldi MJ, Surra JC, Martín-Belloso O, Odriozola-Serrano I, Orman I, Segovia JC, Osada J, Navarro MÁ. Effect of virgin olive oil as spreadable preparation on atherosclerosis compared to dairy butter in Apoe-deficient mice. J Physiol Biochem 2024; 80:671-683. [PMID: 38787512 PMCID: PMC11502577 DOI: 10.1007/s13105-024-01029-8] [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: 07/07/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Olive oil is the main source of lipid energy in the Mediterranean diet and there is strong evidence of its health benefits. The effect of extra virgin olive oil (EVOO) in the form of a preparation of spreadable virgin olive oil (S-VO) on the progression of atheroma plaques was investigated in Apoe-deficient mice, a model of accelerated atherosclerosis. METHODS Two isocaloric Western purified diets containing 20% fat, either as S-VO or as dairy butter, were used to feed 28 males and 16 females of two-month-old Apoe-deficient mice for 12 weeks. S-VO was prepared by blending more than 75% virgin olive oil with other vegetal natural fat to obtain a solid fat. Plasma total cholesterol, triglycerides and HDL cholesterol were measured. Hepatic lipid droplets were analyzed. Areas of atherosclerotic aortic lesions were quantified in cross-sectional images of the proximal aorta and en face analysis of the whole aorta. RESULTS Total plasma cholesterol was increased in mice on the butter-supplemented diet in both female and male mice compared to S-VO, and the ratio of TC/HDL-cholesterol was significantly lower in S-VO than in the butter diet, although only in males, and no differences in plasma triglycerides were observed. No significant differences in hepatic lipid droplets were observed between diets in either sex. Aortic lesion areas were significantly higher in mice consuming the butter versus the S-VO diet in both sexes. CONCLUSION Extra virgin olive oil prepared in spreadable form maintained the delay in atheroma plaque progression compared to butter.
Collapse
Affiliation(s)
- Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013, Saragossa, Spain.
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, 50013, Saragossa, Spain.
- CIBER de Fisiopatología de La Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain.
- Oliberus, Campus Iberus, Zaragoza, Spain.
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013, Saragossa, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, 50013, Saragossa, Spain
- CIBER de Fisiopatología de La Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Sonia Gascón
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013, Saragossa, Spain
- Illes Balears, Instituto de Medicina Legal de Las Islas Baleares, E-07003, Palma, Spain
| | - Juan Mariño
- Las Arbequinas de Rosalía, Monesma de San Juan, 22415, Huesca, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, 50013, Saragossa, Spain
- CIBER de Fisiopatología de La Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Departamento de Patología Animal, Facultad de Veterinaria, Universidad de Zaragoza, 50013, Saragossa, Spain
- Oliberus, Campus Iberus, Zaragoza, Spain
| | - Gloria Estopañán
- Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Avda. Montañana 930, 50059, Saragossa, Spain
| | - María Jesús Rodriguez-Yoldi
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, 50013, Saragossa, Spain
- CIBER de Fisiopatología de La Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Departamento de Farmacología , Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013, Saragossa, Spain
- Oliberus, Campus Iberus, Zaragoza, Spain
| | - Joaquín Carlos Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, 50013, Saragossa, Spain
- CIBER de Fisiopatología de La Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Instituto de Investigación Sanitaria de Aragón, Escuela Politécnica Superior de Huesca, Universidad de Zaragoza, 50013, Saragossa, Spain
- Oliberus, Campus Iberus, Zaragoza, Spain
| | - Olga Martín-Belloso
- Department of Food Technology, Engineering and Science, University of Lleida, Av. Alcalde Rovira Roure, 191, 25198, Lleida, Spain
- Agrotecnio-CERCA Center, Av. Rovira Roure, 191, 25198, Lleida, Spain
- Alimentos Funcionales, Campus Iberus, Zaragoza, Spain
| | - Isabel Odriozola-Serrano
- Department of Food Technology, Engineering and Science, University of Lleida, Av. Alcalde Rovira Roure, 191, 25198, Lleida, Spain
- Agrotecnio-CERCA Center, Av. Rovira Roure, 191, 25198, Lleida, Spain
- Alimentos Funcionales, Campus Iberus, Zaragoza, Spain
| | - Israel Orman
- Cell Technology Division. Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CIEMAT/CIBERER, Madrid, Spain
- Advanced Cell Therapy Unit., Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | - Jose Carlos Segovia
- Cell Technology Division. Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CIEMAT/CIBERER, Madrid, Spain
- Advanced Cell Therapy Unit., Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013, Saragossa, Spain.
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, 50013, Saragossa, Spain.
- CIBER de Fisiopatología de La Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - María Ángeles Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013, Saragossa, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, 50013, Saragossa, Spain
- CIBER de Fisiopatología de La Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Oliberus, Campus Iberus, Zaragoza, Spain
| |
Collapse
|
2
|
Sánchez-Marco J, Bidooki SH, Abuobeid R, Barranquero C, Herrero-Continente T, Arnal C, Martínez-Beamonte R, Lasheras R, Surra JC, Navarro MA, Rodríguez-Yoldi MJ, Arruebo M, Sebastian V, Osada J. Thioredoxin domain containing 5 is involved in the hepatic storage of squalene into lipid droplets in a sex-specific way. J Nutr Biochem 2024; 124:109503. [PMID: 37898391 DOI: 10.1016/j.jnutbio.2023.109503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/05/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Hepatic thioredoxin domain-containing 5 (TXNDC5) is a member of the protein disulfide isomerase family found associated with anti-steatotic properties of squalene and located in the endoplasmic reticulum and in lipid droplets. Considering that the latter are involved in hepatic squalene accumulation, the present research was aimed to investigate the role of TXNDC5 on hepatic squalene management in mice and in the AML12 hepatic cell line. Wild-type and TXNDC5-deficient (KO) mice were fed Western diets with or without 1% squalene supplementation for 6 weeks. In males, but not in females, absence of TXNDC5 blocked hepatic, but not duodenal, squalene accumulation. Hepatic lipid droplets were isolated and characterized using label-free LC-MS/MS analysis. TXNDC5 accumulated in this subcellular compartment of mice receiving squalene and was absent in TXNDC5-KO male mice. The latter mice were unable to store squalene in lipid droplets. CALR and APMAP were some of the proteins that responded to the squalene administration in all studied conditions. CALR and APMAP were positively associated with lipid droplets in the presence of squalene and they were decreased by the absence of TXNDC5. The increased squalene content was reproduced in vitro using AML12 cells incubated with squalene-loaded nanoparticles and this effect was not observed in an engineered cell line lacking TXNDC5. The phenomenon was also present when incubated in the presence of a squalene epoxidase inhibitor, suggesting a mechanism of squalene exocytosis involving CALR and APMAP. In conclusion, squalene accumulation in hepatic lipid droplets is sex-dependent on TXNDC5 that blocks its secretion.
Collapse
Affiliation(s)
- Javier Sánchez-Marco
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Seyed Hesamoddin Bidooki
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Roubi Abuobeid
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Tania Herrero-Continente
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Lasheras
- Laboratorio Agroambiental, Servicio de Seguridad Agroalimentaria de la Dirección General de Alimentación y Fomento Agroalimentario, Gobierno de Aragón, Zaragoza, Spain
| | - Joaquín C Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Huesca, Spain
| | - María A Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - María J Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Manuel Arruebo
- Departamento de Ingeniería Química y Tecnologías del Medio Ambiente, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Victor Sebastian
- Departamento de Ingeniería Química y Tecnologías del Medio Ambiente, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Abuobeid R, Herrera-Marcos LV, Arnal C, Bidooki SH, Sánchez-Marco J, Lasheras R, Surra JC, Rodríguez-Yoldi MJ, Martínez-Beamonte R, Osada J. Differentially Expressed Genes in Response to a Squalene-Supplemented Diet Are Accurate Discriminants of Porcine Non-Alcoholic Steatohepatitis. Int J Mol Sci 2023; 24:12552. [PMID: 37628732 PMCID: PMC10454218 DOI: 10.3390/ijms241612552] [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: 06/19/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Squalene is the major unsaponifiable component of virgin olive oil, the fat source of the Mediterranean diet. To evaluate its effect on the hepatic transcriptome, RNA sequencing was carried out in two groups of male Large White x Landrace pigs developing nonalcoholic steatohepatitis by feeding them a high fat/cholesterol/fructose and methionine and choline-deficient steatotic diet or the same diet with 0.5% squalene. Hepatic lipids, squalene content, steatosis, activity (ballooning + inflammation), and SAF (steatosis + activity + fibrosis) scores were analyzed. Pigs receiving the latter diet showed hepatic squalene accumulation and twelve significantly differentially expressed hepatic genes (log2 fold change < 1.5 or <1.5) correlating in a gene network. These pigs also had lower hepatic triglycerides and lipid droplet areas and higher cellular ballooning. Glutamyl aminopeptidase (ENPEP) was correlated with triglyceride content, while alpha-fetoprotein (AFP), neutralized E3 ubiquitin protein ligase 3 (NEURL3), 2'-5'-oligoadenylate synthase-like protein (OASL), and protein phosphatase 1 regulatory inhibitor subunit 1B (PPP1R1B) were correlated with activity reflecting inflammation and ballooning, and NEURL3 with the SAF score. AFP, ENPEP, and PPP1R1B exhibited a remarkably strong discriminant power compared to those pathological parameters in both experimental groups. Moreover, the expression of PPP1R1B, TMEM45B, AFP, and ENPEP followed the same pattern in vitro using human hepatoma (HEPG2) and mouse liver 12 (AML12) cell lines incubated with squalene, indicating a direct effect of squalene on these expressions. These findings suggest that squalene accumulated in the liver is able to modulate gene expression changes that may influence the progression of non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Roubi Abuobeid
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
| | - Luis V. Herrera-Marcos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Seyed Hesamoddin Bidooki
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
| | - Javier Sánchez-Marco
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
| | - Roberto Lasheras
- Laboratorio Agroambiental, Servicio de Seguridad Agroalimentaria de la Dirección General de Alimentación y Fomento Agroalimentario, Gobierno de Aragón, E-50071 Zaragoza, Spain
| | - Joaquín C. Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-22071 Huesca, Spain
| | - María Jesús Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
4
|
Antonopoulou S, Demopoulos CA. Protective Effect of Olive Oil Microconstituents in Atherosclerosis: Emphasis on PAF Implicated Atherosclerosis Theory. Biomolecules 2023; 13:700. [PMID: 37189447 PMCID: PMC10135796 DOI: 10.3390/biom13040700] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Atherosclerosis is a progressive vascular multifactorial process. The mechanisms underlining the initiating event of atheromatous plaque formation are inflammation and oxidation. Among the modifiable risk factors for cardiovascular diseases, diet and especially the Mediterranean diet (MedDiet), has been widely recognized as one of the healthiest dietary patterns. Olive oil (OO), the main source of the fatty components of the MedDiet is superior to the other "Mono-unsaturated fatty acids containing oils" due to the existence of specific microconstituents. In this review, the effects of OO microconstituents in atherosclerosis, based on data from in vitro and in vivo studies with special attention on their inhibitory activity against PAF (Platelet-Activating Factor) actions, are presented and critically discussed. In conclusion, we propose that the anti-atherogenic effect of OO is attributed to the synergistic action of its microconstituents, mainly polar lipids that act as PAF inhibitors, specific polyphenols and α-tocopherol that also exert anti-PAF activity. This beneficial effect, also mediated through anti-PAF action, can occur from microconstituents extracted from olive pomace, a toxic by-product of the OO production process that constitutes a significant ecological problem. Daily intake of moderate amounts of OO consumed in the context of a balanced diet is significant for healthy adults.
Collapse
Affiliation(s)
- Smaragdi Antonopoulou
- Laboratory of Biology, Biochemistry and Microbiology, Department of Nutrition-Dietetics, School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece
| | - Constantinos A. Demopoulos
- Laboratory of Biochemistry, Faculty of Chemistry, National & Kapodistrian University of Athens, 15784 Athens, Greece;
| |
Collapse
|
5
|
Senatus L, Egaña-Gorroño L, López-Díez R, Bergaya S, Aranda JF, Amengual J, Arivazhagan L, Manigrasso MB, Yepuri G, Nimma R, Mangar KN, Bernadin R, Zhou B, Gugger PF, Li H, Friedman RA, Theise ND, Shekhtman A, Fisher EA, Ramasamy R, Schmidt AM. DIAPH1 mediates progression of atherosclerosis and regulates hepatic lipid metabolism in mice. Commun Biol 2023; 6:280. [PMID: 36932214 PMCID: PMC10023694 DOI: 10.1038/s42003-023-04643-2] [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: 01/14/2022] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
Atherosclerosis evolves through dysregulated lipid metabolism interwoven with exaggerated inflammation. Previous work implicating the receptor for advanced glycation end products (RAGE) in atherosclerosis prompted us to explore if Diaphanous 1 (DIAPH1), which binds to the RAGE cytoplasmic domain and is important for RAGE signaling, contributes to these processes. We intercrossed atherosclerosis-prone Ldlr-/- mice with mice devoid of Diaph1 and fed them Western diet for 16 weeks. Compared to male Ldlr-/- mice, male Ldlr-/- Diaph1-/- mice displayed significantly less atherosclerosis, in parallel with lower plasma concentrations of cholesterol and triglycerides. Female Ldlr-/- Diaph1-/- mice displayed significantly less atherosclerosis compared to Ldlr-/- mice and demonstrated lower plasma concentrations of cholesterol, but not plasma triglycerides. Deletion of Diaph1 attenuated expression of genes regulating hepatic lipid metabolism, Acaca, Acacb, Gpat2, Lpin1, Lpin2 and Fasn, without effect on mRNA expression of upstream transcription factors Srebf1, Srebf2 or Mxlipl in male mice. We traced DIAPH1-dependent mechanisms to nuclear translocation of SREBP1 in a manner independent of carbohydrate- or insulin-regulated cues but, at least in part, through the actin cytoskeleton. This work unveils new regulators of atherosclerosis and lipid metabolism through DIAPH1.
Collapse
Affiliation(s)
- Laura Senatus
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Lander Egaña-Gorroño
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Raquel López-Díez
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Sonia Bergaya
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Juan Francisco Aranda
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Jaume Amengual
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Michaele B Manigrasso
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Gautham Yepuri
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Ramesh Nimma
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Kaamashri N Mangar
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Rollanda Bernadin
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Boyan Zhou
- Department of Population Health, Division of Biostatistics, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Paul F Gugger
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Huilin Li
- Department of Population Health, Division of Biostatistics, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center and Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Neil D Theise
- Department of Pathology, NYU Grossman School of Medicine, NYU Langone Health, New York, USA
| | - Alexander Shekhtman
- Department of Chemistry, The State University of New York at Albany, Albany, NY, USA
| | - Edward A Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
6
|
Herrera-Marcos LV, Martínez-Beamonte R, Arnal C, Barranquero C, Puente-Lanzarote JJ, Herrero-Continente T, Lou-Bonafonte JM, Gonzalo-Romeo G, Mocciaro G, Jenkins B, Surra JC, Rodríguez-Yoldi MJ, Burillo JC, Lasheras R, García-Gil A, Güemes A, Koulman A, Osada J. Dietary squalene supplementation decreases triglyceride species and modifies phospholipid lipidomic profile in the liver of a porcine model of non-alcoholic steatohepatitis. J Nutr Biochem 2023; 112:109207. [PMID: 36402249 DOI: 10.1016/j.jnutbio.2022.109207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/07/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022]
Abstract
Squalene is a key minor component of virgin olive oil, the main source of fat in the Mediterranean diet, and had shown to improve the liver metabolism in rabbits and mice. The present research was carried out to find out whether this effect was conserved in a porcine model of hepatic steatohepatitis and to search for the lipidomic changes involved. The current study revealed that a 0.5% squalene supplementation to a steatotic diet for a month led to hepatic accumulation of squalene and decreased triglyceride content as well as area of hepatic lipid droplets without influencing cholesterol content or fiber areas. However, ballooning score was increased and associated with the hepatic squalene content. Of forty hepatic transcripts related to lipid metabolism and hepatic steatosis, only citrate synthase and a non-coding RNA showed decreased expressions. The hepatic lipidome, assessed by liquid chromatography-mass spectrometry in a platform able to analyze 467 lipids, revealed that squalene supplementation increased ceramide, Cer(36:2), and phosphatidylcholine (PC[32:0], PC[33:0] and PC[34:0]) species and decreased cardiolipin, CL(69:5), and triglyceride (TG[54:2], TG[55:0] and TG[55:2]) species. Plasma levels of interleukin 12p40 increased in pigs receiving the squalene diet. The latter also modified plasma lipidome by increasing TG(58:12) and decreasing non-esterified fatty acid (FA 14:0, FA 16:1 and FA 18:0) species without changes in total NEFA levels. Together this shows that squalene-induced changes in hepatic and plasma lipidomic profiles, non-coding RNA and anti-inflammatory interleukin are suggestive of an alleviation of the disease despite the increase in the ballooning score.
Collapse
Affiliation(s)
- Luis V Herrera-Marcos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Juan J Puente-Lanzarote
- Servicio de Bioquímica Clínica. Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Tania Herrero-Continente
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - José M Lou-Bonafonte
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Gonzalo Gonzalo-Romeo
- Servicio General de Apoyo a la Investigación. División de Experimentación Animal, Universidad de Zaragoza, Zaragoza, Spain
| | - Gabriele Mocciaro
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Benjamin Jenkins
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Joaquín C Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Huesca, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - María J Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Juan Carlos Burillo
- Laboratorio Agroambiental, Servicio de Seguridad Agroalimentaria de la Dirección General de Alimentación y Fomento Agroalimentario, Zaragoza, Spain
| | - Roberto Lasheras
- Laboratorio Agroambiental, Servicio de Seguridad Agroalimentaria de la Dirección General de Alimentación y Fomento Agroalimentario, Zaragoza, Spain
| | - Agustín García-Gil
- Departamento de Cirugía, Facultad de Medicina, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Antonio Güemes
- Departamento de Cirugía, Facultad de Medicina, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Albert Koulman
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain.
| |
Collapse
|
7
|
Martínez-Beamonte R, Ripalda M, Herrero-Continente T, Barranquero C, Dávalos A, López de las Hazas MC, Álvarez-Lanzarote I, Sánchez-Gimeno AC, Raso J, Arnal C, Surra JC, Osada J, Navarro MA. Pulsed electric field increases the extraction yield of extra virgin olive oil without loss of its biological properties. Front Nutr 2022; 9:1065543. [PMID: 36483924 PMCID: PMC9722962 DOI: 10.3389/fnut.2022.1065543] [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: 10/09/2022] [Accepted: 11/01/2022] [Indexed: 08/30/2023] Open
Abstract
INTRODUCTION Pulsed electric field (PEF) has been used for improving extraction of extra virgin olive oil (EVOO). However, the biological changes induced by the consumption of pulsed electric field-obtained extra virgin olive oil (PEFEVOO) have not been studied yet. MATERIALS AND METHODS EVOO oils from Empeltre variety were prepared by standard (STD) cold pressure method involving crushing of the olives, malaxation and decanting and by this procedure including an additional step of PEF treatment. Chemical analyses of EVOO oils were done. Male and female Apoe-deficient mice received diets differing in both EVOOs for 12 weeks, and their plasma, aortas and livers were analyzed. RESULTS PEF application resulted in a 17% increase in the oil yield and minimal changes in chemical composition regarding phytosterols, phenolic compounds and microRNA. Only in females mice consuming PEF EVOO, a decreased plasma total cholesterol was observed, without significant changes in atherosclerosis and liver steatosis. CONCLUSION PEF technology applied to EVOO extraction maintains the EVOO quality and improves the oil yield. The equivalent biological effects in atherosclerosis and fatty liver disease of PEF-obtained EVOO further support its safe use as a food.
Collapse
Affiliation(s)
- Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Marina Ripalda
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Tania Herrero-Continente
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Dávalos
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM + CSIC, Madrid, Spain
| | - María Carmen López de las Hazas
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, CEI UAM + CSIC, Madrid, Spain
| | - Ignacio Álvarez-Lanzarote
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Ana Cristina Sánchez-Gimeno
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Javier Raso
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Joaquín C. Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María A. Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Vesnina A, Prosekov A, Atuchin V, Minina V, Ponasenko A. Tackling Atherosclerosis via Selected Nutrition. Int J Mol Sci 2022; 23:8233. [PMID: 35897799 PMCID: PMC9368664 DOI: 10.3390/ijms23158233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/02/2022] Open
Abstract
The development and pathogenesis of atherosclerosis are significantly influenced by lifestyle, particularly nutrition. The modern level of science and technology development promote personalized nutrition as an efficient preventive measure against atherosclerosis. In this survey, the factors were revealed that contribute to the formation of an individual approach to nutrition: genetic characteristics, the state of the microbiota of the gastrointestinal tract (GIT) and environmental factors (diets, bioactive components, cardioprotectors, etc.). In the course of the work, it was found that in order to analyze the predisposition to atherosclerosis associated with nutrition, genetic features affecting the metabolism of nutrients are significant. The genetic features include the presence of single nucleotide polymorphisms (SNP) of genes and epigenetic factors. The influence of telomere length on the pathogenesis of atherosclerosis and circadian rhythms was also considered. Relatively new is the study of the relationship between chrono-nutrition and the development of metabolic diseases. That is, to obtain the relationship between nutrition and atherosclerosis, a large number of genetic markers should be considered. In this relation, the question arises: "How many genetic features need to be analyzed in order to form a personalized diet for the consumer?" Basically, companies engaged in nutrigenetic research and choosing a diet for the prevention of a number of metabolic diseases use SNP analysis of genes that accounts for lipid metabolism, vitamins, the body's antioxidant defense system, taste characteristics, etc. There is no set number of genetic markers. The main diets effective against the development of atherosclerosis were considered, and the most popular were the ketogenic, Mediterranean, and DASH-diets. The advantage of these diets is the content of foods with a low amount of carbohydrates, a high amount of vegetables, fruits and berries, as well as foods rich in antioxidants. However, due to the restrictions associated with climatic, geographical, material features, these diets are not available for a number of consumers. The way out is the use of functional products, dietary supplements. In this approach, the promising biologically active substances (BAS) that exhibit anti-atherosclerotic potential are: baicalin, resveratrol, curcumin, quercetin and other plant metabolites. Among the substances, those of animal origin are popular: squalene, coenzyme Q10, omega-3. For the prevention of atherosclerosis through personalized nutrition, it is necessary to analyze the genetic characteristics (SNP) associated with the metabolism of nutrients, to assess the state of the microbiota of the GIT. Based on the data obtained and food preferences, as well as the individual capabilities of the consumer, the optimal diet can be selected. It is topical to exclude nutrients of which their excess consumption stimulates the occurrence and pathogenesis of atherosclerosis and to enrich the diet with functional foods (FF), BAS containing the necessary anti-atherosclerotic, and stimulating microbiota of the GIT nutrients. Personalized nutrition is a topical preventive measure and there are a number of problems hindering the active use of this approach among consumers. The key factors include weak evidence of the influence of a number of genetic features, the high cost of the approach, and difficulties in the interpretation of the results. Eliminating these deficiencies will contribute to the maintenance of a healthy state of the population through nutrition.
Collapse
Affiliation(s)
- Anna Vesnina
- Laboratory of Natural Nutraceuticals Biotesting, Research Department, Kemerovo State University, 650043 Kemerovo, Russia;
| | - Alexander Prosekov
- Laboratory of Biocatalysis, Kemerovo State University, 650043 Kemerovo, Russia;
| | - Victor Atuchin
- Laboratory of Optical Materials and Structures, Institute of Semiconductor Physics, 630090 Novosibirsk, Russia
- Research and Development Department, Kemerovo State University, 650000 Kemerovo, Russia
- Laboratory of Applied Physics, Novosibirsk State University, 630090 Novosibirsk, Russia
- Department of Industrial Machinery Design, Novosibirsk State Technical University, 630073 Novosibirsk, Russia
- R&D Center “Advanced Electronic Technologies”, Tomsk State University, 634034 Tomsk, Russia
| | - Varvara Minina
- Department of Genetic and Fundamental Medicine, Kemerovo State University, 650000 Kemerovo, Russia;
| | - Anastasia Ponasenko
- Laboratory of Genome Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 650002 Kemerovo, Russia;
| |
Collapse
|
9
|
Thioredoxin Domain Containing 5 Suppression Elicits Serum Amyloid A-Containing High-Density Lipoproteins. Biomedicines 2022; 10:biomedicines10030709. [PMID: 35327511 PMCID: PMC8945230 DOI: 10.3390/biomedicines10030709] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/29/2022] Open
Abstract
Thioredoxin domain containing 5 (TXNDC5) is a protein disulfide isomerase involved in several diseases related to oxidative stress, energy metabolism and cellular inflammation. In a previous manuscript, a negative association between fatty liver development and hepatic Txndc5 expression was observed. To study the role of TXNDC5 in the liver, we generated Txndc5-deficient mice. The absence of the protein caused an increased metabolic need to gain weight along with a bigger and fatter liver. RNAseq was performed to elucidate the putative mechanisms, showing a substantial liver overexpression of serum amyloid genes (Saa1, Saa2) with no changes in hepatic protein, but discrete plasma augmentation by the gene inactivation. Higher levels of malonyldialdehyde, apolipoprotein A1 and platelet activating factor-aryl esterase activity were also found in serum from Txndc5-deficient mice. However, no difference in the distribution of high-density lipoproteins (HDL)-mayor components and SAA was found between groups, and even the reactive oxygen species decreased in HDL coming from Txndc5-deficient mice. These results confirm the relation of this gene with hepatic steatosis and with a fasting metabolic derive remedying an acute phase response. Likewise, they pose a new role in modulating the nature of HDL particles, and SAA-containing HDL particles are not particularly oxidized.
Collapse
|
10
|
Ebrahimi M, Farhadian N, Amiri AR, Hataminia F, Soflaei SS, Karimi M. Evaluating the efficacy of extracted squalene from seed oil in the form of microemulsion for the treatment of COVID-19: A clinical study. J Med Virol 2021; 94:119-130. [PMID: 34403141 PMCID: PMC8427120 DOI: 10.1002/jmv.27273] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/11/2021] [Indexed: 12/31/2022]
Abstract
This study investigates the effect of the nanostructure of squalene in the form of microemulsion on COVID‐19 patients. In this blinded clinical trial, a comparison was made between the efficacy of squalene treatment and controls. A total of 30 COVID‐19 patients admitted to the emergency department, and the infection ward was equally allocated to case (n = 15) and control (n = 15) groups according to their age and underlying diseases. The baseline characteristics of subjects, including age, gender, time of treatment onset, underlying condition, white blood cells count, and lymphocyte count were similar (p < 0.05). Baseline laboratory tests and computed tomography (CT) scans were performed for the study groups. The treatment group received 5 mg of intravenous squalene twice a day and standard treatment for 6 days, while controls received only standard treatment. After 6 days of treatment, clinical and CT scan changes were evaluated and compared in intervention and control groups. The need for oxygen therapy (p = 0.020), 2 days of no fever (p = 0.025), cough alleviation (p = 0.010), and lung high‐resolution computed tomography improvement (p = 0.033) were significantly different between cases and controls within 7 days of admission. No adverse effects were observed in the treatment group. Our data suggest that squalene could be considered as a potential treatment for COVID‐19, and further studies are required to confirm the results.
Collapse
Affiliation(s)
- Mahmoud Ebrahimi
- Cardiovascular Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nafiseh Farhadian
- Chemical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ali Reza Amiri
- Emergency Medicine Department, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh Hataminia
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Saffar Soflaei
- Department of Modern Sciences & Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Karimi
- Emergency Medicine Department, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
11
|
Martínez-Beamonte R, Sánchez-Marco J, Felices MJ, Barranquero C, Gascón S, Arnal C, Burillo JC, Lasheras R, Busto R, Lasunción MA, Rodríguez-Yoldi MJ, Osada J. Dietary squalene modifies plasma lipoproteins and hepatic cholesterol metabolism in rabbits. Food Funct 2021; 12:8141-8153. [PMID: 34291245 DOI: 10.1039/d0fo01836h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To evaluate the effects of squalene, the main unsaponifiable component of virgin olive oil, on lipid metabolism, two groups of male New Zealand rabbits were fed a 1% sunflower oil-enriched regular diet or the same diet containing 0.5% squalene for 4 weeks. Plasma triglycerides, total- and HDL-cholesterol and their lipoproteins were assayed. Analyses of hepatic lipid droplets, triglycerides, total- and non-esterified cholesterol, squalene, protein and gene expression, and cholesterol precursors were carried out. In the jejunum, the squalene content and mRNA and protein APOB expressions were measured. Finally, we studied the effect of cholesterol precursors in AML12 cells. Squalene administration significantly increased plasma total cholesterol, mainly carried as non-esterified cholesterol in IDL and large LDL, and corresponded to an increased number of APOB100-containing particles without accumulation of triglycerides and decreased reactive oxygen species. Despite no significant changes in the APOB content in the jejunum, the latter displayed increased APOB mRNA and squalene levels. Increases in the amounts of non-esterified cholesterol, squalene, lanosterol, dihydrolanosterol, lathosterol, cholestanol, zymostenol, desmosterol and caspase 1 were also observed in the liver. Incubation of AML12 cells in the presence of lanosterol increased caspase 1. In conclusion, squalene administration in rabbits increases the number of modified APOB-containing lipoproteins, and hepatic cholesterol biosynthesis is linked to caspase 1 probably through lanosterol.
Collapse
Affiliation(s)
- Roberto Martínez-Beamonte
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Tutunchi H, Naeini F, Ebrahimi-Mameghani M, Mobasseri M, Naghshi S, Ostadrahimi A. The association of the steatosis severity, NAFLD fibrosis score and FIB-4 index with atherogenic dyslipidaemia in adult patients with NAFLD: A cross-sectional study. Int J Clin Pract 2021; 75:e14131. [PMID: 33683797 DOI: 10.1111/ijcp.14131] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Obesity and dyslipidaemia are the major risk factors for non-alcoholic fatty liver disease (NAFLD), and are known to increase cardiovascular disease (CVD), which is the leading cause of death in NAFLD patients. The present cross-sectional study aimed to investigate associations among severity of hepatic steatosis, NAFLD fibrosis score and atherogenic lipid profile. METHODS A total of 265 patients with NAFLD confirmed by ultrasonographic findings were included. The NAFLD fibrosis score and the fibrosis-4 (FIB-4) index were used to classify the probability of fibrosis as low, intermediate and high probability. Serum lipids including total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) were measured, and then TC/HDL-C, LDL-C/HDL-C, TG/HDL-C and non HDL-C/HDL-C ratios were determined. Fasting blood sugar (FBS), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were also assessed. The homeostatic model assessment for insulin resistance (HOMA-IR) was calculated. RESULTS The severity of hepatic steatosis was positively correlated with TC/HDL-C (r = 0.29, P = .002), LDL-C/HDL-C (r = 0.32, P < .001), TG/HDL-C (r = 0.36, P < .001) and non-HDL-C/HDL-C (r = 0.24, P = .001) ratios. Similarly, these parameters were positively correlated with NAFLD fibrosis score and FIB-4 index (P < .05). In addition, alanine aminotransferase and aspartate aminotransferase levels were positively correlated with TG/HDL-C ratio (r = 0.31, P = .003; and r = 0.27, P = .001 respectively). With increasing the severity of hepatic steatosis and NAFLD fibrosis score, the mean of all lipid ratios increased significantly (P < .01 and P < .05, respectively). Importantly, after controlling for potential confounders including age, gender, physical activity level, body mass index, waist circumference and HOMA-IR, the severity of steatosis, NAFLD fibrosis score and FIB-4 index remained independent predictors of atherogenic lipid profile. CONCLUSIONS Severity of hepatic steatosis, NAFLD fibrosis score and FIB-4 index were significantly correlated with atherogenic lipid profile. As NAFLD is high among patients with metabolic risk factors for CVD, their dyslipidaemia should be aggressively managed.
Collapse
Affiliation(s)
- Helda Tutunchi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran university of medical science, Tehran, Iran
| | - Mehrangiz Ebrahimi-Mameghani
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Mobasseri
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Naghshi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran university of medical science, Tehran, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Martínez-Beamonte R, Sánchez-Marco J, Lázaro G, Barco M, Herrero-Continente T, Serrano-Megías M, Botaya D, Arnal C, Barranquero C, Surra JC, Osada J, Navarro MA. Dietary Avian Proteins Are Comparable to Soybean Proteins on the Atherosclerosis Development and Fatty Liver Disease in Apoe-Deficient Mice. Nutrients 2021; 13:nu13061838. [PMID: 34072167 PMCID: PMC8227708 DOI: 10.3390/nu13061838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/05/2022] Open
Abstract
Background and aim: The type and amount of dietary protein has become a topic of renewed interest in light of their involvement in metabolic diseases, atherosclerosis and thrombosis. However, little attention has been devoted to the effect of avian proteins despite their wide human consumption. The aim was to investigate the influence of chicken and turkey as sources of protein compared with that of soybean on atherosclerosis and fatty liver disease. Methods and results: To this purpose, male and female Apoe-deficient were fed purified Western diets differing in their protein sources for 12 weeks. After this period, blood, liver, aortic tree and heart base samples were taken for analyses of plasma lipids and atherosclerosis. Plasma triglycerides, non-esterified fatty acids, esterified cholesterol levels and radical oxygen species in lipoproteins changed depending on the diet and sex. Females consuming the turkey protein-containing diet showed decreased atherosclerotic foci, as evidenced by the en face atherosclerosis analyses. The presence of macrophages and smooth muscle cells in plaques were not modified, and no changes were observed in hepatic lipid droplets in the studied groups either. Paraoxonase activity was higher in the group consuming turkey protein without sex differences, but only in females, it was significantly associated with aortic lesion areas. Conclusions: Compared to soybean protein, the consumption of avian proteins depending on sex resulted in similar or lower atherosclerosis development and comparable hepatic steatosis.
Collapse
Affiliation(s)
- Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Javier Sánchez-Marco
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
| | - Gonzalo Lázaro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
| | - María Barco
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
| | - Tania Herrero-Continente
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
| | | | - David Botaya
- Aves Nobles y Derivados-Aldelis, E-50197 Zaragoza, Spain; (M.S.-M.); (D.B.)
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Joaquín C. Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-22071 Huesca, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Department of Biochemistry and Molecular Biology, Veterinary School, University of Zaragoza, Miguel Servet, 177, E-50013 Zaragoza, Spain
- Correspondence: ; Tel.: +34-976-761644
| | - María A. Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
14
|
Ibrahim N‘I, Naina Mohamed I. Interdependence of Anti-Inflammatory and Antioxidant Properties of Squalene-Implication for Cardiovascular Health. Life (Basel) 2021; 11:103. [PMID: 33573041 PMCID: PMC7911491 DOI: 10.3390/life11020103] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases (CVD) have been recognized as the leading cause of mortality worldwide, accounting for 31% of global mortality. Among the risk factors of CVD, hyperlipidemia has been established as the most potent risk factor. Statins, a class of drug that reduces lower-density lipoprotein cholesterol (LDL-C), are the preferred medical treatment. However, due to the development of statin-associated muscle symptoms, statins are associated with patients' discontinuation and nonadherence. Other statin-induced side effects, such as hepatotoxicity and gastrointestinal upset, all contribute to patients choosing alternative medicines. Squalene (SQ), an unsaturated hydrocarbon naturally synthesized in plants and animals, could become the alternative treatment or supplementary agent for cardiovascular health. SQ has been shown to exert cardioprotective effect via its antioxidant activity. Oxidative stress and inflammatory responses are closely related to each other, which proposes an interdependence relation between antioxidant and anti-inflammatory. Therefore, this review explores the interdependence between the antioxidant and anti-inflammatory effects of SQ implicated on cardiovascular health.
Collapse
Affiliation(s)
| | - Isa Naina Mohamed
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
15
|
Dietary Erythrodiol Modifies Hepatic Transcriptome in Mice in a Sex and Dose-Dependent Way. Int J Mol Sci 2020; 21:ijms21197331. [PMID: 33020388 PMCID: PMC7582860 DOI: 10.3390/ijms21197331] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 01/15/2023] Open
Abstract
Erythrodiol is a terpenic compound found in a large number of plants. To test the hypotheses that its long-term administration may influence hepatic transcriptome and this could be influenced by the presence of APOA1-containing high-density lipoproteins (HDL), Western diets containing 0.01% of erythrodiol (10 mg/kg dose) were provided to Apoe- and Apoa1-deficient mice. Hepatic RNA-sequencing was carried out in male Apoe-deficient mice fed purified Western diets differing in the erythrodiol content. The administration of this compound significantly up- regulated 68 and down-regulated 124 genes at the level of 2-fold change. These genes belonged to detoxification processes, protein metabolism and nucleic acid related metabolites. Gene expression changes of 21 selected transcripts were verified by RT-qPCR. Ccl19-ps2, Cyp2b10, Rbm14-rbm4, Sec61g, Tmem81, Prtn3, Amy2a5, Cyp2b9 and Mup1 showed significant changes by erythrodiol administration. When Cyp2b10, Dmbt1, Cyp2b13, Prtn3 and Cyp2b9 were analyzed in female Apoe-deficient mice, no change was observed. Likewise, no significant variation was observed in Apoa1- or in Apoe-deficient mice receiving doses ranging from 0.5 to 5 mg/kg erythrodiol. Our results give evidence that erythrodiol exerts a hepatic transcriptional role, but this is selective in terms of sex and requires a threshold dose. Furthermore, it requires an APOA1-containing HDL.
Collapse
|
16
|
Gabás‐Rivera C, Jurado‐Ruiz E, Sánchez‐Ortiz A, Romanos E, Martínez‐Beamonte R, Navarro MA, Surra JC, Arnal C, Rodríguez‐Yoldi MJ, Andrés‐Lacueva C, Osada J. Dietary Squalene Induces CytochromesCyp2b10andCyp2c55Independently of Sex, Dose, and Diet in Several Mouse Models. Mol Nutr Food Res 2020; 64:e2000354. [DOI: 10.1002/mnfr.202000354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Clara Gabás‐Rivera
- Departamento Bioquímica y Biología Molecular y Celular Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | | | | | - Eduardo Romanos
- Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50009 Spain
| | - Roberto Martínez‐Beamonte
- Departamento Bioquímica y Biología Molecular y Celular Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - María A. Navarro
- Departamento Bioquímica y Biología Molecular y Celular Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Joaquín C. Surra
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- Departamento de Producción Animal Instituto de Investigación Sanitaria de Aragón (IISA) Escuela Politécnica Superior de Huesca Huesca 22071 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- Departamento de Patología Animal Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - María J. Rodríguez‐Yoldi
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- Departamento de Farmacología y Fisiología Instituto de Investigación Sanitaria de Aragón (IISA) Facultad de Veterinaria Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Cristina Andrés‐Lacueva
- Biomarkers and Nutrimetabolomics Laboratory Department of Nutrition Food Sciences and Gastronomy Faculty of Pharmacy and Food Sciences University of Barcelona Barcelona 08028 Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Jesús Osada
- Departamento Bioquímica y Biología Molecular y Celular Facultad de Veterinaria Instituto de Investigación Sanitaria de Aragón (IISA) Universidad de Zaragoza Zaragoza 50013 Spain
- Instituto Agroalimentario de Aragón CITA‐Universidad de Zaragoza Zaragoza 50013 Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Instituto de Salud Carlos III Madrid 28029 Spain
| |
Collapse
|
17
|
Oksal E, Pangestika I, Muhammad TST, Mohamad H, Amir H, Kassim MNI, Andriani Y. In vitro and in vivo studies of nanoparticles of chitosan- Pandanus tectorius fruit extract as new alternative treatment for hypercholesterolemia via Scavenger Receptor Class B type 1 pathway. Saudi Pharm J 2020; 28:1263-1275. [PMID: 33132720 PMCID: PMC7584805 DOI: 10.1016/j.jsps.2020.08.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Pandanus tectorius fruit, a natural product rich in tangeretin and ethyl caffeate, has been reported to have potential as anti-hypercholesterolemia agent via Scavenger Receptor Class B type 1 (SR-B1) pathway. However, due to its semi-polar properties, P. tectorius extract exhibits poor solubility when used as a medical remedy. The extract’s solubility can potentially be improved through a synthesis of nanoparticles of chitosan-P. tectorius fruit extract. This can also increase the extract’s SR-B1 gene expression activity. To date, no studies of nanoparticles of chitosan-P. tectorius fruit extract and its pathway via SR-B1 have been published anywhere. In this study, cytotoxicity properties against HepG2 were explored by MTT. Then luciferase assay was used to detect their effectiveness in increasing SR-B1 activity. An in vivo study using Sprague dawley was carried out to observe the extract nanoparticles’ effectiveness in reducing the cholesterol levels and the toxicity property in rat’s liver. As the results showed, the extract nanoparticles had no cytotoxic activity against HepG2 cells and exhibited higher SR-B1 gene expression activity than the non-nanoparticle form. As the in vivo study proved, nanoparticle treatment can reduce the levels of TC (197%), LDL (360%), and TG (109%), as well as increase the level of HDL cholesterol by 150%, in comparison to those for the untreated high-cholesterol diet group. From the toxicity study, it was found that there was non-toxicity in the liver. It can be concluded that nanoparticles of chitosan-P. tectorius fruit extract successfully increased P. tectorius fruit extract’s effectiveness in reducing hypercholesterolemia via SR-B1 pathway. Hence, it can be suggested that nanoparticles of chitosan-P. tectorius fruit extract is safe and suitable as an alternative treatment for controlling hypercholesterolemia via SR-B1 pathway.
Collapse
Affiliation(s)
- Efriyana Oksal
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot 21030, Kuala Nerus, Terengganu, Malaysia
| | - Inten Pangestika
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot 21030, Kuala Nerus, Terengganu, Malaysia
| | - Tengku Sifzizul Tengku Muhammad
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot 21030, Kuala Nerus, Terengganu, Malaysia
- Research Management Center, Universiti Malaysia Terengganu, Mengabang Telipot 21030, Kuala Nerus, Terengganu, Malaysia
| | - Habsah Mohamad
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot 21030, Kuala Nerus, Terengganu, Malaysia
| | - Hermansyah Amir
- Educational Chemistry Program, Faculty of Teacher Training and Education, Bengkulu University, Bengkulu 38371, Indonesia
| | - Murni Nur Islamiah Kassim
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot 21030, Kuala Nerus, Terengganu, Malaysia
| | - Yosie Andriani
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot 21030, Kuala Nerus, Terengganu, Malaysia
- Corresponding author.
| |
Collapse
|
18
|
Micera M, Botto A, Geddo F, Antoniotti S, Bertea CM, Levi R, Gallo MP, Querio G. Squalene: More than a Step toward Sterols. Antioxidants (Basel) 2020; 9:antiox9080688. [PMID: 32748847 PMCID: PMC7464659 DOI: 10.3390/antiox9080688] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/16/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Squalene (SQ) is a natural triterpene widely distributed in nature. It is a metabolic intermediate of the sterol biosynthetic pathway and represents a possible target in different metabolic and oxidative stress-related disorders. Growing interest has been focused on SQ’s antioxidant properties, derived from its chemical structure. Strong evidence provided by ex vivo models underline its scavenging activity towards free radicals, whereas only a few studies have highlighted its effect in cellular models of oxidative stress. Given the role of unbalanced free radicals in both the onset and progression of several cardiovascular diseases, an in depth evaluation of SQ’s contribution to antioxidant defense mechanisms could represent a strategic approach in dealing with these pathological conditions. At present experimental results overall show a double-edged sword role of squalene in cardiovascular diseases and its function has to be better elucidated in order to establish intervention lines focused on its features. This review aims to summarize current knowledge about endogenous and exogenous sources of SQ and to point out the controversial role of SQ in cardiovascular physiology.
Collapse
Affiliation(s)
- Marco Micera
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (M.M.); (F.G.); (S.A.); (C.M.B.); (R.L.); (G.Q.)
- Exenia Group S.r.l., 10064 Pinerolo (TO), Italy;
| | | | - Federica Geddo
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (M.M.); (F.G.); (S.A.); (C.M.B.); (R.L.); (G.Q.)
| | - Susanna Antoniotti
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (M.M.); (F.G.); (S.A.); (C.M.B.); (R.L.); (G.Q.)
| | - Cinzia Margherita Bertea
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (M.M.); (F.G.); (S.A.); (C.M.B.); (R.L.); (G.Q.)
| | - Renzo Levi
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (M.M.); (F.G.); (S.A.); (C.M.B.); (R.L.); (G.Q.)
| | - Maria Pia Gallo
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (M.M.); (F.G.); (S.A.); (C.M.B.); (R.L.); (G.Q.)
- Correspondence:
| | - Giulia Querio
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (M.M.); (F.G.); (S.A.); (C.M.B.); (R.L.); (G.Q.)
| |
Collapse
|
19
|
Shimizu N, Ito J, Kato S, Eitsuka T, Miyazawa T, Nakagawa K. Significance of Squalene in Rice Bran Oil and Perspectives on Squalene Oxidation. J Nutr Sci Vitaminol (Tokyo) 2020; 65:S62-S66. [PMID: 31619649 DOI: 10.3177/jnsv.65.s62] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
As an intermediate metabolite during the biosynthesis of sterols, squalene is found ubiquitously in plants and animals. In rice, squalene is contained in rice bran, and consequently, squalene in rice bran oil has gained attention. Studies have shown that the intake of squalene from food sources demonstrate various physiological benefits such as the prevention of cancer and cardiovascular disease. Squalene is also known as an effective antioxidant in edible oils. However, due to its chemical structure, squalene is susceptible to oxidation, which may cause a decline in the nutraceutical and antioxidative effects of squalene in edible oils. Oxidative degradation of squalene also results in the formation of scission products (i.e., aldehydes and ketones) which may lead to off-flavor. Since the rate of squalene oxidation depends on the factors that induce its oxidation (i.e., light or heat), emphasis on oxidation mechanisms is necessary. It has been demonstrated in previous studies that the oxidation products formed by the singlet oxygen oxidation and free radical oxidation of squalene are different, and more recently, we demonstrated that different squalene monohydroperoxide isomers are formed by each oxidation mechanism. We herein discuss the significance of squalene in rice bran oil as well as the oxidative degradation of squalene in edible oils with focus on oxidation mechanisms.
Collapse
Affiliation(s)
- Naoki Shimizu
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University
| | - Junya Ito
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University
| | - Shunji Kato
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University.,Division of Host Defense Mechanism, Tokai University School of Medicine
| | - Takahiro Eitsuka
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University
| | - Teruo Miyazawa
- Food and Biotechnology Innovation Project, New Industry Creation Hatchery Center (NICHe), Tohoku University.,Food and Health Science Research Unit, Graduate School of Agricultural Science, Tohoku University
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University
| |
Collapse
|
20
|
Ibrahim N‘I, Fairus S, Zulfarina MS, Naina Mohamed I. The Efficacy of Squalene in Cardiovascular Disease Risk-A Systematic Review. Nutrients 2020; 12:E414. [PMID: 32033387 PMCID: PMC7071298 DOI: 10.3390/nu12020414] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/31/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Cardiovascular disease (CVD) is the leading cause of death worldwide. Squalene (SQ), an intermediate for the cholesterol biosynthesis, has been proposed to act similarly to statins via inhibition of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase in the liver. PURPOSE This paper explores the effects of SQ in CVD. METHODS A systematic review of the literature was performed to identify relevant studies about SQ and CVD. A comprehensive search in Medline and Scopus for relevant studies published between the years 1946 and 2019 was performed. The main inclusion criteria were that the study was published in English; that the study reported association or effect of SQ and CVD; and that CVD should be related to lifestyle variables, aging, or experimentally induced conditions. RESULTS The literature searches identified 5562 potentially relevant articles, whereby 21 studies met the inclusion criteria. There were three human studies and 18 animal experimental studies included in this paper. Only one human study reported positive outcome of SQ in CVD. The remaining two studies reported inconsistent and/or no effect. For animal studies, 15 studies reported positive effect while the remaining reported negative and/or no effect of SQ on various related parameters. CONCLUSIONS This evidence-based review emphasizes the potential of SQ being used for cardiovascular-related diseases. The effect of SQ, especially of plant-based warrants further exploration. Controlled human observational studies should be performed to provide comprehensive evidence.
Collapse
Affiliation(s)
- Nurul ‘Izzah Ibrahim
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Syed Fairus
- Malaysian Palm Oil Board (MPOB), No. 6 Persiaran Institusi, Bandar Baru Bangi, Kajang 43000, Selangor, Malaysia
| | - Mohamed S. Zulfarina
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Isa Naina Mohamed
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
Herrera-Marcos LV, Sancho-Knapik S, Gabás-Rivera C, Barranquero C, Gascón S, Romanos E, Martínez-Beamonte R, Navarro MA, Surra JC, Arnal C, García-de-Jalón JA, Rodríguez-Yoldi MJ, Tena-Sempere M, Sánchez-Ramos C, Monsalve M, Osada J. Pgc1a is responsible for the sex differences in hepatic Cidec/Fsp27β mRNA expression in hepatic steatosis of mice fed a Western diet. Am J Physiol Endocrinol Metab 2020; 318:E249-E261. [PMID: 31846369 DOI: 10.1152/ajpendo.00199.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hepatic fat-specific protein 27 [cell death-inducing DNA fragmentation effector protein C (Cidec)/Fsp27] mRNA levels have been associated with hepatic lipid droplet extent under certain circumstances. To address its hepatic expression under different dietary conditions and in both sexes, apolipoprotein E (Apoe)-deficient mice were subjected to different experimental conditions for 11 wk to test the influence of cholesterol, Western diet, squalene, oleanolic acid, sex, and surgical castration on Cidec/Fsp27 mRNA expression. Dietary cholesterol increased hepatic Cidec/Fsp27β expression, an effect that was suppressed when cholesterol was combined with saturated fat as represented by Western diet feeding. Using the latter diet, neither oleanolic acid nor squalene modified its expression. Females showed lower levels of hepatic Cidec/Fsp27β expression than males when they were fed Western diets, a result that was translated into a lesser amount of CIDEC/FSP27 protein in lipid droplets and microsomes. This was also confirmed in low-density lipoprotein receptor (Ldlr)-deficient mice. Incubation with estradiol resulted in decreased Cidec/Fsp27β expression in AML12 cells. Whereas male surgical castration did not modify the expression, ovariectomized females did show increased levels compared with control females. Females also showed increased expression of peroxisome proliferator-activated receptor-γ coactivator 1-α (Pgc1a), suppressed by ovariectomy, and the values were significantly and inversely associated with those of Cidec/Fsp27β. When Pgc1a-deficient mice were used, the sex differences in Cidec/Fsp27β expression disappeared. Therefore, hepatic Cidec/Fsp27β expression has a complex regulation influenced by diet and sex hormonal milieu. The mRNA sex differences are controlled by Pgc1a.
Collapse
Affiliation(s)
- Luis V Herrera-Marcos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Sara Sancho-Knapik
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Clara Gabás-Rivera
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sonia Gascón
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Romanos
- Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María A Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquín C Surra
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Huesca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - José A García-de-Jalón
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - María J Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Tena-Sempere
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba e Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Sánchez-Ramos
- Instituto de Investigaciones Biomedicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - María Monsalve
- Instituto de Investigaciones Biomedicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Abstract
Olive-pomace oil is rich in oleic acid, and thus it can be an interesting dietary fat alternative as it can allow reaching the recommendation of consuming 20% of total diet energy in the form of monounsaturated fatty acids. In addition, olive-pomace oil also contains a wide range of minor components that may contribute to its healthy properties. The major components identified with healthy properties are triterpenic dialcohols and acids, squalene, tocopherols, sterols, fatty alcohols and phenolic compounds. The refining process, that the crude pomace-oil must undergo for commercial purposes, significantly reduces the content of phenolic compounds, while the other minor components remain at concentrations which can induce positive health effects, especially on cardiovascular health, outstanding pentacyclic triterpenes and aliphatic fatty alcohols in olive-pomace oil. Numerous in vitro and preclinical studies support that mainly the pure compounds, or extracts isolated from plant sources, play an important role in preventing cardiovascular disease and risk factors. Likewise, tocopherols, squalene and phytosterols, in addition to the minor fraction of phenolic compounds, have shown high biological activity with particular association to the cardiovascular function. In the light of the foregoing, and taking into consideration the absence of clinical studies with olive-pomace oil, it would be of great interest to develop randomized, crossover, controlled, double-blind studies to extend the knowledge and understanding on the health effects of olive-pomace olive.
Collapse
Affiliation(s)
- Raquel Mateos
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), CSIC, Madrid, Spain
| | - Beatriz Sarria
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), CSIC, Madrid, Spain
| | - Laura Bravo
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), CSIC, Madrid, Spain
| |
Collapse
|
23
|
Squalene Stimulates a Key Innate Immune Cell to Foster Wound Healing and Tissue Repair. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:9473094. [PMID: 30363968 PMCID: PMC6186384 DOI: 10.1155/2018/9473094] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/09/2018] [Indexed: 01/05/2023]
Abstract
Anti-inflammatory effects of virgin olive oil (VOO) have been described recently, along with its wound healing effect. One of the main minor compounds found in VOO is squalene (SQ), which also possesses preventive effects against skin damage and anti-inflammatory properties. The inflammatory response is involved in wound healing and manages the whole process by macrophages, among others, as the main innate cells with a critical role in the promotion and resolution of inflammation for tissue repair. Because of that, this work is claimed to describe the role that squalene exerts in the immunomodulation of M1 proinflammatory macrophages, which are the first cells implicate in recent injuries. Pro- and anti-inflammatory cytokines were analysed using TPH1 cell experimental model. SQ induced an increase in the synthesis of anti-inflammatory cytokines, such as IL-10, IL-13, and IL-4, and a decrease in proinflammatory signals, such as TNF-α and NF-κB in M1 proinflammatory macrophages. Furthermore, SQ enhanced remodelling and repairing signals (TIMP-2) and recruitment signals of eosinophils and neutrophils, responsible for phagocytosis processes. These results suggest that SQ is able to promote wound healing by driving macrophage response in inflammation. Therefore, squalene could be useful at the resolution stage of wound healing.
Collapse
|
24
|
Hepatic subcellular distribution of squalene changes according to the experimental setting. J Physiol Biochem 2018; 74:531-538. [DOI: 10.1007/s13105-018-0616-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/14/2018] [Indexed: 02/07/2023]
|
25
|
New perspectives on bioactivity of olive oil: evidence from animal models, human interventions and the use of urinary proteomic biomarkers. Proc Nutr Soc 2015; 74:268-81. [DOI: 10.1017/s0029665115002323] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Olive oil (OO) is the primary source of fat in the Mediterranean diet and has been associated with longevity and a lower incidence of chronic diseases, particularly CHD. Cardioprotective effects of OO consumption have been widely related with improved lipoprotein profile, endothelial function and inflammation, linked to health claims of oleic acid and phenolic content of OO. With CVD being a leading cause of death worldwide, a review of the potential mechanisms underpinning the impact of OO in the prevention of disease is warranted. The current body of evidence relies on mechanistic studies involving animal and cell-based models, epidemiological studies of OO intake and risk factor, small- and large-scale human interventions, and the emerging use of novel biomarker techniques associated with disease risk. Although model systems are important for mechanistic research nutrition, methodologies and experimental designs with strong translational value are still lacking. The present review critically appraises the available evidence to date, with particular focus on emerging novel biomarkers for disease risk assessment. New perspectives on OO research are outlined, especially those with scope to clarify key mechanisms by which OO consumption exerts health benefits. The use of urinary proteomic biomarkers, as highly specific disease biomarkers, is highlighted towards a higher translational approach involving OO in nutritional recommendations.
Collapse
|
26
|
Boulila A, Bejaoui A. Lycium intricatum Boiss.: An unexploited and rich source of unsaturated fatty acids, 4-desmethylsterols and other valuable phytochemicals. Lipids Health Dis 2015; 14:59. [PMID: 26104186 PMCID: PMC4485344 DOI: 10.1186/s12944-015-0055-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/09/2015] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Lycium intricatum Boiss., a Solanaceous shrubbery is used in Tunisia as a windbreak and medicinal plant. However, it is considered as underexploited specie despite its high potential to serve as source with economic and nutritional value. To date only limited information about its phytochemistry, especially of its oil has been published. This work provides data on fatty acids, phytosterols and vitamin D composition of L. intricatum seed oil. It opens up new possibilities of developing L. intricatum as a new crop that contains phytochemicals with high added value little influenced by selection or commercial breeding. FINDINGS The composition of fatty acids, phytosterols and vitamin D in L. intricatum seed oil was assessed by GC-FID. The main fatty acids of L. intricatum seed oil were linoleic acid (49.47%), palmitoleic acid (27.96%) and erucic acid (13.62%). Palimtic acid was present at low percentage (0.63%). The content of unsaturated fatty acids was high as 94.04%. The sterolic fraction was composed of stigmasterol (18.56 mg/100 g), β-sitosterol (13.04 mg/100 g). L. intricatum oil is an oily matrix that contains hydrocarbons, mainly squalene (63.36 mg/100 g), and two triterpenic alcohol erythrodiol (80.36 mg/100 g) and uvaol (24.06 mg/100 g). provitamin D was present in high quantity (8.12 mg/100 g). CONCLUSIONS From these results it has been shown that L. intricatum seeds have great potential as a source of fatty acids and phytosterols for natural health products.
Collapse
Affiliation(s)
- Abdennacer Boulila
- Laboratory of Natural Substances, National Institute of Research and Physico-chemical Analysis, Biotechpole of Sidi Thabet, Ariana, 2020, Tunisia.
| | - Afef Bejaoui
- Laboratory of Plant Biotechnology, National Institute of Applied Sciences and Technology, BP 676, Centre Urbain Nord, Tunis Cedex, 1080, Tunis, Tunisia.
| |
Collapse
|
27
|
Lou-Bonafonte JM, Gabás-Rivera C, Navarro MA, Osada J. PON1 and Mediterranean Diet. Nutrients 2015; 7:4068-92. [PMID: 26024295 PMCID: PMC4488773 DOI: 10.3390/nu7064068] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/20/2015] [Indexed: 12/18/2022] Open
Abstract
The Mediterranean diet has been proven to be highly effective in the prevention of cardiovascular diseases. Paraoxonase 1 (PON1) has been implicated in the development of those conditions, especially atherosclerosis. The present work describes a systematic review of current evidence supporting the influence of Mediterranean diet and its constituents on this enzyme. Despite the differential response of some genetic polymorphisms, the Mediterranean diet has been shown to exert a protective action on this enzyme. Extra virgin olive oil, the main source of fat, has been particularly effective in increasing PON1 activity, an action that could be due to low saturated fatty acid intake, oleic acid enrichment of phospholipids present in high-density lipoproteins that favor the activity, and increasing hepatic PON1 mRNA and protein expressions induced by minor components present in this oil. Other Mediterranean diet constituents, such as nuts, fruits and vegetables, have been effective in modulating the activity of the enzyme, pomegranate and its compounds being the best characterized items. Ongoing research on compounds isolated from all these natural products, mainly phenolic compounds and carotenoids, indicates that some of them are particularly effective, and this may enhance the use of nutraceuticals and functional foods capable of potentiating PON1 activity.
Collapse
Affiliation(s)
- José M Lou-Bonafonte
- Departamento de Farmacología y Fisiología, Facultad de Ciencias de la Salud y del Deporte, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Huesca, E-22002, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, E-28029, Spain.
| | - Clara Gabás-Rivera
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, E-28029, Spain.
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, E-50013, Spain.
| | - María A Navarro
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, E-28029, Spain.
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, E-50013, Spain.
| | - Jesús Osada
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, E-28029, Spain.
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, E-50013, Spain.
| |
Collapse
|
28
|
Sánchez-Fidalgo S, Villegas I, Rosillo MÁ, Aparicio-Soto M, de la Lastra CA. Dietary squalene supplementation improves DSS-induced acute colitis by downregulating p38 MAPK and NFkB signaling pathways. Mol Nutr Food Res 2014; 59:284-92. [PMID: 25387687 DOI: 10.1002/mnfr.201400518] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/29/2014] [Accepted: 10/31/2014] [Indexed: 12/20/2022]
Abstract
SCOPE Squalene is a polyunsaturated triterpene, which has exhibited anticancer and antioxidant activities among others. We investigated dietary squalene supplementation effect on an acute colitis model induced by dextran sulfate sodium (DSS) in C57BL/6 mice. METHODS AND RESULTS Mice were fed from weaning with squalene at 0.02% and 0.1%. After 4 weeks, mice were exposed to 3% DSS for 5 days developing acute colitis. After DSS removal (5 days), colons were histological and biochemically processed. Our results showed that dietary squalene treatment exerts anti-inflammatory action in DSS-induced acute colitis. Western blot revealed that squalene downregulated COX-2 (where COX is cyclooxygenase) and inducible nitric oxide synthase system by inhibition of mitogen-activated protein kinase p38 and the nuclear factor-kappa B signaling pathways, preventing an increase in the cytokines levels. Under our experimental conditions, STAT3 and FOXP3 (where FOXP3 is forkhead box P3) were not modified and the transcriptional regulation of antioxidant and/or detoxifying enzymes, Nrf2 (where Nrf2 is nuclear factor (erythroid-derived 2)-like 2), was reduced in DSS-induced colitis. However, any change could be observed after squalene supplementation. CONCLUSION Squalene was able to improve the oxidative events and returned proinflammatory proteins expression to basal levels probably through p38 mitogen-activated protein kinase and nuclear factor-kappa B signaling pathways. However, supplementary studies are needed in order to provide a basis for developing a new dietary supplementation strategy.
Collapse
|
29
|
Gabás-Rivera C, Barranquero C, Martínez-Beamonte R, Navarro MA, Surra JC, Osada J. Dietary squalene increases high density lipoprotein-cholesterol and paraoxonase 1 and decreases oxidative stress in mice. PLoS One 2014; 9:e104224. [PMID: 25117703 PMCID: PMC4130590 DOI: 10.1371/journal.pone.0104224] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 06/25/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Squalene, the main hydrocarbon in the unsaponifiable fraction of virgin olive oil, is involved in cholesterol synthesis and it has been reported to own antiatherosclerotic and antiesteatosic effects. However, the squalene's role on lipid plasma parameters and the influence of genotype on this effect need to be addressed. EXPERIMENTAL APPROACHES Three male mouse models (wild-type, Apoa1- and Apoe- deficient) were fed chow semisynthetic diets enriched in squalene to provide a dose of 1 g/kg during 11 weeks. After this period, their plasma parameters and lipoprotein profiles were analyzed. KEY RESULTS Squalene administration at a dose of 1 g/kg showed decreased reactive oxygen species in lipoprotein fractions independently of the animal background and caused an specific increase in high density lipoprotein (HDL)-cholesterol levels, accompanied by an increase in phosphatidylcholine and paraoxonase 1 and no changes in apolipoproteins A1 and A4 in wild-type mice. In these mice, the cholesterol increase was due to its esterified form and associated with an increased hepatic expression of Lcat. These effects were not observed in absence of apolipoprotein A1. The increases in HDL- paraoxonase 1 were translated into decreased plasma malondialdehyde levels depending on the presence of Apolipoprotein A1. CONCLUSIONS AND IMPLICATIONS Dietary squalene promotes changes in HDL- cholesterol and paraoxonase 1 and decreases reactive oxygen species in lipoproteins and plasma malondialdehyde levels, providing new benefits of its intake that might contribute to explain the properties of virgin olive oil, although the phenotype related to apolipoproteins A1 and E may be particularly relevant.
Collapse
Affiliation(s)
- Clara Gabás-Rivera
- Departamento Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Barranquero
- Departamento Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Martínez-Beamonte
- Departamento Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María A. Navarro
- Departamento Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquín C. Surra
- Departamento de Producción Animal, Escuela Politécnica Superior de Huesca, Huesca, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Osada
- Departamento Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
30
|
Alkhouri N, Eng K, Lopez R, Nobili V. Non-high-density lipoprotein cholesterol (non-HDL-C) levels in children with nonalcoholic fatty liver disease (NAFLD). SPRINGERPLUS 2014; 3:407. [PMID: 25126490 PMCID: PMC4130966 DOI: 10.1186/2193-1801-3-407] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 07/16/2014] [Indexed: 12/21/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with increased cardiovascular disease (CVD) risk in children. Non-high density lipoprotein-cholesterol (non-HDL-C) has been shown to be a good predictor of cardiovascular events. Recent data in adults found non-alcoholic steatohepatitis (NASH) to be associated with significantly higher levels of non–HDL-C than simple steatosis, suggestive it might be used as a non-invasive tool to diagnose NASH. The goal of our study was to assess non-HDL-C levels in children with NAFLD. Our cohort consisted of pediatric patients with biopsy-proven NAFLD. Anthropometric, laboratory, and histologic data were obtained on all patients. Univariable analysis was performed to assess differences in clinical characteristics between groups. Spearman rank correlation coefficients were calculated to assess the correlation between non-HDL-C levels and clinical variables. ANCOVA was used to adjust for possible confounders. 302 subjects with NAFLD were included in our study; 203 with NASH and 99 without NASH. Subjects with NASH had significantly higher non-HDL-C levels than those without (p = 0.004). Histologic features of NASH, including ballooning, inflammation, and fibrosis were found to be weakly correlated with non-HDL-C levels, (p < 0.05 for all). After adjusting for the presence of metabolic syndrome (MetS), ALT, and GGT, the association between non-HDL-C and NASH was not significant (p = 0.66). In Conclusion, non-HDL-C levels are higher in children with NASH than those with simple steatosis, suggesting increased CVD risk. This may be a reflection of the higher prevalence of MetS. Non-HDL-C had a positive association with histologic features of NASH.
Collapse
Affiliation(s)
- Naim Alkhouri
- Department of Pediatric Gastroenterology, Cleveland Clinic, 9500 Euclid Avenue, A111, Cleveland, OH 44195 USA ; Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Avenue, A51, Cleveland, OH 44195 USA
| | - Katharien Eng
- Department of Pediatric Gastroenterology, Cleveland Clinic, 9500 Euclid Avenue, A111, Cleveland, OH 44195 USA
| | - Rocio Lopez
- Quantitative Health Sciences, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195 USA
| | - Valerio Nobili
- Liver Unit, Bambino Gesù Children's Hospital and Research Institute, 37 Salita di Sant'onofrio, 00165 Rome, Italy
| |
Collapse
|
31
|
Postprandial changes in high density lipoproteins in rats subjected to gavage administration of virgin olive oil. PLoS One 2013; 8:e55231. [PMID: 23383120 PMCID: PMC3558467 DOI: 10.1371/journal.pone.0055231] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 12/28/2012] [Indexed: 01/30/2023] Open
Abstract
Background and Aims The present study was designed to verify the influence of acute fat loading on high density lipoprotein (HDL) composition, and the involvement of liver and different segments of small intestine in the changes observed. Methods and Results To address these issues, rats were administered a bolus of 5-ml of extra-virgin olive oil and sacrificed 4 and 8 hours after feeding. In these animals, lipoproteins were analyzed and gene expressions of apolipoprotein and HDL enzymes were assessed in duodenum, jejunum, ileum and liver. Using this experimental design, total plasma and HDL phospholipids increased at the 8-hour-time-point due to increased sphingomyelin content. An increase in apolipoprotein A4 was also observed mainly in lipid-poor HDL. Increased expression of intestinal Apoa1, Apoa4 and Sgms1 mRNA was accompanied by hepatic decreases in the first two genes in liver. Hepatic expression of Abcg1, Apoa1bp, Apoa2, Apoe, Ptlp, Pon1 and Scarb1 decreased significantly following fat gavage, while no changes were observed for Abca1, Lcat or Pla2g7. Significant associations were also noted for hepatic expression of apolipoproteins and Pon1. Manipulation of postprandial triglycerides using an inhibitor of microsomal transfer protein -CP-346086- or of lipoprotein lipase –tyloxapol- did not influence hepatic expression of Apoa1 or Apoa4 mRNA. Conclusion All these data indicate that dietary fat modifies the phospholipid composition of rat HDL, suggesting a mechanism of down-regulation of hepatic HDL when intestine is the main source of those particles and a coordinated regulation of hepatic components of these lipoproteins at the mRNA level, independently of plasma postprandial triglycerides.
Collapse
|
32
|
In comparison with palm oil, dietary nut supplementation delays the progression of atherosclerotic lesions in female apoE-deficient mice. Br J Nutr 2012; 109:202-9. [PMID: 23302442 DOI: 10.1017/s000711451200092x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Epidemiological studies have demonstrated the benefits of nut consumption on cardiovascular risk factors and CHD, attributed to their fatty acid profile, rich in unsaturated fatty acids, and also to other nutrients. The effect of nuts on atherosclerotic lesions was studied in female and male apoE-knockout mice fed a diet supplemented with 3 % (w/w) mixed nuts (mix: almonds, hazelnuts and walnuts in a proportion of 0.25:0·25:0.50, respectively), and compared with mice receiving an isoenergetic diet of similar fat content provided as palm oil. After 12 weeks, plasma lipid parameters and aortic lesions were measured. Males receiving nuts had lower plasma cholesterol than the palm oil group, and both sex groups had lower plasma non-HDL-cholesterol and lower content of reactive oxygen species in LDL than mice receiving the palm oil diet, the latter decrease being more pronounced in females than in males. Females consuming the nut diet showed a smaller aortic lesion area than those consuming palm oil, whereas no differences were observed in males. In females, hepatic paraoxonase 2 (Pon2) mRNA increased, and no change was observed in prenylcysteine oxidase 1 (Pcyox1) expression after the consumption of the nut-containing diet. In addition, aortic atherosclerotic lesions correlated directly with total plasma cholesterol and inversely with hepatic Pon2 expression. The results suggest that the beneficial effect of nut intake in female apoE-deficient mice may be attributed to reduced non-HDL-cholesterol levels and enhanced PON2 antioxidant activity.
Collapse
|
33
|
Proteomics and gene expression analyses of squalene-supplemented mice identify microsomal thioredoxin domain-containing protein 5 changes associated with hepatic steatosis. J Proteomics 2012; 77:27-39. [DOI: 10.1016/j.jprot.2012.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 06/28/2012] [Accepted: 07/01/2012] [Indexed: 11/18/2022]
|
34
|
Lou-Bonafonte JM, Arnal C, Navarro MA, Osada J. Efficacy of bioactive compounds from extra virgin olive oil to modulate atherosclerosis development. Mol Nutr Food Res 2012; 56:1043-57. [DOI: 10.1002/mnfr.201100668] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Granados-Principal S, Quiles JL, Ramirez-Tortosa CL, Ochoa-Herrera J, Perez-Lopez P, Pulido-Moran M, Ramirez-Tortosa MC. Squalene ameliorates atherosclerotic lesions through the reduction of CD36 scavenger receptor expression in macrophages. Mol Nutr Food Res 2012; 56:733-40. [DOI: 10.1002/mnfr.201100703] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Sergio Granados-Principal
- Department of Biochemistry and Molecular Biology II, University of Granada; Granada Spain
- “José Mataix” Institute of Nutrition and Food Technology. Biomedical Research Centre, University of Granada; Granada Spain
| | - Jose L. Quiles
- “José Mataix” Institute of Nutrition and Food Technology. Biomedical Research Centre, University of Granada; Granada Spain
- Department of Physiology, University of Granada; Granada Spain
| | | | - Julio Ochoa-Herrera
- “José Mataix” Institute of Nutrition and Food Technology. Biomedical Research Centre, University of Granada; Granada Spain
- Department of Physiology, University of Granada; Granada Spain
| | - Patricia Perez-Lopez
- “José Mataix” Institute of Nutrition and Food Technology. Biomedical Research Centre, University of Granada; Granada Spain
- Department of Physiology, University of Granada; Granada Spain
| | - Mario Pulido-Moran
- Department of Biochemistry and Molecular Biology II, University of Granada; Granada Spain
- “José Mataix” Institute of Nutrition and Food Technology. Biomedical Research Centre, University of Granada; Granada Spain
| | - MCarmen Ramirez-Tortosa
- Department of Biochemistry and Molecular Biology II, University of Granada; Granada Spain
- “José Mataix” Institute of Nutrition and Food Technology. Biomedical Research Centre, University of Granada; Granada Spain
| |
Collapse
|
36
|
Ramírez-Torres A, Barceló-Batllori S, Fernández-Vizarra E, Navarro MA, Arnal C, Guillén N, Acín S, Osada J. Proteomics and gene expression analyses of mitochondria from squalene-treated apoE-deficient mice identify short-chain specific acyl-CoA dehydrogenase changes associated with fatty liver amelioration. J Proteomics 2012; 75:2563-75. [DOI: 10.1016/j.jprot.2012.02.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 02/06/2012] [Accepted: 02/20/2012] [Indexed: 02/07/2023]
|
37
|
Rotllan N, Llaverías G, Julve J, Jauhiainen M, Calpe-Berdiel L, Hernández C, Simó R, Blanco–Vaca F, Escolà-Gil JC. Differential effects of gemfibrozil and fenofibrate on reverse cholesterol transport from macrophages to feces in vivo. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:104-10. [DOI: 10.1016/j.bbalip.2010.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 11/04/2010] [Accepted: 11/19/2010] [Indexed: 10/18/2022]
|
38
|
Horvat S, Mcwhir J, Rozman D. Defects in cholesterol synthesis genes in mouse and in humans: lessons for drug development and safer treatments. Drug Metab Rev 2011; 43:69-90. [DOI: 10.3109/03602532.2010.540580] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
39
|
Chiang CH, Huang CC, Chan WL, Chen JW, Leu HB. The severity of non-alcoholic fatty liver disease correlates with high sensitivity C-reactive protein value and is independently associated with increased cardiovascular risk in healthy population. Clin Biochem 2010; 43:1399-404. [DOI: 10.1016/j.clinbiochem.2010.09.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 09/02/2010] [Accepted: 09/03/2010] [Indexed: 12/22/2022]
|
40
|
Alkhouri N, Tamimi TAR, Yerian L, Lopez R, Zein NN, Feldstein AE. The inflamed liver and atherosclerosis: a link between histologic severity of nonalcoholic fatty liver disease and increased cardiovascular risk. Dig Dis Sci 2010; 55:2644-50. [PMID: 19960252 DOI: 10.1007/s10620-009-1075-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 11/25/2009] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the Western world. It encompasses a spectrum of disease ranging from simple steatosis to nonalcoholic steatohepatitis (NASH). Growing evidence links NAFLD to cardiovascular (CV) disease; however, the association between the histologic severity of NAFLD and CV risk remains poorly understood. AIM To assess the relationship between severity of liver injury and CV risk markers in a large, well-characterized group of patients with biopsy-proven NAFLD. METHODS Our cohort consisted of 83 consecutive patients undergoing liver biopsy for clinical suspicion of NAFLD. Patients were subsequently divided into three groups: normal biopsy (n=11) simple steatosis (n=36), and NASH (n=36). CV risk markers included: triglyceride/high-density lipoprotein (HDL), total cholesterol/HDL, and low-density lipoprotein/HDL ratios. RESULTS All lipid ratios were found to be significantly associated with NAFLD (p<0.05) after adjusting for age and gender. More importantly, there was a stepwise, statistically significant increase in lipid ratios from patients with normal biopsies to patients with simple steatosis to those with NASH (p<0.05). A positive correlation was found between the lipid ratios and NAFLD activity score (NAS) as well as the individual histological features of the NAS (steatosis, inflammation, and ballooning) with the strongest correlation being with NAS (rho (95% CI) 0.41 (0.21, 0.62), p<0.001). CONCLUSION In patients with NAFLD, the histologic severity of liver injury and inflammation is strongly associated with an increased CV risk and an atherogenic lipid profile.
Collapse
Affiliation(s)
- Naim Alkhouri
- Department of Pediatric Gastroenterology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Nobili V, Alkhouri N, Bartuli A, Manco M, Lopez R, Alisi A, Feldstein AE. Severity of liver injury and atherogenic lipid profile in children with nonalcoholic fatty liver disease. Pediatr Res 2010; 67:665-70. [PMID: 20496475 DOI: 10.1203/pdr.0b013e3181da4798] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome. The aim of this study was to assess the relationship between severity of liver injury and atherogenic lipid profile in a large group of children with NAFLD. A total of 118 consecutive children with biopsy-proven NAFLD were included. Patients underwent extensive metabolic profiling. The NAFLD activity and fibrosis scores showed a significant positive correlation with triglyceride/HDL, total cholesterol/HDL, and LDL/HDL ratios (p<0.05) but not with apolipoprotein B/apolipoprotein A-1 ratio (p=0.58). After adjusting for BMI, homeostatic model assessment, impaired glucose tolerance, and presence of metabolic syndrome, both the NAFLD activity score and stage of fibrosis remained independent predictors of proatherogenic lipid profile. All lipid ratios, except for apolipoprotein B/apolipoprotein A-1, were found to be markedly higher in children with nonalcoholic steatohepatitis compared with those with simple steatosis or borderline disease (p<0.05). This study shows for the first time that in children with NAFLD, the severity of liver injury is strongly associated with the presence of a more atherogenic lipid profile, having potential significant diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Valerio Nobili
- Liver Unit, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
42
|
López-Miranda J, Pérez-Jiménez F, Ros E, De Caterina R, Badimón L, Covas MI, Escrich E, Ordovás JM, Soriguer F, Abiá R, de la Lastra CA, Battino M, Corella D, Chamorro-Quirós J, Delgado-Lista J, Giugliano D, Esposito K, Estruch R, Fernandez-Real JM, Gaforio JJ, La Vecchia C, Lairon D, López-Segura F, Mata P, Menéndez JA, Muriana FJ, Osada J, Panagiotakos DB, Paniagua JA, Pérez-Martinez P, Perona J, Peinado MA, Pineda-Priego M, Poulsen HE, Quiles JL, Ramírez-Tortosa MC, Ruano J, Serra-Majem L, Solá R, Solanas M, Solfrizzi V, de la Torre-Fornell R, Trichopoulou A, Uceda M, Villalba-Montoro JM, Villar-Ortiz JR, Visioli F, Yiannakouris N. Olive oil and health: summary of the II international conference on olive oil and health consensus report, Jaén and Córdoba (Spain) 2008. Nutr Metab Cardiovasc Dis 2010; 20:284-294. [PMID: 20303720 DOI: 10.1016/j.numecd.2009.12.007] [Citation(s) in RCA: 408] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 12/13/2009] [Accepted: 12/14/2009] [Indexed: 12/15/2022]
Abstract
Olive oil (OO) is the most representative food of the traditional Mediterranean Diet (MedDiet). Increasing evidence suggests that monounsaturated fatty acids (MUFA) as a nutrient, OO as a food, and the MedDiet as a food pattern are associated with a decreased risk of cardiovascular disease, obesity, metabolic syndrome, type 2 diabetes and hypertension. A MedDiet rich in OO and OO per se has been shown to improve cardiovascular risk factors, such as lipid profiles, blood pressure, postprandial hyperlipidemia, endothelial dysfunction, oxidative stress, and antithrombotic profiles. Some of these beneficial effects can be attributed to the OO minor components. Therefore, the definition of the MedDiet should include OO. Phenolic compounds in OO have shown antioxidant and anti-inflammatory properties, prevent lipoperoxidation, induce favorable changes of lipid profile, improve endothelial function, and disclose antithrombotic properties. Observational studies from Mediterranean cohorts have suggested that dietary MUFA may be protective against age-related cognitive decline and Alzheimer's disease. Recent studies consistently support the concept that the OO-rich MedDiet is compatible with healthier aging and increased longevity. In countries where the population adheres to the MedDiet, such as Spain, Greece and Italy, and OO is the principal source of fat, rates of cancer incidence are lower than in northern European countries. Experimental and human cellular studies have provided new evidence on the potential protective effect of OO on cancer. Furthermore, results of case-control and cohort studies suggest that MUFA intake including OO is associated with a reduction in cancer risk (mainly breast, colorectal and prostate cancers).
Collapse
Affiliation(s)
- J López-Miranda
- Department of Medicine, Reina Sofia University Hospital, School of Medicine, Cordoba, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Gingival vascular damage in atherosclerotic rabbits: Hydroxytyrosol and squalene benefits. Food Chem Toxicol 2009; 47:2327-31. [DOI: 10.1016/j.fct.2009.06.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 06/02/2009] [Accepted: 06/15/2009] [Indexed: 11/16/2022]
|
44
|
Guillén N, Navarro MA, Arnal C, Noone E, Arbonés-Mainar JM, Acín S, Surra JC, Muniesa P, Roche HM, Osada J. Microarray analysis of hepatic gene expression identifies new genes involved in steatotic liver. Physiol Genomics 2009; 37:187-98. [PMID: 19258494 PMCID: PMC2685506 DOI: 10.1152/physiolgenomics.90339.2008] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Trans-10, cis-12-conjugated linoleic acid (CLA)-enriched diets promote fatty liver in mice, while cis-9, trans-11-CLA ameliorates this effect, suggesting regulation of multiple genes. To test this hypothesis, apoE-deficient mice were fed a Western-type diet enriched with linoleic acid isomers, and their hepatic gene expression was analyzed with DNA microarrays. To provide an initial screening of candidate genes, only 12 with remarkably modified expression between both CLA isomers were considered and confirmed by quantitative RT-PCR. Additionally mRNA expression of 15 genes involved in lipid metabolism was also studied. Ten genes (Fsp27, Aqp4, Cd36, Ly6d, Scd1, Hsd3b5, Syt1, Cyp7b1, and Tff3) showed significant associations among their expressions and the degree of hepatic steatosis. Their involvement was also analyzed in other models of steatosis. In hyperhomocysteinemic mice lacking Cbs gene, only Fsp27, Cd36, Scd1, Syt1, and Hsd3b5 hepatic expressions were associated with steatosis. In apoE-deficient mice consuming olive-enriched diet displaying reduction of the fatty liver, only Fsp27 and Syt1 expressions were found associated. Using this strategy, we have shown that expression of these genes is highly associated with hepatic steatosis in a genetic disease such as Cbs deficiency and in two common situations such as Western diets containing CLA isomers or a Mediterranean-type diet. Conclusion: The results highlight new processes involved in lipid handling in liver and will help to understand the complex human pathology providing new proteins and new strategies to cope with hepatic steatosis.
Collapse
Affiliation(s)
- Natalia Guillén
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto Aragonés de Ciencias de la Salud (Universidad de Zaragoza-Salud del Gobierno de Aragón), Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|