1
|
Li X, Wang Y, Liu J, Gao T, Cao L, Yan M, Li N. Dysregulation of the SREBP pathway is associated with poor prognosis and serves as a potential biomarker for the diagnosis of hepatocellular carcinoma. Mol Med Rep 2025; 31:112. [PMID: 40017126 PMCID: PMC11894594 DOI: 10.3892/mmr.2025.13477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 12/20/2024] [Indexed: 03/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a severe disease associated with a poor prognosis. The role of aberrant lipid metabolism in the development and progression of HCC necessitates detailed characterization. Sterol regulatory element‑binding proteins (SREBPs), pivotal transcription factors governing lipogenesis, are central to this process. The present study aimed to assess the regulation of HCC by the SREBP signaling pathway, examining the expression levels of genes in this pathway, the clinical implications and its prognostic value using the Kaplan‑Meier method. Pearson's correlation coefficient was used to identify the co‑expression of SREBP pathway genes in HCC. Genomic analysis examined the frequency of TP53 mutations in groups with and without SREBP pathway alterations. In addition, small interfering RNAs targeting genes of the SREBP pathway were transfected into Huh‑7 and HCC‑LM3 cell lines. Subsequently, Cell Counting Kit‑8 and Transwell assays were carried out to evaluate the viability and invasion of these cells. Reverse transcription‑quantitative PCR and western blotting were performed to investigate the expression of TP53 in response to silencing of SREBP pathway genes. Dysregulation of SREBP pathway genes was detected in HCC tissues compared with in normal liver tissues, and predicted a poor prognosis. Silencing these genes reduced the viability and invasion of HCC cells. Furthermore, abnormal SREBP pathway gene expression was associated with poor survival rates, vascular invasion, advanced tumor stage and an increased incidence of TP53 mutations. By contrast, knockdown of SREBP pathway genes decreased mutant TP53 expression at both the mRNA and protein levels in HCC cells. The findings of the present study suggested that SREBP pathway genes could serve as promising prognostic biomarkers for HCC. The combined analysis of individual gene expression levels offers offer novel insights into the pathogenesis and progression of HCC.
Collapse
Affiliation(s)
- Xiaodan Li
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
- Scientific Research Department, Shanghai University of Medicine amd Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Yuhan Wang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Junchi Liu
- Scientific Research Department, Shanghai University of Medicine amd Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Tianmiao Gao
- Scientific Research Department, Shanghai University of Medicine amd Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Lizhi Cao
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Meng Yan
- Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Na Li
- Scientific Research Department, Shanghai University of Medicine amd Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
2
|
Frankhouser DE, DeWees T, Snodgrass IF, Cole RM, Steck S, Thomas D, Kalu C, Belury MA, Clinton SK, Newman JW, Yee LD. Randomized dose-response trial of n-3 fatty acids in hormone receptor negative breast cancer survivors - impact on breast adipose oxylipin and DNA methylation patterns. Am J Clin Nutr 2025:S0002-9165(25)00239-4. [PMID: 40288580 DOI: 10.1016/j.ajcnut.2025.04.021] [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: 10/31/2024] [Revised: 03/30/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Increasing evidence suggests the unique susceptibility of estrogen receptor and progesterone receptor negative [ERPR(-)] breast cancer to dietary fat amount and type. Dietary n-3 (ω-3) polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), may modulate breast adipose fatty acids and downstream metabolites to counteract procarcinogenic signaling in the mammary microenvironment. OBJECTIVES We aimed to determine effects of ∼1 to 5 g/d EPA+DHA over 12 mo on breast adipose fatty acid and oxylipin profiles in survivors of ERPR(-) breast cancer, a high-risk molecular subtype. METHODS We conducted a proof-of-concept 12-mo randomized double-blind trial comparing ∼5 g/d and ∼1 g/d EPA+DHA supplementation in females within 5 y of completing standard therapy for ERPR(-) breast cancer Stages 0 to III. Blood and breast adipose tissue specimens were collected every 3 mo for fatty acid, oxylipin, and DNA methylation (DNAm) analyses. RESULTS A total of 51 participants completed the 12-mo intervention. Study treatments were generally well tolerated. Although both doses increased n-3 PUFAs from baseline in breast adipose, erythrocytes, and plasma, the 5 g/d supplement was more potent with differences (% total fatty acids) of 0.76 (95% confidence interval [CI]: 0.56, 0.96), 6.25 (95% CI: 5.02, 7.48), and 5.89 (95% CI: 4.53, 7.25), respectively. The 5 g/d dose also reduced plasma triglycerides from baseline, with changes (mg/dL) of 27.38 (95% CI: 10.99, 43.78) and 24.58 (95% CI: 9.05, 40.10) at 6 and 12 months, respectively. Breast adipose oxylipins showed dose-dependent increases in DHA and EPA metabolites. Distinct DNAm patterns in adipose tissue after 12 mo suggest potential downregulation of aberrant lipid metabolism pathways at the 5 g/d dose. CONCLUSIONS Over 1 y, EPA+DHA dose-dependently increased breast adipose concentrations of these fatty acids and their derivative oxylipin metabolites and produced differential DNAm profiles involved in metabolism-related pathways critical to ERPR(-) breast cancer development. This distinct metabolic and epigenetic modulation of the breast microenvironment is achievable with high-dose n-3 PUFA supplementation. This trial was registered at clinicaltrials.gov as NCT02295059.
Collapse
Affiliation(s)
- David E Frankhouser
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, CA
| | - Todd DeWees
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, CA; Department of Surgery, City of Hope, Duarte, CA
| | - Isabel F Snodgrass
- University of California Davis West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA
| | - Rachel M Cole
- Department of Food Science and Technology, The Ohio State University, Columbus, OH
| | - Sarah Steck
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | | | - Martha A Belury
- Department of Food Science and Technology, The Ohio State University, Columbus, OH; The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Steven K Clinton
- The Ohio State University Comprehensive Cancer Center, Columbus, OH; Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - John W Newman
- University of California Davis West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA; Department of Nutrition, University of California Davis, Davis, CA; United States Department of Agriculture Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA
| | - Lisa D Yee
- Department of Surgery, City of Hope, Duarte, CA; City of Hope Comprehensive Cancer Center, Duarte, CA.
| |
Collapse
|
3
|
Sabikunnahar B, Snyder JP, Rodriguez PD, Sessions KJ, Amiel E, Frietze SE, Krementsov DN. Natural genetic variation in wild-derived mice controls host survival and transcriptional responses during endotoxic shock. Immunohorizons 2025; 9:vlaf007. [PMID: 40139977 PMCID: PMC11945298 DOI: 10.1093/immhor/vlaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/22/2025] [Indexed: 03/29/2025] Open
Abstract
Innate immune cells sense microbial danger signals, resulting in dynamic transcriptional reprogramming and rapid inflammatory responses. If not properly regulated, such responses can be detrimental to the host, as is seen in septic shock. A better understanding of the genetic regulation of responses during endotoxemia could provide potential therapeutic insights. However, the majority of animal model studies have been performed using classic inbred laboratory strains of mice, capturing limited genetic diversity. Here, we compared classic inbred C57BL/6 (B6) mice with wild-derived and genetically divergent PWD/PhJ (PWD) mice using in vivo and in vitro models of endotoxemia. Compared with B6 mice, PWD mice were markedly resistant to bacterial lipopolysaccharide (LPS)-induced endotoxic shock. Using LPS stimulation of bone marrow derived dendritic cells (BMDC) and RNA sequencing, we demonstrate that B6 and PWD BMDCs exhibit partially overlapping yet highly divergent transcriptional responses, with B6 skewed toward stereotypical proinflammatory pathway activation, and PWD engaging regulatory or developmental pathways. To dissect genetic regulation of inflammatory responses by allelic variants, we used BMDCs from a sub-consomic strain carrying a ∼50 Mb PWD-derived portion of chromosome 11 on the B6 background. This identified a subset of cis-regulated and a large number of trans-regulated genes. Bioinformatic analyses identified candidate trans regulators encoded in the chromosome 11 locus as transcription factors Irf1, Ncor1, and Srebf1. Our results demonstrate that natural genetic variation controls host survival and transcriptional reprogramming during endotoxemia, suggesting possibilities for prediction of sepsis risk and/or personalized therapeutic interventions.
Collapse
Affiliation(s)
- Bristy Sabikunnahar
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Julia P Snyder
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Princess D Rodriguez
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, United States
| | - Katherine J Sessions
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Eyal Amiel
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Seth E Frietze
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| |
Collapse
|
4
|
Frankhouser DE, DeWess T, Snodgrass IF, Cole RM, Steck S, Thomas D, Kalu C, Belury MA, Clinton SK, Newman JW, Yee LD. Randomized dose-response trial of n-3 fatty acids in hormone receptor negative breast cancer survivors- impact on breast adipose oxylipin and DNA methylation patterns. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.16.24313691. [PMID: 39371146 PMCID: PMC11451633 DOI: 10.1101/2024.09.16.24313691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background Increasing evidence suggests the unique susceptibility of estrogen receptor and progesterone receptor negative (ERPR-) breast cancer to dietary fat amount and type. Dietary n-3 polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), may modulate breast adipose fatty acid profiles and downstream bioactive metabolites to counteract pro-inflammatory, pro-carcinogenic signaling in the mammary microenvironment. Objective To determine effects of ~1 to 5 g/d EPA+DHA over 12 months on breast adipose fatty acid and oxylipin profiles in women with ERPR(-) breast cancer, a high-risk molecular subtype. Methods We conducted a 12-month randomized controlled, double-blind clinical trial of ~5g/d vs ~1g/d DHA+EPA supplementation in women within 5 years of completing standard therapy for ERPR(-) breast cancer Stages 0-III. Blood and breast adipose tissue specimens were collected every 3 months for biomarker analyses including fatty acids by gas chromatography, oxylipins by LC-MS/MS, and DNA methylation by reduced-representation bisulfite sequencing (RRBS). Results A total of 51 participants completed the 12-month intervention. Study treatments were generally well-tolerated. While both doses increased n-3 PUFAs from baseline in breast adipose, erythrocytes, and plasma, the 5g/d supplement was more potent (n =51, p <0.001). The 5g/d dose also reduced plasma triglycerides from baseline (p =0.008). Breast adipose oxylipins at 0, 6, and 12 months showed dose-dependent increases in unesterified and esterified DHA and EPA metabolites (n =28). Distinct DNA methylation patterns in adipose tissue after 12 months were identified, with effects unique to the 5g/d dose group (n =17). Conclusions Over the course of 1 year, EPA+DHA dose-dependently increased concentrations of these fatty acids and their derivative oxylipin metabolites, producing differential DNA methylation profiles of gene promoters involved in metabolism-related pathways critical to ERPR(-) breast cancer development and progression. These data provide evidence of both metabolic and epigenetic effects of n-3 PUFAs in breast adipose tissue, elucidating novel mechanisms of action for high-dose EPA+DHA-mediated prevention of ERPR(-) breast cancer.
Collapse
Affiliation(s)
- David E. Frankhouser
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, CA 91010
| | - Todd DeWess
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, CA 91010
- Department of Surgery, City of Hope, Duarte CA 91010
| | - Isabel F. Snodgrass
- University of California Davis West Coast Metabolomics Center, Genome Center, University of California Davis, Davis CA, 95616
| | - Rachel M. Cole
- Department of Food Science and Technology, The Ohio State University, Columbus OH 43210
| | - Sarah Steck
- The Ohio State University Comprehensive Cancer Center, Columbus OH 43210
| | | | - Chidimma Kalu
- Department of Surgery, City of Hope, Duarte CA 91010
| | - Martha A. Belury
- Department of Food Science and Technology, The Ohio State University, Columbus OH 43210
- The Ohio State University Comprehensive Cancer Center, Columbus OH 43210
| | - Steven K. Clinton
- The Ohio State University Comprehensive Cancer Center, Columbus OH 43210
- Department of Internal Medicine, The Ohio State University, Columbus OH 43210
| | - John W. Newman
- University of California Davis West Coast Metabolomics Center, Genome Center, University of California Davis, Davis CA, 95616
- Department of Nutrition, University of California Davis, Davis CA, 956169
- United States Department of Agriculture Agricultural Research Service, Western Human Nutrition Research Center, Davis CA, 95616
| | - Lisa D. Yee
- Department of Surgery, City of Hope, Duarte CA 91010
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| |
Collapse
|
5
|
Reis LG, de Azevedo Ruiz VL, Massami Kitamura SM, Furugen Cesar Andrade A, de Oliveira Bussiman F, Daiana Poleti M, Coelho da Silveira J, Fukumasu H, Faccioli LH, Marzocchi-Machado CM, de Francisco Strefezzi R, Neves Garcia E, Casey T, Netto AS. Feeding sows milk biofortified with n-6 and n-3 modulates immune status of sows and drives positive transgenerational effects. PLoS One 2024; 19:e0306707. [PMID: 39190668 PMCID: PMC11349115 DOI: 10.1371/journal.pone.0306707] [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: 03/23/2024] [Accepted: 06/21/2024] [Indexed: 08/29/2024] Open
Abstract
The risk of chronic diseases such as cardiovascular disease, cancer, inflammation, obesity, and autoimmune disease is linked to the quality of dietary fats, with lower intake of saturated and higher intake of n-6 and n-3 polyunsaturated fatty acids (PUFA) considered beneficial to health. This study investigated the effect of supplementing sows' diets with cow's milk biofortified with n-6 or n-3 PUFA, at varying n-6/n-3 ratios (8.26, 7.92, and 2.72) during their growing phase and throughout gestation and lactation on their reproductive performance and immune-inflammatory status. Specifically, we analyzed circulating cholesterol and fatty acid profiles of serum, colostrum and milk, sow body weight, and neonate colostrum intake, Apgar scores, muscle composition, and embryo viability. Analysis of circulating immunoglobulins (Ig), interleukins, and eicosanoids and complement system hemolytic activity were used to evaluate inflammatory and immune responses of sows and piglets. Expression of lipolysis and lipogenic genes in the liver were investigated in sows and piglets, with additional investigation of hypothalamus genes regulating appetite in sows. Feeding sows milk biofortified with n-6 and n-3 PUFA altered serum fatty acid profiles, reduced triglycerides (TG), increased embryo total number, increased early gestation backfat, and reduced colostrum IgG. Piglets of biofortified sow had higher circulating IgA, IgM and TNF-α, and lower IL-10. Sows fed n-3 biofortified milk had higher very low-density lipoproteins (VLDL) and TNF-α in circulation. Offspring from sows fed n-6 versus n-3 biofortified milk had lower IL-10 and expression levels of SREBP-1. N-3 versus n-6 also lowered arachidonic acid (ARA) levels in sow's milk and piglet viability 1. Findings offer insights into the potential health benefits of dietary supplementation with biofortified milk in swine, which serve as good model of diet-nutrition studies of humans, and therefore can potentially be considered in dietary recommendations both human and animal populations.
Collapse
Affiliation(s)
- Leriana Garcia Reis
- Department of Animal Science, Purdue University, West Lafayette, Indiana, United States of America
| | - Vera Letticie de Azevedo Ruiz
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Simone Maria Massami Kitamura
- Department of Animal Science, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - André Furugen Cesar Andrade
- Department of Animal Reproduction, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | | | - Mirele Daiana Poleti
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Heidge Fukumasu
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Lúcia Helena Faccioli
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Cleni Mara Marzocchi-Machado
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ricardo de Francisco Strefezzi
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Edna Neves Garcia
- Department of Animal Science, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Theresa Casey
- Department of Animal Science, Purdue University, West Lafayette, Indiana, United States of America
| | - Arlindo Saran Netto
- Department of Animal Science, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Koppula S, Wankhede NL, Sammeta SS, Shende PV, Pawar RS, Chimthanawala N, Umare MD, Taksande BG, Upaganlawar AB, Umekar MJ, Kopalli SR, Kale MB. Modulation of cholesterol metabolism with Phytoremedies in Alzheimer's disease: A comprehensive review. Ageing Res Rev 2024; 99:102389. [PMID: 38906182 DOI: 10.1016/j.arr.2024.102389] [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: 04/20/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Alzheimer's disease (AD) is a complex neurological ailment that causes cognitive decline and memory loss. Cholesterol metabolism dysregulation has emerged as a crucial element in AD pathogenesis, contributing to the formation of amyloid-beta (Aβ) plaques and tau tangles, the disease's hallmark neuropathological characteristics. Thus, targeting cholesterol metabolism has gained attention as a potential therapeutic method for Alzheimer's disease. Phytoremedies, which are generated from plants and herbs, have shown promise as an attainable therapeutic option for Alzheimer's disease. These remedies contain bioactive compounds like phytochemicals, flavonoids, and polyphenols, which have demonstrated potential in modulating cholesterol metabolism and related pathways. This comprehensive review explores the modulation of cholesterol metabolism by phytoremedies in AD. It delves into the role of cholesterol in brain function, highlighting disruptions observed in AD. Additionally, it examines the underlying molecular mechanisms of cholesterol-related pathology in AD. The review emphasizes the significance of phytoremedies as a potential therapeutic intervention for AD. It discusses the drawbacks of current treatments and the need for alternative strategies addressing cholesterol dysregulation and its consequences. Through an in-depth analysis of specific phytoremedies, the review presents compelling evidence of their potential benefits. Molecular mechanisms underlying phytoremedy effects on cholesterol metabolism are examined, including regulation of cholesterol-related pathways, interactions with Aβ pathology, influence on tau pathology, and anti-inflammatory effects. The review also highlights challenges and future perspectives, emphasizing standardization, clinical evidence, and personalized medicine approaches to maximize therapeutic potential in AD treatment. Overall, phytoremedies offer promise as a potential avenue for AD management, but further research and collaboration are necessary to fully explore their efficacy, safety, and mechanisms of action.
Collapse
Affiliation(s)
- Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Shivkumar S Sammeta
- National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037, India.
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Rupali S Pawar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | | | - Mohit D Umare
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India.
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea.
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
7
|
Chandrasekaran P, Weiskirchen R. The Role of SCAP/SREBP as Central Regulators of Lipid Metabolism in Hepatic Steatosis. Int J Mol Sci 2024; 25:1109. [PMID: 38256181 PMCID: PMC10815951 DOI: 10.3390/ijms25021109] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly increasing worldwide at an alarming pace, due to an increase in obesity, sedentary and unhealthy lifestyles, and unbalanced dietary habits. MASLD is a unique, multi-factorial condition with several phases of progression including steatosis, steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Sterol element binding protein 1c (SREBP1c) is the main transcription factor involved in regulating hepatic de novo lipogenesis. This transcription factor is synthesized as an inactive precursor, and its proteolytic maturation is initiated in the membrane of the endoplasmic reticulum upon stimulation by insulin. SREBP cleavage activating protein (SCAP) is required as a chaperon protein to escort SREBP from the endoplasmic reticulum and to facilitate the proteolytic release of the N-terminal domain of SREBP into the Golgi. SCAP inhibition prevents activation of SREBP and inhibits the expression of genes involved in triglyceride and fatty acid synthesis, resulting in the inhibition of de novo lipogenesis. In line, previous studies have shown that SCAP inhibition can resolve hepatic steatosis in animal models and intensive research is going on to understand the effects of SCAP in the pathogenesis of human disease. This review focuses on the versatile roles of SCAP/SREBP regulation in de novo lipogenesis and the structure and molecular features of SCAP/SREBP in the progression of hepatic steatosis. In addition, recent studies that attempt to target the SCAP/SREBP axis as a therapeutic option to interfere with MASLD are discussed.
Collapse
Affiliation(s)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital Aachen, D-52074 Aachen, Germany
| |
Collapse
|
8
|
Wang T, Soundararajan A, Rabinowitz J, Jaiswal A, Osborne T, Pattabiraman PP. Identification of the novel role of sterol regulatory element binding proteins (SREBPs) in mechanotransduction and intraocular pressure regulation. FASEB J 2023; 37:e23248. [PMID: 37823226 PMCID: PMC10826798 DOI: 10.1096/fj.202301185r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
Trabecular meshwork (TM) cells are contractile and mechanosensitive, and they aid in maintaining intraocular pressure (IOP) homeostasis. Lipids are attributed to modulating TM contractility, with poor mechanistic understanding. In this study using human TM cells, we identify the mechanosensing role of the transcription factors sterol regulatory element binding proteins (SREBPs) involved in lipogenesis. By constitutively activating SREBPs and pharmacologically inactivating SREBPs, we have mechanistically deciphered the attributes of SREBPs in regulating the contractile properties of TM. The pharmacological inhibition of SREBPs by fatostatin and molecular inactivation of SREBPs ex vivo and in vivo, respectively, results in significant IOP lowering. As a proof of concept, fatostatin significantly decreased the SREBPs responsive genes and enzymes involved in lipogenic pathways as well as the levels of the phospholipid, cholesterol, and triglyceride. Further, we show that fatostatin mitigated actin polymerization machinery and stabilization, and decreased ECM synthesis and secretion. We thus postulate that lowering lipogenesis in the TM outflow pathway can hold the key to lowering IOP by modifying the TM biomechanics.
Collapse
Affiliation(s)
- Ting Wang
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, Indiana, 46202, United States of America
- Stark Neuroscience Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, 320 W. 15th Street, Indiana, 46202, United States of America
| | - Avinash Soundararajan
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, Indiana, 46202, United States of America
| | - Jeffrey Rabinowitz
- Department of Ophthalmology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Anant Jaiswal
- Institute for Fundamental Biomedical Research, Department of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, Florida, 33701, United States of America
| | - Timothy Osborne
- Institute for Fundamental Biomedical Research, Department of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, Florida, 33701, United States of America
| | - Padmanabhan Paranji Pattabiraman
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, Indiana, 46202, United States of America
- Stark Neuroscience Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, 320 W. 15th Street, Indiana, 46202, United States of America
| |
Collapse
|
9
|
Hong E, Kang H, Yang G, Oh S, Kim E. The PKA-SREBP1c Pathway Plays a Key Role in the Protective Effects of Lactobacillus johnsonii JNU3402 Against Diet-Induced Fatty Liver in Mice. Mol Nutr Food Res 2023; 67:e2200496. [PMID: 37650271 DOI: 10.1002/mnfr.202200496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 05/24/2023] [Indexed: 09/01/2023]
Abstract
SCOPE The present study aims to assess the protective effect of Lactobacillus johnsonii JNU3402 (LJ3402) against diet-induced non-alcoholic fatty liver disease (NAFLD) and determine the mechanism underlying its beneficial effect on the liver in mice. METHODS AND RESULTS Seven-week-old male mice are fed a high-fat diet (HFD) with or without oral supplementation of LJ3402 for 14 weeks. In mice fed an HFD, LJ3402 administration alleviates liver steatosis, diet-induced obesity, and insulin resistance with a decreased hepatic expression of sterol-regulatory element-binding protein-1c (SREBP-1c), fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC), and an increased phosphorylation of SREBP-1c. The mechanistic study shows that LJ3402 inhibits SREBP-1c transcriptional activity by enhancing protein kinase A (PKA)-mediated phosphorylation and reduces the expression of its lipogenic target genes in AML12 and HepG2 cells, thereby attenuating hepatic lipid accumulation. Moreover, silencing the PKA α catalytic subunit or the inhibition of PKA activity by H89 abolishes LJ3402 suppression of free fatty acid (FFA)-induced SREBP-1c activity in hepatocytes. In addition, LJ3402 administration elevates the plasma lactate levels in mice fed an HFD; this lactate increases PKA-mediated SREBP-1c phosphorylation in AML12 cells with a decreased expression of its target genes, reducing hepatic lipid accumulation. CONCLUSION LJ3402 attenuates HFD-induced fatty liver in mice through the lactate-PKA-SREBP-1c pathway.
Collapse
Affiliation(s)
- Eunjeong Hong
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Hyuno Kang
- Division of Analytical Science, Korea Basic Science Institute, 169-148, Gwahak-ro, Yuseong-gu, Daejeon, 34133, Republic of Korea
| | - Garam Yang
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Sejong Oh
- Division of Animal Science, College of Agriculture & Life Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Eungseok Kim
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| |
Collapse
|
10
|
Wang T, Soundararajan A, Rabinowitz J, Jaiswal A, Osborne T, Pattabiraman PP. Identification of the novel role of sterol regulatory element binding proteins (SREBPs) in mechanotransduction and intraocular pressure regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.05.527136. [PMID: 37214961 PMCID: PMC10197526 DOI: 10.1101/2023.02.05.527136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Trabecular meshwork (TM) cells are highly contractile and mechanosensitive to aid in maintaining intraocular pressure (IOP) homeostasis. Lipids are attributed to modulating TM contractility with poor mechanistic understanding. In this study using human TM cells, we identify the mechanosensing role of the transcription factors sterol regulatory element binding proteins (SREBPs) involved in lipogenesis. By constitutively activating SREBPs and pharmacologically inactivating SREBPs, we have mechanistically deciphered the attributes of SREBPs in regulating the contractile properties of TM. The pharmacological inhibition of SREBPs by fatostatin and molecular inactivation of SREBPs ex vivo and in vivo respectively results in significant IOP lowering. As a proof of concept, fatostatin significantly decreased the SREBPs responsive genes and enzymes involved in lipogenic pathways as well as the levels of the phospholipid, cholesterol, and triglyceride. Further, we show that fatostatin mitigated actin polymerization machinery and stabilization, and decreased ECM synthesis and secretion. We thus postulate that lowering lipogenesis in the TM outflow pathway can hold the key to lowering IOP by modifying the TM biomechanics. Synopsis In this study, we show the role of lipogenic transcription factors sterol regulatory element binding proteins (SREBPs) in the regulation of intraocular pressure (IOP). ( Synopsis Figure - Created using Biorender.com ) SREBPs are involved in the sensing of changes in mechanical stress on the trabecular meshwork (TM). SREBPs aid in transducing the mechanical signals to induce actin polymerization and filopodia/lamellipodia formation.SREBPs inactivation lowered genes and enzymes involved in lipogenesis and modified lipid levels in TM.SREBPs activity is a critical regulator of ECM engagement to the matrix sites.Inactivation of SCAP-SREBP pathway lowered IOP via actin relaxation and decreasing ECM production and deposition in TM outflow pathway signifying a novel relationship between SREBP activation status and achieving IOP homeostasis.
Collapse
|
11
|
Reiss AB, Ahmed S, Johnson M, Saeedullah U, De Leon J. Exosomes in Cardiovascular Disease: From Mechanism to Therapeutic Target. Metabolites 2023; 13:479. [PMID: 37110138 PMCID: PMC10142472 DOI: 10.3390/metabo13040479] [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: 03/01/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality globally. In recent decades, clinical research has made significant advances, resulting in improved survival and recovery rates for patients with CVD. Despite this progress, there is substantial residual CVD risk and an unmet need for better treatment. The complex and multifaceted pathophysiological mechanisms underlying the development of CVD pose a challenge for researchers seeking effective therapeutic interventions. Consequently, exosomes have emerged as a new focus for CVD research because their role as intercellular communicators gives them the potential to act as noninvasive diagnostic biomarkers and therapeutic nanocarriers. In the heart and vasculature, cell types such as cardiomyocytes, endothelial cells, vascular smooth muscle, cardiac fibroblasts, inflammatory cells, and resident stem cells are involved in cardiac homeostasis via the release of exosomes. Exosomes encapsulate cell-type specific miRNAs, and this miRNA content fluctuates in response to the pathophysiological setting of the heart, indicating that the pathways affected by these differentially expressed miRNAs may be targets for new treatments. This review discusses a number of miRNAs and the evidence that supports their clinical relevance in CVD. The latest technologies in applying exosomal vesicles as cargo delivery vehicles for gene therapy, tissue regeneration, and cell repair are described.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA
| | | | | | | | | |
Collapse
|
12
|
Bioactive Compounds as Inhibitors of Inflammation, Oxidative Stress and Metabolic Dysfunctions via Regulation of Cellular Redox Balance and Histone Acetylation State. Foods 2023; 12:foods12050925. [PMID: 36900446 PMCID: PMC10000917 DOI: 10.3390/foods12050925] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
Bioactive compounds (BCs) are known to exhibit antioxidant, anti-inflammatory, and anti-cancer properties by regulating the cellular redox balance and histone acetylation state. BCs can control chronic oxidative states caused by dietary stress, i.e., alcohol, high-fat, or high-glycemic diet, and adjust the redox balance to recover physiological conditions. Unique functions of BCs to scavenge reactive oxygen species (ROS) can resolve the redox imbalance due to the excessive generation of ROS. The ability of BCs to regulate the histone acetylation state contributes to the activation of transcription factors involved in immunity and metabolism against dietary stress. The protective properties of BCs are mainly ascribed to the roles of sirtuin 1 (SIRT1) and nuclear factor erythroid 2-related factor 2 (NRF2). As a histone deacetylase (HDAC), SIRT1 modulates the cellular redox balance and histone acetylation state by mediating ROS generation, regulating nicotinamide adenine dinucleotide (NAD+)/NADH ratio, and activating NRF2 in metabolic progression. In this study, the unique functions of BCs against diet-induced inflammation, oxidative stress, and metabolic dysfunction have been considered by focusing on the cellular redox balance and histone acetylation state. This work may provide evidence for the development of effective therapeutic agents from BCs.
Collapse
|
13
|
Fung C, Wilding B, Schittenhelm RB, Bryson-Richardson RJ, Bird PI. Expression of the Z Variant of α1-Antitrypsin Suppresses Hepatic Cholesterol Biosynthesis in Transgenic Zebrafish. Int J Mol Sci 2023; 24:ijms24032475. [PMID: 36768797 PMCID: PMC9917206 DOI: 10.3390/ijms24032475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/31/2023] Open
Abstract
Individuals homozygous for the Pi*Z allele of SERPINA1 (ZAAT) are susceptible to lung disease due to insufficient α1-antitrypsin secretion into the circulation and may develop liver disease due to compromised protein folding that leads to inclusion body formation in the endoplasmic reticulum (ER) of hepatocytes. Transgenic zebrafish expressing human ZAAT show no signs of hepatic accumulation despite displaying serum insufficiency, suggesting the defect in ZAAT secretion occurs independently of its tendency to form inclusion bodies. In this study, proteomic, transcriptomic, and biochemical analysis provided evidence of suppressed Srebp2-mediated cholesterol biosynthesis in the liver of ZAAT-expressing zebrafish. To investigate the basis for this perturbation, CRISPR/Cas9 gene editing was used to manipulate ER protein quality control factors. Mutation of erlec1 resulted in a further suppression in the cholesterol biosynthesis pathway, confirming a role for this ER lectin in targeting misfolded ZAAT for ER-associated degradation (ERAD). Mutation of the two ER mannosidase homologs enhanced ZAAT secretion without inducing hepatic accumulation. These insights into hepatic ZAAT processing suggest potential therapeutic targets to improve secretion and alleviate serum insufficiency in this form of the α1-antitrypsin disease.
Collapse
Affiliation(s)
- Connie Fung
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
- Correspondence: (C.F.); (P.I.B.)
| | - Brendan Wilding
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Ralf B. Schittenhelm
- Monash Proteomics and Metabolomics Facility, Monash University, Melbourne 3800, Australia
| | | | - Phillip I. Bird
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
- Correspondence: (C.F.); (P.I.B.)
| |
Collapse
|
14
|
Laval T, Pedró-Cos L, Malaga W, Guenin-Macé L, Pawlik A, Mayau V, Yahia-Cherbal H, Delos O, Frigui W, Bertrand-Michel J, Guilhot C, Demangel C. De novo synthesized polyunsaturated fatty acids operate as both host immunomodulators and nutrients for Mycobacterium tuberculosis. eLife 2021; 10:71946. [PMID: 34951591 PMCID: PMC8752091 DOI: 10.7554/elife.71946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/24/2021] [Indexed: 11/30/2022] Open
Abstract
Successful control of Mycobacterium tuberculosis (Mtb) infection by macrophages relies on immunometabolic reprogramming, where the role of fatty acids (FAs) remains poorly understood. Recent studies unraveled Mtb’s capacity to acquire saturated and monounsaturated FAs via the Mce1 importer. However, upon activation, macrophages produce polyunsaturated fatty acids (PUFAs), mammal-specific FAs mediating the generation of immunomodulatory eicosanoids. Here, we asked how Mtb modulates de novo synthesis of PUFAs in primary mouse macrophages and whether this benefits host or pathogen. Quantitative lipidomics revealed that Mtb infection selectively activates the biosynthesis of ω6 PUFAs upstream of the eicosanoid precursor arachidonic acid (AA) via transcriptional activation of Fads2. Inhibiting FADS2 in infected macrophages impaired their inflammatory and antimicrobial responses but had no effect on Mtb growth in host cells nor mice. Using a click-chemistry approach, we found that Mtb efficiently imports ω6 PUFAs via Mce1 in axenic culture, including AA. Further, Mtb preferentially internalized AA over all other FAs within infected macrophages by mechanisms partially depending on Mce1 and supporting intracellular persistence. Notably, IFNγ repressed de novo synthesis of AA by infected mouse macrophages and restricted AA import by intracellular Mtb. Together, these findings identify AA as a major FA substrate for intracellular Mtb, whose mobilization by innate immune responses is opportunistically hijacked by the pathogen and downregulated by IFNγ.
Collapse
Affiliation(s)
- Thomas Laval
- Immunobiology of Infection Unit, Institut Pasteur, Paris, France
| | - Laura Pedró-Cos
- Immunobiology of Infection Unit, Institut Pasteur, Paris, France
| | - Wladimir Malaga
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS UMR5089, Toulouse, France
| | | | - Alexandre Pawlik
- Integrated Mycobacterial Pathogenomics Unit, Institut Pasteur, Paris, France
| | - Véronique Mayau
- Immunobiology of Infection Unit, Institut Pasteur, Paris, France
| | | | | | - Wafa Frigui
- Integrated Mycobacterial Pathogenomics Unit, Institut Pasteur, Paris, France
| | | | - Christophe Guilhot
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS UMR5089, Toulouse, France
| | | |
Collapse
|
15
|
Ahluwalia MK. Nutrigenetics and nutrigenomics-A personalized approach to nutrition. ADVANCES IN GENETICS 2021; 108:277-340. [PMID: 34844714 DOI: 10.1016/bs.adgen.2021.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The prevalence of non-communicable diseases has been on an upward trajectory for some time and this puts an enormous burden on the healthcare expenditure. Lifestyle modifications including dietary interventions hold an immense promise to manage and prevent these diseases. Recent advances in genomic research provide evidence that focussing these efforts on individual variations in abilities to metabolize nutrients (nutrigenetics) and exploring the role of dietary compounds on gene expression (nutrigenomics and nutri-epigenomics) can lead to more meaningful personalized dietary strategies to promote optimal health. This chapter aims to provide examples on these gene-diet interactions at multiple levels to support the need of embedding targeted dietary interventions as a way forward to prevent, avoid and manage diseases.
Collapse
|
16
|
Rogers CJ, Kyubwa EM, Lukaszewicz AI, Starbird MA, Nguyen M, Copeland BT, Yamada-Hanff J, Menon N. Observation of Unique Circulating miRNA Signatures in Non-Human Primates Exposed to Total-Body vs. Whole Thorax Lung Irradiation. Radiat Res 2021; 196:547-559. [PMID: 34525208 DOI: 10.1667/rade-21-00043.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/26/2021] [Indexed: 11/03/2022]
Abstract
A radiological/nuclear (RAD-NUC) incident, especially in an urban setting, results in diverse radiation-induced injuries due to heterogeneities in dose, the extent of partial-body shielding, human biodiversity and pre-existing health conditions. For example, acute radiation syndrome (ARS) can result in death within days to weeks of exposure to 0.7-10 Gy doses and is associated with destruction of the bone marrow, known as hematopoietic ARS (H-ARS). However, partial-body shielding that spares a portion of the bone marrow from exposure can significantly reduce the occurrence of H-ARS, but delayed effects of acute radiation exposure (DEARE) can still occur within months or years after exposure depending on the individual. In a mass casualty event, ideal triage must be able to pre-symptomatically identify individuals likely to develop radiation-induced injuries and provide an appropriate treatment plan. Today, while there are FDA approved treatments for hematopoietic ARS, there are no approved diagnosis for radiation injury and no approved treatments for the broad spectra of injuries associated with radiation. This has resulted in a major capability gap in the nations preparedness to a potentially catastrophic RAD-NUC event. Circulating microRNA (miRNA) are a promising class of biomarkers for this application because the molecules are accessible via a routine blood draw and are excreted by various tissues throughout the body. To test if miRNA can be used to predict distinct tissue-specific radiation-induced injuries, we compared the changes to the circulating miRNA profiles after total-body irradiation (TBI) and whole thorax lung irradiation (WTLI) in non-human primates at doses designed to induce ARS (day 2 postirradiation; 2-6.5 Gy) and DEARE (day 15 postirradiation; 9.8 or 10.7 Gy), respectively. In both models, miRNA sequences were identified that correlated with the onset of severe neutropenia (counts <500 µL-1; TBI) or survival (WTLI). This method identified panels of eleven miRNA for both model and assigned functional roles for the panel members using gene ontology enrichment analysis. A common signature of radiation-induced injury was observed in both models: apoptosis, DNA damage repair, p53 signaling, pro-inflammatory response, and growth factor/cytokine signaling pathways were predicted to be disrupted. In addition, injury-specific pathways were identified. In TBI, pathways associated with ubiquitination, specifically of histone H2A, were enriched, suggesting more impact to DNA damage repair mechanisms and apoptosis. In WTLI, pro-fibrotic pathways including transforming growth factor (TGF-β) and bone morphogenetic protein (BMP) signaling pathways were enriched, consistent with the onset of late lung injury. These results suggest that miRNA may indeed be able to predict the onset of distinct types of radiation-induced injuries.
Collapse
|
17
|
A network-based approach to identify protein kinases critical for regulating srebf1 in lipid deposition causing obesity. Funct Integr Genomics 2021; 21:557-570. [PMID: 34327622 DOI: 10.1007/s10142-021-00798-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022]
Abstract
Obesity is a rapidly growing health pandemic, underlying a wide variety of disease conditions leading to increases in global mortality. It is known that the phosphorylation of various proteins regulates sterol regulatory element-binding transcription factors 1 (srebf1), a key lipogenic transcription factor, to cause the development of obesity. To detect the key protein kinases for regulating srebf1 in lipid deposition, we established the srebf1 knockout model in zebrafish (KO, srebf1-/-) by CRISPR/Cas9. The KO zebrafish exhibited a significant reduction of total free fatty acid content (fell 60.5%) and lipid deposition decrease compared with wild-type (WT) zebrafish. Meanwhile, srebf1 deletion in zebrafish eliminated lipid deposition induced by high-fat diet feeding. Compared with WT zebrafish, a total of 697 differentially expressed proteins and 316 differentially expressed phosphoproteins with 439 sites were identified in KO by differential proteomic and phosphoproteomic analyses. A significant number of proteins identified were involved in lipid and glucose metabolism. Moreover, some protein kinases critical for regulating srebf1 in lipid deposition, including Cdk2, Pkc, Prkceb, mTORC1, Mapk12, and Wnk1, were determined by network analyses. An in vitro study was performed to verify the network analysis results. Our findings provide potential targets (kinases) for human obesity treatments.
Collapse
|
18
|
Taqi MO, Saeed-Zidane M, Gebremedhn S, Salilew-Wondim D, Tholen E, Neuhoff C, Hoelker M, Schellander K, Tesfaye D. NRF2-mediated signaling is a master regulator of transcription factors in bovine granulosa cells under oxidative stress condition. Cell Tissue Res 2021; 385:769-783. [PMID: 34008050 PMCID: PMC8526460 DOI: 10.1007/s00441-021-03445-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 03/01/2021] [Indexed: 11/30/2022]
Abstract
Transcription factors (TFs) are known to be involved in regulating the expression of several classes of genes during folliculogenesis. However, the regulatory role of TFs during oxidative stress (OS) is not fully understood. The current study was aimed to investigate the regulation of the TFs in bovine granulosa cells (bGCs) during exposure to OS induced by H2O2 in vitro. For this, bGCs derived from ovarian follicles were cultured in vitro till their confluency and then treated with H2O2 for 40 min. Twenty-four hours later, cells were subjected to various phenotypic and gene expression analyses for genes related to TFs, endoplasmic reticulum stress, apoptosis, cell proliferation, and differentiation markers. The bGCs exhibited higher reactive oxygen species accumulation, DNA fragmentation, and endoplasmic reticulum stress accompanied by reduction of mitochondrial activity after exposure to OS. In addition, higher lipid accumulation and lower cell proliferation were noticed in H2O2-challenged cells. The mRNA level of TFs including NRF2, E2F1, KLF6, KLF9, FOS, SREBF1, SREBF2, and NOTCH1 was increased in H2O2-treated cells compared with non-treated controls. However, the expression level of KLF4 and its downstream gene, CCNB1, were downregulated in the H2O2-challenged group. Moreover, targeted inhibition of NRF2 using small interference RNA resulted in reduced expression of KLF9, FOS, SREBF2, and NOTCH1 genes, while the expression of KLF4 was upregulated. Taken together, bovine granulosa cells exposed to OS exhibited differential expression of various transcription factors, which are mediated by the NRF2 signaling pathway.
Collapse
Affiliation(s)
- Mohamed Omar Taqi
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany.,Central Laboratory for Agricultural Climate, Agricultural Research Center, Giza, Egypt
| | - Mohammed Saeed-Zidane
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany.,Institute of Animal Breeding and Husbandry, Animal Breeding and Genetics Group, University of Kiel, Kiel, Germany
| | - Samuel Gebremedhn
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany.,Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory (ARBL), Colorado State University, Fort Collins, CO, USA
| | - Dessie Salilew-Wondim
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany
| | - Ernst Tholen
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany
| | - Christiane Neuhoff
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany
| | - Michael Hoelker
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany.,Teaching and Research Station Frankenforst, University of Bonn, Koenigswinter, Germany
| | - Karl Schellander
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany
| | - Dawit Tesfaye
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany. .,Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory (ARBL), Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
19
|
Zhang X, Xia H, Wang J, Leng R, Zhou X, Gao Q, He K, Liu D, Huang B. Effect of selenium-enriched kiwifruit on body fat reduction and liver protection in hyperlipidaemic mice. Food Funct 2021; 12:2044-2057. [PMID: 33532813 DOI: 10.1039/d0fo02410d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
This study aimed to investigate the effects and mechanism of selenium-enriched kiwifruit (Se-Kiwi) on lipid-lowering and liver protection in hyperlipidaemic mice induced by consuming a long-term high-fat diet. Selenium-enriched cultivation can significantly improve the contents of vitamins and functional elements in kiwifruits, especially vitamin C, selenium, and manganese, thus enhancing the activity of antioxidant enzymes in Se-Kiwi. Se-Kiwi can significantly improve the activity of antioxidant enzymes in the liver of hyperlipidaemic mice, restore the liver morphology of mice close to normal, reduce the fat content in the liver, and inhibit the accumulation of abdominal fat cells. Meanwhile, the expression levels of inflammation-related factors (TNF-α and NF-κB) and lipid synthesis related genes (SREBP-1c and FAS) are inhibited at the gene transcription and protein expression levels, and the expression levels of energy expenditure related genes (PPAR-α and CPT1) are increased, resulting in lipid reductions and liver protection. In conclusion, our results indicate that the protective mechanism of Se-Kiwi on high-fat diet mice is associated with enhancing the activity of antioxidant enzymes, reducing the degree of the inflammatory reaction, inhibiting the fat synthesis, and accelerating body energy consumption.
Collapse
Affiliation(s)
- Xiaoni Zhang
- School of Life Sciences, Anhui University, Hefei 230601, China. and Center for Stem Cell and Translational Medicine, Anhui University, Hefei 230601, China
| | - Haidong Xia
- School of Life Sciences, Anhui University, Hefei 230601, China. and Center for Stem Cell and Translational Medicine, Anhui University, Hefei 230601, China
| | - Jie Wang
- School of Life Sciences, Anhui University, Hefei 230601, China. and Center for Stem Cell and Translational Medicine, Anhui University, Hefei 230601, China
| | - Ruyue Leng
- School of Life Sciences, Anhui University, Hefei 230601, China. and Center for Stem Cell and Translational Medicine, Anhui University, Hefei 230601, China
| | - Xiaojing Zhou
- School of Life Sciences, Anhui University, Hefei 230601, China. and Center for Stem Cell and Translational Medicine, Anhui University, Hefei 230601, China
| | - Qian Gao
- School of Life Sciences, Anhui University, Hefei 230601, China. and Center for Stem Cell and Translational Medicine, Anhui University, Hefei 230601, China
| | - Kan He
- School of Life Sciences, Anhui University, Hefei 230601, China. and Center for Stem Cell and Translational Medicine, Anhui University, Hefei 230601, China
| | - Dahai Liu
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, China
| | - Bei Huang
- School of Life Sciences, Anhui University, Hefei 230601, China. and Center for Stem Cell and Translational Medicine, Anhui University, Hefei 230601, China
| |
Collapse
|
20
|
Haubold S, Kröger-Koch C, Tuchscherer A, Kanitz E, Weitzel JM, Hoeflich A, Starke A, Tröscher A, Sauerwein H, Hammon HM. Effects of a combined essential fatty acid and conjugated linoleic acid abomasal infusion on metabolic and endocrine traits, including the somatotropic axis, in dairy cows. J Dairy Sci 2020; 103:12069-12082. [PMID: 32981718 DOI: 10.3168/jds.2020-18569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/17/2020] [Indexed: 01/04/2023]
Abstract
The objective of this study was to test the effects of essential fatty acids (EFA), particularly α-linolenic acid (ALA), and conjugated linoleic acid (CLA) supplementation on metabolic and endocrine traits related to energy metabolism, including the somatotropic axis, in mid-lactation dairy cows. Four cows (126 ± 4 d in milk) were used in a dose-escalation study design and were abomasally infused with coconut oil (CTRL; 38.3 g/d; providing saturated fatty acids), linseed and safflower oils (EFA; 39.1 and 1.6 g/d; n-6:n-3 FA ratio = 1:3), Lutalin (CLA; cis-9,trans-11 and trans-10,cis-12 CLA, 4.6 g/d of each), or EFA and CLA (EFA+CLA) for 6 wk. The initial dosage was doubled twice after 2 wk, resulting in 3 dosages (dosages 1, 2, and 3). Each cow received each fat treatment at different times. Cows were fed with a corn silage-based total mixed ration providing a low-fat content and a high n-6:n-3 fatty acid ratio. Plasma concentrations of metabolites and hormones (insulin-like growth factor-binding proteins only on wk 0 and 6) were analyzed at wk 0, 2, 4, and 6 of each treatment period. Liver biopsies were taken before starting the trial and at wk 6 of each treatment period to measure hepatic mRNA abundance of genes linked to glucose, cholesterol and lipid metabolism, and the somatotropic axis. The changes in the milk and blood fatty acid patterns and lactation performance of these cows have already been published in a companion paper. The plasma concentration of total cholesterol increased with dosage in all groups, except CLA, reaching the highest levels in EFA+CLA and CTRL compared with CLA. The high-density lipoprotein cholesterol plasma concentration increased in CTRL and was higher than that in EFA and CLA, whereas the concentration of low-density lipoprotein cholesterol increased in a dose-dependent manner in EFA and EFA+CLA, and was higher than that in CLA. Hepatic mRNA expression of 3-hydroxy-3-methyl-glutaryl-CoA synthase 1 was upregulated in all groups but was highest in EFA+CLA. Expression of sterol regulatory element-binding factor 1 tended to be lowest due to EFA treatment, whereas expression of long chain acyl-CoA-synthetase was lower in EFA than in CTRL. Hepatic mRNA expression of GHR1A tended to be higher in EFA+CLA than in CTRL. The plasma concentration of insulin-like growth factor I increased in CLA, and the plasma IGFBP-2 concentration was lower in EFA+CLA than in CTRL at wk 6. The plasma concentration of adiponectin decreased in EFA+CLA up to dosage 2. Plasma concentrations of albumin and urea were lower in CLA than in CTRL throughout the experimental period. Supplementation with EFA and CLA affected cholesterol and lipid metabolism and their regulation differently, indicating distinct stimulation after the combined EFA and CLA treatment. The decreased IGFBP-2 plasma concentration and upregulated hepatic mRNA abundance of GHR1A in EFA+CLA-supplemented cows indicated the beneficial effect of the combined EFA and CLA treatment on the somatotropic axis in mid-lactation dairy cows. Moreover, supplementation with CLA might affect protein metabolism in dairy cows.
Collapse
Affiliation(s)
- S Haubold
- Institute of Nutritional Physiology "Oskar Kellner," Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - C Kröger-Koch
- Institute of Nutritional Physiology "Oskar Kellner," Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - A Tuchscherer
- Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - E Kanitz
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - J M Weitzel
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - A Hoeflich
- Institute of Genome Biology of Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - A Starke
- Clinic for Ruminants and Swine, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | | | - H Sauerwein
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, 53115 Bonn, Germany
| | - H M Hammon
- Institute of Nutritional Physiology "Oskar Kellner," Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany.
| |
Collapse
|
21
|
Ganoderic acid A attenuates high-fat-diet-induced liver injury in rats by regulating the lipid oxidation and liver inflammation. Arch Pharm Res 2020; 43:744-754. [PMID: 32715385 DOI: 10.1007/s12272-020-01256-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/24/2020] [Indexed: 01/04/2023]
Abstract
Ganoderic Acid A (GA) has many pharmacological effects such as anti-tumor, antibacterial, anti-inflammatory, and immunosuppressive effects. However, the protective effect of GA on liver injury has not been reported. This study aimed to investigate the action of GA on insufficient methionine and choline combined with high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) in rats. NAFLD model was established by insufficient methionine and choline combined with high fat feeding to rats. The levels of Acetyl-CoA carboxylase, fatty acid synthase, sterol regulatory element binding protein, liver X receptors, AMP-activated protein kinase, peroxisome proliferator-activated receptor α, PPARg coactivator 1α and NF-κB pathway in the liver were detected by western blot. The results of this study demonstrated that the expression of GA can not only significantly decrease the live weight and liver weight per body weight of HFD mice, but also restore the alanine aminotransferase, aspartate aminotransferase, total bilirubin levels, triglyceride and cholesterol in serum. In addition, the expression of GA increased the levels of high-density lipoprotein cholesterol in serum, ameliorated pathological changes and decreased NAS score of mice's liver. In conclusion, the treatment with GA could improve NAFLD in rats by regulating the levels of signaling events involved in free fatty acid production, lipid oxidation and liver inflammation.
Collapse
|
22
|
Jiang W, Xu S, Guo H, Lu L, Liu J, Wang G, Hao K. Magnesium isoglycyrrhizinate prevents the nonalcoholic hepatic steatosis via regulating energy homeostasis. J Cell Mol Med 2020; 24:7201-7213. [PMID: 32410294 PMCID: PMC7339216 DOI: 10.1111/jcmm.15230] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/22/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
Non-alcoholic fatty liver disease is a public health problem worldwide associated with high morbidity and hepatic steatosis, but no effective therapeutic interventions. Magnesium isoglycyrrhizinate (MGIG), a derivative of an active component of Glycyrrhiza glabra, is widely used for the treatment of inflammatory liver diseases due to its potent anti-inflammatory and hepatoprotective activities. Hence, this study aimed to study the effects of MGIG on hepatic steatosis in mice fed a high-fat diet (HFD). Oil Red O staining and transmission electron microscopy revealed a decrease in lipid accumulation in the liver after MGIG treatment along with improved mitochondrial ultramicrostructures. Metabonomic analysis demonstrated that MGIG intervention increased glutamate utilization in mitochondria by promoting the uptake of glutamate into the tricarboxylic acid (TCA) cycle. The NAD+ /NADH ratio and the expression of other lipid-metabolism-related genes were increased in MGIG-treated livers. Transcriptome sequencing showed that the expression of TLR4, an isoform of the innate immunity Toll-like receptors (TLRs), was significantly decreased after MGIG treatment, suggesting a link between the anti-inflammatory effects of MGIG and its suppression of lipidation. Our results reveal the potent effects of MGIG on lipid metabolism and suggest that hepatic TLR4 might be a crucial therapeutic target to regulate energy homeostasis in hepatic steatosis.
Collapse
Affiliation(s)
- Wenjiao Jiang
- Key Laboratory of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Shiyu Xu
- Key Laboratory of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Huijie Guo
- Key Laboratory of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Li Lu
- Key Laboratory of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Jie Liu
- Key Laboratory of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Kun Hao
- Key Laboratory of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
23
|
Ruiz CF, Montal ED, Haley JA, Bott AJ, Haley JD. SREBP1 regulates mitochondrial metabolism in oncogenic KRAS expressing NSCLC. FASEB J 2020; 34:10574-10589. [PMID: 32568455 DOI: 10.1096/fj.202000052r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/08/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
Cancer cells require extensive metabolic reprograming in order to provide the bioenergetics and macromolecular precursors needed to sustain a malignant phenotype. Mutant KRAS is a driver oncogene that is well-known for its ability to regulate the ERK and PI3K signaling pathways. However, it is now appreciated that KRAS can promote the tumor growth via upregulation of anabolic metabolism. We recently reported that oncogenic KRAS promotes a gene expression program of de novo lipogenesis in non-small cell lung cancer (NSCLC). To define the mechanism(s) responsible, we focused on the lipogenic transcription factor SREBP1. We observed that KRAS increases SREBP1 expression and genetic knockdown of SREBP1 significantly inhibited the cell proliferation of mutant KRAS-expressing cells. Unexpectedly, lipogenesis was not significantly altered in cells subject to SREBP1 knockdown. Carbon tracing metabolic studies showed a significant decrease in oxidative phosphorylation and RNA-seq data revealed a significant decrease in mitochondrial encoded subunits of the electron transport chain (ETC). Taken together, these data support a novel role, distinct from lipogenesis, of SREBP1 on mitochondrial function in mutant KRAS NSCLC.
Collapse
Affiliation(s)
- Christian F Ruiz
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Emily D Montal
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John A Haley
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alex J Bott
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - John D Haley
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, NY, USA
| |
Collapse
|
24
|
Nguyen HT, Yamamoto K, Iida M, Agusa T, Ochiai M, Guo J, Karthikraj R, Kannan K, Kim EY, Iwata H. Effects of prenatal bisphenol A exposure on the hepatic transcriptome and proteome in rat offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 720:137568. [PMID: 32145629 DOI: 10.1016/j.scitotenv.2020.137568] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/06/2020] [Accepted: 02/24/2020] [Indexed: 06/10/2023]
Abstract
Developmental exposure to bisphenol A (BPA) is associated with liver dysfunction and diseases in adulthood. The aims of this study were to assess the effects of prenatal BPA exposure on the hepatic transcriptome and proteome in female and male offspring and to understand adverse outcome pathways (AOPs) to observed phenotypic effects. Pregnant Wistar rats were exposed to 50 or 5000 μg BPA/kg bw/day, or 17β-estradiol (E2, 50 μg/kg bw/day) from embryonic day 3 to 18. The liver transcriptome and proteome profiles were analyzed in the newborn (postnatal day 1; PND1) and weaning (PND21) rat offspring. Based on the differentially expressed genes/proteins derived from transcriptome and proteome profiles, we performed pathway, transcription factor, and disease enrichment analyses. A principal component analysis of transcriptome data demonstrated that prenatal BPA exposure caused masculinization of the hepatic transcriptome in females. Both of transcriptomic and proteomic data showed that prenatal BPA exposure led to the disruption of cell cycle, lipid homeostasis, and hormone balance in offspring. Most of the effects at the transcript level were extended from newborn to weaning in males, but were moderated until weaning in females. The alterations at the transcript and protein levels were accordant with the observation of increases in body weight and anogenital distance and changes in hepatosomatic index in the offspring. Collectively, we constructed AOPs with evidence of sex- and age-specific actions of prenatal BPA exposure in the offspring.
Collapse
Affiliation(s)
- Hoa Thanh Nguyen
- Center for Marine Environmental Studies, Ehime University, Matsuyama, 790-8577, Japan
| | - Kimika Yamamoto
- Center for Marine Environmental Studies, Ehime University, Matsuyama, 790-8577, Japan
| | - Midori Iida
- Graduate School of Computer Science and System Engineering, Kyushu Institute of Technology, Iizuka, 820-0067, Japan
| | - Tetsuro Agusa
- Center for Marine Environmental Studies, Ehime University, Matsuyama, 790-8577, Japan
| | - Mari Ochiai
- Center for Marine Environmental Studies, Ehime University, Matsuyama, 790-8577, Japan
| | - Jiahua Guo
- Center for Marine Environmental Studies, Ehime University, Matsuyama, 790-8577, Japan
| | - Rajendiran Karthikraj
- Wadsworth Center, New York State Department of Health, Albany, NY, 12201-0509, United States
| | - Kurunthachalam Kannan
- Wadsworth Center, New York State Department of Health, Albany, NY, 12201-0509, United States
| | - Eun-Young Kim
- Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Hisato Iwata
- Center for Marine Environmental Studies, Ehime University, Matsuyama, 790-8577, Japan.
| |
Collapse
|
25
|
Ning Q, Wang Y, Zhang Y, Shen G, Xie Z, Pang J. Nuciferine Prevents Hepatic Steatosis by Regulating Lipid Metabolismin Diabetic Rat Model. Open Life Sci 2019; 14:699-706. [PMID: 33817209 PMCID: PMC7874802 DOI: 10.1515/biol-2019-0079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
Objective This study investigatesthe nuciferine capacity to regulate the liver’s lipid metabolism regarding steatosis and injury in STZ-induced diabetic rats. Materials and Methods The rats were randomly divided into groups control, diabetic and nuciferine 200 mg/kg/ day treatment. After 4 days of STZ injection, the nuciferine group was treated and administered via oral gavages for 4 weeks. At the end of experiment, blood, liver, myocardial and muscular samples were collected. Results Nuciferine-treated significantly increased the body weight from 339.4g to 367.8g, but significantly decreased the food and water intake compared with diabetic rats. Also, the nuciferine-treated rats had significantly decreased TC, TG, and FFAs in the liver compared with the diabetic group, especially the serum markers of blood glucose. These were associated with the gene expression related to lipogenesis which was significantly down-regulated; the gene expression involved in lipolysis and fatty acid β-oxidation was significantly up-regulated. Discussion and Conclusion The data provide evidence that nuciferine supplementation could protect the liver by regulating lipid metabolism gene expression resulting in decreasing the steatosis and injury in diabetic rat. Thus, nuciferine could be developed as a diabetic adjuvant food additive in future.
Collapse
Affiliation(s)
- Qian Ning
- Jinshan College of Fujian Agriculture and Forestry University, Fujian Province, 350001, P. R. China
| | - Yang Wang
- Jinshan College of Fujian Agriculture and Forestry University, Fujian Province, 350001, P. R. China
| | - Yi Zhang
- Jinshan College of Fujian Agriculture and Forestry University, Fujian Province, 350001, P. R. China
| | - Guozhi Shen
- Jinshan College of Fujian Agriculture and Forestry University, Fujian Province, 350001, P. R. China
| | - Zhenglu Xie
- Jinshan College of Fujian Agriculture and Forestry University, Fujian Province, 350001, P. R. China.,Collaborative Innovation Center of Animal Health and Food Safety Application Technology in Fujian, Fujian Vocational College of Agriculture, Fuzhou City, Fujian Province, 350002, P.R. China
| | - Jie Pang
- College of Food Science, Fujian Agriculture and Forestry University, Fujian Province, 350001, P. R. China
| |
Collapse
|
26
|
Shahzad K, Lopreiato V, Liang Y, Trevisi E, Osorio JS, Xu C, Loor JJ. Hepatic metabolomics and transcriptomics to study susceptibility to ketosis in response to prepartal nutritional management. J Anim Sci Biotechnol 2019; 10:96. [PMID: 31867104 PMCID: PMC6918647 DOI: 10.1186/s40104-019-0404-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022] Open
Abstract
Background Ketosis in dairy cows is associated with body fat mobilization during the peripartal period. Sub-clinical and clinical ketosis arise more frequently in cows that are overfed energy during the entire dry (last 50 to 45 days prior to parturition) or close-up period (last ~ 28 days prepartum). Methods A retrospective analysis was performed on 12 cows from a larger cohort that were fed a higher-energy diet [1.54 Mcal/kg of dry matter (DM); 35.9% of DM corn silage and 13% of DM ground corn] during the close-up dry period, of which 6 did not develop clinical ketosis (OVE, 0.83 mmol/L plasma hydroxybutyrate; BHB) and 6 were diagnosed with clinical ketosis (KET, 1.4 mmol/L BHB) during the first week postpartum. A whole-transcriptome bovine microarray (Agilent Technologies) and metabolomics (GC-MS, LC-MS; Metabolon® Inc.) were used to perform transcript and metabolite profiling of liver tissue harvested at − 10 days relative to parturition which allowed to establish potential associations between prepartal transcriptome/metabolome profiles and susceptibility to clinical ketosis postpartum. Results Cows in KET had greater (P = 0.01) overall body weight between − 2 and 1 week around parturition, but similar body condition score than OVE. Although dry matter intake (DMI) did not differ prepartum, KET cows had lower (P < 0.01) DMI and similar milk yield as OVE cows during the first week postpartum. Transcriptome analysis revealed a total of 3065 differentially expressed genes (DEG; P ≤ 0.05) in KET. Metabolomics identified 15 out of 313 total biochemical compounds significantly affected (P ≤ 0.10) in KET. Among those, greater concentrations (P ≤ 0.06, + 2.3-fold) of glycochenodeoxycholate in KET cows also have been detected in humans developing non-alcoholic fatty liver disease. Bioinformatics analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database and the DEG revealed that, among the top 20 most-impacted metabolic pathway categories in KET, 65% were overall downregulated. Those included ‘Metabolism of cofactors and vitamins’, ‘Biosynthesis of other secondary metabolites’, ‘Lipid’, ‘Carbohydrate’, and ‘Glycan biosynthesis and metabolism’. The lower relative concentration of glucose-6-phosphate and marked downregulation of fructose-1,6-bisphosphatase 2 and pyruvate dehydrogenase kinase 4 support a strong impairment in gluconeogenesis in prepartal liver of cows developing KET postpartum. Among the top 20 most-impacted non-metabolic pathways, 85% were downregulated. Pathways such as ‘mTOR signalling’ and ‘Insulin signalling’ were among those. ‘Ribosome’, ‘Nucleotide excision repair’, and ‘Adherens junctions’ were the only upregulated pathways in cows with KET. Conclusions The combined data analyses revealed more extensive alterations of the prepartal liver transcriptome than metabolome in cows overfed energy and developing ketosis postpartum. The causative link between these tissue-level adaptations and onset of clinical ketosis needs to be studied further.
Collapse
Affiliation(s)
- Khuram Shahzad
- 1COMSATS Institute of Information Technology, ChakShahzad, Islamabad, 44000 Pakistan.,2Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Vincenzo Lopreiato
- 3Istituto di Zootecnica, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Yusheng Liang
- 2Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Erminio Trevisi
- 3Istituto di Zootecnica, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Johan S Osorio
- 4Department of Dairy Science, South Dakota State University, Brookings, SD 57006 USA
| | - Chuang Xu
- 5College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Xinyang Rd. 5, Daqing, 163319 China
| | - Juan J Loor
- 2Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| |
Collapse
|
27
|
Taqi MO, Saeed‐Zidane M, Gebremedhn S, Salilew‐Wondim D, Khdrawy O, Rings F, Neuhoff C, Hoelker M, Schellander K, Tesfaye D. Sexual dimorphic expression and release of transcription factors in bovine embryos exposed to oxidative stress. Mol Reprod Dev 2019; 86:2005-2019. [DOI: 10.1002/mrd.23272] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Mohamed O. Taqi
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Mohammed Saeed‐Zidane
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Samuel Gebremedhn
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Dessie Salilew‐Wondim
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Omar Khdrawy
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Franca Rings
- Teaching and Research Station Frankenforst, Faculty of AgricultureUniversity of Bonn Königswinter Germany
- Center of Integrated Dairy Research, Faculty of AgricultureUniversity of Bonn Bonn Germany
| | - Christiane Neuhoff
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
| | - Michael Hoelker
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
- Teaching and Research Station Frankenforst, Faculty of AgricultureUniversity of Bonn Königswinter Germany
- Center of Integrated Dairy Research, Faculty of AgricultureUniversity of Bonn Bonn Germany
| | - Karl Schellander
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
- Center of Integrated Dairy Research, Faculty of AgricultureUniversity of Bonn Bonn Germany
| | - Dawit Tesfaye
- Department of Animal Breeding and Husbandry, Institute of Animal ScienceUniversity of Bonn Bonn Germany
- Center of Integrated Dairy Research, Faculty of AgricultureUniversity of Bonn Bonn Germany
| |
Collapse
|
28
|
Oat fiber inhibits atherosclerotic progression through improving lipid metabolism in ApoE−/− mice. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
29
|
Han S, Zhang W, Zhang R, Jiao J, Fu C, Tong X, Zhang W, Qin L. Cereal fiber improves blood cholesterol profiles and modulates intestinal cholesterol metabolism in C57BL/6 mice fed a high-fat, high-cholesterol diet. Food Nutr Res 2019; 63:1591. [PMID: 30863273 PMCID: PMC6403461 DOI: 10.29219/fnr.v63.1591] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/24/2018] [Accepted: 12/29/2018] [Indexed: 01/06/2023] Open
Abstract
Background Dietary intake of cereal fiber has been reported to benefit lipid metabolism through multiple mechanisms. The present study aimed to discover the potential mechanisms by which cereal fiber could modify the intestinal cholesterol metabolism. Design Male C57BL/6 mice were fed a reference chow (RC) diet; high-fat, high-cholesterol (HFC) diet; HFC plus oat fiber diet; or HFC plus wheat bran fiber diet for 24 weeks. Serum lipids were measured by enzymatic methods. Western blot was used to determine the protein expressions involved in intestinal cholesterol metabolism. Results Our results showed that HFC-induced elevations of serum triglycerides, total cholesterol, and low-density lipoprotein cholesterol were normalized in both groups that received cereal fiber. At the protein level, compared with the HFC diet group, the two cereal fibers, especially the oat fiber, significantly increased the protein expression of peroxisome proliferator-activated receptor alpha, liver X receptor alpha, sterol regulatory element-binding protein (SREBP) 2, low-density lipoprotein receptor, adenosine triphosphate (ATP)-binding cassette A1, and ATP-binding cassette G1, while decreasing the protein expression of Niemann-Pick C1-like protein 1, SREBP-1, fatty acid synthase, and acetyl-coenzyme A carboxylase, which were involved in intestinal cholesterol metabolism. Conclusion Taken together, increased intake of cereal fiber improved blood cholesterol profiles and increased the intestinal cholesterol efflux and cholesterol clearance in C57BL/6 mice fed a HFC diet. Oat fiber had a stronger effect than wheat bran fiber on cholesterol metabolism by modulating the PPARα, LXRα, and SREBP signaling pathways.
Collapse
Affiliation(s)
- Shufen Han
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Wei Zhang
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China.,Suzhou Maternal and Child Health Care and Family Planning Service Center, Suzhou, China
| | - Ru Zhang
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Jun Jiao
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Chunling Fu
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Xing Tong
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | | | - Liqiang Qin
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| |
Collapse
|
30
|
Park HY, Kang HS, Im SS. Recent insight into the correlation of SREBP-mediated lipid metabolism and innate immune response. J Mol Endocrinol 2018; 61:R123-R131. [PMID: 30307160 DOI: 10.1530/jme-17-0289] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fatty acids are essential nutrients that contribute to several intracellular functions. Fatty acid synthesis and oxidation are known to be regulated by sterol regulatory element-binding proteins (SREBPs), which play a pivotal role in the regulation of cellular triglyceride synthesis and cholesterol biogenesis. Recent studies point to a multifunctional role of SREBPs in the pathogenesis of metabolic diseases, such as obesity, type II diabetes and cancer as well as in immune responses. Notably, fatty acid metabolic intermediates are involved in energy homeostasis and pathophysiological conditions. In particular, intracellular fatty acid metabolism affects an inflammatory response, thereby influencing metabolic diseases. The objective of this review is to summarize the recent advances in our understanding of the dual role of SREBPs in both lipid metabolism and inflammation-mediated metabolic diseases.
Collapse
Affiliation(s)
- Hyeon Young Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Hye Suk Kang
- Department of Physiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, South Korea
| |
Collapse
|
31
|
Sterol regulatory element binding protein 1 trans-activates 25-hydroxy vitamin D3 24-hydroxylase gene expression in renal proximal tubular cells. Biochem Biophys Res Commun 2018; 500:275-282. [DOI: 10.1016/j.bbrc.2018.04.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/09/2018] [Indexed: 01/25/2023]
|
32
|
Menon B, Guo X, Garcia N, Gulappa T, Menon KMJ. miR-122 Regulates LHR Expression in Rat Granulosa Cells by Targeting Insig1 mRNA. Endocrinology 2018; 159:2075-2082. [PMID: 29579170 PMCID: PMC5905391 DOI: 10.1210/en.2017-03270] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/13/2018] [Indexed: 11/19/2022]
Abstract
Luteinizing hormone/chorionic gonadotropin receptor (LHR) expression in the ovary is regulated by a messenger RNA (mRNA) binding protein, which specifically binds to the coding region of LHR mRNA. We have shown that miR-122, a short noncoding RNA, mediates LHR mRNA levels by modulating the expression of LHR mRNA-binding protein (LRBP) through the regulation of sterol regulatory element binding protein (SREBP) activation. The present results show that miR-122 regulates LRBP levels by increasing the processing of SREBP through the degradation of Insig1, the anchoring protein of SREBP. We present evidence showing that mRNA and protein levels of Insig1 undergo a time-dependent increase following the treatment of rat granulosa cells with follicle-stimulating hormone (FSH), which leads to a decrease in LRBP levels. Furthermore, overexpression of miR-122 using an adenoviral vector (AdmiR-122) abolished FSH-induced increases in Insig1 mRNA and protein. We further confirmed the role of Insig1 by showing that inhibition of Insig1 using a specific small interfering RNA prior to FSH treatment resulted in the abrogation of LHR upregulation. Silencing of Insig1 also reversed FSH-mediated decreases in SREBP and LRBP activation. These results show that decreased levels of miR-122 increase Insig1 and suppress SREBP processing in response to FSH stimulation of rat granulosa cells.
Collapse
Affiliation(s)
- Bindu Menon
- Department of Obstetrics/Gynecology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
- Correspondence: K. M. J. Menon, PhD, 6428 Medical Science Building I, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, Michigan 48109. E-mail: ; or Bindu Menon, PhD, 6436 Medical Sciences Building 1, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, Michigan 48109. E-mail:
| | - Xingzi Guo
- Department of Obstetrics/Gynecology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
| | - Natalia Garcia
- Department of Obstetrics/Gynecology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
| | - Thippeswamy Gulappa
- Department of Obstetrics/Gynecology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
| | - K M J Menon
- Department of Obstetrics/Gynecology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
- Correspondence: K. M. J. Menon, PhD, 6428 Medical Science Building I, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, Michigan 48109. E-mail: ; or Bindu Menon, PhD, 6436 Medical Sciences Building 1, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, Michigan 48109. E-mail:
| |
Collapse
|
33
|
Golic M, Stojanovska V, Bendix I, Wehner A, Herse F, Haase N, Kräker K, Fischer C, Alenina N, Bader M, Schütte T, Schuchardt M, van der Giet M, Henrich W, Muller DN, Felderhoff-Müser U, Scherjon S, Plösch T, Dechend R. Diabetes Mellitus in Pregnancy Leads to Growth Restriction and Epigenetic Modification of the
Srebf2
Gene in Rat Fetuses. Hypertension 2018; 71:911-920. [DOI: 10.1161/hypertensionaha.117.10782] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/08/2018] [Accepted: 02/06/2018] [Indexed: 11/16/2022]
Abstract
Diabetic pregnancy is correlated with increased risk of metabolic and neurological disorders in the offspring putatively mediated epigenetically. Little is known about epigenetic changes already present in fetuses of diabetic pregnancies. We aimed at characterizing the perinatal environment after preexisting maternal diabetes mellitus and at identifying relevant epigenetic changes in the fetus. We focused on the transcription factor
Srebf2
(sterol regulatory element binding transcription factor 2), a master gene in regulation of cholesterol metabolism. We tested whether diabetic pregnancy induces epigenetic changes in the
Srebf2
promoter and if they become manifest in altered
Srebf2
gene expression. We worked with a transgenic rat model of type 2 diabetes mellitus (Tet29) in which the insulin receptor is knocked down by doxycycline-induced RNA interference. Doxycycline was administered preconceptionally to Tet29 and wild-type control rats. Only Tet29 doxycycline dams were hyperglycemic, hyperinsulinemic, and hyperlipidemic. Gene expression was analyzed with quantitative real-time reverse transcriptase polymerase chain reaction and CpG promoter methylation with pyrosequencing. Immunohistochemistry was performed on fetal brains. Fetuses from diabetic Tet29 dams were hyperglycemic and growth restricted at the end of pregnancy. They further displayed decreased liver and brain weight with concomitant decreased microglial activation in the hippocampus in comparison to fetuses of normoglycemic mothers. Importantly, diabetic pregnancy induced CpG hypermethylation of the
Srebf2
promoter in the fetal liver and brain, which was associated with decreased
Srebf2
gene expression. In conclusion, diabetic and hyperlipidemic pregnancy induces neurological, metabolic, and epigenetic alterations in the rat fetus.
Srebf2
is a potential candidate mediating intrauterine environment-driven epigenetic changes and later diabetic offspring health.
Collapse
Affiliation(s)
- Michaela Golic
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Violeta Stojanovska
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Ivo Bendix
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Anika Wehner
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Florian Herse
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Nadine Haase
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Kristin Kräker
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Caroline Fischer
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Natalia Alenina
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Michael Bader
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Till Schütte
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Mirjam Schuchardt
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Markus van der Giet
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Wolfgang Henrich
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Dominik N. Muller
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Ursula Felderhoff-Müser
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Sicco Scherjon
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Torsten Plösch
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| | - Ralf Dechend
- From the Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Germany (M.G., W.H.); Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Breast Center, Campus Charité Mitte, Germany (M.G.); Experimental and Clinical Research Center, a cooperation between the
| |
Collapse
|
34
|
The compositional and metabolic responses of gilthead seabream (Sparus aurata) to a gradient of dietary fish oil and associatedn-3 long-chain PUFA content. Br J Nutr 2017; 118:1010-1022. [DOI: 10.1017/s0007114517002975] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractThe replacement of fish oil (FO) with vegetable oil (VO) in feed formulations reduces the availability ofn-3 long-chain PUFA (LC-PUFA) to marine fish such as gilthead seabream. The aim of this study was to examine compositional and physiological responses to a dietary gradient ofn-3 LC-PUFA. Six iso-energetic and iso-nitrogenous diets (D1–D6) were fed to seabream, with the added oil being a blend of FO and VO to achieve a dietary gradient ofn-3 LC-PUFA. Fish were sampled after 4 months feeding, to determine biochemical composition, tissue fatty acid concentrations and lipid metabolic gene expression. The results indicated a disturbance to lipid metabolism, with fat in the liver increased and fat deposits in the viscera reduced. Tissue fatty acid profiles were altered towards the fatty acid compositions of the diets. There was evidence of endogenous modification of dietary PUFA in the liver which correlated with the expression of fatty acid desaturase 2 (fads2). Expression of sterol regulatory element binding protein 1 (srebp1), fads2and fatty acid synthase increased in the liver, whereas PPARα1 pathways appeared to be supressed by dietary VO in a concentration-dependent manner. The effects in lipogenic genes appear to become measurable in D1–D3, which agrees with the weight gain data suggesting that disturbances to energy metabolism and lipogenesis may be related to performance differences. These findings suggested that suppression ofβ-oxidation and stimulation ofsrebp1-mediated lipogenesis may play a role in contributing toward steatosis in fish fedn-3 LC-PUFA deficient diets.
Collapse
|
35
|
Sun JH, Liu X, Cong LX, Li H, Zhang CY, Chen JG, Wang CM. Metabolomics study of the therapeutic mechanism of Schisandra Chinensis lignans in diet-induced hyperlipidemia mice. Lipids Health Dis 2017; 16:145. [PMID: 28764799 PMCID: PMC5537938 DOI: 10.1186/s12944-017-0533-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/04/2017] [Indexed: 12/31/2022] Open
Abstract
Background Schisandra, a globally distributed plant, has been widely applied for the treatment of diseases such as hyperlipidemia, fatty liver and obesity in China. In the present work, a rapid resolution liquid chromatography coupled with quadruple-time-of-flight mass spectrometry (RRLC-Q-TOF-MS)-based metabolomics was conducted to investigate the intervention effect of Schisandra chinensis lignans (SCL) on hyperlipidemia mice induced by high-fat diet (HFD). Methods Hyperlipidemia mice were orally administered with SCL (100 mg/kg) once a day for 4 weeks. Serum biochemistry assay of triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-c) and high-density lipoprotein cholesterol (HDL-c) was conducted to confirm the treatment of SCL on lipid regulation. Metabolomics analysis on serum samples was carried out, and principal component analysis (PCA) and partial least squares-discriminant analysis (PLS-DA) were carried out for the pattern recognition and characteristic metabolites identification. The relative levels of critical regulatory factors of liver lipid metabolism, sterol regulatory element-binding proteins (SREBPs) and its related gene expressions were measured by quantitative real-time polymerase chain reaction (RT-PCR) for investigating the underlying mechanism. Results Oral administration of SCL significantly decreased the serum levels of TC, TG and LDL-c and increased the serum level of HDL-c in the hyperlipidemia mice, and no effect of SCL on blood lipid levels was observed in control mice. Serum samples were scattered in the PCA scores plots in response to the control, HFD and SCL group. Totally, thirteen biomarkers were identified and nine of them were recovered to the normal levels after SCL treatment. Based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis, the anti-hyperlipidemia mechanisms of SCL may be involved in the following metabolic pathways: tricarboxylic acid (TCA) cycle, synthesis of ketone body and cholesterol, choline metabolism and fatty acid metabolism. Meanwhile, SCL significantly inhibited the mRNA expression level of hepatic lipogenesis genes such as SREBP-1c, fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC), and decreased the mRNA expression of liver X receptor α (LXRα). Moreover, SCL also significantly decreased the expression level of SREBP-2 and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) in the liver of hyperlipidemia mice. Conclusion Anti-hyperlipidemia effect of SCL was confirmed by both serum biochemistry and metabolomics analysis. The mechanism may be related to the down-regulation of LXRα/SREBP-1c/FAS/ACC and SREBP2/HMGCR signaling pathways.
Collapse
Affiliation(s)
- Jing-Hui Sun
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, 132013, China
| | - Xu Liu
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, 132013, China
| | - Li-Xin Cong
- Second Treatment Area of Senile Disease, First Affiliated Hospital of Changchun University of Traditional Chinese Medicine, 1478, Gongnong Road, Changchun, 130021, China
| | - He Li
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, 132013, China
| | - Cheng-Yi Zhang
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, 132013, China
| | - Jian-Guang Chen
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, 132013, China.
| | - Chun-Mei Wang
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, 132013, China.
| |
Collapse
|
36
|
Siddiqa A, Cirillo E, Tareen SHK, Ali A, Kutmon M, Eijssen LMT, Ahmad J, Evelo CT, Coort SL. Visualizing the regulatory role of Angiopoietin-like protein 8 (ANGPTL8) in glucose and lipid metabolic pathways. Genomics 2017; 109:408-418. [PMID: 28684091 DOI: 10.1016/j.ygeno.2017.06.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 01/23/2023]
Abstract
ANGPTL8 (Angiopoietin-like protein 8) is a newly identified hormone emerging as a novel drug target for treatment of diabetes mellitus and dyslipidemia due to its unique metabolic nature. With increasing number of studies targeting the regulation of ANGPTL8, integration of their findings becomes indispensable. This study has been conducted with the aim to collect, analyze, integrate and visualize the available knowledge in the literature about ANGPTL8 and its regulation. We utilized this knowledge to construct a regulatory pathway of ANGPTL8 which is available at WikiPathways, an open source pathways database. It allows us to visualize ANGPTL8's regulation with respect to other genes/proteins in different pathways helping us to understand the complex interplay of novel hormones/genes/proteins in metabolic disorders. To the best of our knowledge, this is the first attempt to present an integrated pathway view of ANGPTL8's regulation and its associated pathways and is important resource for future omics-based studies.
Collapse
Affiliation(s)
- Amnah Siddiqa
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology, Pakistan; Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, The Netherlands
| | - Elisa Cirillo
- Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, The Netherlands
| | - Samar H K Tareen
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, The Netherlands
| | - Amjad Ali
- Atta-ur-Rahman School of Applied Biosciences - ASAB, National University of Sciences and Technology, Pakistan
| | - Martina Kutmon
- Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, The Netherlands; Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, The Netherlands
| | - Lars M T Eijssen
- Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, The Netherlands
| | - Jamil Ahmad
- Research Centre for Modeling and Simulation - RCMS, National University of Sciences and Technology, Pakistan.
| | - Chris T Evelo
- Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, The Netherlands; Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, The Netherlands
| | - Susan L Coort
- Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, The Netherlands
| |
Collapse
|
37
|
Li Y, Ma Z, Jiang S, Hu W, Li T, Di S, Wang D, Yang Y. A global perspective on FOXO1 in lipid metabolism and lipid-related diseases. Prog Lipid Res 2017; 66:42-49. [PMID: 28392404 DOI: 10.1016/j.plipres.2017.04.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023]
Abstract
Lipid metabolism is a complex physiological process that is involved in nutrient adjustment, hormone regulation, and homeostasis. An unhealthy lifestyle and chronic nutrient overload can cause lipid metabolism disorders, which may lead to serious lipid-related diseases, including obesity, non-alcoholic fatty liver disease (NAFLD), and type 2 diabetes mellitus (T2DM). Therefore, tools for preventing dysfunctional lipid metabolism are urgently needed. The transcription factor forkhead box protein O1 (FOXO1) is involved in lipid metabolism and plays a critical role in the development of lipid-related diseases. In this review, we provide a global perspective on the role of FOXO1 in lipid metabolism and lipid-related diseases. The information included here may be useful for the design of future studies and advancing investigations of FOXO1 as a therapeutic target.
Collapse
Affiliation(s)
- Yue Li
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China.
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
38
|
Jongbloed F, Saat TC, Verweij M, Payan-Gomez C, Hoeijmakers JHJ, van den Engel S, van Oostrom CT, Ambagtsheer G, Imholz S, Pennings JLA, van Steeg H, IJzermans JNM, Dollé MET, de Bruin RWF. A signature of renal stress resistance induced by short-term dietary restriction, fasting, and protein restriction. Sci Rep 2017; 7:40901. [PMID: 28102354 PMCID: PMC5244361 DOI: 10.1038/srep40901] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 12/14/2016] [Indexed: 11/09/2022] Open
Abstract
During kidney transplantation, ischemia-reperfusion injury (IRI) induces oxidative stress. Short-term preoperative 30% dietary restriction (DR) and 3-day fasting protect against renal IRI. We investigated the contribution of macronutrients to this protection on both phenotypical and transcriptional levels. Male C57BL/6 mice were fed control food ad libitum, underwent two weeks of 30%DR, 3-day fasting, or received a protein-, carbohydrate- or fat-free diet for various periods of time. After completion of each diet, renal gene expression was investigated using microarrays. After induction of renal IRI by clamping the renal pedicles, animals were monitored seven days postoperatively for signs of IRI. In addition to 3-day fasting and two weeks 30%DR, three days of a protein-free diet protected against renal IRI as well, whereas the other diets did not. Gene expression patterns significantly overlapped between all diets except the fat-free diet. Detailed meta-analysis showed involvement of nuclear receptor signaling via transcription factors, including FOXO3, HNF4A and HMGA1. In conclusion, three days of a protein-free diet is sufficient to induce protection against renal IRI similar to 3-day fasting and two weeks of 30%DR. The elucidated network of common protective pathways and transcription factors further improves our mechanistic insight into the increased stress resistance induced by short-term DR.
Collapse
Affiliation(s)
- F Jongbloed
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands.,Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - T C Saat
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M Verweij
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - C Payan-Gomez
- Department of Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - J H J Hoeijmakers
- Department of Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - S van den Engel
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - C T van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - G Ambagtsheer
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - S Imholz
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - J L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - H van Steeg
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands.,Department of Toxicogenetics, Leiden University Medical Center, Leiden, the Netherlands
| | - J N M IJzermans
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - R W F de Bruin
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
39
|
Flow signaling and atherosclerosis. Cell Mol Life Sci 2016; 74:1835-1858. [PMID: 28039525 PMCID: PMC5391278 DOI: 10.1007/s00018-016-2442-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 12/26/2022]
Abstract
Atherosclerosis rarely develops in the region of arteries exposed to undisturbed flow (u-flow, unidirectional flow). Instead, atherogenesis occurs in the area exposed to disturbed flow (d-flow, multidirectional flow). Based on these general pathohistological observations, u-flow is considered to be athero-protective, while d-flow is atherogenic. The fact that u-flow and d-flow induce such clearly different biological responses in the wall of large arteries indicates that these two types of flow activate each distinct intracellular signaling cascade in vascular endothelial cells (ECs), which are directly exposed to blood flow. The ability of ECs to differentially respond to the two types of flow provides an opportunity to identify molecular events that lead to endothelial dysfunction and atherosclerosis. In this review, we will focus on various molecular events, which are differentially regulated by these two flow types. We will discuss how various kinases, ER stress, inflammasome, SUMOylation, and DNA methylation play roles in the differential flow response, endothelial dysfunction, and atherosclerosis. We will also discuss the interplay among the molecular events and how they coordinately regulate flow-dependent signaling and cellular responses. It is hoped that clear understanding of the way how the two flow types beget each unique phenotype in ECs will lead us to possible points of intervention against endothelial dysfunction and cardiovascular diseases.
Collapse
|
40
|
Wang CM, Yuan RS, Zhuang WY, Sun JH, Wu JY, Li H, Chen JG. Schisandra polysaccharide inhibits hepatic lipid accumulation by downregulating expression of SREBPs in NAFLD mice. Lipids Health Dis 2016; 15:195. [PMID: 27852305 PMCID: PMC5112637 DOI: 10.1186/s12944-016-0358-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 10/28/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Hepatoprotective effects of Chinese herbal medicine Schisandra Chinensis (Schisandra) have been widely investigated. However, most studies were focused on its lignan extracts. We investigated the effects of Schisandra polysaccharide (SCP) in a mouse model of non-alcoholic fatty liver disease (NAFLD), and studied its effect on sterol regulatory element binding proteins (SREBPs) and the related genes. METHODS The mouse model of NAFLD was established by feeding mice with a high-fat diet for 16 weeks. Effect of SCP-treatment (100 mg/kg, once daily for 12 weeks) on biochemical parameters and liver histopathology was assessed. Relative levels of sterol regulatory element-binding proteins (SREBPs) and their gene expressions were determined by quantitative real-time polymerase chain reaction and Western Blot. RESULTS SCP significantly reduced the liver index by 12.0%. Serum levels of triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol, alanine aminotransferase and aspartate aminotransferase were decreased by 31.3, 28.3, 42.8, 20.1 and 15.5%, respectively. Serum high-density lipoprotein cholesterol was increased by 26.9%. Further, SCP lowered hepatic TC and TG content by 27.0% and 28.3%, respectively, and alleviated fatty degeneration and necrosis of liver cells. A significant downregulation of mRNA and protein expressions of hepatic lipogenesis genes, SREBP-1c, fatty acid synthase and acetyl-CoA carboxylase, and the mRNA expression of liver X receptor α (LXRα) was observed in NAFLD mice treated with SCP. SCP also significantly reduced the hepatic expression of SREBP-2 and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). CONCLUSION These findings demonstrate the hepatoprotective effects of SCP in a mouse model of NAFLD; the effects may be mediated via downregulation of LXRα/SREBP-1c/FAS/ACC and SREBP-2/HMGCR signaling pathways in the liver.
Collapse
Affiliation(s)
- Chun-Mei Wang
- Department of Pharmacology, College of Pharmacy, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, People's Republic of China
| | - Rong-Shuang Yuan
- Department of Pharmacology, College of Pharmacy, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, People's Republic of China
| | - Wen-Yue Zhuang
- Department of Molecular Biology, College of Laboratory Medicine, Beihua University, Jilin, Jilin, 132013, China
| | - Jing-Hui Sun
- Department of Pharmacology, College of Pharmacy, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, People's Republic of China
| | - Jin-Ying Wu
- Department of Pharmacology, College of Pharmacy, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, People's Republic of China
| | - He Li
- Department of Pharmacology, College of Pharmacy, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, People's Republic of China.
| | - Jian-Guang Chen
- Department of Pharmacology, College of Pharmacy, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, People's Republic of China.
| |
Collapse
|
41
|
Sun S, Wen J, Qiu F, Yin Y, Xu G, Li T, Nie J, Xiong G, Zhang C, Liao D, Chen J, Tuo Q. Identification of the C-terminal domain of Daxx acts as a potential regulator of intracellular cholesterol synthesis in HepG2 cells. Biochem Biophys Res Commun 2016; 480:139-145. [PMID: 27671201 DOI: 10.1016/j.bbrc.2016.09.102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 12/12/2022]
Abstract
Daxx is a highly conserved nuclear transcriptional factor, which has been implicated in many nuclear processes including transcription and cell cycle regulation. Our previous study demonstrated Daxx also plays a role in regulation of intracellular cholesterol content. Daxx contains several domains that are essential for interaction with a growing number of proteins. To delineate the underlying mechanism of hypocholesterolemic activity of Daxx, we constructed a set of plasmids which can be used to overexpress different fragments of Daxx and transfected to HepG2 cells. We found that the C- terminal region Daxx626-740 clearly reduced intracellular cholesterol levels and inhibited the expression of SREBPs and SCAP. In GST pull-down experiments and Double immunofluorescence assays, Daxx626-740 was demonstrated to bind directly to androgen receptor (AR). Our findings suggest that the interaction of Daxx626-740 and AR abolishes the AR-mediated activation of SCAP/SREBPs pathway, which suppresses the de novo cholesterol synthesis. Thus, C-terminal domain of Daxx acts as a potential regulator of intracellular cholesterol content in HepG2 cells.
Collapse
Affiliation(s)
- Shaowei Sun
- Institute of Pharmacy and Pharmacology, School of Life Science and Technology, University of South China, Hengyang 421001, Hunan, China; Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Juan Wen
- Institute of Pharmacy and Pharmacology, School of Life Science and Technology, University of South China, Hengyang 421001, Hunan, China
| | - Fei Qiu
- Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Yufang Yin
- Department of Pharmacology, Medical School of Southern Illinois University, Springfield, IL 62702, USA
| | - Guina Xu
- Institute of Pharmacy and Pharmacology, School of Life Science and Technology, University of South China, Hengyang 421001, Hunan, China
| | - Tianping Li
- Institute of Pharmacy and Pharmacology, School of Life Science and Technology, University of South China, Hengyang 421001, Hunan, China
| | - Juan Nie
- Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Guozuo Xiong
- Institute of Pharmacy and Pharmacology, School of Life Science and Technology, University of South China, Hengyang 421001, Hunan, China
| | - Caiping Zhang
- Institute of Pharmacy and Pharmacology, School of Life Science and Technology, University of South China, Hengyang 421001, Hunan, China
| | - Duangfang Liao
- Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Jianxiong Chen
- Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Qinhui Tuo
- Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| |
Collapse
|
42
|
Leskinen H, Viitala S, Mutikainen M, Kairenius P, Tapio I, Taponen J, Bernard L, Vilkki J, Shingfield KJ. Ruminal Infusions of Cobalt EDTA Modify Milk Fatty Acid Composition via Decreases in Fatty Acid Desaturation and Altered Gene Expression in the Mammary Gland of Lactating Cows. J Nutr 2016; 146:976-85. [PMID: 27075908 DOI: 10.3945/jn.115.226100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 03/01/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Intravenous or ruminal infusion of lithium salt of cobalt EDTA (Co-EDTA) or cobalt-acetate alters milk fat composition in cattle, but the mechanisms involved are not known. OBJECTIVE The present study evaluated the effect of ruminal Co-EDTA infusion on milk FA composition, mammary lipid metabolism, and mammary lipogenic gene expression. METHODS For the experiment, 4 cows in midlactation and fitted with rumen cannulae were used in a 4 × 4 Latin square with 28-d periods. Co-EDTA was administered in the rumen to supply 0, 1.5, 3.0, or 4.5 g Co/d over an 18-d interval with a 10-d washout between experimental periods. Milk production was recorded daily, and milk FA composition was determined on alternate days. Mammary tissue was biopsied on day 16, and arteriovenous differences of circulating lipid fractions and FA uptake across the mammary gland were measured on day 18. RESULTS Co-EDTA had no effect on intake, proportions of rumen volatile FA, or milk production but caused dose-dependent changes in milk FA composition. Alterations in milk fat composition were evident within 3 d of infusion and characterized by linear or quadratic decreases (P < 0.05) in FAs containing a cis-9 double bond, an increase in 4:0 and 16:0, and linear decreases in milk 8:0, 10:0, 12:0, and 14:0 concentrations. Co-EDTA progressively decreased (P < 0.05) the stearoyl-CoA desaturase (SCD)-catalyzed desaturation of FAs in the mammary gland by up to 72% but had no effect on mammary SCD1 mRNA or SCD protein abundance. Changes in milk FA composition were accompanied by altered expression of specific genes involved in de novo FA and triacylglycerol synthesis. CONCLUSION Ruminal infusion of Co-EDTA alters milk FA composition in cattle via a mechanism that involves decreases in the desaturation of FAs synthesized de novo or extracted from blood and alterations in mammary lipogenic gene expression, without affecting milk fat yield.
Collapse
Affiliation(s)
- Heidi Leskinen
- Green Technology, Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Sirja Viitala
- Green Technology, Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Mervi Mutikainen
- Green Technology, Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Piia Kairenius
- Green Technology, Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Ilma Tapio
- Green Technology, Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Juhani Taponen
- Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Laurence Bernard
- Adipose Tissue and Milk Lipid Laboratory, Herbivore Research Unit, INRA-Theix, Saint-Genès-Champanelle, France; and
| | - Johanna Vilkki
- Green Technology, Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Kevin J Shingfield
- Green Technology, Natural Resources Institute Finland (Luke), Jokioinen, Finland; Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| |
Collapse
|
43
|
Li M, Ye T, Wang XX, Li X, Qiang O, Yu T, Tang CW, Liu R. Effect of Octreotide on Hepatic Steatosis in Diet-Induced Obesity in Rats. PLoS One 2016; 11:e0152085. [PMID: 27002331 PMCID: PMC4803296 DOI: 10.1371/journal.pone.0152085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 03/08/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) caused by liver lipid dysregulation is linked to obesity. Somatostatin (SST) and its analogs have been used to treat pediatric hypothalamic obesity. However, the application of such drugs for the treatment of NAFLD has not been evaluated. OBJECTIVE This study aimed to investigate the expression levels of important regulators of hepatic lipid metabolism and the possible effect of the SST analog octreotide on these regulators. METHODS SD rats were assigned to a control group and a high-fat diet group. Obese rats from the high-fat diet group were further divided into the obese and octreotide-treated groups. The body weight, plasma SST, fasting plasma glucose (FPG), insulin, triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C) and free fatty acid (FFA) levels were measured. Hepatic steatosis was evaluated based on the liver TG content, HE staining and oil red O staining. The SREBP-1c, ACC1, FAS, MTP, apoB and ADRP expression levels in the liver were also determined by RT-PCR, qRT-PCR, western blot or ELISA. RESULTS The obese rats induced by high-fat diet expressed more SREBP-1c, FAS and ADRP but less MTP protein in the liver than those of control rats, whereas octreotide intervention reversed these changes and increased the level of apoB protein. Compared to the control group, obese rats showed increased liver ACC1, SREBP-1c and apoB mRNA levels, whereas octreotide-treated rats showed decreased mRNA levels of apoB and SREBP-1c. This was accompanied by increased body weight, liver TG contents, FPG, TG, TC, LDL-C, FFA, insulin and derived homeostatic model assessment (HOMA) values. Octreotide intervention significantly decreased these parameters. Compared to the control group, the obese group showed a decreasing trend on plasma SST levels, which were significantly increased by the octreotide intervention. CONCLUSION Octreotide can ameliorate hepatic steatosis in obese rats, possibly by decreasing hepatic lipogenesis and increasing TG export from hepatocytes.
Collapse
Affiliation(s)
- Mao Li
- Division of Peptides Related to Human Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Ye
- Division of Peptides Related to Human Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao-Xia Wang
- Division of Peptides Related to Human Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Li
- Division of Peptides Related to Human Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ou Qiang
- Division of Peptides Related to Human Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Yu
- Division of Peptides Related to Human Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng-Wei Tang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Liu
- Division of Peptides Related to Human Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Han S, Jiao J, Zhang W, Xu J, Wan Z, Zhang W, Gao X, Qin L. Dietary fiber prevents obesity-related liver lipotoxicity by modulating sterol-regulatory element binding protein pathway in C57BL/6J mice fed a high-fat/cholesterol diet. Sci Rep 2015; 5:15256. [PMID: 26510459 PMCID: PMC4625144 DOI: 10.1038/srep15256] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 09/16/2015] [Indexed: 01/27/2023] Open
Abstract
Adequate intake of dietary fibers has proven metabolic and cardiovascular benefits, molecular mechanisms remain still limited. This study was aimed to investigate the effects of cereal dietary fiber on obesity-related liver lipotoxicity in C57BL/6J mice fed a high-fat/cholesterol (HFC) diet and underlying mechanism. Forty-eight adult male C57BL/6J mice were randomly given a reference chow diet, or a high fat/cholesterol (HFC) diet supplemented with or without oat fiber or wheat bran fiber for 24 weeks. Our results showed mice fed oat or wheat bran fiber exhibited lower weight gain, lipid profiles and insulin resistance, compared with HFC diet. The two cereal dietary fibers potently decreased protein expressions of sterol regulatory element binding protein-1 and key factors involved in lipogenesis, including fatty acid synthase and acetyl-CoA carboxylase in target tissues. At molecular level, the two cereal dietary fibers augmented protein expressions of peroxisome proliferator-activated receptor alpha and gamma, liver X receptor alpha, and ATP-binding cassette transporter A1 in target tissues. Our findings indicated that cereal dietary fiber supplementation abrogated obesity-related liver lipotoxicity and dyslipidemia in C57BL/6J mice fed a HFC diet. In addition, the efficacy of oat fiber is greater than wheat bran fiber in normalizing these metabolic disorders and pathological profiles.
Collapse
Affiliation(s)
- Shufen Han
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, 199 Renai Road, Suzhou City 215123, China
| | - Jun Jiao
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, 199 Renai Road, Suzhou City 215123, China
| | - Wei Zhang
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, 199 Renai Road, Suzhou City 215123, China
| | - Jiaying Xu
- Key Laboratory of Radiation Biology, School of Radiation Medicine and Protection, Soochow University, 199 Renai Road, Suzhou City 215123, China
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, 199 Renai Road, Suzhou City 215123, China
| | - Weiguo Zhang
- DSM Nutritional Products Human Nutrition and Health, Unite 5-7, Floor 8, Tower C, Parkview Green, 9 Dongdaqiao Road, Beijing 100020, China
| | - Xiaoran Gao
- DSM Nutritional Products Human Nutrition and Health, 476 Libing Road, Shanghai 201203, China
| | - Liqiang Qin
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, 199 Renai Road, Suzhou City 215123, China
| |
Collapse
|
45
|
Feng RB, Fan CL, Liu Q, Liu Z, Zhang W, Li YL, Tang W, Wang Y, Li MM, Ye WC. Crude triterpenoid saponins from Ilex latifolia (Da Ye Dong Qing) ameliorate lipid accumulation by inhibiting SREBP expression via activation of AMPK in a non-alcoholic fatty liver disease model. Chin Med 2015; 10:23. [PMID: 26300958 PMCID: PMC4544818 DOI: 10.1186/s13020-015-0054-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 07/28/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Ilex latifolia Thunb. (Da Ye Dong Qing) is used for weight loss and for its antidiabetic effects. This study aims to investigate the beneficial effects and potential mechanisms of action of crude triterpenoid saponins (CTS) from I. latifolia in a mouse model of high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD). METHODS Male C57BL/6 mice (n = 50), were arbitrarily divided into five groups (n = 10 in each group): a control group, HFD group, simvastatin group (10 mg/kg/day), and two CTS treatment groups (100 and 200 mg/kg/day). All mice except those in the control group were fed an HFD for 4 weeks. Animals in the treatment groups were orally administered simvastatin or CTS for 8 weeks. Oral glucose tolerance tests and insulin tolerance tests were performed. At the end of treatment, plasma lipid levels, and oxidative parameters in the liver were measured using commercial test kits. Western blotting was used to evaluate whether CTS induced AMP-activated protein kinase (AMPK) and acetyl CoA carboxylase activation, and the expression of transcription factors and their target genes was evaluated in a quantitative PCR assay. RESULTS Compared with the HFD group, the CTS (200 mg/kg/day) treatment group showed significantly decreased plasma lipid parameters (P < 0.001, P = 0.018, and P = 0.005 for triglycerides, total cholesterol and low-density lipoprotein cholesterol, respectively), and improved insulin resistance (P = 0.006). CTS (100 and 200 mg/kg/day) supplementation also reduced hepatic lipids and protected the liver from oxidative stress by attenuating malondialdehyde content (P < 0.001 and P < 0.001, respectively) and restoring aspartate aminotransferase levels (P < 0.001 and P < 0.001, respectively). Moreover, CTS (200 mg/kg/day) reduced lipid accumulation by enhancing AMPK phosphorylation and inhibiting expression of sterol regulatory element-binding proteins (SREBPs) and their target genes SREBP-1c, SREBP-2, fatty acid synthase, and stearoyl-CoA desaturase (P = 0.013, P = 0.007, P = 0.011, and P = 0.014, respectively). CONCLUSION CTS from I. latifolia improved insulin resistance and liver injury in HFD-fed mice, and attenuated NAFLD via the activation of AMPK and inhibition of the gene expression of SREBPs and some of their target molecules.
Collapse
Affiliation(s)
- Rui-Bing Feng
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Chun-Lin Fan
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Qing Liu
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Zhong Liu
- />Guangzhou Jinan Biomedicine Research and Development Center, National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Wei Zhang
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Yao-Lan Li
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Wei Tang
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Ying Wang
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Man-Mei Li
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| | - Wen-Cai Ye
- />College of Pharmacy, Jinan University, Guangzhou, 510632 People’s Republic of China
- />Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632 People’s Republic of China
| |
Collapse
|
46
|
Apodaca G, Brown WJ. Membrane traffic research: challenges for the next decade. Front Cell Dev Biol 2014; 2:52. [PMID: 25364759 PMCID: PMC4207031 DOI: 10.3389/fcell.2014.00052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/02/2014] [Indexed: 01/26/2023] Open
Affiliation(s)
- Gerard Apodaca
- The Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh Pittsburgh, PA, USA
| | - William J Brown
- Molecular Biology and Genetics, Cornell University Ithaca, NY, USA
| |
Collapse
|
47
|
Ezetimibe suppresses cholesterol accumulation in lipid-loaded vascular smooth muscle cells in vitro via MAPK signaling. Acta Pharmacol Sin 2014; 35:1129-36. [PMID: 25087996 DOI: 10.1038/aps.2014.10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 07/14/2014] [Indexed: 01/30/2023]
Abstract
AIM To investigate the mechanisms of anti-atherosclerotic action of ezetimibe in rat vascular smooth muscle cells (VSMCs) in vitro. METHODS VSMCs of SD rats were cultured in the presence of Chol:MβCD (10 μg/mL) for 72 h, and intracellular lipid droplets and cholesterol levels were evaluated using Oil Red O staining, HPLC and Enzymatic Fluorescence Assay, respectively. The expression of caveolin-1, sterol response element-binding protein-1 (SREBP-1) and ERK1/2 were analyzed using Western blot assays. Translocation of SREBP-1 and ERK1/2 was detected with immunofluorescence. RESULTS Treatment with Chol:MβCD dramatically increased the cellular levels of total cholesterol (TC), cholesterol ester (CE) and free cholesterol (FC) in VSMCs, which led to the formation of foam cells. Furthermore, Chol:MβCD treatment significantly decreased the expression of caveolin-1, and stimulated the expression and nuclear translocation of SREBP-1 in VSMCs. Co-treatment with ezetimibe (3 μmol/L) significantly decreased the cellular levels of TC, CE and FC, which was accompanied by elevation of caveolin-1 expression, and by a reduction of SREBP-1 expression and nuclear translocation. Co-treatment with ezetimibe dose-dependently decreased the expression of phosphor-ERK1/2 (p-ERK1/2) in VSMCs. The ERK1/2 inhibitor PD98059 (50 μmol/L) altered the cholesterol level and the expression of p-ERK1/2, SREBP-1 and caveolin-1 in the same manner as ezetimibe did. CONCLUSION Ezetimibe suppresses cholesterol accumulation in rat VSMCs in vitro by regulating SREBP-1 and caveolin-1 expression, possibly via the MAPK signaling pathway.
Collapse
|
48
|
Yang Y, Li W, Liu Y, Sun Y, Li Y, Yao Q, Li J, Zhang Q, Gao Y, Gao L, Zhao J. Alpha-lipoic acid improves high-fat diet-induced hepatic steatosis by modulating the transcription factors SREBP-1, FoxO1 and Nrf2 via the SIRT1/LKB1/AMPK pathway. J Nutr Biochem 2014; 25:1207-1217. [PMID: 25123628 DOI: 10.1016/j.jnutbio.2014.06.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/27/2014] [Accepted: 06/01/2014] [Indexed: 02/07/2023]
Abstract
Understanding the mechanism by which alpha-lipoic acid supplementation has a protective effect upon nonalcoholic fatty liver disease in vivo and in vitro may lead to targets for preventing hepatic steatosis. Male C57BL/6J mice were fed a normal diet, high-fat diet or high-fat diet supplemented with alpha-lipoic acid for 24 weeks. HepG2 cells were incubated with normal medium, palmitate or alpha-lipoic acid. The lipid-lowering effects were measured. The protein expression and distribution were analyzed by Western blot, immunoprecipitation and immunofluorescence, respectively. We found that alpha-lipoic acid enhanced sirtuin 1 deacetylase activity through liver kinase B1 and stimulated AMP-activated protein kinase. By activating the sirtuin 1/liver kinase B1/AMP-activated protein kinase pathway, the translocation of sterol regulatory element-binding protein-1 into the nucleus and forkhead box O1 into the cytoplasm was prevented. Alpha-lipoic acid increased adipose triacylglycerol lipase expression and decreased fatty acid synthase abundance. In in vivo and in vitro studies, alpha-lipoic acid also increased nuclear NF-E2-related factor 2 levels and downstream target amounts via the sirtuin 1 pathway. Alpha-lipoic acid eventually reduced intrahepatic and serum triglyceride content. The protective effects of alpha-lipoic acid on hepatic steatosis appear to be associated with the transcription factors sterol regulatory element-binding protein-1, forkhead box O1 and NF-E2-related factor 2.
Collapse
Affiliation(s)
- Yi Yang
- Department of Endocrinology, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China.
| | - Wang Li
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yang Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yuning Sun
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yan Li
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Qing Yao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Jianning Li
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Qian Zhang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yujing Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Ling Gao
- Central Lab. Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Jiajun Zhao
- Department of Endocrinology, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Institute of Endocrinology, Shandong Academy of Clinical Medicine, Jinan 250021, China.
| |
Collapse
|
49
|
Kessler EC, Gross JJ, Bruckmaier RM, Albrecht C. Cholesterol metabolism, transport, and hepatic regulation in dairy cows during transition and early lactation. J Dairy Sci 2014; 97:5481-90. [PMID: 24952770 DOI: 10.3168/jds.2014-7926] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 05/07/2014] [Indexed: 11/19/2022]
Abstract
The transition from the nonlactating to the lactating state represents a critical period for dairy cow lipid metabolism because body reserves have to be mobilized to meet the increasing energy requirements for the initiation of milk production. The purpose of this study was to provide a comprehensive overview on cholesterol homeostasis in transition dairy cows by assessing in parallel plasma, milk, and hepatic tissue for key factors of cholesterol metabolism, transport, and regulation. Blood samples and liver biopsies were taken from 50 multiparous Holstein dairy cows in wk 3 antepartum (a.p.), wk 1 postpartum (p.p.), wk 4 p.p., and wk 14 p.p. Milk sampling was performed in wk 1, 4, and 14 p.p. Blood and milk lipid concentrations [triglycerides (TG), cholesterol, and lipoproteins], enzyme activities (phospholipid transfer protein and lecithin:cholesterol acyltransferase) were analyzed using enzymatic assays. Hepatic gene expression patterns of 3-hydroxy-3-methylglutaryl-coenzyme A (HMGC) synthase 1 (HMGCS1) and HMGC reductase (HMGCR), sterol regulatory element-binding factor (SREBF)-1 and -2, microsomal triglyceride transfer protein (MTTP), ATP-binding cassette transporter (ABC) A1 and ABCG1, liver X receptor (LXR) α and peroxisome proliferator activated receptor (PPAR) α and γ were measured using quantitative RT-PCR. Plasma TG, cholesterol, and lipoprotein concentrations decreased from wk 3 a.p. to a minimum in wk 1 p.p., and then gradually increased until wk 14 p.p. Compared with wk 4 p.p., phospholipid transfer protein activity was increased in wk 1 p.p., whereas lecithin:cholesterol acyltransferase activity was lowest at this period. Total cholesterol concentration and mass, and cholesterol concentration in the milk fat fraction decreased from wk 1 p.p. to wk 4 p.p. Both total and milk fat cholesterol concentration were decreased in wk 4 p.p. compared with wk 1 and 14 p.p. The mRNA abundance of genes involved in cholesterol synthesis (SREBF-2, HMGCS1, and HMGCR) markedly increased from wk 3 a.p. to wk 1 p.p., whereas SREBF-1 was downregulated. The expression of ABCA1 increased from wk 3 a.p. to wk 1 p.p., whereas ABCG1 was increased in wk 14 p.p. compared with other time points. In conclusion, hepatic expression of genes involved in the biosynthesis of cholesterol as well as the ABCA1 transporter were upregulated at the onset of lactation, whereas plasma concentrations of total cholesterol, phospholipids, lipoprotein-cholesterol, and TG were at a minimum. Thus, at the gene expression level, the liver seems to react to the increased demand for cholesterol after parturition. Whether the low plasma cholesterol and TG levels are due to impaired hepatic export mechanisms or reflect an enhanced transfer of these compounds into the milk to provide essential nutrients for the newborn remains to be elucidated.
Collapse
Affiliation(s)
- E C Kessler
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland
| | - J J Gross
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland
| | - R M Bruckmaier
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland
| | - C Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland; Swiss National Center of Competence in Research (NCCR) TransCure, University of Bern, CH-3012 Bern, Switzerland.
| |
Collapse
|
50
|
Leonard MO, Limonciel A, Jennings P. Stress Response Pathways. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2014. [DOI: 10.1007/978-1-4939-0521-8_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|