1
|
Pareek CS, Sachajko M, Kalra G, Sultana S, Szostak A, Chalaskiewicz K, Kepka-Borkowska K, Poławska E, Ogłuszka M, Pierzchała D, Starzyński R, Taniguchi H, Juszczuk-Kubiak E, Lepczyński A, Ślaska B, Kozera W, Czarnik U, Wysocki P, Kadarmideen HN, Te Pas MFW, Szyda J, Pierzchała M. Identification of trait-associated microRNA modules in liver transcriptome of pig fed with PUFAs-enriched supplementary diet. J Appl Genet 2025; 66:389-407. [PMID: 39546271 PMCID: PMC12000271 DOI: 10.1007/s13353-024-00912-w] [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: 04/14/2024] [Revised: 08/16/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024]
Abstract
Dietary lipids provide energy, are cellular structural components, and are involved in physiological processes. Lipids are the dietary source in supplementary diet experiments in pigs. This study aims to investigate the dietary effects of PUFAs on the hepatic transcriptome and physiological pathways of two diets on two pig breeds. Polish Landrace (PL: n = 6) and six PLxDuroc (PLxD: n = 6) pigs were fed with a normal diet (n = 3) or PUFAs-enriched healthy diet (n = 3), and the hepatic miRNA profiles were studied for weighted gene co-expression network analysis biological interactions between gene networks and metabolic pathways of DE miRNA genes. The study identified trait-associated modules that were significantly associated with four phenotypic traits in the dietary groups of PL and PLxD: meat colour (a*), shoulder subcutaneous fat thickness, conductivity 24 h post-mortem (PE24), and ashes. Trait-wise, a large set of co-expressed miRNAs of porcine liver were identified in these trait-associated significant modules (9, 7, 2, and 8) in PL and PLxD. Each module is represented by a module eigengene (ME). Forty-four miRNAs out of 94 miRNAs interacted with 6719 statistically significant target genes with a target score > 90. The GO/pathway analysis showed association with pathways including regulation of metallopeptidase activity, sebaceous gland development, collagen fibril organization, WNT signalling, epithelial tube morphogenesis, etc. The study showed the differences in miRNA expression between the dietary groups of PL and PLxD breeds. Hub genes of discovered miRNA clusters can be considered predicted miRNA genes associated with PE24, meat colour, shoulder subcutaneous fat thickness, and ashes. Discovered target genes for miRNA clusters play significant roles in biological functions such as (i) muscle and body growth development, (ii) different cellular processes and developments, (iii) system development, and (iv) metabolic processes.
Collapse
Affiliation(s)
- C S Pareek
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100, Toruń, Poland
- Division of Functional Genomics in Biological and Biomedical Research, Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100, Torun, Poland
| | - M Sachajko
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100, Toruń, Poland
| | - G Kalra
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100, Toruń, Poland
| | - S Sultana
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100, Toruń, Poland
| | - A Szostak
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - K Chalaskiewicz
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - K Kepka-Borkowska
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - E Poławska
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - M Ogłuszka
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - D Pierzchała
- Maria Sklodowska-Curie National Research Institute of Oncology, W.K. Roentgena 5 Str, 02-781, Warsaw, Poland
| | - R Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
| | - H Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland
- African Genome Center, Mohammed VI Polytechnic University, UM6P, Lot 660, Hay Moulay Rachid Ben Guerir, 43150, Morocco
| | - E Juszczuk-Kubiak
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology Prof. Wacław, Dąbrowski Institute of Agriculture and Food Biotechnology - State Research Institute (IBPRS-PIB), Rakowiecka 36 Str, 02-532, Warsaw, Poland
| | - A Lepczyński
- Department of Physiology, Cytobiology and Proteomics, West Pomeranian University of Technology, K. Janickiego 32 Str, 71-270, Szczecin, Poland
| | - B Ślaska
- Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13 Str, 20-950, Lublin, Poland
| | - W Kozera
- Department of Pig Breeding, Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bio-Engineering, University of Warmia and Mazury in Olsztyn, Ul. M. Oczapowskiego 5 Str, 10-719, Olsztyn, Poland
| | - U Czarnik
- Department of Pig Breeding, Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bio-Engineering, University of Warmia and Mazury in Olsztyn, Ul. M. Oczapowskiego 5 Str, 10-719, Olsztyn, Poland
| | - P Wysocki
- Department of Pig Breeding, Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bio-Engineering, University of Warmia and Mazury in Olsztyn, Ul. M. Oczapowskiego 5 Str, 10-719, Olsztyn, Poland
| | - H N Kadarmideen
- Department of Animal and Veterinary Sciences, Aarhus University, Blichers Alle 20, 8830, Tjele, Denmark
| | - M F W Te Pas
- Wageningen Livestock Research, Wageningen University and Research, 6708 WD, Wageningen, The Netherlands
| | - J Szyda
- Biostatistics Group, Department of Genetics, Wrocław University of Environmental and Life Sciences, Kozuchowska 7, 51-631, Wrocław, Poland
| | - M Pierzchała
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A Str, 05-552, Jastrzebiec, Magdalenka, Poland.
| |
Collapse
|
2
|
Yang J, Shrestha A, Ramalingam L. Fishing for Solutions: How Pre-Conceptional Fish Oil Supplementation in Obese Fathers Reduces Risk of Non-Alcoholic Fatty Liver Disease in Offspring Mice. Mol Nutr Food Res 2025; 69:e202400452. [PMID: 39910853 PMCID: PMC11874265 DOI: 10.1002/mnfr.202400452] [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: 08/12/2024] [Revised: 11/30/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025]
Abstract
Metabolic dysfunction associated fatty liver disease (MAFLD) is a chronic condition with hepatic fat accumulation. The intergenerational effect of obesity has predominantly focused on mothers, with limited studies on paternal obesity. Nutritional intervention with fish oil (FO) has beneficial effects in reducing markers of obesity. We hypothesized that supplementing obese fathers with FO before conception could enhance the metabolic health of their offspring liver. Male mice were assigned to low-fat (LF), high fat (HF), or HF supplemented with FO for 10 weeks. Subsequently, these males were mated with females on a chow diet. Offspring were sacrificed at 8 weeks, and liver tissues were analyzed for gene expression and histology. Offspring body weight was not significantly impacted by paternal diet. However, male offspring of HF fathers had higher levels of markers of inflammation and fatty acid synthesis compared to offspring of LF fed fathers. Paternal FO supplementation significantly reduced fatty acid synthesis and glucose metabolism, while increasing fatty acid oxidation in male offspring, with a less pronounced effect in female offspring. These findings suggest that FO supplementation in obese fathers prior to conception attenuates the development of MAFLD in male offspring. This data underscores the significance of paternal nutritional intervention in promoting offspring health.
Collapse
Affiliation(s)
- Junhui Yang
- Department of Nutrition and Food StudiesSyracuse UniversitySyracuseNew YorkUSA
| | - Akriti Shrestha
- Department of Nutrition and Food StudiesSyracuse UniversitySyracuseNew YorkUSA
| | - Latha Ramalingam
- Department of Nutrition and Food StudiesSyracuse UniversitySyracuseNew YorkUSA
| |
Collapse
|
3
|
Vouilloz A, Bourgeois T, Diedisheim M, Pilot T, Jalil A, Le Guern N, Bergas V, Rohmer N, Castelli F, Leleu D, Varin A, de Barros JPP, Degrace P, Rialland M, Blériot C, Venteclef N, Thomas C, Masson D. Impaired unsaturated fatty acid elongation alters mitochondrial function and accelerates metabolic dysfunction-associated steatohepatitis progression. Metabolism 2025; 162:156051. [PMID: 39454822 DOI: 10.1016/j.metabol.2024.156051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND AND AIMS Although qualitative and quantitative alterations in liver Polyunsaturated Fatty Acids (PUFAs) are observed in MASH in humans, a causal relationship of PUFAs biosynthetic pathways is yet to be clarified. ELOVL5, an essential enzyme in PUFA elongation regulates hepatic triglyceride metabolism. Nonetheless, the long-term consequences of elongase disruption, particularly in murine models of MASH, have not been evaluated. APPROACH & RESULTS In humans, transcriptomic data indicated that PUFAs biosynthesis enzymes and notably ELOVL5 were induced during MASH progression. Moreover, gene module association determination revealed that ELOVL5 expression was associated with mitochondrial function in both humans and mice. WT and Elovl5-deficient mice were fed a high-fat, high-sucrose (HF/HS) diet for four months. Elovl5 deficiency led to limited systemic metabolic alterations but significant hepatic phenotype was observed in Elovl5-/- mice after the HF/HS diet, including hepatomegaly, pronounced macrovesicular and microvesicular steatosis, hepatocyte ballooning, immune cell infiltration, and fibrosis. Lipid analysis confirmed hepatic triglyceride accumulation and a reshaping of FA profile. Transcriptomic analysis indicated significant upregulation of genes involved in immune cell recruitment and fibrosis, and downregulation of genes involved in oxidative phosphorylation in Elovl5-/- mice. Alterations of FA oxidation and energy metabolism were confirmed by non-targeted metabolomic approach. Analysis of mitochondrial function in Elovl5-/- mice showed morphological alterations, qualitative cardiolipin changes with an enrichment in species containing shorter unsaturated FAs, and decreased activity of I and III respiratory chain complexes. CONCLUSION Enhanced susceptibility to diet-induced MASH and fibrosis in Elovl5-/- mice is intricately associated with disruptions in mitochondrial homeostasis, stemming from a profound reshaping of mitochondrial lipids, notably cardiolipins.
Collapse
Affiliation(s)
- Adrien Vouilloz
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Thibaut Bourgeois
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Marc Diedisheim
- Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France; Clinique Saint Gatien Alliance (NCT+), Saint-Cyr-sur-Loire, France
| | - Thomas Pilot
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Antoine Jalil
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Naig Le Guern
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Victoria Bergas
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; Lipidomic Analytical Facility, 21000 Dijon, France
| | - Noéline Rohmer
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour La Santé (DMTS), MetaboHUB, F-91191 Gif-sur-Yvette, France
| | - Florence Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour La Santé (DMTS), MetaboHUB, F-91191 Gif-sur-Yvette, France
| | - Damien Leleu
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, 21000 Dijon, France
| | - Alexis Varin
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; Lipidomic Analytical Facility, 21000 Dijon, France
| | - Jean-Paul Pais de Barros
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; Lipidomic Analytical Facility, 21000 Dijon, France
| | - Pascal Degrace
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Mickael Rialland
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - Camille Blériot
- Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France
| | - Nicolas Venteclef
- Institut Necker-Enfants Malades, INSERM UMR-S1151, Université Paris Cité, 75015 Paris, France
| | - Charles Thomas
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France
| | - David Masson
- Université de Bourgogne, 21000 Dijon, France; INSERM, LNC UMR1231, 21000 Dijon, France; LipSTIC LabEx, 21000 Dijon, France; CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, 21000 Dijon, France.
| |
Collapse
|
4
|
Omaña-Guzmán I, Rosas-Diaz M, Martínez-López YE, Perez-Navarro LM, Diaz-Badillo A, Alanis A, Bustamante A, Castillo-Ruiz O, Del Toro-Cisneros N, Esquivel-Hernandez DA, Garcia-Villalobos G, Garibay-Nieto N, Garcia-Oropesa EM, Hernandez-Martinez JC, Lopez-Sosa EB, Maldonado C, Martinez D, Membreno J, Moctezuma-Chavez OO, Munguia-Cisneros CX, Nava-González EJ, Perales-Torres AL, Pérez-García A, Rivera-Marrero H, Valdez A, Vázquez-Chávez AA, Ramirez-Pfeiffer C, Carter KV, Tapia B, Vela L, Lopez-Alvarenga JC. Strategic interventions in clinical randomized trials for metabolic dysfunction-associated steatotic liver disease (MASLD) and obesity in the pediatric population: a systematic review with meta-analysis and bibliometric analysis. BMC Med 2024; 22:548. [PMID: 39574069 PMCID: PMC11580631 DOI: 10.1186/s12916-024-03744-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is a prevalent hepatic condition linked to metabolic alterations. It gradually causes liver damage and potentially progresses to cirrhosis. Despite its significance, research, especially in the pediatric population, is limited, leading to contradictory findings in diagnosis and treatment. This meta-analysis aims to synthesize existing literature on therapeutic interventions for MASLD in children and adolescents. METHODS A comprehensive search of randomized controlled clinical trials yielded 634 entries from PubMed, Scopus, and Web of Science up to 2023. Interventions included medications, behavioral modifications, dietary changes, probiotics, supplements, surgical procedures, or combinations. The analysis focused on studies with treatment duration of at least 3 months, employing a random-effects REML meta-analysis model. Treatment effects on anthropometric measurements and biochemical components were examined and adjusted for heterogeneity factors analysis. A bibliometric analysis for insights into research contributors was performed. RESULTS The systematic review incorporated 31 clinical trials, with 24 meeting criteria for meta-analysis. These comprised 3 medication studies, 20 with supplements, 4 focusing on lifestyle, and 4 centered on diets. Significant overall treatment effects were observed for ALT, AST, BMI, and HOMA-IR mainly by supplements and lifestyle. Meta-regression identified age, BMI changes, and treatment duration as factors modifying ALT concentrations. Bibliometric analysis involving 31 linked studies highlighted contributions from 13 countries, with the USA, Spain, and Chile being the most influential. CONCLUSIONS We conclude that supplementation and lifestyle changes can effectively impact ALT and AST levels, which can help address liver issues in obese children. However, the evaluation of risk bias, the high heterogeneity, and the bibliometric analysis emphasize the need for more high-quality studies and broader inclusion of diverse child populations to provide better therapeutic recommendations. TRIAL REGISTRATION PROSPERO, CRD42023393952. Registered on January 25, 2023.
Collapse
Affiliation(s)
- Isabel Omaña-Guzmán
- Pediatric Obesity Clinic and Wellness Unit, Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico
| | - Marisol Rosas-Diaz
- Laboratorio de Biologia Molecular, Universidad Autónoma de Tamaulipas, Tamaulipas, Mexico
| | | | | | - Alvaro Diaz-Badillo
- Department of Health and Behavioral Sciences, Texas A&M University, San Antonio, TX, USA
| | - Anthony Alanis
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | | | | | - Noemi Del Toro-Cisneros
- Departamento de Nefrología y Metabolismo Mineral, Instituto Nacional de La Nutricion Salvador Zubiran, Mexico City, Mexico
| | | | | | - Nayely Garibay-Nieto
- Pediatric Obesity Clinic and Wellness Unit, Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico
| | | | | | - Elena Beatriz Lopez-Sosa
- Angiologia y Cirugía Vascular, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado de México (ISSTE), Hospital 20 de Noviembre, Mexico City, Mexico
| | - Carlos Maldonado
- Instituto Nacional de Enfermedaes Respiratorias, Mexico City, Mexico
| | - David Martinez
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Joshua Membreno
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | | | - Claudia X Munguia-Cisneros
- Centro Especializado de Metabolismo y Diabetes (CEDIAMET), Universidad México Americana del Norte, Tamaulipas, Mexico
| | - Edna J Nava-González
- Facultad de Salud Pública y Nutrición, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | | | - Adolfo Pérez-García
- Research Department, Hospital General de México "Dr. Eduardo Liceaga", Mexico City, Mexico
| | | | - Alisha Valdez
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | | | | | - Kathleen V Carter
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Beatriz Tapia
- Faculty Affairs, Asst Dean Faculty Development, The University of Texas Rio Grande Valley, Edinburg, TX, USA
- Division of Population Health & Biostatistics, School of Medicine, University of Texas Rio Grande Valley, 2102 Treasure Hills Boulevard, Edinburg, TX, 78550, USA
| | - Leonel Vela
- Division of Population Health & Biostatistics, School of Medicine, University of Texas Rio Grande Valley, 2102 Treasure Hills Boulevard, Edinburg, TX, 78550, USA
| | - Juan Carlos Lopez-Alvarenga
- Escuela de Medicina, Universidad México Americana del Norte, Reynosa, Tamaulipas, Mexico.
- Division of Population Health & Biostatistics, School of Medicine, University of Texas Rio Grande Valley, 2102 Treasure Hills Boulevard, Edinburg, TX, 78550, USA.
| |
Collapse
|
5
|
Yi Zhang, Feng R, Li H, Wu W, Ma W, Chen X, Chen J, Liang C. CXCR4 influences PUFA desaturation and oxidative stress injury in experimental prostatitis mice by activating Fads2 via PPARγ. Free Radic Biol Med 2024; 223:237-249. [PMID: 39094710 DOI: 10.1016/j.freeradbiomed.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/29/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
Chronic prostatitis-induced excessive inflammation and oxidative stress (OS) damage substantially affect men's quality of life. However, its treatment remains a major clinical challenge. Therefore, the identification of drugs that can decrease chronic prostatitis and oxidative stress targets is urgent and essential. CXCR4 is a classic chemokine receptor that is crucially associated with the occurrence and development of inflammation. This investigation aimed to elucidate how CXCR4 affects prostatitis regression and progression. The effect of CXCR4 on chronic prostatitis was evaluated by HE staining, immunohistochemistry, immunofluorescence, PCR, and TUNEL analyses. Furthermore, CXCR4 influence on metabolism was also evaluated by monitoring body weight, body temperature, food intake, and LC/MS. Additionally, chromatin immunoprecipitation, Western blot, and double luciferase reporter gene assays were carried out to elucidate the mechanism by which CXCR4 modulates Fads2 transcription by PPARγ. Lastly, ROS, DHE, mito-tracker, and ATP were utilized to validate the α-linolenic acid's protective effect against OS in prostate epithelial cells. It was revealed that the inhibition of CXCR4 can effectively alleviate prostatitis in mice. Furthermore, downregulating CXCR4 expression can markedly reduce the inflammatory cell infiltration in mouse prostates, decrease the elevated levels of DNA damage markers,MDA and 4-HNE, and mitigate apoptosis of prostatic epithelial cells. Moreover, treatment of CXCR4 knockdown mice with a PPARγ inhibitor revealed different degrees of changes in the above phenotypes. Mechanistically, the PPARγ protein translocates to the nucleus and serves as a transcription factor to regulate Fads2 expression, thereby altering PUFA metabolism. Additionally, in vitro experiments indicated that α-linolenic acid can effectively alleviate OS damage and RWPE-1 cell apoptosis by protecting mitochondrial function and enhancing the antioxidant capacity of prostatic epithelial cells. In conclusion, reducing the levels of CXCR4 can alleviate inflammation and OS damage in chronic prostatitis.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, PR China; Institute of Urology, Anhui Medical University, Hefei, Anhui, PR China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, PR China
| | - Rui Feng
- Department of Urology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Haolin Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, PR China; Institute of Urology, Anhui Medical University, Hefei, Anhui, PR China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, PR China
| | - Weikang Wu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, PR China; Institute of Urology, Anhui Medical University, Hefei, Anhui, PR China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, PR China
| | - Wenming Ma
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, PR China; Institute of Urology, Anhui Medical University, Hefei, Anhui, PR China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, PR China
| | - Xianguo Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, PR China; Institute of Urology, Anhui Medical University, Hefei, Anhui, PR China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, PR China.
| | - Jing Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, PR China; Institute of Urology, Anhui Medical University, Hefei, Anhui, PR China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, PR China.
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, PR China; Institute of Urology, Anhui Medical University, Hefei, Anhui, PR China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, PR China.
| |
Collapse
|
6
|
Guo C, Liu Z, Fan H, Wang H, Zhang X, Zhao S, Li Y, Wang T, Dai L, Huang J, Chen X, Zhang T. Nonlinear relationships of circulating polyunsaturated fatty acids with the complications of liver cirrhosis: A prospective, longitudinal cohort study. Clin Nutr 2024; 43:2083-2091. [PMID: 39094473 DOI: 10.1016/j.clnu.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND & AIMS The role of circulating polyunsaturated fatty acids (PUFAs) in preventing liver cirrhosis complications remains unclear. METHODS Between 2006 and 2010, 273,834 UK Biobank participants with plasma PUFA quantification data were enrolled and followed up until October 31, 2022. Plasma PUFAs were quantified using a high-throughput nuclear magnetic resonance-based metabolic profiling platform. Liver cirrhosis complications were defined as hospitalization for liver cirrhosis or presentation with hepatocellular carcinoma. RESULTS During a median follow-up of 13.9 years, 2026 participants developed liver cirrhosis complications. Total plasma PUFAs, omega-3 PUFAs, docosahexaenoic acid (DHA), omega-6 PUFAs, and linoleic acid (LA) were inversely associated with the risk of liver cirrhosis complications, whereas the plasma omega-6/omega-3 ratio was positively associated. Nonparametrically restricted cubic spline regression showed nonlinear associations of plasma PUFAs with liver cirrhosis complications. The inflection points were 4.78 mmol/L for total PUFAs, 0.73 mmol/L for omega-3 PUFAs, 0.25 mmol/L for DHA, 4.07 mmol/L for omega-6 PUFAs, and 2.99 mmol/L for LA. Plasma omega-3 PUFAs were negatively associated with the risk of liver cirrhosis complications when omega-3 PUFAs were <0.73 mmol/L (adjusted hazard ratio [HR], 0.11 [0.08-0.16]), whereas the association was inverted when omega-3 PUFAs were ≥0.73 mmol/L (adjusted HR, 1.87 [1.20-2.92]). CONCLUSIONS The protective effect of plasma omega-3 PUFAs on liver cirrhosis complications is reversed after passing the corresponding inflection point, suggesting an optimal dietary omega-3 PUFA supplementation dose.
Collapse
Affiliation(s)
- Chengnan Guo
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China; Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Hong Fan
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Haili Wang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Xin Zhang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Shuzhen Zhao
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Yi Li
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Tianye Wang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Luojia Dai
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Jiayi Huang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China.
| | - Tiejun Zhang
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China; Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China.
| |
Collapse
|
7
|
Serbis A, Polyzos SA, Paschou SA, Siomou E, Kiortsis DN. Diet, exercise, and supplements: what is their role in the management of the metabolic dysfunction-associated steatotic liver disease in children? Endocrine 2024; 85:988-1006. [PMID: 38519764 DOI: 10.1007/s12020-024-03783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as nonalcoholic fatty liver disease (NAFLD), is the main cause of chronic liver disease in children and adolescents. Indeed, epidemiological studies have shown that MASLD affects up to 40% of children with obesity. Despite the recent approval of medications that target weight loss in adolescents that could have benefits on pediatric MASLD, lifestyle interventions, such as diet and exercise, remain the mainstay of our therapeutic approach. More specifically, studies on diet alone have focused on the possible role of carbohydrate or fat restriction, albeit without a definite answer on the best approach. Weight loss after dietary intervention in children with obesity and MASLD has a beneficial effect, regardless of the diet used. In relation to the role of exercise in MASLD reversal, indirect evidence comes from studies showing that a sedentary lifestyle leading to poor fitness, and low muscle mass is associated with MASLD. However, research on the direct effect of exercise on MASLD in children is scarce. A combination of diet and exercise seems to be beneficial with several studies showing improvement in surrogate markers of MASLD, such as serum alanine aminotransferase and hepatic fat fraction, the latter evaluated with imaging studies. Several dietary supplements, such as vitamin E, probiotics, and omega-3 fatty acid supplements have also been studied in children and adolescents with MASLD, but with equivocal results. This review aims to critically present available data on the effects of lifestyle interventions, including diet, exercise, and dietary supplements, on pediatric MASLD, thus suggesting a frame for future research that could enhance our knowledge on pediatric MASLD management and optimize clinicians' approach to this vexing medical condition.
Collapse
Affiliation(s)
- Anastasios Serbis
- Department of Pediatrics, School of Medicine, University of Ioannina, Ioannina, Greece.
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ekaterini Siomou
- Department of Pediatrics, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Dimitrios N Kiortsis
- Laboratory of Physiology, Medical School, University of Ioannina, Ioannina, Greece
| |
Collapse
|
8
|
Sabinari I, Horakova O, Cajka T, Kleinova V, Wieckowski MR, Rossmeisl M. Influence of Lipid Class Used for Omega-3 Fatty Acid Supplementation on Liver Fat Accumulation in MASLD. Physiol Res 2024; 73:S295-S320. [PMID: 39016154 PMCID: PMC11412347 DOI: 10.33549/physiolres.935396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) occurs in subjects with obesity and metabolic syndrome. MASLD may progress from simple steatosis (i.e., hepatic steatosis) to steatohepatitis, characterized by inflammatory changes and liver cell damage, substantially increasing mortality. Lifestyle measures associated with weight loss and/or appropriate diet help reduce liver fat accumulation, thereby potentially limiting progression to steatohepatitis. As for diet, both total energy and macronutrient composition significantly influence the liver's fat content. For example, the type of dietary fatty acids can affect the metabolism of lipids and hence their tissue accumulation, with saturated fatty acids having a greater ability to promote fat storage in the liver than polyunsaturated ones. In particular, polyunsaturated fatty acids of n-3 series (omega-3), such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), have been intensively studied for their antisteatotic effects, both in preclinical animal models of obesity and hepatic steatosis and in overweight/obese patients. Their effects may depend not only on the dose and duration of administration of omega-3, or DHA/EPA ratio, but also on the lipid class used for their supplementation. This review summarizes the available evidence from recent comparative studies using omega-3 supplementation via different lipid classes. Albeit the evidence is mainly limited to preclinical studies, it suggests that phospholipids and possibly wax esters could provide greater efficacy against MASLD compared to traditional chemical forms of omega-3 supplementation (i.e., triacylglycerols, ethyl esters). This cannot be attributed solely to improved EPA and/or DHA bioavailability, but other mechanisms may be involved. Keywords: MASLD • Metabolic dysfunction-associated steatotic liver disease • NAFLD • Non-alcoholic fatty liver disease • n-3 polyunsaturated fatty acids.
Collapse
Affiliation(s)
- I Sabinari
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
9
|
Pompili S, Cappariello A, Vetuschi A, Sferra R. G-Protein-Coupled Receptor 120 Agonist Mitigates Steatotic and Fibrotic Features Triggered in Obese Mice by the Administration of a High-Fat and High-Carbohydrate Diet. ACS OMEGA 2024; 9:31899-31909. [PMID: 39072106 PMCID: PMC11270546 DOI: 10.1021/acsomega.4c03507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024]
Abstract
Nonalcoholic fatty liver disease (NALFD) represents a complex condition ranging from simple steatosis (nonalcoholic fatty liver, NAFL) to inflammation, and fibrosis is one of the main features of nonalcoholic steatohepatitis (NASH). The pathogenesis of NAFLD is not well established but involves several factors (i.e., predisposition of genetic variants, obesity, and unhealthy lifestyle as unbalanced diets) that lead to an alteration of lipid homeostasis and consequently to an abnormal accumulation of triglycerides and other lipids in the liver parenchyma. Currently, no resolutive pharmacological treatment for NAFLD is available, and the only therapeutic approach is a healthy diet and physical exercise. In this study, we investigated the potential beneficial effect of GprA, a new synthetic agonist of G-protein-coupled receptor 120/free fatty acid receptor 4 (GPR120/FFAR4), in the progression of NAFL/NASH in mice fed for different periods (26 weeks and 30 weeks), with a high-fat (40% kcal) and high-carbohydrate diet, also called a Western-style diet (WSD). In our experimental model, the histological, protein, and transcriptomic analyses highlighted that the GprA can reduce signs of steatosis in WSD-fed mice. Furthermore, in 30 week-treated mice, GprA is also effective in the reduction of collagen deposition and fibrosis development. Altogether, our data validate the central role of FFAR4 in the context of NAFL/NASH onset and progression and reveal that GprA could represent an interesting candidate for the development of a new therapeutic approach in NAFLD treatment.
Collapse
Affiliation(s)
- Simona Pompili
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Alfredo Cappariello
- Department
of Life, Health and Experimental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Antonella Vetuschi
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Roberta Sferra
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
10
|
Li X, Liu C, Zhang R, Li Y, Ye D, Wang H, He M, Sun Y. Biosynthetic deficiency of docosahexaenoic acid causes nonalcoholic fatty liver disease and ferroptosis-mediated hepatocyte injury. J Biol Chem 2024; 300:107405. [PMID: 38788853 PMCID: PMC11231757 DOI: 10.1016/j.jbc.2024.107405] [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: 11/03/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Exogenous omega-3 fatty acids, particularly docosahexaenoic acid (DHA), have shown to exert beneficial effects on nonalcoholic fatty liver disease (NAFLD), which is characterized by the excessive accumulation of lipids and chronic injury in the liver. However, the effect of endogenous DHA biosynthesis on the lipid homeostasis of liver is poorly understood. In this study, we used a DHA biosynthesis-deficient zebrafish model, elovl2 mutant, to explore the effect of endogenously biosynthesized DHA on hepatic lipid homeostasis. We found the pathways of lipogenesis and lipid uptake were strongly activated, while the pathways of lipid oxidation and lipid transport were inhibited in the liver of elovl2 mutants, leading to lipid droplet accumulation in the mutant hepatocytes and NAFLD. Furthermore, the elovl2 mutant hepatocytes exhibited disrupted mitochondrial structure and function, activated endoplasmic reticulum stress, and hepatic injury. We further unveiled that the hepatic cell death and injury was mainly mediated by ferroptosis, rather than apoptosis, in elovl2 mutants. Elevating DHA content in elovl2 mutants, either by the introduction of an omega-3 desaturase (fat1) transgene or by feeding with a DHA-rich diet, could strongly alleviate NAFLD features and ferroptosis-mediated hepatic injury. Together, our study elucidates the essential role of endogenous DHA biosynthesis in maintaining hepatic lipid homeostasis and liver health, highlighting that DHA deficiency can lead to NAFLD and ferroptosis-mediated hepatic injury.
Collapse
Affiliation(s)
- Xuehui Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chengjie Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ru Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ding Ye
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Houpeng Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Mudan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghua Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.
| |
Collapse
|
11
|
Xue X, Wang L, Wu R, Li Y, Liu R, Ma Z, Jia K, Zhang Y, Li X. Si-Wu-Tang alleviates metabolic dysfunction-associated fatty liver disease by inhibiting ACSL4-mediated arachidonic acid metabolism and ferroptosis in MCD diet-fed mice. Chin Med 2024; 19:79. [PMID: 38844978 PMCID: PMC11157816 DOI: 10.1186/s13020-024-00953-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) is a prevalent chronic liver disease worldwide. Si-Wu-Tang (SWT), a traditional Chinese medicine decoction has shown therapeutic effects on various liver diseases. However, the hepatoprotective effects and underlying mechanism of SWT on MAFLD remain unclear. METHODS First, a methionine-choline-deficient (MCD) diet-fed mice model was used and lipidomic analysis and transcriptomic analysis were performed. The contents of total iron ions, ferrous ions, and lipid peroxidation were detected and Prussian blue staining was performed to confirm the protective effects of SWT against ferroptosis. Finally, chemical characterization and network pharmacological analysis were employed to identify the potential active ingredients. RESULTS Serological and hepatic histopathological findings indicated SWT's discernible therapeutic impact on MCD diet-induced MAFLD. Lipidomic analysis revealed that SWT improved intrahepatic lipid accumulation by inhibiting TG synthesis and promoting TG transport. Transcriptomic analysis suggested that SWT ameliorated abnormal FA metabolism by inhibiting FA synthesis and promoting FA β-oxidation. Then, ferroptosis phenotype experiments revealed that SWT could effectively impede hepatocyte ferroptosis, which was induced by long-chain acyl-CoA synthetase 4 (ACSL4)-mediated esterification of arachidonic acid (AA). Finally, chemical characterization and network pharmacological analysis identified that paeoniflorin and other active ingredients might be responsible for the regulative effects against ferroptosis and MAFLD. CONCLUSION In conclusion, our study revealed the intricate mechanism through which SWT improved MCD diet-induced MAFLD by targeting FA metabolism and ferroptosis in hepatocytes, thus offering a novel therapeutic approach for the treatment of MAFLD and its complications.
Collapse
Affiliation(s)
- Xiaoyong Xue
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Le Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ruiyu Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Yufei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Zhi Ma
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Kexin Jia
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yinhao Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
12
|
Bednarski TK, Rahim M, Hasenour CM, Banerjee DR, Trenary IA, Wasserman DH, Young JD. Pharmacological SERCA activation limits diet-induced steatohepatitis and restores liver metabolic function in mice. J Lipid Res 2024; 65:100558. [PMID: 38729350 PMCID: PMC11179628 DOI: 10.1016/j.jlr.2024.100558] [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: 01/25/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/12/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease is the most common form of liver disease and poses significant health risks to patients who progress to metabolic dysfunction-associated steatohepatitis. Fatty acid overload alters endoplasmic reticulum (ER) calcium stores and induces mitochondrial oxidative stress in hepatocytes, leading to hepatocellular inflammation and apoptosis. Obese mice have impaired liver sarco/ER Ca2+-ATPase (SERCA) function, which normally maintains intracellular calcium homeostasis by transporting Ca2+ ions from the cytoplasm to the ER. We hypothesized that restoration of SERCA activity would improve diet-induced steatohepatitis in mice by limiting ER stress and mitochondrial dysfunction. WT and melanocortin-4 receptor KO (Mc4r-/-) mice were placed on either chow or Western diet (WD) for 8 weeks. Half of the WD-fed mice were administered CDN1163 to activate SERCA, which reduced liver fibrosis and inflammation. SERCA activation also restored glucose tolerance and insulin sensitivity, improved histological markers of metabolic dysfunction-associated steatohepatitis, increased expression of antioxidant enzymes, and decreased expression of oxidative stress and ER stress genes. CDN1163 decreased hepatic citric acid cycle flux and liver pyruvate cycling, enhanced expression of mitochondrial respiratory genes, and shifted hepatocellular [NADH]/[NAD+] and [NADPH]/[NADP+] ratios to a less oxidized state, which was associated with elevated PUFA content of liver lipids. In sum, the data demonstrate that pharmacological SERCA activation limits metabolic dysfunction-associated steatotic liver disease progression and prevents metabolic dysfunction induced by WD feeding in mice.
Collapse
Affiliation(s)
- Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina A Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
13
|
Merenda T, Juszczak F, Ferier E, Duez P, Patris S, Declèves AÉ, Nachtergael A. Natural compounds proposed for the management of non-alcoholic fatty liver disease. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:24. [PMID: 38556609 PMCID: PMC10982245 DOI: 10.1007/s13659-024-00445-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
Although non-alcoholic fatty liver disease (NAFLD) presents as an intricate condition characterized by a growing prevalence, the often-recommended lifestyle interventions mostly lack high-level evidence of efficacy and there are currently no effective drugs proposed for this indication. The present review delves into NAFLD pathology, its diverse underlying physiopathological mechanisms and the available in vitro, in vivo, and clinical evidence regarding the use of natural compounds for its management, through three pivotal targets (oxidative stress, cellular inflammation, and insulin resistance). The promising perspectives that natural compounds offer for NAFLD management underscore the need for additional clinical and lifestyle intervention trials. Encouraging further research will contribute to establishing more robust evidence and practical recommendations tailored to patients with varying NAFLD grades.
Collapse
Affiliation(s)
- Théodora Merenda
- Unit of Clinical Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Florian Juszczak
- Department of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Elisabeth Ferier
- Department of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
- Unit of Therapeutic Chemistry and Pharmacognosy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Pierre Duez
- Unit of Therapeutic Chemistry and Pharmacognosy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Stéphanie Patris
- Unit of Clinical Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Anne-Émilie Declèves
- Department of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Amandine Nachtergael
- Unit of Therapeutic Chemistry and Pharmacognosy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium.
| |
Collapse
|
14
|
Balouei F, Stefanon B, Martello E, Atuahene D, Sandri M, Meineri G. Supplementation with Silybum marianum Extract, Synbiotics, Omega-3 Fatty Acids, Vitamins, and Minerals: Impact on Biochemical Markers and Fecal Microbiome in Overweight Dogs. Animals (Basel) 2024; 14:579. [PMID: 38396547 PMCID: PMC10886211 DOI: 10.3390/ani14040579] [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: 11/28/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Overweight and obese dogs can develop metabolic dysfunction, characterized by an inflammatory response and involvement of liver functions. If a modulation of the gut microbiome and its interaction with the gut-liver axis is implicated in the development of metabolic dysfunction, exploration becomes necessary. Over the past decade, diverse therapeutic approaches have emerged to target pathogenic factors involved in metabolic dysfunction. This study investigated the impact of a supplement with hepatoprotective activity, containing extracts of Silybum marianum, prebiotics, probiotics, n-3 polyunsaturated fatty acids, vitamins, and minerals on hematological markers of liver functions and inflammation, as well as on the intestinal microbiota of 10 overweight adult dogs over a 35-day time span. Animals underwent clinical and laboratory evaluations every 7 days, both before the administration of the supplement (T0) and after 7, 14, 21, 28, and 35 days (T1, T2, T3, T4, and T5). In comparison to T0, a significant (p < 0.05) decrease in ALP, glucose, direct bilirubin, and CRP was observed from T3 to T5. The alpha diversity of the fecal microbiota significantly decreased (p < 0.05) only at T1, with high variability observed between dogs. Total short-chain fatty acid and lactic acid were also lower at T1 (p < 0.05) compared to the other times of sampling. The beta diversity of the fecal microbiota failed to show a clear pattern in relation to the sampling times. These results of blood parameters in overweight dogs show a reduction of the inflammation and an improvement of metabolic status during the study period, but the effective contribution of the supplement in this clinical outcome deserves further investigation. Furthermore, the considerable individual variability observed in the microbiome hinders the confident detection of supplement effects.
Collapse
Affiliation(s)
- Fatemeh Balouei
- Department of Agrifood, Environmental and Animal Science, University of Udine, Via delle Scienze 206, 33100 Udine, Italy; (F.B.); (M.S.)
| | - Bruno Stefanon
- Department of Agrifood, Environmental and Animal Science, University of Udine, Via delle Scienze 206, 33100 Udine, Italy; (F.B.); (M.S.)
| | - Elisa Martello
- Division of Epidemiology and Public Health, School of Medicine, University of Nottingham, Nottingham City Hospital Campus, Nottingham NG5 1PB, UK;
| | - David Atuahene
- Department of Veterinary Sciences, School of Agriculture and Veterinary Medicine, University of Turin, Grugliasco, 10095 Turin, Italy; (D.A.); (G.M.)
| | - Misa Sandri
- Department of Agrifood, Environmental and Animal Science, University of Udine, Via delle Scienze 206, 33100 Udine, Italy; (F.B.); (M.S.)
| | - Giorgia Meineri
- Department of Veterinary Sciences, School of Agriculture and Veterinary Medicine, University of Turin, Grugliasco, 10095 Turin, Italy; (D.A.); (G.M.)
| |
Collapse
|
15
|
Liu H, Li N, Kuang G, Gong X, Wang T, Hu J, Du H, Zhong M, Guo J, Xie Y, Xiang Y, Wu S, Yuan Y, Yin X, Wan J, Li K. Protectin D1 inhibits TLR4 signaling pathway to alleviate non-alcoholic steatohepatitis via upregulating IRAK-M. Free Radic Biol Med 2024; 210:42-53. [PMID: 37984750 DOI: 10.1016/j.freeradbiomed.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a prevalent metabolic disease, characterized by the hepatic steatosis, inflammation, and fibrosis, which is lack of effective treatment currently. Protectin D1 (PTD1), a lipid mediator from omega-3 fatty acid docosahexaenoic acid (DHA), has displayed wide pharmacological actions including anti-inflammation in a variety of diseases, but the role of PTD1 on NASH remains unclear. In this study, using the methionine and choline deficient (MCD) fed NASH model, we explored the effect and underlying mechanism of PTD1 on NASH in mice. Our results showed PTD1 improved MCD-induced steatosis, hepatocellular injury, inflammation and fibrosis. Furthermore, PTD1 inhibited MCD-induced activation of TLR4 downstream molecules (TAK1, p38 and p65) without affecting the levels of TLR4 and phosphorylated IRAK-1. Notably, the levels of IRAK-M protein and the binding between IRAK-M and TRAF6 in the liver were also increased by PTD1 in NASH mice. Moreover, IRAK-M knockout remarkedly reverted the beneficial effects of PTD1 on the NASH in mice. Thus, these results demonstrated that PTD1 could protect mice from NASH by inhibiting the activation of TLR4 downstream signaling pathway, which might be related to the upregulation of IRAK-M, indicating that PTD1 may provide a new treatment for NASH.
Collapse
Affiliation(s)
- Hao Liu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Nana Li
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Ge Kuang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Ting Wang
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Hu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Hui Du
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Minxuan Zhong
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Jiashi Guo
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yao Xie
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yang Xiang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Shengwang Wu
- Department of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yiling Yuan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinru Yin
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Jingyuan Wan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China.
| | - Ke Li
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
16
|
Chen YF, Fan ZK, Gao X, Zhou F, Guo XF, Sinclair AJ, Li D. n-3 polyunsaturated fatty acids in phospholipid or triacylglycerol form attenuate nonalcoholic fatty liver disease via mediating cannabinoid receptor 1/adiponectin/ceramide pathway. J Nutr Biochem 2024; 123:109484. [PMID: 37866428 DOI: 10.1016/j.jnutbio.2023.109484] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/24/2023]
Abstract
n-3 polyunsaturated fatty acids (PUFA) have shown to exert beneficial effects in the treatment of nonalcoholic fatty liver disease (NAFLD). Supplements of n-3 PUFA occur in either phospholipid or triacylglycerol form. The present study aimed to compare whether the different n-3 PUFA of marine-origin, namely krill oil, DHA/EPA-phospholipid (PL), and EPA/DHA-triacylglycerol (TAG) forms had differential abilities to ameliorate NAFLD. The NAFLD model was established in mice fed a high-fat and high-cholesterol diet (HFD). The mice showed evidence of weight gain, dyslipidemia, insulin resistance and hepatic steatosis after 9 weeks of HFD, while the three forms of the n-3 PUFA reduced hepatic TAG accumulation, fatty liver and improved insulin instance, and hepatic biomarkers after 9 weeks of intervention. Of these, krill oil intervention significantly reduced adipocyte hypertrophy and hepatic steatosis in comparison with DHA/EPA-PL and EPA/DHA-TAG groups. Importantly, only krill oil intervention significantly reduced serum alanine transaminase, aspartate transaminase concentrations and low-density lipoprotein-cholesterol, compared with the HFD group. Supplemental n-3 PUFA lowered circulating anandamide (AEA) and 2-arachidonoylglycerol (2-AG) concentrations, compared with the HFD group, which was associated with down-regulating CB1 and upregulating adiponectin expressions in adipose tissue. Besides, targeted lipidomic analyses indicated that the increased adiponectin levels were accompanied by reductions in hepatic ceramide levels. The reduced ceramide levels were associated with inhibiting lipid synthesis and increasing fatty acid β-oxidation, finally inhibiting TAG accumulation in the liver. Through mediating CB1/adiponectin/ceramide pathway, the present study suggested that administration of krill oil had superior health effects in the therapy of NAFLD in comparison with DHA/EPA-PL and EPA/DHA-TAG.
Collapse
Affiliation(s)
- Yan-Fang Chen
- Institute of Nutrition & Health, Qingdao University, Qingdao, China; School of Public Health, Qingdao University, Qingdao, China
| | - Ze-Kai Fan
- Institute of Nutrition & Health, Qingdao University, Qingdao, China; School of Public Health, Qingdao University, Qingdao, China
| | - Xiang Gao
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Fang Zhou
- Qingdao University Function Center of Medical Nutrition, Qingdao, China
| | - Xiao-Fei Guo
- Institute of Nutrition & Health, Qingdao University, Qingdao, China; School of Public Health, Qingdao University, Qingdao, China.
| | - Andrew J Sinclair
- Department of Nutrition, Dietetics and Food, Monash University, Melbourne, Australia
| | - Duo Li
- Institute of Nutrition & Health, Qingdao University, Qingdao, China; Qingdao University Function Center of Medical Nutrition, Qingdao, China
| |
Collapse
|
17
|
Šmíd V, Dvořák K, Stehnová K, Strnad H, Rubert J, Stříteský J, Staňková B, Stránská M, Hajšlová J, Brůha R, Vítek L. The Ameliorating Effects of n-3 Polyunsaturated Fatty Acids on Liver Steatosis Induced by a High-Fat Methionine Choline-Deficient Diet in Mice. Int J Mol Sci 2023; 24:17226. [PMID: 38139055 PMCID: PMC10743075 DOI: 10.3390/ijms242417226] [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: 10/31/2023] [Revised: 12/02/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
The pathogenesis of non-alcoholic fatty liver disease (NAFLD) is associated with abnormalities of liver lipid metabolism. On the contrary, a diet enriched with n-3 polyunsaturated fatty acids (n-3-PUFAs) has been reported to ameliorate the progression of NAFLD. The aim of our study was to investigate the impact of dietary n-3-PUFA enrichment on the development of NAFLD and liver lipidome. Mice were fed for 6 weeks either a high-fat methionine choline-deficient diet (MCD) or standard chow with or without n-3-PUFAs. Liver histology, serum biochemistry, detailed plasma and liver lipidomic analyses, and genome-wide transcriptome analysis were performed. Mice fed an MCD developed histopathological changes characteristic of NAFLD, and these changes were ameliorated with n-3-PUFAs. Simultaneously, n-3-PUFAs decreased serum triacylglycerol and cholesterol concentrations as well as ALT and AST activities. N-3-PUFAs decreased serum concentrations of saturated and monounsaturated free fatty acids (FAs), while increasing serum concentrations of long-chain PUFAs. Furthermore, in the liver, the MCD significantly increased the hepatic triacylglycerol content, while the administration of n-3-PUFAs eliminated this effect. Administration of n-3-PUFAs led to significant beneficial differences in gene expression within biosynthetic pathways of cholesterol, FAs, and pro-inflammatory cytokines (IL-1 and TNF-α). To conclude, n-3-PUFA supplementation appears to represent a promising nutraceutical approach for the restoration of abnormalities in liver lipid metabolism and the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Václav Šmíd
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
| | - Karel Dvořák
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
| | - Kamila Stehnová
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, 166 28 Prague, Czech Republic; (K.S.); (J.R.); (J.H.)
| | - Hynek Strnad
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Josep Rubert
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, 166 28 Prague, Czech Republic; (K.S.); (J.R.); (J.H.)
| | - Jan Stříteský
- Institute of Pathology, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 00 Prague, Czech Republic;
| | - Barbora Staňková
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic
| | - Milena Stránská
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, 166 28 Prague, Czech Republic; (K.S.); (J.R.); (J.H.)
| | - Jana Hajšlová
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, 166 28 Prague, Czech Republic; (K.S.); (J.R.); (J.H.)
| | - Radan Brůha
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
| | - Libor Vítek
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic
| |
Collapse
|
18
|
Lakshimi VI, Kavitha M. New Insights into Prospective Health Potential of ω-3 PUFAs. Curr Nutr Rep 2023; 12:813-829. [PMID: 37996669 DOI: 10.1007/s13668-023-00508-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 11/25/2023]
Abstract
PURPOSE OF REVIEW Docosahexaenoic acid and eicosapentaenoic acid are the two essential long-chain ω-3 polyunsaturated fatty acids (ω-3 PUFAs) promoting human health which are obtained from diet or supplementation. The eicosanoids derived from ω-6 and ω-3 PUFAs have opposite characteristics of pro- and anti-inflammatory activities. The proinflammatory effects of ω-6 PUFAs are behind the pathology of the adverse health conditions of PUFA metabolism like cardiovascular diseases, neurological disorders, and inflammatory diseases. A balanced ω-6 to ω-3 ratio of 1-4:1 is critical to prevent the associated disorders. But due to modern agricultural practices, there is a disastrous shift in this ratio to 10-20:1. This review primarily aims to discuss the myriad health potentials of ω-3 PUFAs uncovered through recent research. It further manifests the importance of maintaining a balanced ω-6 to ω-3 PUFA ratio. RECENT FINDINGS ω-3 PUFAs exhibit protective effects against diabetes mellitus-associated complications including diabetic retinopathy, diabetic nephropathy, and proteinuria. COVID-19 is also not an exception to the health benefits of ω-3 PUFAs. Supplementation of ω-3 PUFAs improved the respiratory and clinical symptoms in COVID-19 patients. ω-3 PUFAs exhibit a variety of health benefits including anti-inflammatory property and antimicrobial property and are effective in protecting against various health conditions like atherosclerosis, cardiovascular diseases, diabetes mellitus, COVID-19, and neurological disorders. In the present review, various health potentials of ω-3 PUFAs are extensively reviewed and summarized. Further, the importance of a balanced ω-6 to ω-3 PUFA ratio has been emphasized besides stating the diverse sources of ω-3 PUFA.
Collapse
Affiliation(s)
- V Iswareya Lakshimi
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| | - M Kavitha
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
19
|
Padiadpu J, Garcia‐Jaramillo M, Newman NK, Pederson JW, Rodrigues R, Li Z, Singh S, Monnier P, Trinchieri G, Brown K, Dzutsev AK, Shulzhenko N, Jump DB, Morgun A. Multi-omic network analysis identified betacellulin as a novel target of omega-3 fatty acid attenuation of western diet-induced nonalcoholic steatohepatitis. EMBO Mol Med 2023; 15:e18367. [PMID: 37859621 PMCID: PMC10630881 DOI: 10.15252/emmm.202318367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023] Open
Abstract
Clinical and preclinical studies established that supplementing diets with ω3 polyunsaturated fatty acids (PUFA) can reduce hepatic dysfunction in nonalcoholic steatohepatitis (NASH) but molecular underpinnings of this action were elusive. Herein, we used multi-omic network analysis that unveiled critical molecular pathways involved in ω3 PUFA effects in a preclinical mouse model of western diet induced NASH. Since NASH is a precursor of liver cancer, we also performed meta-analysis of human liver cancer transcriptomes that uncovered betacellulin as a key EGFR-binding protein upregulated in liver cancer and downregulated by ω3 PUFAs in animals and humans with NASH. We then confirmed that betacellulin acts by promoting proliferation of quiescent hepatic stellate cells, inducing transforming growth factor-β2 and increasing collagen production. When used in combination with TLR2/4 agonists, betacellulin upregulated integrins in macrophages thereby potentiating inflammation and fibrosis. Taken together, our results suggest that suppression of betacellulin is one of the key mechanisms associated with anti-inflammatory and anti-fibrotic effects of ω3 PUFA on NASH.
Collapse
Affiliation(s)
| | | | - Nolan K Newman
- College of PharmacyOregon State UniversityCorvallisORUSA
| | - Jacob W Pederson
- Carlson College of Veterinary MedicineOregon State UniversityCorvallisORUSA
| | - Richard Rodrigues
- College of PharmacyOregon State UniversityCorvallisORUSA
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Zhipeng Li
- Carlson College of Veterinary MedicineOregon State UniversityCorvallisORUSA
| | - Sehajvir Singh
- College of PharmacyOregon State UniversityCorvallisORUSA
| | - Philip Monnier
- College of PharmacyOregon State UniversityCorvallisORUSA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Kevin Brown
- College of PharmacyOregon State UniversityCorvallisORUSA
- School of Chemical, Biological, and Environmental EngineeringOregon State UniversityCorvallisORUSA
| | - Amiran K Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Natalia Shulzhenko
- Carlson College of Veterinary MedicineOregon State UniversityCorvallisORUSA
| | - Donald B Jump
- Nutrition Program, School of Biological and Population Health Sciences, Linus Pauling InstituteOregon State UniversityCorvallisORUSA
| | - Andrey Morgun
- College of PharmacyOregon State UniversityCorvallisORUSA
| |
Collapse
|
20
|
Myint M, Oppedisano F, De Giorgi V, Kim BM, Marincola FM, Alter HJ, Nesci S. Inflammatory signaling in NASH driven by hepatocyte mitochondrial dysfunctions. J Transl Med 2023; 21:757. [PMID: 37884933 PMCID: PMC10605416 DOI: 10.1186/s12967-023-04627-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/14/2023] [Indexed: 10/28/2023] Open
Abstract
Liver steatosis, inflammation, and variable degrees of fibrosis are the pathological manifestations of nonalcoholic steatohepatitis (NASH), an aggressive presentation of the most prevalent chronic liver disease in the Western world known as nonalcoholic fatty liver (NAFL). Mitochondrial hepatocyte dysfunction is a primary event that triggers inflammation, affecting Kupffer and hepatic stellate cell behaviour. Here, we consider the role of impaired mitochondrial function caused by lipotoxicity during oxidative stress in hepatocytes. Dysfunction in oxidative phosphorylation and mitochondrial ROS production cause the release of damage-associated molecular patterns from dying hepatocytes, leading to activation of innate immunity and trans-differentiation of hepatic stellate cells, thereby driving fibrosis in NASH.
Collapse
Affiliation(s)
| | - Francesca Oppedisano
- Department of Health Sciences, Institute of Research for Food Safety and Health, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Valeria De Giorgi
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, USA
| | | | | | - Harvey J Alter
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, USA
| | - Salvatore Nesci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Italy.
| |
Collapse
|
21
|
Spooner MH, Garcia-Jaramillo M, Apperson KD, Löhr CV, Jump DB. Time course of western diet (WD) induced nonalcoholic steatohepatitis (NASH) in female and male Ldlr-/- mice. PLoS One 2023; 18:e0292432. [PMID: 37819925 PMCID: PMC10566735 DOI: 10.1371/journal.pone.0292432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a global health problem. Identification of factors contributing to the onset and progression of NAFLD have the potential to direct novel strategies to combat NAFLD. METHODS We examined the time course of western diet (WD)-induced NAFLD and its progression to nonalcoholic steatohepatitis (NASH) in age-matched female and male Ldlr-/- mice, with time-points at 1, 4, 8, 20 and 40 weeks on the WD. Controls included Ldlr-/- mice maintained on a purified low-fat diet (LFD) for 1 and 40 weeks. The approach included quantitation of anthropometric, plasma and liver markers of disease, plus hepatic histology, lipids, oxylipins, gene expression and selected metabolites. RESULTS One week of feeding the WD caused a significant reduction in hepatic essential fatty acids (EFAs: 18:2, ω6, 18:3, ω3) which preceded the decline in many C20-22 ω3 and ω6 polyunsaturated fatty acids (PUFA) and PUFA-derived oxylipins after 4 weeks on the WD. In addition, expression of hepatic inflammation markers (CD40, CD44, Mcp1, Nlrp3, TLR2, TLR4, Trem2) increased significantly in both female & male mice after one week on the WD. These markers continued to increase over the 40-week WD feeding study. WD effects on hepatic EFA and inflammation preceded all significant WD-induced changes in body weight, insulin resistance (HOMA-IR), oxidative stress status (GSH/GSSG ratio) and histological and gene expression markers of macrosteatosis, extracellular matrix remodeling and fibrosis. CONCLUSIONS Our findings establish that feeding Ldlr-/- mice the WD rapidly lowered hepatic EFAs and induced key inflammatory markers linked to NASH. Since EFAs have an established role in inflammation and hepatic inflammation plays a major role in NASH, we suggest that early clinical assessment of EFA status and correcting EFA deficiencies may be useful in reducing NASH severity.
Collapse
Affiliation(s)
- Melinda H. Spooner
- Nutrition Program, College of Health, Oregon State University, Corvallis, OR, United States of America
| | - Manuel Garcia-Jaramillo
- Environmental and Molecular Toxicology, Oregon State University, Corvallis OR, United States of America
| | - K. Denise Apperson
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States of America
| | - Christiane V. Löhr
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States of America
| | - Donald B. Jump
- Nutrition Program, College of Health, Oregon State University, Corvallis, OR, United States of America
| |
Collapse
|
22
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic fatty liver disease worldwide, particularly in obese and type 2 diabetic individuals. Currently, there are no therapies for NAFLD that have been approved by the US Food and Drug Administration. Herein, we examine the rationale for using ω3 polyunsaturated fatty acids (PUFAs) in NAFLD therapy. This focus is based on the finding that NAFLD severity is associated with a reduction of hepatic C20-22 ω3 PUFAs. Because C20-22 ω3 PUFAs are pleiotropic regulators of cell function, loss of C20-22 ω3 PUFAs has the potential to significantly impact hepatic function. We describe NAFLD prevalence and pathophysiology as well as current NAFLD therapies. We also present evidence from clinical and preclinical studies that evaluated the capacity of C20-22 ω3 PUFAs to treat NAFLD. Given the clinical and preclinical evidence, dietary C20-22 ω3 PUFA supplementation has the potential to decrease human NAFLD severity by reducing hepatosteatosis and liver injury.
Collapse
Affiliation(s)
- Melinda H Spooner
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA;
| | - Donald B Jump
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA;
| |
Collapse
|
23
|
Padiadpu J, Spooner MH, Li Z, Newman N, Löhr CV, Apperson KD, Dzutsev A, Trinchieri G, Shulzhenko N, Morgun A, Jump DB. Early transcriptome changes associated with western diet induced NASH in Ldlr-/- mice points to activation of hepatic macrophages and an acute phase response. Front Nutr 2023; 10:1147602. [PMID: 37609485 PMCID: PMC10440380 DOI: 10.3389/fnut.2023.1147602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/21/2023] [Indexed: 08/24/2023] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is a global health problem. Identifying early gene indicators contributing to the onset and progression of NAFLD has the potential to develop novel targets for early therapeutic intervention. We report on the early and late transcriptomic signatures of western diet (WD)-induced nonalcoholic steatohepatitis (NASH) in female and male Ldlr-/- mice, with time-points at 1 week and 40 weeks on the WD. Control Ldlr-/- mice were maintained on a low-fat diet (LFD) for 1 and 40 weeks. Methods The approach included quantitation of anthropometric and hepatic histology markers of disease as well as the hepatic transcriptome. Results Only mice fed the WD for 40 weeks revealed evidence of NASH, i.e., hepatic steatosis and fibrosis. RNASeq transcriptome analysis, however, revealed multiple cell-specific changes in gene expression after 1 week that persisted to 40 weeks on the WD. These early markers of disease include induction of acute phase response (Saa1-2, Orm2), fibrosis (Col1A1, Col1A2, TGFβ) and NASH associated macrophage (NAM, i.e., Trem2 high, Mmp12 low). We also noted the induction of transcripts associated with metabolic syndrome, including Mmp12, Trem2, Gpnmb, Lgals3 and Lpl. Finally, 1 week of WD feeding was sufficient to significantly induce TNFα, a cytokine involved in both hepatic and systemic inflammation. Conclusion This study revealed early onset changes in the hepatic transcriptome that develop well before any anthropometric or histological evidence of NALFD or NASH and pointed to cell-specific targeting for the prevention of disease progression.
Collapse
Affiliation(s)
- Jyothi Padiadpu
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Melinda H. Spooner
- Nutrition Program, Colleges of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| | - Zhipeng Li
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - Nolan Newman
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Christiane V. Löhr
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - K. Denise Apperson
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - Amiran Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NCI-NIH), Bethesda, MD, United States
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NCI-NIH), Bethesda, MD, United States
| | - Natalia Shulzhenko
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Donald B. Jump
- Nutrition Program, Colleges of Public Health and Human Sciences, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
24
|
Tabesh MR, Eghtesadi M, Abolhasani M, Maleklou F, Ejtehadi F, Alizadeh Z. Nutrition, Physical Activity, and Prescription of Supplements in Pre- and Post-bariatric Surgery Patients: An Updated Comprehensive Practical Guideline. Obes Surg 2023; 33:2557-2572. [PMID: 37389806 DOI: 10.1007/s11695-023-06703-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 07/01/2023]
Abstract
Only in the USA, 315 billion dollars are spent annually on the medical cost of obesity in adult patients. Till now, bariatric surgery is the most effective method for treating obesity and can play an essential role in reducing the direct and indirect costs of obesity treatment. Nonetheless, there are few comprehensive guidelines which include nutrition, physical activity, and supplements, before and after surgery. The purpose of the present narrative review is to provide an updated and comprehensive practical guideline to help multidisciplinary teams. The core keywords include nutrition, diet, physical activity, exercise, supplements, macronutrients, micronutrients, weight reduction, bariatric surgery, Roux-en-Y Gastric Bypass, Sleeve Gastrostomy, Laparoscopic Adjustable Gastric Banding, and Biliopancreatic diversion with duodenal switch which were searched in databases including PubMed/Medline, Cochrane, and some other sources such as Google Scholar. We answered questions in five important areas: (a) nutritional strategies before bariatric surgery, (b) nutrition after bariatric surgery, (c) physical activity before and after bariatric surgery, (d) weight regain after bariatric surgery, and (e) micronutrient assessments and recommendations before and after bariatric surgery. Some new items were added in this updated guideline including "weight regain" and "pregnancy after bariatric surgery." Other fields were updated based on new evidence and guidelines.
Collapse
Affiliation(s)
| | - Maryam Eghtesadi
- Sports and Exercise Medicine Department, Tehran University of Medical Sciences, No. 7, Ale-ahmad Highway, Tehran, 14395-578, Iran
| | - Maryam Abolhasani
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Maleklou
- Sports Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ejtehadi
- Sports Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Alizadeh
- Sports and Exercise Medicine Department, Tehran University of Medical Sciences, No. 7, Ale-ahmad Highway, Tehran, 14395-578, Iran.
| |
Collapse
|
25
|
Sandel P, Ma L, Wang H, Pasman EA. You Are What You Eat: A Review on Dietary Interventions for Treating Pediatric Nonalcoholic Fatty Liver Disease. Nutrients 2023; 15:3350. [PMID: 37571287 PMCID: PMC10421125 DOI: 10.3390/nu15153350] [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: 06/26/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
As the obesity pandemic worsens, cases of pediatric nonalcoholic fatty liver disease (NAFLD) and complications of this disease, such as progressive liver failure, in young adults will continue to rise. Lifestyle changes in the form of dietary modifications and exercise are currently first-line treatments. Large pediatric-specific randomized controlled trials to support specific interventions are currently lacking. A variety of dietary modifications in children with NAFLD have been suggested and studied with mixed results, including low-sugar and high-protein diets, the Mediterranean diet, and the Dietary Approach to Stop Hypertension (DASH). The roles of dietary supplements such as Vitamin E, polyunsaturated fatty acids (PUFAs), ginger, and probiotics have also been investigated. A further understanding of specific dietary interventions and supplements is needed to provide both generalizable and sustainable dietary recommendations to reverse the progression of NAFLD in the pediatric population.
Collapse
Affiliation(s)
- Piper Sandel
- Section of Academic General Pediatrics, Department of Pediatrics, University of California San Diego, San Diego, CA 92123, USA; (L.M.); (H.W.)
| | - Lawrence Ma
- Section of Academic General Pediatrics, Department of Pediatrics, University of California San Diego, San Diego, CA 92123, USA; (L.M.); (H.W.)
| | - Helen Wang
- Section of Academic General Pediatrics, Department of Pediatrics, University of California San Diego, San Diego, CA 92123, USA; (L.M.); (H.W.)
| | - Eric A. Pasman
- Division of Pediatric Gastroenterology, Department of Pediatrics, Naval Medical Center San Diego, San Diego, CA 92134, USA;
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
26
|
Šarac I, Debeljak-Martačić J, Takić M, Stevanović V, Milešević J, Zeković M, Popović T, Jovanović J, Vidović NK. Associations of fatty acids composition and estimated desaturase activities in erythrocyte phospholipids with biochemical and clinical indicators of cardiometabolic risk in non-diabetic Serbian women: the role of level of adiposity. Front Nutr 2023; 10:1065578. [PMID: 37545582 PMCID: PMC10397414 DOI: 10.3389/fnut.2023.1065578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Fatty acids (FAs) composition and desaturase activities can be altered in different metabolic conditions, but the adiposity-independent associations with clinical and biochemical indicators of cardiometabolic risk are still unclear. This study aimed to analyze the associations of FAs composition and estimated desaturase activities with anthropometric, clinical, and biochemical cardiometabolic risk indicators in non-diabetic Serbian women, and to investigate if these associations were independent of the level of adiposity and other confounders. Methods In 76 non-diabetic, otherwise healthy Serbian women, aged 24-68 years, with or without metabolic syndrome or obesity (BMI=23.6±5.6 kg/m2), FA composition in erythrocyte phospholipids was measured by gas-liquid chromatography. Desaturase activities were estimated from product/precursor FAs ratios (D9D:16:1n-7/16:0; D6D:20:3n-6/18:2n-6; D5D:20:4n-6/20:3n-6). Correlations were made with anthropometric, biochemical (serum glucose, triacylglycerols, LDL-C, HDL-C, ALT, AST, and their ratios) and clinical (blood pressure) indicators of cardiometabolic risk. Linear regression models were performed to test the independence of these associations. Results Estimated desaturase activities and certain FAs were associated with anthropometric, clinical and biochemical indicators of cardiometabolic risk: D9D, D6D, 16:1n-7 and 20:3n-6 were directly associated, while D5D and 18:0 were inversely associated. However, the associations with clinical and biochemical indicators were not independent of the associations with the level of adiposity, since they were lost after controlling for anthropometric indices. After controlling for multiple confounders (age, postmenopausal status, education, smoking, physical activity, dietary macronutrient intakes, use of supplements, alcohol consumption), the level of adiposity was the most significant predictor of desaturase activities and aforementioned FAs levels, and mediated their association with biochemical/clinical indicators. Vice versa, desaturase activities predicted the level of adiposity, but not other components of cardiometabolic risk (if the level of adiposity was accounted). While the associations of anthropometric indices with 16:1n-7, 20:3n-6, 18:0 and D9D and D6D activities were linear, the associations with D5D activity were the inverse U-shaped. The only adiposity-independent association of FAs profiles with the indicators of cardiometabolic risk was a positive association of 20:5n-3 with ALT/AST ratio, which requires further exploration. Discussion Additional studies are needed to explore the mechanisms of the observed associations.
Collapse
Affiliation(s)
- Ivana Šarac
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jasmina Debeljak-Martačić
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marija Takić
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vuk Stevanović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Milešević
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Zeković
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Popović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jovica Jovanović
- Department of Occupational Health, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Nevena Kardum Vidović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
27
|
Zhang P, Pan Y, Wu S, He Y, Wang J, Chen L, Zhang S, Zhang H, Zhao Y, Niu L, Gan M, Wang Y, Shen L, Zhu L. n-3 PUFA Promotes Ferroptosis in PCOS GCs by Inhibiting YAP1 through Activation of the Hippo Pathway. Nutrients 2023; 15:nu15081927. [PMID: 37111146 PMCID: PMC10145554 DOI: 10.3390/nu15081927] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/06/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder characterized by hyperandrogenemia with multiple suspended sinus follicles, thickened cortical tissue, and excessive proliferation of ovarian granulosa cells that severely affects the fertility and quality of life of women. The addition of n-3 PUFA to the diet may slightly reduce body weight and greatly alleviate disturbed blood hormone levels in PCOS mice. We treated KGN as a cell model for n-3 PUFA addition and showed that n-3 PUFA inhibited the proliferation of GCs and promoted ferroptosis in ovarian granulosa cells. We used CCK-8, fluorescence quantitative transmission electron microscopy experiments and ferroptosis marker gene detection and other methods. Furthermore, n-3 PUFA was found to promote YAP1 exocytosis by activating Hippo and weakening the cross-talk between YAP1 and Nrf2 by activating the Hippo signaling pathway. In this study, we found that n-3 PUFA inhibited the over proliferation of granulosa cells in ovarian follicles by activating Hippo, promoting YAP1 exocytosis, weakening the cross-talk between YAP1 and Nrf2, and ultimately activating the ferroptosis sensitivity of ovarian granulosa cells. We demonstrate that n-3 PUFA can alleviate the hormonal and estrous cycle disorder with PCOS by inhibiting the YAP1-Nrf2 crosstalk that suppresses over proliferating ovarian granulosa cells and promotes iron death in GCs. These findings reveal the molecular mechanisms by which n-3 PUFA attenuates PCOS and identify YAP1-Nrf2 as a potential therapeutic target for regulation granulosa cells in PCOS.
Collapse
Affiliation(s)
- Peiwen Zhang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuheng Pan
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shuang Wu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuxu He
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jinyong Wang
- Chongqing Academy of Animal Science, Chongqing 402460, China
| | - Lei Chen
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shunhua Zhang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hui Zhang
- Sichaun Center for Animal Disease Control, Chengdu 610041, China
| | - Ye Zhao
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lili Niu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mailin Gan
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Wang
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Linyuan Shen
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Zhu
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
28
|
Gong M, Lu H, Li L, Feng M, Zou Z. Integration of transcriptomics and metabonomics revealed the protective effects of hemp seed oil against methionine-choline-deficient diet-induced non-alcoholic steatohepatitis in mice. Food Funct 2023; 14:2096-2111. [PMID: 36734470 DOI: 10.1039/d2fo03054c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic liver disease with few therapeutic options available currently. Hemp seed oil extracted from the seeds of hemp (Cannabis sativa L.) has significant nutritional and biological properties due to the unique composition of polyunsaturated fatty acids and various antioxidant compounds. However, little is known about the beneficial effects and molecular mechanisms of hemp seed oil on NASH. Here, the hepatoprotective effects of hemp seed oil on methionine-choline-deficient (MCD) diet-induced NASH in C57BL/6 mice were explored via integration of transcriptomics and metabolomics. Hemp seed oil could improve hepatic steatosis, inflammation and fibrosis in mice with MCD diet-induced NASH. In a nuclear magnetic resonance (NMR)-based metabonomic study, the hepatic and urinary metabolic profiles of mice supplemented with hemp seed oil showed a tendency to recover to healthy controls compared to those of NASH mice. Eight potential biomarkers associated with NASH in both liver tissue and urine were restored to near normal levels by administration of hemp seed oil. The proposed pathways were mainly involved in pyrimidine metabolism, one-carbon metabolism, amino acid metabolism, glycolysis and the tricarboxylic acid (TCA) cycle. Hepatic transcriptomics based on Illumina RNA-Seq sequencing showed that hemp seed oil exerted anti-NASH activities by regulating multiple signaling pathways, e.g., downregulation of the TNF signaling pathway, the IL-17 signaling pathway, the MAPK signaling pathway and the NF-κB signaling pathway, which played a pivotal role in the pathogenesis of NASH. In particular, integration of metabonomic and transcriptomic results suggested that hemp seed oil could attenuate NASH-related liver fibrosis by inhibition of glutaminolysis. These results provided new insights into the hepatoprotective effects of hemp seed oil against MCD diet-induced NASH and hemp seed oil might have potential as an effective therapy for NASH.
Collapse
Affiliation(s)
- Mengjuan Gong
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Hailong Lu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lixi Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Meiqi Feng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhongjie Zou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
29
|
Monirujjaman M, Renani LB, Isesele P, Dunichand-Hoedl AR, Mazurak VC. Increased Expression of Hepatic Stearoyl-CoA Desaturase (SCD)-1 and Depletion of Eicosapentaenoic Acid (EPA) Content following Cytotoxic Cancer Therapy Are Reversed by Dietary Fish Oil. Int J Mol Sci 2023; 24:ijms24043547. [PMID: 36834959 PMCID: PMC9962117 DOI: 10.3390/ijms24043547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Cancer treatment evokes impediments to liver metabolism that culminate in fatty liver. This study determined hepatic fatty acid composition and expression of genes and mediators involved in lipid metabolism following chemotherapy treatment. Female rats bearing the Ward colon tumor were administered Irinotecan (CPT-11) +5-fluorouracil (5-FU) and maintained on a control diet or a diet containing eicosapentaenoic acid (EPA) + docosahexaenoic acid (DHA) (2.3 g/100 g fish oil). Healthy animals provided with a control diet served as a reference group. Livers were collected one week after chemotherapy. Triacylglycerol (TG), phospholipid (PL), ten lipid metabolism genes, leptin, and IL-4 were measured. Chemotherapy increased TG content and reduced EPA content in the liver. Expression of SCD1 was upregulated by chemotherapy, while dietary fish oil downregulated its expression. Dietary fish oil down-regulated expression of the fatty acid synthesis gene FASN, while restoring the long chain fatty acid converting genes FADS2 and ELOVL2, and genes involved in mitochondrial β-oxidation (CPT1α) and lipid transport (MTTP1), to values similar to reference animals. Neither leptin nor IL-4 were affected by chemotherapy or diet. Depletion of EPA is associated with pathways evoking enhanced TG accumulation in the liver. Restoring EPA through diet may pose a dietary strategy to attenuate chemotherapy-associated impediments in liver fatty acid metabolism.
Collapse
|
30
|
Chen J, Ruan X, Sun Y, Li X, Yuan S, Larsson SC. Plasma phospholipid arachidonic acid in relation to non-alcoholic fatty liver disease: Mendelian randomization study. Nutrition 2023; 106:111910. [PMID: 36459845 DOI: 10.1016/j.nut.2022.111910] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/11/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVES The role of plasma phospholipid arachidonic acid (AA) in the development of non-alcoholic fatty liver disease (NALFD), cirrhosis, and liver cancer remains unclear. This study aimed to determine the causality of the associations of plasma phospholipid AA with NALFD, cirrhosis, and liver cancer using Mendelian randomization analysis. METHODS Nine independent single-nucleotide polymorphisms associated with plasma phospholipid AA at the genome-wide significance were used as instrumental variables. Summary-level data for three outcomes were obtained from 1) a genome-wide association study for NAFLD, 2) the UK Biobank study, and 3) the FinnGen study. The sensitivity analysis excluding the pleiotropic variant rs174547 in the FADS1 gene was performed. Estimates from different sources were combined using the fixed-effects meta-analysis method. RESULTS Per standard deviation increase in AA levels, the combined odds ratio was 1.06 (95% confidence interval, 1.02-1.11; P = 0.008) for NAFLD, 1.05 (95% confidence interval, 1.01-1.09; P = 0.009) for cirrhosis, and 0.99 (95% confidence interval, 0.94-1.05; P = 0.765) for liver cancer. The associations remained stable in the sensitivity analysis excluding rs174547. CONCLUSIONS This study suggests potential causal associations of high levels of plasma phospholipid AA with the risk of NAFLD and cirrhosis.
Collapse
Affiliation(s)
- Jie Chen
- Center for Global Health, Zhejiang University School of Medicine, Hangzhou, China; Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xixian Ruan
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuhao Sun
- Center for Global Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Center for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
31
|
Jiang X, Yang Q, Qu H, Chen Y, Zhu S. Endogenous n-3 PUFAs Improve Non-Alcoholic Fatty Liver Disease through FFAR4-Mediated Gut-Liver Crosstalk. Nutrients 2023; 15:nu15030586. [PMID: 36771292 PMCID: PMC9919706 DOI: 10.3390/nu15030586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
The gut-liver axis plays a key role in the development and progression of non-alcoholic fatty liver disease (NAFLD). Due to the complexity and incomplete understanding of the cross-talk between the gut and liver, effective therapeutic targets are largely unknown. Free fatty acid receptors (FFARs) may bridge the cross-talk between the gut and liver. FFAR4 has received considerable attention due to its important role in lipid metabolism. However, the role of FFAR4 in this cross talk in NAFLD remains unclear. In this study, mice with high endogenous n-3 PUFAs but FFAR4 deficiency were generated by crossbreeding Fat-1 and FFAR4 knockout mice. FFAR4 deficiency blocked the protective effects of high endogenous n-3 PUFAs on intestinal barrier dysfunction and hepatic steatosis. In addition, FFAR4 deficiency decreased gut microbiota diversity and enriched Rikenella, Anaerotruncus, and Enterococcus, and reduced Dubosiella, Ruminococcaceae UCG-010, Ruminococcaceae UCG-014, Coriobacteriaceae UCG-002, Faecalibaculum, Ruminococcaceae UCG-009, and Akkermansia. Notably, FFAR4 deficiency co-regulated pantothenic acid and CoA biosynthesis, β-alanine metabolism, and sphingolipid metabolism pathways in the gut and liver, potentially associated with the aggravation of NAFLD. Together, the beneficial effects of n-3 PUFAs on the gut and liver were mediated by FFAR4, providing insights on the role of FFAR4 in the treatment of NAFLD through the gut-liver axis.
Collapse
Affiliation(s)
- Xuan Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qin Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Hongyan Qu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yongquan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and School of Translational Medicine, Jiangnan University, Wuxi 214122, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and School of Translational Medicine, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
32
|
Mao YJ, Ying MM, Xu G. Identification of hub genes and small molecule therapeutic drugs related to simple steatosis with secondary analysis of existing microarray data. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2114550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Yi-Jie Mao
- Department of Clinical Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Miao-Miao Ying
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Gang Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| |
Collapse
|
33
|
Ramadan NM, Elmasry K, Elsayed HRH, El-Mesery A, Eraky SM. The hepatoprotective effects of n3-polyunsaturated fatty acids against non-alcoholic fatty liver disease in diabetic rats through the FOXO1/PPARα/GABARAPL1 signalling pathway. Life Sci 2022; 311:121145. [DOI: 10.1016/j.lfs.2022.121145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
|
34
|
Xie F, Xu HF, Zhang J, Liu XN, Kou BX, Cai MY, Wu J, Dong JL, Meng QH, Wang Y, Chen D, Zhang Y. Dysregulated hepatic lipid metabolism and gut microbiota associated with early-stage NAFLD in ASPP2-deficiency mice. Front Immunol 2022; 13:974872. [PMID: 36466835 PMCID: PMC9716097 DOI: 10.3389/fimmu.2022.974872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/02/2022] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Growing evidence indicates that lipid metabolism disorders and gut microbiota dysbiosis were related to the progression of non-alcoholic fatty liver disease (NAFLD). Apoptosis-stimulating p53 protein 2 (ASPP2) has been reported to protect against hepatocyte injury by regulating the lipid metabolism, but the mechanisms remain largely unknown. In this study, we investigate the effect of ASPP2 deficiency on NAFLD, lipid metabolism and gut microbiota using ASPP2 globally heterozygous knockout (ASPP2+/-) mice. METHODS ASPP2+/- Balb/c mice were fed with methionine and choline deficient diet for 3, 10 and 40 day to induce an early and later-stage of NAFLD, respectively. Fresh fecal samples were collected and followed by 16S rRNA sequencing. HPLC-MRM relative quantification analysis was used to identify changes in hepatic lipid profiles. The expression level of innate immunity-, lipid metabolism- and intestinal permeability-related genes were determined. A spearman's rank correlation analysis was performed to identify possible correlation between hepatic medium and long-chain fatty acid and gut microbiota in ASPP2-deficiency mice. RESULTS Compared with the WT control, ASPP2-deficiency mice developed moderate steatosis at day 10 and severe steatosis at day 40. The levels of hepatic long chain omega-3 fatty acid, eicosapentaenoic (EPA, 20:5 n-3) and docosahexaenoic (DHA, 22:6 n-3), were decreased at day 10 and increased at day 40 in ASPP+/- mice. Fecal microbiota analysis showed significantly increased alpha and beta diversity, as well as the composition of gut microbiota at the phylum, class, order, family, genus, species levels in ASPP2+/- mice. Moreover, ASPP-deficiency mice exhibited impaired intestinal barrier function, reduced expression of genes associated with chemical barrier (REG3B, REG3G, Lysozyme and IAP), and increased expression of innate immune components (TLR4 and TLR2). Furthermore, correlation analysis between gut microbiota and fatty acids revealed that EPA was significantly negatively correlated with Bifidobacterium family. CONCLUSION Our findings suggested that ASPP2-deficiency promotes the progression of NAFLD, alterations in fatty acid metabolism and gut microbiota dysbiosis. The long chain fatty acid EPA was significantly negatively correlated with Bifidobacterial abundance, which is a specific feature of NAFLD in ASPP2-deficiency mice. Totally, the results provide evidence for a mechanism of ASPP2 on dysregulation of fatty acid metabolism and gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Fang Xie
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Hang-fei Xu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jing Zhang
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiao-ni Liu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Bu-xin Kou
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Meng-yin Cai
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Jing Wu
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jin-ling Dong
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Qing-hua Meng
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Yang Zhang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| |
Collapse
|
35
|
Sodum N, Rao V, Cheruku SP, Kumar G, Sankhe R, Kishore A, Kumar N, Rao CM. Amelioration of high-fat diet (HFD) + CCl4 induced NASH/NAFLD in CF-1 mice by activation of SIRT-1 using cinnamoyl sulfonamide hydroxamate derivatives: in-silico molecular modelling and in-vivo prediction. 3 Biotech 2022; 12:147. [PMID: 35720958 PMCID: PMC9200928 DOI: 10.1007/s13205-022-03192-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/28/2022] [Indexed: 11/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the major hepatic metabolic disorders that occurs because of the accumulation of lipids in hepatocytes in the form of free fatty acids (FFA) and triglycerides (TG) which become non-alcoholic steatohepatitis (NASH). NOTCH-1 receptors act as novel targets for the development of NAFLD/NASH, where overexpression of NOTCH-1 receptor alters the lipid metabolism in hepatocytes leading to NAFLD. SIRT-1 deacetylates the NOTCH-1 receptor and inhibits NAFLD. Hence, computer-aided drug design (CADD) was used to check the SIRT-1 activation ability of cinnamic sulfonyl hydroxamate derivatives (NMJ 1–8), resveratrol, and vorinostat. SIRT-1 (PDB ID: 5BTR) was docked with eight hydroxamate derivatives and vorinostat using Schrödinger software. Based on binding energy obtained (– 26.31 to – 47.34 kcal/mol), vorinostat, NMJ-2, NMJ-3, NMJ-5 were selected for induced-fit docking (IFD) and results were within – 750.70 to – 753.22 kcal/mol. Qikprop tool was used to analyse the pre pharmacokinetic parameters (ADME analysis) of all hydroxamate compounds. As observed in the molecular dynamic (MD) study, NMJ-2, NMJ-3 were showing acceptable results for activation of SIRT-1. Based on these predictions, in-vivo studies were conducted in CF1 mice, where NMJ-3 showed significant (p < 0.05) changes in lipid profile and anti-oxidant parameters (Catalase, SOD, GSH, nitrite, and LPO) and plasma insulin levels. NMJ-3 treatment also reduced inflammation, fibrosis, and necrosis in liver samples.
Collapse
Affiliation(s)
- Nalini Sodum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104 Karnataka India
| | - Vanishree Rao
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104 Karnataka India
| | - Sri Pragnya Cheruku
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104 Karnataka India
| | - Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104 Karnataka India
| | - Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104 Karnataka India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104 Karnataka India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Export Promotions Industrial Park (EPIP), Industrial Area Hajipur, Vaishali District, Hajipur, 844102 Bihar India
| | - C Mallikarjuna Rao
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104 Karnataka India
| |
Collapse
|
36
|
The Role of Insulin Resistance in Fueling NAFLD Pathogenesis: From Molecular Mechanisms to Clinical Implications. J Clin Med 2022; 11:jcm11133649. [PMID: 35806934 PMCID: PMC9267803 DOI: 10.3390/jcm11133649] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a predominant hepatopathy that is rapidly becoming the most common cause of hepatocellular carcinoma worldwide. The close association with metabolic syndrome’s extrahepatic components has suggested the nature of the systemic metabolic-related disorder based on the interplay between genetic, nutritional, and environmental factors, creating a complex network of yet-unclarified pathogenetic mechanisms in which the role of insulin resistance (IR) could be crucial. This review detailed the clinical and pathogenetic evidence involved in the NAFLD–IR relationship, presenting both the classic and more innovative models. In particular, we focused on the reciprocal effects of IR, oxidative stress, and systemic inflammation on insulin-sensitivity disruption in critical regions such as the hepatic and the adipose tissue, while considering the impact of genetics/epigenetics on the regulation of IR mechanisms as well as nutrients on specific insulin-related gene expression (nutrigenetics and nutrigenomics). In addition, we discussed the emerging capability of the gut microbiota to interfere with physiological signaling of the hormonal pathways responsible for maintaining metabolic homeostasis and by inducing an abnormal activation of the immune system. The translation of these novel findings into clinical practice could promote the expansion of accurate diagnostic/prognostic stratification tools and tailored pharmacological approaches.
Collapse
|
37
|
Šmíd V, Dvořák K, Šedivý P, Kosek V, Leníček M, Dezortová M, Hajšlová J, Hájek M, Vítek L, Bechyňská K, Brůha R. Effect of Omega-3 Polyunsaturated Fatty Acids on Lipid Metabolism in Patients With Metabolic Syndrome and NAFLD. Hepatol Commun 2022; 6:1336-1349. [PMID: 35147302 PMCID: PMC9134818 DOI: 10.1002/hep4.1906] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 12/15/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. n-3 polyunsaturated fatty acids (n-3-PUFAs) have been reported to ameliorate the progression of NAFLD in experimental studies; however, clinical trials have yielded contradictory results. The aim of our study was to assess the effects of n-3-PUFA administration on lipid metabolism and the progression of NAFLD in patients with metabolic syndrome. Sixty patients with metabolic syndrome and NAFLD were randomized in a double-blind placebo-controlled trial (3.6 g/day n-3-PUFA vs. placebo). During the 1-year follow-up, the patients underwent periodic clinical and laboratory examinations, liver stiffness measurements, magnetic resonance spectroscopy of the liver, and plasma lipidomic analyses. After 12 months of n-3-PUFA administration, a significant decrease in serum GGT activity was recorded compared with the placebo group (2.03 ± 2.8 vs. 1.43 ± 1.6; P < 0.05). Although no significant changes in anthropometric parameters were recorded, a significant correlation between the reduction of liver fat after 12 months of treatment-and weight reduction-was observed; furthermore, this effect was clearly potentiated by n-3-PUFA treatment (P < 0.005). In addition, n-3-PUFA treatment resulted in substantial changes in the plasma lipidome, with n-3-PUFA-enriched triacylglycerols and phospholipids being the most expressed lipid signatures. Conclusion: Twelve months of n-3-PUFA treatment of patients with NAFLD patients was associated with a significant decrease in GGT activity, the liver fat reduction in those who reduced their weight, and beneficial changes in the plasma lipid profile.
Collapse
Affiliation(s)
- Václav Šmíd
- Fourth Department of Internal MedicineFirst Faculty of Medicine and General University Hospital in PragueCharles UniversityPragueCzech Republic
| | - Karel Dvořák
- Fourth Department of Internal MedicineFirst Faculty of Medicine and General University Hospital in PragueCharles UniversityPragueCzech Republic
| | - Petr Šedivý
- Department of Diagnostic and Interventional RadiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Vít Kosek
- Department of Food Analysis and NutritionUniversity of Chemistry and TechnologyPragueCzech Republic
| | - Martin Leníček
- Institute of Medical Biochemistry and Laboratory DiagnosticsFirst Faculty of Medicine and General University Hospital in PragueCharles UniversityPragueCzech Republic
| | - Monika Dezortová
- Department of Diagnostic and Interventional RadiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Jana Hajšlová
- Department of Food Analysis and NutritionUniversity of Chemistry and TechnologyPragueCzech Republic
| | - Milan Hájek
- Department of Diagnostic and Interventional RadiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Libor Vítek
- Fourth Department of Internal MedicineFirst Faculty of Medicine and General University Hospital in PragueCharles UniversityPragueCzech Republic
- Institute of Medical Biochemistry and Laboratory DiagnosticsFirst Faculty of Medicine and General University Hospital in PragueCharles UniversityPragueCzech Republic
| | - Kamila Bechyňská
- Department of Food Analysis and NutritionUniversity of Chemistry and TechnologyPragueCzech Republic
| | - Radan Brůha
- Fourth Department of Internal MedicineFirst Faculty of Medicine and General University Hospital in PragueCharles UniversityPragueCzech Republic
| |
Collapse
|
38
|
Mitrovic M, Sistilli G, Horakova O, Rossmeisl M. Omega-3 phospholipids and obesity-associated NAFLD: Potential mechanisms and therapeutic perspectives. Eur J Clin Invest 2022; 52:e13650. [PMID: 34291454 DOI: 10.1111/eci.13650] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 02/06/2023]
Abstract
Prevalence of non-alcoholic fatty liver disease (NAFLD) increases in line with obesity and type 2 diabetes, and there is no approved drug therapy. Polyunsaturated fatty acids of n-3 series (omega-3) are known for their hypolipidaemic and anti-inflammatory effects. Existing clinical trials suggest varying effectiveness of triacylglycerol- or ethyl ester-bound omega-3 in the treatment of NAFLD, without affecting advanced stages such as non-alcoholic steatohepatitis. Preclinical studies suggest that the lipid class used to supplement omega-3 may determine the extent and nature of their effects on metabolism. Phospholipids of marine origin represent an alternative source of omega-3. The aim of this review is to summarise the available evidence on the use of omega-3 phospholipids, primarily in obesity-related NAFLD, and to outline perspectives of their use in the prevention/treatment of NAFLD. A PubMed literature search was conducted in May 2021. In total, 1088 articles were identified, but based on selection criteria, 38 original papers were included in the review. Selected articles describing the potential mechanisms of action of omega-3 phospholipids have also been included. Preclinical evidence clearly indicates that omega-3 phospholipids have strong antisteatotic effects in the liver, which are stronger compared to omega-3 administered as triacylglycerols. Multiple mechanisms are likely involved in the overall antisteatotic effects, involving not only the liver but also adipose tissue and the gut. Robust preclinical evidence for strong antisteatotic effects of omega-3 phospholipids in the liver should be confirmed in clinical trials. Further research is needed on the possible effects of omega-3 phospholipids on advanced NAFLD.
Collapse
Affiliation(s)
- Marko Mitrovic
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Gabriella Sistilli
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Olga Horakova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Rossmeisl
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
39
|
Tetrahydrocurcumin protects against nonalcoholic fatty liver disease by improving lipid metabolism and redox homeostasis. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
40
|
Jiang W, Li FR, Yang HH, Chen GC, Hua YF. Relationship Between Fish Oil Use and Incidence of Primary Liver Cancer: Findings From a Population-Based Prospective Cohort Study. Front Nutr 2022; 8:771984. [PMID: 35036409 PMCID: PMC8759152 DOI: 10.3389/fnut.2021.771984] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Background: N-3 long-chain polyunsaturated fatty acids (LCPUFAs) prevented non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) in studies of mouse models. We examined prospective relationships between fish oil use and risk of primary liver cancer and the major histological subtypes, such as HCC and intrahepatic cholangiocarcinoma (ICC). Methods: We included 434,584 middle-aged and older men and women who were free of cancer at recruitment of the UK Biobank (2006–2010). Information on fish oil use and other dietary habits was collected via questionnaires. Cox proportional hazards models were used to compute the hazard ratio (HR) and 95% CI of liver cancer associated with fish oil use, with adjustment for socio-demographic, lifestyle, dietary, and other clinical risk factors. Results: At baseline, 31.4% of participants reported regular use of fish oil supplements. During a median of 7.8 years of follow-up, 262 incident liver cancer cases were identified, among which 127 were HCC and 110 were ICC cases. As compared with non-users, fish oil users had a significantly 44% (95% CI: 25–59%) lower risk of total liver cancer, and 52% (95% CI: 24–70%) and 40% (95% CI: 7–61%) lower risk of HCC and ICC, respectively. Higher intake of oily fish also was associated with a lower risk of HCC (≥2 vs. <1 serving/week: HR = 0.46; 95% CI: 0.23–0.96; P-trend = 0.027) but not ICC (P-trend = 0.96). Conclusion: Habitual use of fish oil supplements was associated lower risk of primary liver cancer regardless of cancer histological subtypes, potentially supporting a beneficial role of dietary n-3 LCPUFAs in liver cancer prevention.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Hepatopancreatobiliary Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, China
| | - Fu-Rong Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.,Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Huan-Huan Yang
- Wanke School of Public Health, Tsinghua University, Beijing, China
| | - Guo-Chong Chen
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Yong-Fei Hua
- Department of Hepatopancreatobiliary Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, China
| |
Collapse
|
41
|
Cespiati A, Meroni M, Lombardi R, Oberti G, Dongiovanni P, Fracanzani AL. Impact of Sarcopenia and Myosteatosis in Non-Cirrhotic Stages of Liver Diseases: Similarities and Differences across Aetiologies and Possible Therapeutic Strategies. Biomedicines 2022; 10:biomedicines10010182. [PMID: 35052859 PMCID: PMC8773740 DOI: 10.3390/biomedicines10010182] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia is defined as a loss of muscle strength, mass and function and it is a predictor of mortality. Sarcopenia is not only a geriatric disease, but it is related to several chronic conditions, including liver diseases in both its early and advanced stages. Despite the increasing number of studies exploring the role of sarcopenia in the early stages of chronic liver disease (CLD), its prevalence and the relationship between these two clinical entities are still controversial. Myosteatosis is characterized by fat accumulation in the muscles and it is related to advanced liver disease, although its role in the early stages is still under researched. Therefore, in this narrative review, we firstly aimed to evaluate the prevalence and the pathogenetic mechanisms underlying sarcopenia and myosteatosis in the early stage of CLD across different aetiologies (mainly non-alcoholic fatty liver disease, alcohol-related liver disease and viral hepatitis). Secondly, due to the increasing prevalence of sarcopenia worldwide, we aimed to revise the current and the future therapeutic approaches for the management of sarcopenia in CLD.
Collapse
Affiliation(s)
- Annalisa Cespiati
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (A.C.); (M.M.); (G.O.); (P.D.); (A.L.F.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (A.C.); (M.M.); (G.O.); (P.D.); (A.L.F.)
| | - Rosa Lombardi
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (A.C.); (M.M.); (G.O.); (P.D.); (A.L.F.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-5503-4192; Fax: +39-02-5503-3509
| | - Giovanna Oberti
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (A.C.); (M.M.); (G.O.); (P.D.); (A.L.F.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (A.C.); (M.M.); (G.O.); (P.D.); (A.L.F.)
| | - Anna Ludovica Fracanzani
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (A.C.); (M.M.); (G.O.); (P.D.); (A.L.F.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
42
|
Mir RA, Nazir M, Naik S, Mukhtar S, Ganai BA, Zargar SM. Utilizing the underutilized plant resources for development of life style foods: Putting nutrigenomics to use. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2022; 171:128-138. [PMID: 34998100 DOI: 10.1016/j.plaphy.2021.12.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/26/2021] [Accepted: 12/30/2021] [Indexed: 06/14/2023]
Abstract
Sufficient amount of minerals, vitamins, and proteins in human diet play indispensable role in maintaining the active metabolism for better human health. All the essential nutrients that are requisite for an individual's survival are acquired from plants as well as animals. Micronutrients and macronutrients directly influence the metabolic pathways and their deficiencies play a substantial role in development of manifold disorders. In addition to environmental factors, quality and quantity of foods are key factors in maintaining the human health. Transition from healthy to diseased state is concurrent with the pattern of gene expression that is largely influenced by nutrition and environment. A combined approach to study the influence of nutrition on expression of numerous genes can be well explored through nutrigenomic studies. Nutrigenomics includes studies wherein applied genomics is used to investigate nutritional science to understand the compartmentalization of genes that influence the cause of diet-related complications. This review describes the role of underutilized crops as frontline foods to circumvent the health complications through the nutrigenomic studies. Further dynamics of nutrigenomic tools to study the impact of nutrition on the changing pattern of genome stability and gene expression for developing precise safety measures against wide range of health ailments linked to metabolic networks. Additionally, this review provides detailed information on nutrigenomic studies undertaken to unravel the potential of underutilized crops to augment the human health and to carry the agronomic/genomic approaches to enhance nutritional profile of underutilized crops to overcome diet-related disorders.
Collapse
Affiliation(s)
- Rakeeb Ahmad Mir
- Department of Biotechnology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185131, India
| | - Muslima Nazir
- Centre of Research for Development, University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India
| | - Samiullah Naik
- Proteomics Laboratory, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Jammu and Kashmir, 190025, India
| | - Shazia Mukhtar
- Proteomics Laboratory, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Jammu and Kashmir, 190025, India
| | - Bashir Ahmad Ganai
- Centre of Research for Development, University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India
| | - Sajad Majeed Zargar
- Proteomics Laboratory, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Jammu and Kashmir, 190025, India.
| |
Collapse
|
43
|
Plant and marine N3-PUFA regulation of fatty acid trafficking along the adipose tissue-liver axis varies according to nutritional state. J Nutr Biochem 2022; 102:108940. [PMID: 35017005 DOI: 10.1016/j.jnutbio.2022.108940] [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: 08/10/2021] [Revised: 11/26/2021] [Accepted: 12/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Marine sourced N3-PUFA regulate lipid metabolism in adipose tissue and liver; however, less is known about plant sourced N3-PUFA. The goal of this study was to investigate plant and marine N3-PUFA regulation of fatty acid trafficking along the adipose tissue-liver axis according to nutritional state. METHODS Mice were fed low-fat diets (7% w/w) containing either lard, flaxseed, or menhaden oils for 8 weeks, and were euthanized in either fed or fasted states. Substrate utilization and physical activity were assessed during the transition from a fed to fasted state. Plasma biomarkers (triglycerides (TAG), non-esterified fatty acids (NEFA)), as well as liver and epididymal adipose tissue (eWAT) lipogenic and lipolytic markers, were measured. RESULTS Neither plant nor marine N3-PUFA influenced substrate utilization or activity during the transition from a fed to fasted state. In the fed state, marine N3-PUFA reduced plasma TAG levels compared to the other diets, with no further reduction seen in fasted mice. Hepatic lipogenic markers (Fasn, Acc, Scd1, and Elovl6) were reduced in the fed state with marine N3-PUFA, but not plant N3-PUFA. In the fasted state, mice fed either N3-PUFA accumulated less liver TAG, had lower plasma NEFA, and suppressed eWAT HSL activity compared to lard. CONCLUSION Marine N3-PUFA are more potent regulators of lipogenesis than plant N3-PUFA in the fed state, whereas both N3-PUFA influence eWAT lipolysis and plasma NEFA in the fasted state. This work provides novel insights regarding N3-PUFA regulation of fatty acid trafficking along the adipose tissue-liver axis according to nutritional state.
Collapse
|
44
|
Yang Y, Li S, Xu Y, Ke J, Zhao D. The Perirenal Fat Thickness Was Associated with Nonalcoholic Fatty Liver Disease in Patients with Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2022; 15:1505-1515. [PMID: 35586202 PMCID: PMC9109981 DOI: 10.2147/dmso.s350579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Obesity is an important risk factor for nonalcoholic fatty liver disease (NAFLD). Perirenal fat and paranephric fat were seldom studied in NAFLD. We aimed to explore the relationship between perirenal fat thickness (PrFT) and paranephric fat thickness (PnFT) and NAFLD in patients with type 2 diabetes mellitus (T2DM). PATIENTS AND METHODS A total of 493 diabetic patients including 231 NAFLD patients were enrolled in our study from September 2019 to December 2020. Patients with NAFLD were categorized into three subgroups according to the severity and fibrosis risk of NAFLD. Anthropometric indices and clinical characteristics were collected from clinical records. PrFT and PnFT were measured via ultrasound. Multivariate logistic regression analysis was used to assess the association between PrFT, PnFT and presence, severity and advanced fibrosis risk of NAFLD. RESULTS Compared with non-NAFLD patients, those with NAFLD had significantly higher PrFT and PnFT. The PrFT and PnFT were independently associated with presence of NAFLD and the PrFT was independently associated with the advanced fibrosis risk of NAFLD after adjusting confounding factors. CONCLUSION The PrFT was independently associated with the presence and advanced fibrosis risk of NAFLD in patients with T2DM.
Collapse
Affiliation(s)
- Yuxian Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Shuting Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yuechao Xu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Jing Ke; Dong Zhao, Email ;
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
45
|
Kytikova OY, Novgorodtseva TP, Denisenko YK, Antonyuk MV, Gvozdenko TA. Associations Of Delta Fatty Acid Desaturase Gene Polymorphisms With Lipid Metabolism Disorders. RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Overweight, obesity, type 2 diabetes mellitus, metabolic syndrome, cardiovascular diseases, and non-alcoholic fatty liver disease are common chronic ailments associated with lipid metabolism disorders. One of the mechanisms of these disorders is related to the deficiency and/or change in the balance of essential fatty acids (FAs). At the same time, the provision of ω3 and ω6 polyunsaturated fatty acids (PUFAs) depends, besides sufficient dietary intake, on efficiency of their endogenous biosynthesis by desaturation and elongation processes regulated by FA elongases and FA desaturases. Desaturases are encoded by PUFA desaturase genes (FADSs): FADS1 and FADS2. Alteration of FA desaturase activity and single nucleotide polymorphisms (SNPs) in the FADS1 and FADS2 gene cluster are associated with lipid metabolism dysfunction and may affect the pathogenesis of lipid-related diseases. People of different ages, from different ethnic backgrounds and countries may exhibit varying degrees of response to dietary supplements of ω3 and ω6 PUFAs. The study of the relationship between lipid metabolism disorders and genetic factors controlling FA metabolism is an important research area since the health effects of alimentary ω3 and ω6 PUFAs can depend on genetic variants of the FADS genes. This review summarizes the literature data on the association of FADS gene polymorphisms with lipid metabolism disorders and their role in the development of chronic non-communicable pathologies associated with changes in lipid metabolism.
Collapse
Affiliation(s)
- Oksana Yu. Kytikova
- Research Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | | | - Yulia K. Denisenko
- Research Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | - Marina V. Antonyuk
- Research Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | - Tatyana A. Gvozdenko
- Research Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| |
Collapse
|
46
|
Oppedisano F, Bulotta RM, Maiuolo J, Gliozzi M, Musolino V, Carresi C, Ilari S, Serra M, Muscoli C, Gratteri S, Palma E, Mollace V. The Role of Nutraceuticals in Osteoarthritis Prevention and Treatment: Focus on n-3 PUFAs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4878562. [PMID: 34925695 PMCID: PMC8683171 DOI: 10.1155/2021/4878562] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/30/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a disease caused by joint degeneration with massive cartilage loss, and obesity is among the risk factors for its onset, though the pathophysiological mechanisms underlying the disease and better therapeutic approach still remain to be assessed. In recent years, several nutraceutical interventions have been investigated in order to define better solutions for preventing and treating OA. Among them, polyunsaturated fatty acids (n-3 PUFAs) appear to represent potential candidates in counteracting OA and its consequences, due to their anti-inflammatory, antioxidant, and chondroinductive effects. PUFAs have been found to counteract the onset and progression of OA by reducing bone and cartilage destruction, inhibiting proinflammatory cytokine release, reactive oxygen species (ROS) generation, and the NF-κB pathway's activation. Moreover, a diet rich in n-3 PUFAs and their derivatives (maresins and resolvins) demonstrates beneficial effects on associated pain reduction. Finally, it has been shown that together with the anti-inflammatory and antioxidant properties of eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids, their antiapoptotic and antiangiogenic effects contribute in reducing OA development. The present review is aimed at assessing evidence suggesting the potential benefit of nutraceutical supplementation with PUFAs in OA management according to their efficacy in targeting relevant pathophysiological mechanisms responsible for inflammation and joint destruction processes, and this may represent a novel and potentially useful approach in OA prevention and treatment. For that purpose, a PubMed literature survey was conducted with a focus on some in vitro and in vivo studies and clinical trials from 2015 to 2020.
Collapse
Affiliation(s)
- Francesca Oppedisano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Rosa Maria Bulotta
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Micaela Gliozzi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Cristina Carresi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Sara Ilari
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Maria Serra
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Carolina Muscoli
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Santo Gratteri
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Ernesto Palma
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy
| |
Collapse
|
47
|
Mirashrafi S, Kafeshani M, Hassanzadeh A, Entezari MH. Is any association between alternate healthy eating index (AHEI) with lipid profile and liver enzymes? A cross-sectional Study. J Diabetes Metab Disord 2021; 20:1537-1544. [PMID: 34900806 PMCID: PMC8630323 DOI: 10.1007/s40200-021-00898-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 09/06/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Based on evidence, there is a significant relationship between diet quality and the chronic conditions such as dyslipidemia and liver disorders. In the current study, we investigated the relationship between alternative healthy eating index (AHEI) and some biochemical measurements (lipid profile and liver enzymes). METHODS In this cross-sectional study, 127 hospital employees were randomly selected (of 250 screened volunteers). AHEI was estimated based on a 168 items food frequency questionnaire. The laboratory data [triglyceride (TG), total cholesterol (TC), high density lipoprotein (HDL), low density lipoprotein (LDL), alanine transaminase (ALT), aspartate transaminase (AST)] were extracted from periodic examinations. The physical activity was also estimated with international physical activity questionnaire (IPAQ). RESULTS Among 127 completed data, the mean AHEI score was 47.1. There was a direct relationship between AHEI score and HDL (P = 0.02). Among the AHEI components, the score of nuts and soybeans was directly associated with HDL level (P = 0.007). There was no association between the other AHEI components and selected biomarkers. CONCLUSION Nuts and soybeans consumption-one unit per day-may increase HDL levels. More studies are needed to evaluate the diet quality with the other dietary indices. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s40200-021-00898-w.
Collapse
Affiliation(s)
- Shahrzad Mirashrafi
- Department of Clinical Nutrition, School of Nutrition & Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Kafeshani
- Department of Clinical Nutrition, School of Nutrition & Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Hezar-Jerib Ave, P.O. Box 319, 8174673461 Isfahan, Iran
| | - Akbar Hassanzadeh
- Department of Epidemiology and Biostatistics, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hassan Entezari
- Department of Clinical Nutrition, School of Nutrition & Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Hezar-Jerib Ave, P.O. Box 319, 8174673461 Isfahan, Iran
| |
Collapse
|
48
|
Shan Z, Nisar MF, Li M, Zhang C, Wan C(C. Theaflavin Chemistry and Its Health Benefits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6256618. [PMID: 34804369 PMCID: PMC8601833 DOI: 10.1155/2021/6256618] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 02/07/2023]
Abstract
Huge epidemiological and clinical studies have confirmed that black tea is a rich source of health-promoting ingredients, such as catechins and theaflavins (TFs). Furthermore, TF derivatives mainly include theaflavin (TF1), theaflavin-3-gallate (TF2A), theaflavin-3'-gallate (TF2B), and theaflavin-3,3'-digallate (TF3). All of these TFs exhibit extensive usages in pharmaceutics, foods, and traditional medication systems. Various indepth studies reported that how TFs modulates health effects in cellular and molecular mechanisms. The available literature regarding the pharmacological activities of TFs has revealed that TF3 has remarkable anti-inflammatory, antioxidant, anticancer, antiobesity, antiosteoporotic, and antimicrobial properties, thus posing significant effects on human health. The current manuscript summarizes both the chemistry and various pharmacological effects of TFs on human health, lifestyle or aging associated diseases, and populations of gut microbiota. Furthermore, the biological potential of TFs has also been focused to provide a deeper understanding of its mechanism of action.
Collapse
Affiliation(s)
- Zhiguo Shan
- College of Agriculture and Forestry, Pu'er University, Pu'er 665099, China
| | - Muhammad Farrukh Nisar
- Jiangxi Key Laboratory for Postharvest Technology and Nondestructive Testing of Fruits & Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur 63100, Pakistan
| | - Mingxi Li
- Research Center of Tea and Tea Culture, College of Agronomy, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Chunhua Zhang
- College of Agriculture and Forestry, Pu'er University, Pu'er 665099, China
| | - Chunpeng (Craig) Wan
- Jiangxi Key Laboratory for Postharvest Technology and Nondestructive Testing of Fruits & Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
49
|
Yki-Järvinen H, Luukkonen PK, Hodson L, Moore JB. Dietary carbohydrates and fats in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2021; 18:770-786. [PMID: 34257427 DOI: 10.1038/s41575-021-00472-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2021] [Indexed: 02/06/2023]
Abstract
The global prevalence of nonalcoholic fatty liver disease (NAFLD) has dramatically increased in parallel with the epidemic of obesity. Controversy has emerged around dietary guidelines recommending low-fat-high-carbohydrate diets and the roles of dietary macronutrients in the pathogenesis of metabolic disease. In this Review, the topical questions of whether and how dietary fats and carbohydrates, including free sugars, differentially influence the accumulation of liver fat (specifically, intrahepatic triglyceride (IHTG) content) are addressed. Focusing on evidence from humans, we examine data from stable isotope studies elucidating how macronutrients regulate IHTG synthesis and disposal, alter pools of bioactive lipids and influence insulin sensitivity. In addition, we review cross-sectional studies on dietary habits of patients with NAFLD and randomized controlled trials on the effects of altering dietary macronutrients on IHTG. Perhaps surprisingly, evidence to date shows no differential effects between free sugars, with both glucose and fructose increasing IHTG in the context of excess energy. Moreover, saturated fat raises IHTG more than polyunsaturated or monounsaturated fats, with adverse effects on insulin sensitivity, which are probably mediated in part by increased ceramide synthesis. Taken together, the data support the use of diets that have a reduced content of free sugars, refined carbohydrates and saturated fat in the treatment of NAFLD.
Collapse
Affiliation(s)
- Hannele Yki-Järvinen
- Department of Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland. .,Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Panu K Luukkonen
- Department of Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland.,Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
| | | |
Collapse
|
50
|
Li Y, Gan M, Tang T, Shao J, Lai T, Ma Y, Elzo MA, Jia X, Hu S, Wang J, Lai S. Intramuscular adipocyte and fatty acid differences between high-fat and control rabbit groups subject to a restricted diet. Vet Med Sci 2021; 7:2051-2060. [PMID: 34273256 PMCID: PMC8464271 DOI: 10.1002/vms3.576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Fatty acids of intramuscular fat (IMF) in rabbits can influence meat quality, but it is unclear which fatty acids benefit to human health. A rabbit model of weight gain and weight loss was constructed using two rabbit groups and two growth stages. Stage 1 included control group1 fed a commercial diet(CG1) and experimental group1 fed a high fat diet (EG1). Stage 2 include control group2(CG2) and experimental group2 (EG2) both fed a restricted commercial diet. We detected differences in blood biochemical indicators as well as changes in intramuscular adipose cells and intramuscular fatty acid content in control and experiment groups at two stages. High fat induction can make rabbits become obese, have higher concentrations of glucose (GLU), total cholesterol (TC), triglyceride (TG), low density lipoprotein-cholesterol (LDL-C) and free fatty acid (FFA), and lower concentrations of insulin (INS). In addition, a high-fat diet promotes hypertrophy of precursor adipocytes in femoral muscles. Conversely, a restricted diet causes weight loss, decreases the concentration of TG, FFA, and INS in CG2 and EG2, and increases the deposition of unsaturated fatty acids in the femoral muscle. The content of monounsaturated trans oleic acid (C18:1n-9T) in EG2 was significantly higher than in CG2, whereas oleic acid (C18:1n-9C) was significantly lower in EG2 than in CG2. The polyunsaturated fatty acids Linolenate (C18:3 n-3) and cis-5,8,11,14,17-Eicosapentaenoate (C20:5 n-3) increased in CG2 and EG2. The content of Linoleate (C18:2 n-6) and γ-Linolenic acid (C18:3 n-6) significantly increased in CG2. The content of cis-11,14-Eicosatrienoic acid (C20:2) decreased significantly in CG2, but increased significantly in EG2.Thus, a high-fat diet can increase the formation of unhealthy fatty acids. Conversely, weight loss due to a restricted diet leads to an increase in unsaturated fatty acids in the femoral muscle, indicating that it reduces obesity symptoms and it may improve meat quality in rabbit.
Collapse
Affiliation(s)
- YanHong Li
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - Mingchuan Gan
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - Tao Tang
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - Jiahao Shao
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - Tianfu Lai
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - Yuan Ma
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - Mauricio A Elzo
- Department of Animal Sciences, University of Florida, Gainesville, Florida, USA
| | - Xianbo Jia
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - Shenqiang Hu
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - Jie Wang
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| | - SongJia Lai
- College of Animal Science and Technology, Sichuan Agricultural University-Chengdu Campus, Chengdu, Sichuan, P. R. China
| |
Collapse
|