1
|
Chen H, Wan X, He Q, Xiao G, Zheng Y, Luo M, Yang C, Ren D, Lu L, Peng H, Lin H. Single-cell RNA sequencing reveals cellular dynamics and therapeutic effects of astragaloside IV in slow transit constipation. BIOMOLECULES & BIOMEDICINE 2024; 24:871-887. [PMID: 38289380 PMCID: PMC11293215 DOI: 10.17305/bb.2024.10187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 07/31/2024]
Abstract
The cellular characteristics of intestinal cells involved in the therapeutic effects of astragaloside IV (AS-IV) for treating slow transit constipation (STC) remain unclear. This study aimed to determine the dynamics of colon tissue cells in the STC model and investigate the effects of AS-IV treatment by single-cell RNA sequencing (scRNA-seq). STC mouse models were developed using loperamide, with subsequent treatment using AS-IV. Colon tissues and feces were collected for scRNA-seq and targeted short-chain fatty acid quantification. We integrated scRNA-seq data with network pharmacology to analyze the effect of AS-IV on constipation. AS-IV showed improvement in defecation for STC mice induced by loperamide. Notably, in STC mice, epithelial cells, T cells, B cells, and fibroblasts demonstrated alterations in cell proportions and aberrant functions, which AS-IV partially rectified. AS-IV has the potential to modulate the metabolic pathway of epithelial cells through its interaction with peroxisome proliferator-activated receptor gamma (PPARγ). AS-IV reinstated fecal butyrate levels and improved energy metabolism in epithelial cells. The proportion of naïve CD4+T cells is elevated in STC, and the differentiation of these cells into regulatory T cells (Treg) is regulated by B cells and fibroblasts through the interaction of ligand-receptor pairs. AS-IV treatment can partially alleviate this trend. The status of fibroblasts in STC undergoes alterations, and the FB_C4_Adamdec1 subset, associated with angiogenesis and the Wingless-related integration (Wnt) pathway, emerges. Our comprehensive analysis identifies perturbations of epithelial cells and tissue microenvironment cells in STC and elucidates mechanisms underlying the therapeutic efficacy of AS-IV.
Collapse
Affiliation(s)
- Huaxian Chen
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xingyang Wan
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiulan He
- Department of Anaesthesiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guozhong Xiao
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yihui Zheng
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minyi Luo
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chaoxin Yang
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Donglin Ren
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Lu
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Peng
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongcheng Lin
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Roussel C, Sola M, Lessard-Lord J, Nallabelli N, Généreux P, Cavestri C, Azeggouar Wallen O, Villano R, Raymond F, Flamand N, Silvestri C, Di Marzo V. Human gut microbiota and their production of endocannabinoid-like mediators are directly affected by a dietary oil. Gut Microbes 2024; 16:2335879. [PMID: 38695302 PMCID: PMC11067990 DOI: 10.1080/19490976.2024.2335879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
Dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) and the gut microbiome affect each other. We investigated the impact of supplementation with Buglossoides arvensis oil (BO), rich in stearidonic acid (SDA), on the human gut microbiome. Employing the Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME), we simulated the ileal and ascending colon microbiomes of four donors. Our results reveal two distinct microbiota clusters influenced by BO, exhibiting shared and contrasting shifts. Notably, Bacteroides and Clostridia abundance underwent similar changes in both clusters, accompanied by increased propionate production in the colon. However, in the ileum, cluster 2 displayed a higher metabolic activity in terms of BO-induced propionate levels. Accordingly, a triad of bacterial members involved in propionate production through the succinate pathway, namely Bacteroides, Parabacteroides, and Phascolarctobacterium, was identified particularly in this cluster, which also showed a surge of second-generation probiotics, such as Akkermansia, in the colon. Finally, we describe for the first time the capability of gut bacteria to produce N-acyl-ethanolamines, and particularly the SDA-derived N-stearidonoyl-ethanolamine, following BO supplementation, which also stimulated the production of another bioactive endocannabinoid-like molecule, commendamide, in both cases with variations across individuals. Spearman correlations enabled the identification of bacterial genera potentially involved in endocannabinoid-like molecule production, such as, in agreement with previous reports, Bacteroides in the case of commendamide. This study suggests that the potential health benefits on the human microbiome of certain dietary oils may be amenable to stratified nutrition strategies and extend beyond n-3 PUFAs to include microbiota-derived endocannabinoid-like mediators.
Collapse
Affiliation(s)
- Charlène Roussel
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Mathilde Sola
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Jacob Lessard-Lord
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
| | - Nayudu Nallabelli
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Pamela Généreux
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
| | - Camille Cavestri
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
| | - Oumaima Azeggouar Wallen
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Rosaria Villano
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli (Napoli), Italy
| | - Frédéric Raymond
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Nicolas Flamand
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Cristoforo Silvestri
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Vincenzo Di Marzo
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| |
Collapse
|
3
|
Crawford CK, Beltran A, Castillo D, Matloob MS, Uehara ME, Quilici ML, Cervantes VL, Kol A. Fenofibrate reduces glucose-induced barrier dysfunction in feline enteroids. Sci Rep 2023; 13:22558. [PMID: 38110453 PMCID: PMC10728136 DOI: 10.1038/s41598-023-49874-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023] Open
Abstract
Diabetes mellitus (DM) is a common chronic metabolic disease in humans and household cats that is characterized by persistent hyperglycemia. DM is associated with dysfunction of the intestinal barrier. This barrier is comprised of an epithelial monolayer that contains a network of tight junctions that adjoin cells and regulate paracellular movement of water and solutes. The mechanisms driving DM-associated barrier dysfunction are multifaceted, and the direct effects of hyperglycemia on the epithelium are poorly understood. Preliminary data suggest that fenofibrate, An FDA-approved peroxisome proliferator-activated receptor-alpha (PPARα) agonist drug attenuates intestinal barrier dysfunction in dogs with experimentally-induced DM. We investigated the effects of hyperglycemia-like conditions and fenofibrate treatment on epithelial barrier function using feline intestinal organoids. We hypothesized that glucose treatment directly increases barrier permeability and alters tight junction morphology, and that fenofibrate administration can ameliorate these deleterious effects. We show that hyperglycemia-like conditions directly increase intestinal epithelial permeability, which is mitigated by fenofibrate. Moreover, increased permeability is caused by disruption of tight junctions, as evident by increased junctional tortuosity. Finally, we found that increased junctional tortuosity and barrier permeability in hyperglycemic conditions were associated with increased protein kinase C-α (PKCα) activity, and that fenofibrate treatment restored PKCα activity to baseline levels. We conclude that hyperglycemia directly induces barrier dysfunction by disrupting tight junction structure, a process that is mitigated by fenofibrate. We further propose that counteracting modulation of PKCα activation by increased intracellular glucose levels and fenofibrate is a key candidate regulatory pathway of tight junction structure and epithelial permeability.
Collapse
Affiliation(s)
- Charles K Crawford
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Aeelin Beltran
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Diego Castillo
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Muhammad S Matloob
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Mimoli E Uehara
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Mary L Quilici
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Veronica Lopez Cervantes
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Amir Kol
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
4
|
Gora AH, Rehman S, Dias J, Fernandes JMO, Olsvik PA, Sørensen M, Kiron V. Microbial oil, alone or paired with β-glucans, can control hypercholesterolemia in a zebrafish model. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159383. [PMID: 37657755 DOI: 10.1016/j.bbalip.2023.159383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/12/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
Dyslipidemia is often associated with unhealthy dietary habits, and many mammalian studies have explored the mode of action of certain bioactive compounds such as β-glucans and n-3 PUFAs to understand their potential to normalize the lipid metabolism. There are only a few investigations that adopted omic approaches to unveil their combined effect on hypercholesterolemia. Zebrafish (Danio rerio) was used as a model organism to reveal the efficacy of Schizochytrium oil and β-glucans (from Euglena gracilis and Phaeodactylum tricornutum) against cholesterol-rich diet induced dyslipidemia. One of the folowing four diets was fed to a particular group of fish: a control high-cholesterol diet, a Schizochytrium oil diet or one of the two diets containing the oil and β-glucan. The plasma HDL, expression of hepatic genes linked to, among others, ferric ion binding and plasma phosphatidylcholines were higher and plasma cholesterol esters and triacylglycerols were lower in the microbial oil-fed fish compared to the fish fed high cholesterol diet. While the fish fed a mix of microbial oil and Euglena β-glucan had lower plasma triacylglycerols and expression of hepatic genes linked to PPAR signaling pathway and enriched biosynthesis of plasma unsaturated fatty acids, the fish fed microbial oil-Phaeodactylum β-glucan combination had lower abundance of triacylglycerols rich in saturated and mono-unsaturated fatty acids and cholesterol esters in the plasma.
Collapse
Affiliation(s)
- Adnan H Gora
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Saima Rehman
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | | | - Pål A Olsvik
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Mette Sørensen
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway.
| |
Collapse
|
5
|
Yu J, Hu G, Guo X, Cao H, Zhang C. Quercetin Alleviates Inflammation and Energy Deficiency Induced by Lipopolysaccharide in Chicken Embryos. Animals (Basel) 2023; 13:2051. [PMID: 37443849 DOI: 10.3390/ani13132051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
Energy deficiency causes multiple organ dysfunctions after LPS induction. Quercetin is a phenolic compound found in herbal medicines. However, the effects of quercetin in alleviating LPS-induced energy deficiency remain unclear. In the present study, an in vivo LPS-induced inflammation model was established in chicken embryos. Specific pathogen-free chicken embryos (n = 120) were allocated to control, PBS with or without ethanol, quercetin (10, 20, or 40 nmol, respectively), and LPS (125 ng/egg) with or without quercetin groups. Fifteen day old embryonated eggs were injected with the abovementioned solutions via the allantoic cavity. On embryonic day 19, the tissues of the embryos were collected for histopathological examination using frozen oil red O staining, RNA extraction, real-time quantitative polymerase chain reaction, and immunohistochemical investigations. The glycogen and lipid contents in the liver increased after LPS stimulation as compared with the PBS group, whereas quercetin decreased the accumulation as compared with the LPS group. The mRNA expressions of AMPKα1 and AMPKα2 in the duodena, ceca, and livers were upregulated after LPS induction as compared with the PBS group, while quercetin could downregulate these expressions as compared with the LPS group. The immunopositivity of AMPKα2 in the villus, crypt, lamina propria, tunica muscularis, and myenteric plexus in the duodena and in the cytoplasms of hepatocytes significantly increased after LPS induction when compared with the PBS group (p < 0.01), whereas the immunopositivity to AMPKα2 in the quercetin treatment group significantly decreased when compared with the LPS group (p < 0.01 or p < 0.05). The LPS-induced high expressions of transcription factor PPARα and glucose transporter (SGLT1) were blocked by quercetin in the duodena, ceca, and livers. Quercetin treatment improved the LPS-induced decrease in APOA4 in the duodena, ceca, and livers. The mRNA expression of PEPT1 in the duodena and ceca increased after LPS challenge, whereas quercetin could downregulate PEPT1 gene expression. These data demonstrate that quercetin improved the energy deficiency induced by LPS in chicken embryos. The LPS-induced inflammation model was established to avoid the effect of LPS exposure from the environment and intestinal flora. The results form the basis the administration of quercetin pretreatment (in ovo infection) to improve the energy state of chicken embryos and improve the inflammation response.
Collapse
Affiliation(s)
- Jinhai Yu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
6
|
Czigle S, Bittner Fialová S, Tóth J, Mučaji P, Nagy M. Treatment of Gastrointestinal Disorders-Plants and Potential Mechanisms of Action of Their Constituents. Molecules 2022; 27:2881. [PMID: 35566230 PMCID: PMC9105531 DOI: 10.3390/molecules27092881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
The worldwide prevalence of gastrointestinal diseases is about 40%, with standard pharmacotherapy being long-lasting and economically challenging. Of the dozens of diseases listed by the Rome IV Foundation criteria, for five of them (heartburn, dyspepsia, nausea and vomiting disorder, constipation, and diarrhoea), treatment with herbals is an official alternative, legislatively supported by the European Medicines Agency (EMA). However, for most plants, the Directive does not require a description of the mechanisms of action, which should be related to the therapeutic effect of the European plant in question. This review article, therefore, summarizes the basic pharmacological knowledge of synthetic drugs used in selected functional gastrointestinal disorders (FGIDs) and correlates them with the constituents of medicinal plants. Therefore, the information presented here is intended as a starting point to support the claim that both empirical folk medicine and current and decades-old treatments with official herbal remedies have a rational basis in modern pharmacology.
Collapse
Affiliation(s)
- Szilvia Czigle
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, SK-832 32 Bratislava, Slovakia; (S.B.F.); (J.T.); (P.M.); (M.N.)
| | | | | | | | | | | |
Collapse
|
7
|
Role of Peroxisome Proliferator-Activated Receptors (PPARs) in Energy Homeostasis of Dairy Animals: Exploiting Their Modulation through Nutrigenomic Interventions. Int J Mol Sci 2021; 22:ijms222212463. [PMID: 34830341 PMCID: PMC8619600 DOI: 10.3390/ijms222212463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/31/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are the nuclear receptors that could mediate the nutrient-dependent transcriptional activation and regulate metabolic networks through energy homeostasis. However, these receptors cannot work properly under metabolic stress. PPARs and their subtypes can be modulated by nutrigenomic interventions, particularly under stress conditions to restore cellular homeostasis. Many nutrients such as polyunsaturated fatty acids, vitamins, dietary amino acids and phytochemicals have shown their ability for potential activation or inhibition of PPARs. Thus, through different mechanisms, all these nutrients can modulate PPARs and are ultimately helpful to prevent various metabolic disorders, particularly in transition dairy cows. This review aims to provide insights into the crucial role of PPARs in energy metabolism and their potential modulation through nutrigenomic interventions to improve energy homeostasis in dairy animals.
Collapse
|
8
|
Vecka M, Dušejovská M, Staňková B, Rychlík I, Žák A. A Matched Case-Control Study of Noncholesterol Sterols and Fatty Acids in Chronic Hemodialysis Patients. Metabolites 2021; 11:774. [PMID: 34822432 PMCID: PMC8618803 DOI: 10.3390/metabo11110774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/17/2022] Open
Abstract
Dyslipidemia is common among patients on hemodialysis, but its etiology is not fully understood. Although changes in cholesterol homeostasis and fatty acid metabolism play an important role during dialysis, the interaction of these metabolic pathways has yet to be studied in sufficient detail. In this study, we enrolled 26 patients on maintenance hemodialysis treatment (high-volume hemodiafiltration, HV HDF) without statin therapy (17 men/9 women) and an age/gender-matched group of 26 individuals without signs of nephropathy. The HV-HDF group exhibited more frequent signs of cardiovascular disease, disturbed saccharide metabolism, and altered lipoprotein profiles, manifesting in lower HDL-C, and raised concentrations of IDL-C and apoB-48 (all p < 0.01). HV-HDF patients had higher levels of campesterol (p < 0.01) and β-sitosterol (p = 0.06), both surrogate markers of cholesterol absorption and unchanged lathosterol concentrations. Fatty acid (FA) profiles were changed mostly in cholesteryl esters, with a higher content of saturated and n-3 polyunsaturated fatty acids (PUFA) in the HV-HDF group. However, n-6 PUFA in cholesteryl esters were less abundant (p < 0.001) in the HV-HDF group. Hemodialysis during end-stage kidney disease induces changes associated with higher absorption of cholesterol and disturbed lipoprotein metabolism. Changes in fatty acid metabolism reflect the combined effect of renal insufficiency and its comorbidities, mostly insulin resistance.
Collapse
Affiliation(s)
- Marek Vecka
- Fourth Department of Internal Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08 Prague, Czech Republic; (M.D.); (B.S.); (A.Ž.)
- Institute of Clinical Chemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Na Bojišti 3, 121 08 Prague, Czech Republic
| | - Magdalena Dušejovská
- Fourth Department of Internal Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08 Prague, Czech Republic; (M.D.); (B.S.); (A.Ž.)
| | - Barbora Staňková
- Fourth Department of Internal Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08 Prague, Czech Republic; (M.D.); (B.S.); (A.Ž.)
| | - Ivan Rychlík
- Department of Internal Medicine, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Šrobárova 50, 100 34 Prague, Czech Republic;
| | - Aleš Žák
- Fourth Department of Internal Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08 Prague, Czech Republic; (M.D.); (B.S.); (A.Ž.)
| |
Collapse
|
9
|
Faizo N, Narasimhulu CA, Forsman A, Yooseph S, Parthasarathy S. Peroxidized Linoleic Acid, 13-HPODE, Alters Gene Expression Profile in Intestinal Epithelial Cells. Foods 2021; 10:foods10020314. [PMID: 33546321 PMCID: PMC7913489 DOI: 10.3390/foods10020314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/20/2022] Open
Abstract
Lipid peroxides (LOOHs) abound in processed food and have been implicated in the pathology of diverse diseases including gut, cardiovascular, and cancer diseases. Recently, RNA Sequencing (RNA-seq) has been widely used to profile gene expression. To characterize gene expression and pathway dysregulation upon exposure to peroxidized linoleic acid, we incubated intestinal epithelial cells (Caco-2) with 100 µM of 13-hydroperoxyoctadecadienoic acid (13-HPODE) or linoleic acid (LA) for 24 h. Total RNA was extracted for library preparation and Illumina HiSeq sequencing. We identified 3094 differentially expressed genes (DEGs) in 13-HPODE-treated cells and 2862 DEGs in LA-treated cells relative to untreated cells. We show that 13-HPODE enhanced lipid metabolic pathways, including steroid hormone biosynthesis, PPAR signaling, and bile secretion, which alter lipid uptake and transport. 13-HPODE and LA treatments promoted detoxification mechanisms including cytochrome-P450. Conversely, both treatments suppressed oxidative phosphorylation. We also show that both treatments may promote absorptive cell differentiation and reduce proliferation by suppressing pathways involved in the cell cycle, DNA synthesis/repair and ribosomes, and enhancing focal adhesion. A qRT-PCR analysis of representative DEGs validated the RNA-seq analysis. This study provides insights into mechanisms by which 13-HPODE alters cellular processes and its possible involvement in mitochondrial dysfunction-related disorders and proposes potential therapeutic strategies to treat LOOH-related pathologies.
Collapse
Affiliation(s)
- Nisreen Faizo
- Burnett School of Biomedical Sciences, Genomics and Bioinformatics Cluster, College of Medicine, University of Central Florida, Orlando, FL 32816, USA;
| | - Chandrakala Aluganti Narasimhulu
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA; (C.A.N.); (S.P.)
| | - Anna Forsman
- Department of Biology, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL 32816, USA;
| | - Shibu Yooseph
- Department of Computer Science, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL 32816, USA
- Correspondence: ; Tel.: +1-407-823-5307
| | - Sampath Parthasarathy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA; (C.A.N.); (S.P.)
| |
Collapse
|
10
|
Hua H, Zhang Y, Zhao F, Chen K, Wu T, Liu Q, Huang S, Zhang A, Jia Z. Celastrol inhibits intestinal lipid absorption by reprofiling the gut microbiota to attenuate high-fat diet-induced obesity. iScience 2021; 24:102077. [PMID: 33598642 PMCID: PMC7868996 DOI: 10.1016/j.isci.2021.102077] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022] Open
Abstract
Celastrol, a compound extracted from traditional Chinese medicine, has been reported as a potent anti-obesity agent with controversial mechanisms. Here both C57BL/6J and leptin-deficient (ob/ob) mice fed a high-fat diet (HFD) displayed body weight loss after celastrol therapy, opposing the previous viewpoint that celastrol improves obesity by sensitizing leptin signaling. More importantly, celastrol downregulated lipid transporters in the intestine, increased lipid excretion in feces, and reduced body weight gain in HFD mice. Meanwhile, analysis of gut microbiota revealed that celastrol altered the gut microbiota composition in HFD-fed mice, and modulating gut microbiota by antibiotics or fecal microbiota transplantation blocked the celastrol effect on intestinal lipid transport and body weight gain, suggesting a critical role of the gut microbiota composition in mediating the anti-obesity role of celastrol under HFD. Together, the findings revealed that celastrol reduces intestinal lipid absorption to antagonize obesity by resetting the gut microbiota profile under HFD feeding. Celastrol reduced intestinal lipid transporters and lipids absorption Celastrol reset gut microbiota profile to modulate intestinal lipid transport Celastrol attenuated obesity in leptin-deficient (ob/ob) mice fed high fat diet
Collapse
Affiliation(s)
- Hu Hua
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| | - Yue Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| | - Fei Zhao
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China
| | - Ke Chen
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China.,Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China
| | - Tong Wu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China.,Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China
| | - Qianqi Liu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China.,Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| |
Collapse
|
11
|
Durkin LA, Childs CE, Calder PC. Omega-3 Polyunsaturated Fatty Acids and the Intestinal Epithelium-A Review. Foods 2021; 10:foods10010199. [PMID: 33478161 PMCID: PMC7835870 DOI: 10.3390/foods10010199] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Epithelial cells (enterocytes) form part of the intestinal barrier, the largest human interface between the internal and external environments, and responsible for maintaining regulated intestinal absorption and immunological control. Under inflammatory conditions, the intestinal barrier and its component enterocytes become inflamed, leading to changes in barrier histology, permeability, and chemical mediator production. Omega-3 (ω-3) polyunsaturated fatty acids (PUFAs) can influence the inflammatory state of a range of cell types, including endothelial cells, monocytes, and macrophages. This review aims to assess the current literature detailing the effects of ω-3 PUFAs on epithelial cells. Marine-derived ω-3 PUFAs, eicosapentaenoic acid and docosahexaenoic acid, as well as plant-derived alpha-linolenic acid, are incorporated into intestinal epithelial cell membranes, prevent changes to epithelial permeability, inhibit the production of pro-inflammatory cytokines and eicosanoids and induce the production of anti-inflammatory eicosanoids and docosanoids. Altered inflammatory markers have been attributed to changes in activity and/or expression of proteins involved in inflammatory signalling including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), peroxisome proliferator activated receptor (PPAR) α and γ, G-protein coupled receptor (GPR) 120 and cyclooxygenase (COX)-2. Effective doses for each ω-3 PUFA are difficult to determine due to inconsistencies in dose and time of exposure between different in vitro models and between in vivo and in vitro models. Further research is needed to determine the anti-inflammatory potential of less-studied ω-3 PUFAs, including docosapentaenoic acid and stearidonic acid.
Collapse
Affiliation(s)
- Luke A. Durkin
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Correspondence:
| | - Caroline E. Childs
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
12
|
Seiler KM, Goo WH, Zhang Q, Courtney C, Bajinting A, Guo J, Warner BW. Adaptation of extracellular matrix to massive small bowel resection in mice. J Pediatr Surg 2020; 55:1107-1112. [PMID: 32164986 PMCID: PMC7299777 DOI: 10.1016/j.jpedsurg.2020.02.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Extracellular matrix (ECM) affects cell behavior, and vice versa. How ECM changes after small bowel resection (SBR) to support adaptive cellular processes has not been described. Here we characterize changes in ECM following SBR and integrate this with concomitant transcriptional perturbations. METHODS A 50% proximal SBR or sham surgery was performed on mice. On postoperative day 7, ileal tissue was sequentially depleted of protein components to generate an ECM-enriched fraction. ECM was analyzed for protein composition using mass spectrometry with subsequent Ingenuity Pathway Analysis (IPA) to identify predicted pathways and upstream regulators. qPCR and RNA-sequencing (RNA-Seq) were performed to corroborate these predicted pathways. RESULTS 3034 proteins were differentially regulated between sham and SBR, of which 95 were significant (P < 0.05). IPA analysis predicted PPARα agonism to be an upstream regulator of the observed proteomic changes (P < 0.001). qPCR and RNA-Seq with KEGG analysis confirmed significant engagement of the PPAR pathway (P < 0.05). CONCLUSION Transcriptional signatures of adapting bowel predict subsequent ECM changes after SBR. How ECM communicates with surrounding cells to drive adaptation and vice versa merits further investigation. Our findings thus far suggest ECM supports tissue hyperplasia and altered metabolic demand following SBR.
Collapse
Affiliation(s)
- Kristen M. Seiler
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | | | - Qiang Zhang
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO
| | - Cathleen Courtney
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Adam Bajinting
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Jun Guo
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Brad W. Warner
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
13
|
Yu B, Peng XH, Wang LY, Wang AB, Su YY, Chen JH, Zhang XW, Zhao DZ, Wang H, Pang DX, Ouyang HS, Tang XC, Zhang MJ. Abnormality of intestinal cholesterol absorption in ApcMin/+ mice with colon cancer cachexia. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:759-767. [PMID: 31933883 PMCID: PMC6945172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 01/24/2019] [Indexed: 06/10/2023]
Abstract
Colorectal cancer syndrome has been one of the greatest concerns in the world, particularly in developed countries. Several epidemiological studies have shown that dyslipidemia may be associated with the progression of intestinal cachexia, but there is little research on the function of the small intestine, which is involved in blood lipid metabolism, in dyslipidemia. In the present study, we aimed to explore the function of intestinal cholesterol absorption in the ApcMin/+ mouse model using an intestinal lipid absorption test. We found that both triglyceride (TG) and total cholesterol (TC) uptake were inhibited in the intestine of ApcMin/+ mice with age and the intestinal peroxisome proliferator-activated receptor α (PPARα) downregulated the processes of β-oxidation, oxidative stress response, and cholesterol absorption in APC-deficient mice. In addition, reduced expression levels of farnesoid X receptor (FXR) and apical sodium-dependent bile acid transporter (ASBT) indicated that bile acid metabolism might be associated with intestinal cholesterol absorption in ApcMin/+ mice. Thus, our data suggested that the intestine plays an essential role in cholesterol uptake and that bile acid metabolism seems to cause a decrease in intestinal cholesterol uptake in ApcMin/+ mice.
Collapse
Affiliation(s)
- Biao Yu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Xiao-Huan Peng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Ling-Yu Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - An-Bei Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Yan-Yan Su
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Jia-Huan Chen
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Xin-Wei Zhang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Da-Zhong Zhao
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - He Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Da-Xin Pang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Hong-Sheng Ouyang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Xiao-Chun Tang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| | - Ming-Jun Zhang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University Changchun, Jilin Province, China
| |
Collapse
|
14
|
Clostridium ramosum regulates enterochromaffin cell development and serotonin release. Sci Rep 2019; 9:1177. [PMID: 30718836 PMCID: PMC6362283 DOI: 10.1038/s41598-018-38018-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/14/2018] [Indexed: 12/22/2022] Open
Abstract
Peripheral serotonin (5-hydroxytryptamine: 5-HT) synthesized in the intestine by enterochromaffin cells (ECs), plays an important role in the regulation of peristaltic of the gut, epithelial secretion and promotes the development and maintenance of the enteric neurons. Recent studies showed that the indigenous gut microbiota modulates 5-HT signalling and that ECs use sensory receptors to detect dietary and microbiota-derived signals from the lumen to subsequently transduce the information to the nervous system. We hypothesized that Clostridium ramosum by increasing gut 5-HT availability consequently contributes to high-fat diet-induced obesity. Using germ-free mice and mice monoassociated with C. ramosum, intestinal cell lines and mouse organoids, we demonstrated that bacterial cell components stimulate host 5-HT secretion and program the differentiation of colonic intestinal stem progenitors toward the secretory 5-HT-producing lineage. An elevated 5-HT level regulates the expression of major proteins involved in intestinal fatty acid absorption in vitro, suggesting that the presence of C. ramosum in the gut promotes 5-HT secretion and thereby could facilitates intestinal lipid absorption and the development of obesity.
Collapse
|
15
|
Losacco MC, de Almeida CFT, Hijo AHT, Bargi-Souza P, Gama P, Nunes MT, Goulart-Silva F. High-fat diet affects gut nutrients transporters in hypo and hyperthyroid mice by PPAR-a independent mechanism. Life Sci 2018; 202:35-43. [DOI: 10.1016/j.lfs.2018.03.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/22/2018] [Accepted: 03/27/2018] [Indexed: 10/17/2022]
|
16
|
Liu C, Han T, Stachura DL, Wang H, Vaisman BL, Kim J, Klemke RL, Remaley AT, Rana TM, Traver D, Miller YI. Lipoprotein lipase regulates hematopoietic stem progenitor cell maintenance through DHA supply. Nat Commun 2018; 9:1310. [PMID: 29615667 PMCID: PMC5882990 DOI: 10.1038/s41467-018-03775-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 03/07/2018] [Indexed: 01/15/2023] Open
Abstract
Lipoprotein lipase (LPL) mediates hydrolysis of triglycerides (TGs) to supply free fatty acids (FFAs) to tissues. Here, we show that LPL activity is also required for hematopoietic stem progenitor cell (HSPC) maintenance. Knockout of Lpl or its obligatory cofactor Apoc2 results in significantly reduced HSPC expansion during definitive hematopoiesis in zebrafish. A human APOC2 mimetic peptide or the human very low-density lipoprotein, which carries APOC2, rescues the phenotype in apoc2 but not in lpl mutant zebrafish. Creating parabiotic apoc2 and lpl mutant zebrafish rescues the hematopoietic defect in both. Docosahexaenoic acid (DHA) is identified as an important factor in HSPC expansion. FFA-DHA, but not TG-DHA, rescues the HSPC defects in apoc2 and lpl mutant zebrafish. Reduced blood cell counts are also observed in Apoc2 mutant mice at the time of weaning. These results indicate that LPL-mediated release of the essential fatty acid DHA regulates HSPC expansion and definitive hematopoiesis.
Collapse
Affiliation(s)
- Chao Liu
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Tianxu Han
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - David L Stachura
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Huawei Wang
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Boris L Vaisman
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, 31 Center St, Bethesda, MD, 20892, USA
| | - Jungsu Kim
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Richard L Klemke
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, 31 Center St, Bethesda, MD, 20892, USA
| | - Tariq M Rana
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - David Traver
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
17
|
Lamichane S, Dahal Lamichane B, Kwon SM. Pivotal Roles of Peroxisome Proliferator-Activated Receptors (PPARs) and Their Signal Cascade for Cellular and Whole-Body Energy Homeostasis. Int J Mol Sci 2018; 19:ijms19040949. [PMID: 29565812 PMCID: PMC5979443 DOI: 10.3390/ijms19040949] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs), members of the nuclear receptor superfamily, are important in whole-body energy metabolism. PPARs are classified into three isoforms, namely, PPARα, β/δ, and γ. They are collectively involved in fatty acid oxidation, as well as glucose and lipid metabolism throughout the body. Importantly, the three isoforms of PPARs have complementary and distinct metabolic activities for energy balance at a cellular and whole-body level. PPARs also act with other co-regulators to maintain energy homeostasis. When endogenous ligands bind with these receptors, they regulate the transcription of genes involved in energy homeostasis. However, the exact molecular mechanism of PPARs in energy metabolism remains unclear. In this review, we summarize the importance of PPAR signals in multiple organs and focus on the pivotal roles of PPAR signals in cellular and whole-body energy homeostasis.
Collapse
Affiliation(s)
- Shreekrishna Lamichane
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea.
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea.
| | - Babita Dahal Lamichane
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea.
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea.
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea.
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea.
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea.
| |
Collapse
|
18
|
Lehnert K, Weirup L, Harding KC, Härkönen T, Karlsson O, Teilmann J. Antarctic seals: Molecular biomarkers as indicators for pollutant exposure, health effects and diet. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 599-600:1693-1704. [PMID: 28535598 DOI: 10.1016/j.scitotenv.2017.04.224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 06/07/2023]
Abstract
Weddell (Leptonychotes weddellii), Ross (Ommatophoca rossii) and crabeater seals (Lobodon carcinophaga) are phocid seals with a circumpolar distribution around Antarctica. As long-lived and large top predators, they bioaccumulate contaminants and are considered as sentinels of ecosystem health. Antarctic seals are increasingly exposed to climate change, pollution, shipping and fisheries. To reveal and understand possible anthropogenic impacts on their immune and health status, this study investigates sensitive biomarkers of the xenobiotic metabolism and immune system in relation to mercury (Hg) burden. Gene-transcription studies using minimally-invasive blood samples are useful to monitor physiological processes in wildlife that can be related to different stressors. Blood samples of 72 wild-caught seals (Weddell n=33; Ross n=12; crabeater n=27) in the Amundsen and Ross Seas in 2008-2011 were investigated. Copy numbers per μl mRNA transcription of xenobiotic biomarkers (aryl hydrocarbon receptor (AHR)), aryl hydrocarbon receptor nuclear translocator (ARNT) and peroxisome proliferator-activated receptor (PPARα) and immune relevant cell mediators (cytokines interleukin-2 (IL-2), interleukin-10 (IL-10) and heat-shock-protein 70 (HSP70)) were measured using reference genes Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide (YWHAZ) and ribosomal protein L4 (RPL4) by real time RT-qPCR. Hg concentration was analysed in fur. Hg concentration increased with body weight and standard length in all species. Crabeater seals showed a lower Hg concentration than Ross and Weddell seals. Species-specific differences in gene-transcription were found between all species with highest levels of AHR, ARNT and PPARα in crabeater seals. Ross seals showed highest IL-10 and HSP70 transcription, while HSP70 was exceptionally low in crabeater seals. Between Hg and HSP70 a clear negative relationship was found in all species. The species-specific, age and sex-dependent gene-transcription probably reflect dietary habits, pollutant exposure and immune status.
Collapse
Affiliation(s)
- K Lehnert
- Institute for Terrestrial and Aquatic Wildlife Research, University of Veterinary Medicine Hannover, 25761 Büsum, Germany.
| | - L Weirup
- Institute for Terrestrial and Aquatic Wildlife Research, University of Veterinary Medicine Hannover, 25761 Büsum, Germany
| | - K C Harding
- University of Gothenburg, Department of Biological and Environmental Sciences, Box 463, SE-405 30 Gothenburg, Sweden
| | - T Härkönen
- Swedish Museum of Natural History, Department of Environmental Research and Monitoring, P.O. Box 50007, 104 05 Stockholm, Sweden
| | - O Karlsson
- Swedish Museum of Natural History, Department of Environmental Research and Monitoring, P.O. Box 50007, 104 05 Stockholm, Sweden
| | - J Teilmann
- Aarhus University, Department of Bioscience, Frederiksborgvej 399, P.O. Box 358, 4000 Roskilde, Denmark
| |
Collapse
|
19
|
PPARγ Modulates Long Chain Fatty Acid Processing in the Intestinal Epithelium. Int J Mol Sci 2017; 18:ijms18122559. [PMID: 29182565 PMCID: PMC5751162 DOI: 10.3390/ijms18122559] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/26/2022] Open
Abstract
Nuclear receptor PPARγ affects lipid metabolism in several tissues, but its role in intestinal lipid metabolism has not been explored. As alterations have been observed in the plasma lipid profile of ad libitum fed intestinal epithelium-specific PPARγ knockout mice (iePPARγKO), we submitted these mice to lipid gavage challenges. Within hours after gavage with long chain unsaturated fatty acid (FA)-rich canola oil, the iePPARγKO mice had higher plasma free FA levels and lower gastric inhibitory polypeptide levels than their wild-type (WT) littermates, and altered expression of incretin genes and lipid metabolism-associated genes in the intestinal epithelium. Gavage with the medium chain saturated FA-rich coconut oil did not result in differences between the two genotypes. Furthermore, the iePPARγKO mice did not exhibit defective lipid uptake and stomach emptying; however, their intestinal transit was more rapid than in WT mice. When fed a canola oil-rich diet for 4.5 months, iePPARγKO mice had higher body lean mass than the WT mice. We conclude that intestinal epithelium PPARγ is activated preferentially by long chain unsaturated FAs compared to medium chain saturated FAs. Furthermore, we hypothesize that the iePPARγKO phenotype originates from altered lipid metabolism and release in epithelial cells, as well as changes in intestinal motility.
Collapse
|
20
|
Identification of the principal transcriptional regulators for low-fat and high-fat meal responsive genes in small intestine. Nutr Metab (Lond) 2017; 14:66. [PMID: 29075307 PMCID: PMC5654052 DOI: 10.1186/s12986-017-0221-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/16/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND High-fat (HF) diet is a well-known cause of obesity. To identify principle transcriptional regulators that could be therapeutic targets of obesity, we investigated transcriptomic modulation in the duodenal mucosa following low-fat (LF) and HF meal ingestion. METHODS Whereas one group of mice was sacrificed after fasting, the others were fed ad libitum with LF or HF meal, and sacrificed 30 min, 1 h and 3 h after the beginning of the meal. A transcriptome analysis of the duodenal mucosa of the 7 groups was conducted using both microarray and serial analysis of gene expression (SAGE) method followed by an Ingenuity Pathways Analysis (IPA). RESULTS SAGE and microarray showed that the modulation of a total of 896 transcripts in the duodenal mucosa after LF and/or HF meal, compared to the fasting condition. The IPA identified lipid metabolism, molecular transport, and small molecule biochemistry as top three molecular and cellular functions for the HF-responsive, HF-specific, HF-delay, and LF-HF different genes. Moreover, the top transcriptional regulator for the HF-responsive and HF-specific genes was peroxisome proliferator-activated receptor alpha (PPARα). On the other hand, the LF-responsive and LF-specific genes were related to carbohydrate metabolism, cellular function and maintenance, and cell death/cellular growth and proliferation, and the top transcriptional regulators were forkhead box protein O1 (FOXO1) and cAMP response element binding protein 1 (CREB1), respectively. CONCLUSIONS These results will help to understand the molecular mechanisms of intestinal response after LF and HF ingestions, and contribute to identify therapeutic targets for obesity and obesity-related diseases.
Collapse
|
21
|
Matsumoto Y, Mochizuki W, Akiyama S, Matsumoto T, Nozaki K, Watanabe M, Nakamura T. Distinct intestinal adaptation for vitamin B12 and bile acid absorption revealed in a new mouse model of massive ileocecal resection. Biol Open 2017; 6:1364-1374. [PMID: 28818841 PMCID: PMC5612230 DOI: 10.1242/bio.024927] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ileocecal resection (ICR), one of several types of intestinal resection that results in short bowel syndrome (SBS), causes severe clinical disease in humans. We here describe a mouse model of massive ICR in which 75% of the distal small intestine is removed. We demonstrate that mice underwent 75% ICR show severe clinical signs and high mortality, which may recapitulate severe forms of human SBS, despite an adaptive response throughout the remnant intestine. By using this model, we also investigated whether the epithelium of the remnant intestine shows enhanced expression of factors involved in region-specific functions of the ileum. Cubn mRNA and its protein product, which play an essential role in vitamin B12 absorption in the ileum, are not compensatory up-regulated in any part of the remnant intestine, demonstrating a clear contrast with post-operative up-regulation of genes involved in bile acid absorption. Our study suggests that functional adaptation by phenotypical changes in the intestinal epithelium is not a general feature for nutrient absorption systems that are confined to the ileum. We also propose that the mouse model developed in this study will become a unique system to facilitate studies on SBS with ICR in humans. Summary: Genes involved in region-specific functions of the distal small intestine show distinct adaptive response following massive ileocecal resection in mice.
Collapse
Affiliation(s)
- Yuka Matsumoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Wakana Mochizuki
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Shintaro Akiyama
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Taichi Matsumoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Kengo Nozaki
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Tetsuya Nakamura
- Department of Advanced Therapeutics for GI Diseases, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| |
Collapse
|
22
|
Oleic Acid Uptake Reveals the Rescued Enterocyte Phenotype of Colon Cancer Caco-2 by HT29-MTX Cells in Co-Culture Mode. Int J Mol Sci 2017; 18:ijms18071573. [PMID: 28726765 PMCID: PMC5536061 DOI: 10.3390/ijms18071573] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/07/2017] [Accepted: 07/16/2017] [Indexed: 12/14/2022] Open
Abstract
Gastrointestinal epithelium is the unique route for nutrients and for many pharmaceuticals to enter the body. The present study aimed to analyze precisely whether co-culture of two colon cancer cell lines, mucus-producing cells HT29-MTX and enterocyte-like Caco-2 cells, ameliorate differentiation into an in vitro intestinal barrier model and the signaling pathways involved. Differentiated Caco-2 cells gene datasets were compared first to intestinal or cancer phenotypes and second to signaling pathway gene datasets. Experimental validations were performed in real-time experiments, immunochemistry, and gene expression analyses on Caco-2 versus co-cultures of Caco-2 and HT29-MTX (10%) cells. Partial maintenance of cancer-cell phenotype in differentiated Caco-2 cells was confirmed and fatty acids merged as potential regulators of cancer signaling pathways. HT29-MTX cells induced morphological changes in Caco-2 cells, slightly increased their proliferation rate and profoundly modified gene transcription of phenotype markers, fatty acid receptors, intracellular transporters, and lipid droplet components as well as functional responses to oleic acid. In vitro, enterocyte phenotype was rescued partially by co-culture of cancer cells with goblet cells and completed through oleic acid interaction with signaling pathways dysregulated in cancer cells.
Collapse
|
23
|
Manoharan I, Suryawanshi A, Hong Y, Ranganathan P, Shanmugam A, Ahmad S, Swafford D, Manicassamy B, Ramesh G, Koni PA, Thangaraju M, Manicassamy S. Homeostatic PPARα Signaling Limits Inflammatory Responses to Commensal Microbiota in the Intestine. THE JOURNAL OF IMMUNOLOGY 2016; 196:4739-49. [PMID: 27183583 DOI: 10.4049/jimmunol.1501489] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 03/25/2016] [Indexed: 12/28/2022]
Abstract
Dietary lipids and their metabolites activate members of the peroxisome proliferative-activated receptor (PPAR) family of transcription factors and are critical for colonic health. The PPARα isoform plays a vital role in regulating inflammation in various disease settings, but its role in intestinal inflammation, commensal homeostasis, and mucosal immunity in the gut are unclear. In this study, we demonstrate that the PPARα pathway in innate immune cells orchestrates gut mucosal immunity and commensal homeostasis by regulating the expression of IL-22 and the antimicrobial peptides RegIIIβ, RegIIIγ, and calprotectin. Additionally, the PPARα pathway is critical for imparting regulatory phenotype in intestinal macrophages. PPARα deficiency in mice led to commensal dysbiosis in the gut, resulting in a microbiota-dependent increase in the expression of inflammatory cytokines and enhanced susceptibility to intestinal inflammation. Pharmacological activation of this pathway decreased the expression of inflammatory cytokines and ameliorated colonic inflammation. Taken together, these findings identify a new important innate immune function for the PPARα signaling pathway in regulating intestinal inflammation, mucosal immunity, and commensal homeostasis. Thus, the manipulation of the PPARα pathway could provide novel opportunities for enhancing mucosal immunity and treating intestinal inflammation.
Collapse
Affiliation(s)
| | | | - Yuan Hong
- Cancer Center, Augusta University, Augusta, GA 30912
| | | | | | - Shamim Ahmad
- Cancer Center, Augusta University, Augusta, GA 30912
| | | | | | - Ganesan Ramesh
- Vascular Biology Center, Augusta University, Augusta, GA 30912
| | - Pandelakis A Koni
- Cancer Center, Augusta University, Augusta, GA 30912; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912; and
| | - Muthusamy Thangaraju
- Cancer Center, Augusta University, Augusta, GA 30912; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Santhakumar Manicassamy
- Cancer Center, Augusta University, Augusta, GA 30912; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912; and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912
| |
Collapse
|
24
|
D'Aquila T, Hung YH, Carreiro A, Buhman KK. Recent discoveries on absorption of dietary fat: Presence, synthesis, and metabolism of cytoplasmic lipid droplets within enterocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:730-47. [PMID: 27108063 DOI: 10.1016/j.bbalip.2016.04.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/16/2016] [Accepted: 04/16/2016] [Indexed: 02/07/2023]
Abstract
Dietary fat provides essential nutrients, contributes to energy balance, and regulates blood lipid concentrations. These functions are important to health, but can also become dysregulated and contribute to diseases such as obesity, diabetes, cardiovascular disease, and cancer. Within enterocytes, the digestive products of dietary fat are re-synthesized into triacylglycerol, which is either secreted on chylomicrons or stored within cytoplasmic lipid droplets (CLDs). CLDs were originally thought to be inert stores of neutral lipids, but are now recognized as dynamic organelles that function in multiple cellular processes in addition to lipid metabolism. This review will highlight recent discoveries related to dietary fat absorption with an emphasis on the presence, synthesis, and metabolism of CLDs within this process.
Collapse
Affiliation(s)
- Theresa D'Aquila
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Yu-Han Hung
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Alicia Carreiro
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
25
|
Eicosapentaenoic acid inhibits intestinal β-carotene absorption by downregulation of lipid transporter expression via PPAR-α dependent mechanism. Arch Biochem Biophys 2016; 590:118-124. [DOI: 10.1016/j.abb.2015.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/28/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022]
|
26
|
Abstract
BACKGROUND Dyslipidemia is a common metabolic complication in patients with chronic kidney disease and is detected as changes in lipoprotein concentrations in serum. SUMMARY Recently, other features of lipid abnormalities have been described, such as alterations in serum n-3 polyunsaturated fatty acids, cholesterol metabolism (proportion of hepatic synthesis and intestinal absorption), and dysfunctional high-density lipoprotein. KEY MESSAGES Since abnormalities of these new aspects predict adverse outcomes in hemodialysis populations, they may be helpful in risk stratification of patients and could also be new targets for prevention.
Collapse
Affiliation(s)
- Tetsuo Shoji
- Department of Geriatrics and Vascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
27
|
Silvennoinen R, Quesada H, Kareinen I, Julve J, Kaipiainen L, Gylling H, Blanco-Vaca F, Escola-Gil JC, Kovanen PT, Lee-Rueckert M. Chronic intermittent psychological stress promotes macrophage reverse cholesterol transport by impairing bile acid absorption in mice. Physiol Rep 2015; 3:3/5/e12402. [PMID: 25969465 PMCID: PMC4463831 DOI: 10.14814/phy2.12402] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Psychological stress is a risk factor for atherosclerosis, yet the pathophysiological mechanisms involved remain elusive. The transfer of cholesterol from macrophage foam cells to liver and feces (the macrophage-specific reverse cholesterol transport, m-RCT) is an important antiatherogenic pathway. Because exposure of mice to physical restraint, a model of psychological stress, increases serum levels of corticosterone, and as bile acid homeostasis is disrupted in glucocorticoid-treated animals, we investigated if chronic intermittent restraint stress would modify m-RCT by altering the enterohepatic circulation of bile acids. C57Bl/6J mice exposed to intermittent stress for 5 days exhibited increased transit through the large intestine and enhanced fecal bile acid excretion. Of the transcription factors and transporters that regulate bile acid homeostasis, the mRNA expression levels of the hepatic farnesoid X receptor (FXR), the bile salt export pump (BSEP), and the intestinal fibroblast growth factor 15 (FGF15) were reduced, whereas those of the ileal apical sodium-dependent bile acid transporter (ASBT), responsible for active bile acid absorption, remained unchanged. Neither did the hepatic expression of cholesterol 7α-hydroxylase (CYP7A1), the key enzyme regulating bile acid synthesis, change in the stressed mice. Evaluation of the functionality of the m-RCT pathway revealed increased fecal excretion of bile acids that had been synthesized from macrophage-derived cholesterol. Overall, our study reveals that chronic intermittent stress in mice accelerates m-RCT specifically by increasing fecal excretion of bile acids. This novel mechanism of m-RCT induction could have antiatherogenic potential under conditions of chronic stress.
Collapse
Affiliation(s)
| | - Helena Quesada
- IIB Sant Pau, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona-CIBER de Diabetes y Enfermedades Metabolicas Asociadas, Barcelona, Spain
| | | | - Josep Julve
- IIB Sant Pau, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona-CIBER de Diabetes y Enfermedades Metabolicas Asociadas, Barcelona, Spain
| | - Leena Kaipiainen
- Internal Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Helena Gylling
- Internal Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Francisco Blanco-Vaca
- IIB Sant Pau, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona-CIBER de Diabetes y Enfermedades Metabolicas Asociadas, Barcelona, Spain
| | - Joan Carles Escola-Gil
- IIB Sant Pau, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona-CIBER de Diabetes y Enfermedades Metabolicas Asociadas, Barcelona, Spain
| | | | | |
Collapse
|
28
|
Shoji T. [Kidney diseases and metabolic disorders--Basics and applications required for general physicians. Topics: III. Abnormalities in lipid metabolism in chronic kidney disease]. NIHON NAIKA GAKKAI ZASSHI. THE JOURNAL OF THE JAPANESE SOCIETY OF INTERNAL MEDICINE 2015; 104:923-930. [PMID: 26591341 DOI: 10.2169/naika.104.923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
|
29
|
Rideout TC, Marinangeli CPF, Harding SV. Triglyceride-Lowering Response to Plant Sterol and Stanol Consumption. J AOAC Int 2015; 98:707-715. [PMID: 25941890 DOI: 10.5740/jaoacint.sgerideout] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Phytosterols (PS) have long been recognized for their cholesterol-lowering action, however, recent work has highlighted triglyceride (TG)-lowering responses to PS that may have been overlooked in previous human interventions and mechanistic animal model studies. This review assesses the current state of knowledge regarding the effect of dietary PS supplementation on blood TG concentrations by examining the average therapeutic response, potential mechanisms, and metabolic and genetic factors that may contribute to inter-individual variability. Data from human intervention trials demonstrates that, compared to baseline concentrations, PS supplementation results in a variable TG-lowering response ranging from 0.8 to 28%. It is evident that hypertriglyceridemic individuals (>1.7 mmol/L) have a greater TG-lowering response to PS (11-28%) than subjects with normal plasma TG concentrations (0.8-7%). Although a genetic basis for the variable TG-lowering effects of PS is probable, there are only limited studies to draw on. The available data suggest that polymorphisms in the apolipoprotein E (apoE) gene may affect responsiveness, with PS-induced reductions in TG more readily evident in apoE2 than apoE3 or E4 subjects. Although only a minimal number of animal model studies have been conducted to specifically examine the mechanisms whereby PS may reduce blood TG concentrations, it appears that there may be multiple mechanisms involved including interruption of intestinal fatty acid absorption and modulation of hepatic lipogenesis and very low density lipoprotein packaging and secretion. In summary, the available data suggest that PS may be an effective therapy to lower blood TG, particularly in hypertriglyceridemic individuals. However, before PS can be widely recommended as a TG-lowering therapy, studies that are specifically powered and designed to fully access therapeutic responses and the mechanisms involved are required.
Collapse
Affiliation(s)
- Todd C Rideout
- University at Buffalo, School of Public Health and Health Professions, Department of Exercise and Nutrition Sciences, Buffalo, NY 14214, USA
| | | | | |
Collapse
|
30
|
Hung YH, Linden MA, Gordon A, Rector RS, Buhman KK. Endurance exercise training programs intestinal lipid metabolism in a rat model of obesity and type 2 diabetes. Physiol Rep 2015; 3:3/1/e12232. [PMID: 25602012 PMCID: PMC4387752 DOI: 10.14814/phy2.12232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Endurance exercise has been shown to improve metabolic outcomes in obesity and type 2 diabetes; however, the physiological and molecular mechanisms for these benefits are not completely understood. Although endurance exercise has been shown to decrease lipogenesis, promote fatty acid oxidation (FAO), and increase mitochondrial biosynthesis in adipose tissue, muscle, and liver, its effects on intestinal lipid metabolism remain unknown. The absorptive cells of the small intestine, enterocytes, mediate the highly efficient absorption and processing of nutrients, including dietary fat for delivery throughout the body. We investigated how endurance exercise altered intestinal lipid metabolism in obesity and type 2 diabetes using Otsuka Long‐Evans Tokushima Fatty (OLETF) rats. We assessed mRNA levels of genes associated with intestinal lipid metabolism in nonhyperphagic, sedentary Long‐Evans Tokushima Otsuka (LETO) rats (L‐Sed), hyperphagic, sedentary OLETF rats (O‐Sed), and endurance exercised OLETF rats (O‐EndEx). O‐Sed rats developed hyperphagia‐induced obesity (HIO) and type 2 diabetes compared with L‐Sed rats. O‐EndEx rats gained significantly less weight and fat pad mass, and had improved serum metabolic parameters without change in food consumption compared to O‐Sed rats. Endurance exercise resulted in dramatic up‐regulation of a number of genes in intestinal lipid metabolism and mitochondrial content compared with sedentary rats. Overall, this study provides evidence that endurance exercise programs intestinal lipid metabolism, likely contributing to its role in improving metabolic outcomes in obesity and type 2 diabetes. Endurance exercise has been shown to improve metabolic outcomes in obesity and type 2 diabetes; however, the physiological and molecular mechanisms for these benefits are not completely understood. Although endurance exercise has been shown to decrease lipogenesis, promote fatty acid oxidation (FAO), and increase mitochondrial biosynthesis in adipose tissue, muscle, and liver, its effects on intestinal lipid metabolism remain unknown. Endurance exercise resulted in dramatic up‐regulation of a number of genes in intestinal lipid metabolism and mitochondrial content compared with sedentary rats. Overall, this study provides evidence that endurance exercise programs intestinal lipid metabolism, likely contributing to its role in improving metabolic outcomes in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Yu-Han Hung
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Melissa A Linden
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Alicia Gordon
- Department of Clinical Medicine, University of Dublin, Dublin, Ireland School of Biological Sciences, Dublin Institute of Technology, Dublin, Ireland
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, Missouri Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| |
Collapse
|
31
|
Cizkova K, Rajdova A, Ehrmann J. Spatio-temporal expression of peroxisome proliferator-activated receptor α during human prenatal development. Basic Clin Pharmacol Toxicol 2014; 116:361-6. [PMID: 25225039 DOI: 10.1111/bcpt.12326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/15/2014] [Indexed: 11/30/2022]
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a ligand-dependent transcription factor which is activated by various endogenous as well as exogenous compounds. It is involved in the regulation of a variety of biological processes, such as nutrient metabolism, energy homoeostasis, immunological response and xenobiotic metabolism. Little is known about its expression during human prenatal development. We examined the spatio-temporal expression pattern of PPARα in human embryonic/foetal intestines, liver and kidney from the 5th to 20th week of prenatal life by indirect two-step immunohistochemistry. PPARα expression can already be detected in the early stages of prenatal development; as early as the 7th week of intrauterine development (IUD) in the intestines, 5th week of IUD in the liver and 6th week of IUD in the kidney. We found age-dependent changes in the PPARα expression pattern in the intestines and kidney. These events occur approximately at the commencement of function of these organs. In the intestines, we detected an obvious change of the PPARα expression pattern along the crypt-villous axis in the 11th week of IUD. In the kidney, the most apparent change was increased expression of PPARα in glomeruli in the 12th week of IUD. Moreover, in the liver, we detected a strong positivity in part of the developing blood elements. Information about the spatio-temporal expression pattern of PPARα could be the first step in evaluating the potential harmful impact of a wide range of environmental or pharmaceutical compounds which serve as PPARα ligands on the developing human organism.
Collapse
Affiliation(s)
- Katerina Cizkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | | | | |
Collapse
|
32
|
Blanc V, Park E, Schaefer S, Miller M, Lin Y, Kennedy S, Billing AM, Hamidane HB, Graumann J, Mortazavi A, Nadeau JH, Davidson NO. Genome-wide identification and functional analysis of Apobec-1-mediated C-to-U RNA editing in mouse small intestine and liver. Genome Biol 2014; 15:R79. [PMID: 24946870 PMCID: PMC4197816 DOI: 10.1186/gb-2014-15-6-r79] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 06/19/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND RNA editing encompasses a post-transcriptional process in which the genomically templated sequence is enzymatically altered and introduces a modified base into the edited transcript. Mammalian C-to-U RNA editing represents a distinct subtype of base modification, whose prototype is intestinal apolipoprotein B mRNA, mediated by the catalytic deaminase Apobec-1. However, the genome-wide identification, tissue-specificity and functional implications of Apobec-1-mediated C-to-U RNA editing remain incompletely explored. RESULTS Deep sequencing, data filtering and Sanger-sequence validation of intestinal and hepatic RNA from wild-type and Apobec-1-deficient mice revealed 56 novel editing sites in 54 intestinal mRNAs and 22 novel sites in 17 liver mRNAs, all within 3' untranslated regions. Eleven of 17 liver RNAs shared editing sites with intestinal RNAs, while 6 sites are unique to liver. Changes in RNA editing lead to corresponding changes in intestinal mRNA and protein levels for 11 genes. Analysis of RNA editing in vivo following tissue-specific Apobec-1 adenoviral or transgenic Apobec-1 overexpression reveals that a subset of targets identified in wild-type mice are restored in Apobec-1-deficient mouse intestine and liver following Apobec-1 rescue. We find distinctive polysome profiles for several RNA editing targets and demonstrate novel exonic editing sites in nuclear preparations from intestine but not hepatic apolipoprotein B RNA. RNA editing is validated using cell-free extracts from wild-type but not Apobec-1-deficient mice, demonstrating that Apobec-1 is required. CONCLUSIONS These studies define selective, tissue-specific targets of Apobec-1-dependent RNA editing and show the functional consequences of editing are both transcript- and tissue-specific.
Collapse
Affiliation(s)
- Valerie Blanc
- Department of Medicine, Washington University St Louis, St Louis, MO 63110, USA
| | - Eddie Park
- Department of Developmental and Cell Biology and Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
| | - Sabine Schaefer
- Pacific Northwest Research Institute, Seattle, WA 98122, USA
| | - Melanie Miller
- Department of Medicine, Washington University St Louis, St Louis, MO 63110, USA
| | - Yiing Lin
- Departments of Surgery, Washington University St Louis, St Louis, MO 63110, USA
| | - Susan Kennedy
- Department of Medicine, Washington University St Louis, St Louis, MO 63110, USA
| | - Anja M Billing
- Proteomics Core, Weill Cornell Medical College in Qatar, Doha, Qatar
| | | | - Johannes Graumann
- Proteomics Core, Weill Cornell Medical College in Qatar, Doha, Qatar
| | - Ali Mortazavi
- Department of Developmental and Cell Biology and Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
| | - Joseph H Nadeau
- Pacific Northwest Research Institute, Seattle, WA 98122, USA
| | - Nicholas O Davidson
- Department of Medicine, Washington University St Louis, St Louis, MO 63110, USA
| |
Collapse
|
33
|
Sonoda M, Shoji T, Kimoto E, Okute Y, Shima H, Naganuma T, Motoyama K, Morioka T, Mori K, Fukumoto S, Shioi A, Koyama H, Emoto M, Inaba M. Kidney Function, Cholesterol Absorption and Remnant Lipoprotein Accumulation in Patients with Diabetes Mellitus. J Atheroscler Thromb 2014; 21:346-54. [DOI: 10.5551/jat.20594] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
34
|
Kimura R, Takahashi N, Lin S, Goto T, Murota K, Nakata R, Inoue H, Kawada T. DHA attenuates postprandial hyperlipidemia via activating PPARα in intestinal epithelial cells. J Lipid Res 2013; 54:3258-68. [PMID: 24133194 DOI: 10.1194/jlr.m034942] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
It is known that peroxisome proliferator-activated receptor (PPAR)α, whose activation reduces hyperlipidemia, is highly expressed in intestinal epithelial cells. Docosahexaenoic acid (DHA) could improve postprandial hyperlipidemia, however, its relationship with intestinal PPARα activation is not revealed. In this study, we investigated whether DHA can affect postprandial hyperlipidemia by activating intestinal PPARα using Caco-2 cells and C57BL/6 mice. The genes involved in fatty acid (FA) oxidation and oxygen consumption rate were increased, and the secretion of triacylglyceride (TG) and apolipoprotein B (apoB) was decreased in DHA-treated Caco-2 cells. Additionally, intestinal FA oxidation was induced, and TG and apoB secretion from intestinal epithelial cells was reduced, resulting in the attenuation of plasma TG and apoB levels after oral administration of olive oil in DHA-rich oil-fed mice compared with controls. However, no increase in genes involved in FA oxidation was observed in the liver. Furthermore, the effects of DHA on intestinal lipid secretion and postprandial hyperlipidemia were abolished in PPARα knockout mice. In conclusion, the present work suggests that DHA can inhibit the secretion of TG from intestinal epithelial cells via PPARα activation, which attenuates postprandial hyperlipidemia.
Collapse
Affiliation(s)
- Rino Kimura
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Karimian Azari E, Leitner C, Jaggi T, Langhans W, Mansouri A. Possible role of intestinal fatty acid oxidation in the eating-inhibitory effect of the PPAR-α agonist Wy-14643 in high-fat diet fed rats. PLoS One 2013; 8:e74869. [PMID: 24069361 PMCID: PMC3775792 DOI: 10.1371/journal.pone.0074869] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/09/2013] [Indexed: 12/21/2022] Open
Abstract
PPAR-α plays a key role in lipid metabolism; it enhances fatty acid oxidation (FAO) and ketogenesis. Pharmacological PPAR-α activation improves insulin sensitivity and reduces food intake, but its mechanisms of action remain unknown. We here report that intraperitoneal (IP) administration of the PPAR-α agonist Wy-14643 (40 mg/kg BW) reduced food intake in adult male rats fed a high-fat diet (HFD, 49% of the energy) mainly through an increase in the latency to eat after injection, and without inducing a conditioned taste avoidance. Also, IP administered Wy-14643 caused an acute (the first 60 min) decrease in the respiratory quotient (RQ) and an increase in hepatic portal vein β-hydroxybutyrate level (at 35 min) without affecting plasma non-esterified fatty acids. Given the known stimulatory effect of PPAR-α on FAO and ketogenesis, we measured the protein expression level of carnitine palmitoyltransferase-1 (CPT 1A) and mitochondrial 3-hydroxy-3-methylglutaryl-coenzyme A synthase (HMG-CoAS2), two key enzymes for FAO and ketogenesis, respectively, in liver, duodenum and jejunum. Wy-14643 induced a significant increase in the expression of CPT 1A in the jejunum and duodenum and of HMG-CoAS2 in the jejunum, but neither CPT 1A nor HMG-CoAS2 expression was increased in the liver. The induction of CPT 1A and HMG-CoAS2 expression was associated with a decrease in the lipid droplet content selectively in the jejunum. Our findings indicate that Wy-14643 stimulates FAO and ketogenesis in the intestine, in particular in the jejunum, rather than in the liver, thus supporting the hypothesis that PPAR-α activation inhibits eating by stimulating intestinal FAO.
Collapse
Affiliation(s)
| | - Claudia Leitner
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Thomas Jaggi
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Abdelhak Mansouri
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
- * E-mail:
| |
Collapse
|
36
|
Uchida A, Slipchenko MN, Eustaquio T, Leary JF, Cheng JX, Buhman KK. Intestinal acyl-CoA:diacylglycerol acyltransferase 2 overexpression enhances postprandial triglyceridemic response and exacerbates high fat diet-induced hepatic triacylglycerol storage. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1377-85. [PMID: 23643496 DOI: 10.1016/j.bbalip.2013.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 04/24/2013] [Accepted: 04/25/2013] [Indexed: 02/06/2023]
Abstract
Intestinal acyl-CoA:diacylglycerol acyltransferase 2 (DGAT2) is important in the cellular and physiological responses to dietary fat. To determine the effect of increased intestinal DGAT2 on cellular and physiological responses to acute and chronic dietary fat challenges, we generated mice with intestine-specific overexpression of DGAT2 and compared them with intestine-specific overexpression of DGAT1 and wild-type (WT) mice. We found that when intestinal DGAT2 is present in excess, triacylglycerol (TG) secretion from enterocytes is enhanced compared to WT mice; however, TG storage within enterocytes is similar compared to WT mice. We found that when intestinal DGAT2 is present in excess, mRNA levels of genes involved in fatty acid oxidation were reduced. This result suggests that reduced fatty acid oxidation may contribute to increased TG secretion by overexpression of DGAT2 in intestine. Furthermore, this enhanced supply of TG for secretion in Dgat2(Int) mice may be a significant contributing factor to the elevated fasting plasma TG and exacerbated hepatic TG storage in response to a chronic HFD. These results highlight that altering fatty acid and TG metabolism within enterocytes has the capacity to alter systemic delivery of dietary fat and may serve as an effective target for preventing and treating metabolic diseases such as hepatic steatosis.
Collapse
Affiliation(s)
- Aki Uchida
- Purdue University, West Lafayette, IN, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Hettne KM, Boorsma A, van Dartel DAM, Goeman JJ, de Jong E, Piersma AH, Stierum RH, Kleinjans JC, Kors JA. Next-generation text-mining mediated generation of chemical response-specific gene sets for interpretation of gene expression data. BMC Med Genomics 2013; 6:2. [PMID: 23356878 PMCID: PMC3572439 DOI: 10.1186/1755-8794-6-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 01/25/2013] [Indexed: 11/10/2022] Open
Abstract
Background Availability of chemical response-specific lists of genes (gene sets) for pharmacological and/or toxic effect prediction for compounds is limited. We hypothesize that more gene sets can be created by next-generation text mining (next-gen TM), and that these can be used with gene set analysis (GSA) methods for chemical treatment identification, for pharmacological mechanism elucidation, and for comparing compound toxicity profiles. Methods We created 30,211 chemical response-specific gene sets for human and mouse by next-gen TM, and derived 1,189 (human) and 588 (mouse) gene sets from the Comparative Toxicogenomics Database (CTD). We tested for significant differential expression (SDE) (false discovery rate -corrected p-values < 0.05) of the next-gen TM-derived gene sets and the CTD-derived gene sets in gene expression (GE) data sets of five chemicals (from experimental models). We tested for SDE of gene sets for six fibrates in a peroxisome proliferator-activated receptor alpha (PPARA) knock-out GE dataset and compared to results from the Connectivity Map. We tested for SDE of 319 next-gen TM-derived gene sets for environmental toxicants in three GE data sets of triazoles, and tested for SDE of 442 gene sets associated with embryonic structures. We compared the gene sets to triazole effects seen in the Whole Embryo Culture (WEC), and used principal component analysis (PCA) to discriminate triazoles from other chemicals. Results Next-gen TM-derived gene sets matching the chemical treatment were significantly altered in three GE data sets, and the corresponding CTD-derived gene sets were significantly altered in five GE data sets. Six next-gen TM-derived and four CTD-derived fibrate gene sets were significantly altered in the PPARA knock-out GE dataset. None of the fibrate signatures in cMap scored significant against the PPARA GE signature. 33 environmental toxicant gene sets were significantly altered in the triazole GE data sets. 21 of these toxicants had a similar toxicity pattern as the triazoles. We confirmed embryotoxic effects, and discriminated triazoles from other chemicals. Conclusions Gene set analysis with next-gen TM-derived chemical response-specific gene sets is a scalable method for identifying similarities in gene responses to other chemicals, from which one may infer potential mode of action and/or toxic effect.
Collapse
Affiliation(s)
- Kristina M Hettne
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Obrowsky S, Chandak PG, Patankar JV, Povoden S, Schlager S, Kershaw EE, Bogner-Strauss JG, Hoefler G, Levak-Frank S, Kratky D. Adipose triglyceride lipase is a TG hydrolase of the small intestine and regulates intestinal PPARα signaling. J Lipid Res 2012; 54:425-35. [PMID: 23220585 PMCID: PMC3541705 DOI: 10.1194/jlr.m031716] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme mediating
triglyceride (TG) hydrolysis. The lack of ATGL results in TG accumulation in
multiple tissues, underscoring the critical role of ATGL in maintaining lipid
homeostasis. Recent evidence suggests that ATGL affects TG metabolism via
activation of peroxisome proliferator-activated receptor α (PPARα).
To investigate specific effects of intestinal ATGL on lipid metabolism we
generated mice lacking ATGL exclusively in the intestine (ATGLiKO). We found
decreased TG hydrolase activity and increased intracellular TG content in
ATGLiKO small intestines. Intragastric administration of
[3H]trioleate resulted in the accumulation of radioactive TG in the
intestine, whereas absorption into the systemic circulation was unchanged.
Intraperitoneally injected [3H]oleate also accumulated within TG in
ATGLiKO intestines, indicating that ATGL mobilizes fatty acids from the systemic
circulation absorbed by the basolateral side from the blood. Down-regulation of
PPARα target genes suggested modulation of cholesterol absorption by
intestinal ATGL. Accordingly, ATGL deficiency in the intestine resulted in
delayed cholesterol absorption. Importantly, this study provides evidence that
ATGL has no impact on intestinal TG absorption but hydrolyzes TGs taken up from
the intestinal lumen and systemic circulation. Our data support the role of ATGL
in modulating PPARα-dependent processes also in the small intestine.
Collapse
Affiliation(s)
- Sascha Obrowsky
- Institute of Molecular Biology, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
de Wit NJ, IJssennagger N, Oosterink E, Keshtkar S, Hooiveld GJ, Mensink RP, Hammer S, Smit JW, Müller M, der Meer RV. Oit1/Fam3D, a gut-secreted protein displaying nutritional status-dependent regulation. J Nutr Biochem 2012; 23:1425-33. [DOI: 10.1016/j.jnutbio.2011.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 09/08/2011] [Accepted: 09/08/2011] [Indexed: 11/26/2022]
|
40
|
Thomson ABR, Chopra A, Clandinin MT, Freeman H. Recent advances in small bowel diseases: Part II. World J Gastroenterol 2012; 18:3353-74. [PMID: 22807605 PMCID: PMC3396188 DOI: 10.3748/wjg.v18.i26.3353] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 04/05/2012] [Accepted: 04/13/2012] [Indexed: 02/06/2023] Open
Abstract
As is the case in all areas of gastroenterology and hepatology, in 2009 and 2010 there were many advances in our knowledge and understanding of small intestinal diseases. Over 1000 publications were reviewed, and the important advances in basic science as well as clinical applications were considered. In Part II we review six topics: absorption, short bowel syndrome, smooth muscle function and intestinal motility, tumors, diagnostic imaging, and cystic fibrosis.
Collapse
|
41
|
Uchida A, Whitsitt MC, Eustaquio T, Slipchenko MN, Leary JF, Cheng JX, Buhman KK. Reduced triglyceride secretion in response to an acute dietary fat challenge in obese compared to lean mice. Front Physiol 2012; 3:26. [PMID: 22375122 PMCID: PMC3285805 DOI: 10.3389/fphys.2012.00026] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 02/03/2012] [Indexed: 11/13/2022] Open
Abstract
Obesity results in abnormally high levels of triglyceride (TG) storage in tissues such as liver, heart, and muscle, which disrupts their normal functions. Recently, we found that lean mice challenged with high levels of dietary fat store TGs in cytoplasmic lipid droplets in the absorptive cells of the intestine, enterocytes, and that this storage increases and then decreases over time after an acute dietary fat challenge. The goal of this study was to investigate the effects of obesity on intestinal TG metabolism. More specifically we asked whether TG storage in and secretion from the intestine are altered in obesity. We investigated these questions in diet-induced obese (DIO) and leptin-deficient (ob/ob) mice. We found greater levels of TG storage in the intestine of DIO mice compared to lean mice in the fed state, but similar levels of TG storage after a 6-h fast. In addition, we found similar TG storage in the intestine of lean and DIO mice at multiple time points after an acute dietary fat challenge. Surprisingly, we found remarkably lower TG secretion from both DIO and ob/ob mice compared to lean controls in response to an acute dietary fat challenge. Furthermore, we found altered mRNA levels for genes involved in regulation of intestinal TG metabolism in lean and DIO mice at 6 h fasting and in response to an acute dietary fat challenge. More specifically, we found that many of the genes related to TG synthesis, chylomicron synthesis, TG storage, and lipolysis were induced in response to an acute dietary fat challenge in lean mice, but this induction was not observed in DIO mice. In fact, we found a significant decrease in intestinal mRNA levels of genes related to lipolysis and fatty acid oxidation in DIO mice in response to an acute dietary fat challenge. Our findings demonstrate altered TG handling by the small intestine of obese compared to lean mice.
Collapse
Affiliation(s)
- Aki Uchida
- Interdisciplinary Life Science Program, Purdue University West Lafayette, IN, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Quesada H, Pajuelo D, Fernández-Iglesias A, Díaz S, Ardevol A, Blay M, Salvadó M, Arola L, Blade C. Proanthocyanidins modulate triglyceride secretion by repressing the expression of long chain acyl-CoA synthetases in Caco2 intestinal cells. Food Chem 2011. [DOI: 10.1016/j.foodchem.2011.05.125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
43
|
Roy NC, Altermann E, Park ZA, McNabb WC. A comparison of analog and Next-Generation transcriptomic tools for mammalian studies. Brief Funct Genomics 2011; 10:135-50. [DOI: 10.1093/bfgp/elr005] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
44
|
Involvement of CD36 and intestinal alkaline phosphatases in fatty acid transport in enterocytes, and the response to a high-fat diet. Life Sci 2011; 88:384-91. [DOI: 10.1016/j.lfs.2010.12.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 11/17/2010] [Accepted: 12/06/2010] [Indexed: 11/23/2022]
|
45
|
Martins dos Santos V, Müller M, de Vos WM. Systems biology of the gut: the interplay of food, microbiota and host at the mucosal interface. Curr Opin Biotechnol 2011; 21:539-50. [PMID: 20817507 DOI: 10.1016/j.copbio.2010.08.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 08/05/2010] [Indexed: 01/06/2023]
Abstract
Our intestinal tract is colonized since birth by complex and subject-specific microbial communities that interact with the host. The human adult microbiota has recently been characterized by deep metagenomic sequencing and several hundreds of intestinal genomes have been characterized at the sequence level. Moreover, the transcriptional response of the host and selected microbes has been identified both in animal model systems and in human. Similarly, the transcriptional response of the host to different diets has been determined in humans, germ-free and gene knockout animals. These developments bring the intestinal tract in the realm of systems biology. An integrated, modular modelling framework that cross-links top-down and bottom-up approaches for the various levels of biological organization is paramount for the understanding of intestinal function.
Collapse
Affiliation(s)
- Vítor Martins dos Santos
- Laboratory of Systems & Synthetic Biology, Wageningen University, Dreijenplein 10, 6710 HB Wageningen, The Netherlands.
| | | | | |
Collapse
|
46
|
Uchida A, Slipchenko MN, Cheng JX, Buhman KK. Fenofibrate, a peroxisome proliferator-activated receptor α agonist, alters triglyceride metabolism in enterocytes of mice. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:170-6. [PMID: 21215818 DOI: 10.1016/j.bbalip.2010.12.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 12/17/2010] [Accepted: 12/21/2010] [Indexed: 11/17/2022]
Abstract
Fenofibrate, a drug in the fibrate class of amphiphathic carboxylic acids, has multiple blood lipid modifying actions, which are beneficial to the prevention of atherosclerosis. One of its benefits is in lowering fasting and postprandial blood triglyceride (TG) concentrations. The goal of this study was to determine whether the hypotriglyceridemic actions of fenofibrate in the postprandial state include alterations in TG and fatty acid metabolism in the small intestine. We found that the hypotriglyceridemic actions of fenofibrate in the postprandial state of high-fat (HF) fed mice include a decrease in supply of TG for secretion by the small intestine. A decreased supply of TG for secretion was due in part to the decreased dietary fat absorption and increased intestinal fatty acid oxidation in fenofibrate compared to vehicle treated HF fed mice. These results suggest that the effects of fenofibrate on the small intestine play a critical role in the hypotriglyceridemic effects of fenofibrate.
Collapse
Affiliation(s)
- Aki Uchida
- Department of Foods and Nutrition, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | |
Collapse
|
47
|
Jelier R, Goeman JJ, Hettne KM, Schuemie MJ, den Dunnen JT, 't Hoen PAC. Literature-aided interpretation of gene expression data with the weighted global test. Brief Bioinform 2010; 12:518-29. [PMID: 21183478 DOI: 10.1093/bib/bbq082] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Most methods for the interpretation of gene expression profiling experiments rely on the categorization of genes, as provided by the Gene Ontology (GO) and pathway databases. Due to the manual curation process, such databases are never up-to-date and tend to be limited in focus and coverage. Automated literature mining tools provide an attractive, alternative approach. We review how they can be employed for the interpretation of gene expression profiling experiments. We illustrate that their comprehensive scope aids the interpretation of data from domains poorly covered by GO or alternative databases, and allows for the linking of gene expression with diseases, drugs, tissues and other types of concepts. A framework for proper statistical evaluation of the associations between gene expression values and literature concepts was lacking and is now implemented in a weighted extension of global test. The weights are the literature association scores and reflect the importance of a gene for the concept of interest. In a direct comparison with classical GO-based gene sets, we show that use of literature-based associations results in the identification of much more specific GO categories. We demonstrate the possibilities for linking of gene expression data to patient survival in breast cancer and the action and metabolism of drugs. Coupling with online literature mining tools ensures transparency and allows further study of the identified associations. Literature mining tools are therefore powerful additions to the toolbox for the interpretation of high-throughput genomics data.
Collapse
Affiliation(s)
- Rob Jelier
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
48
|
Diep TA, Madsen AN, Holst B, Kristiansen MM, Wellner N, Hansen SH, Hansen HS. Dietary fat decreases intestinal levels of the anorectic lipids through a fat sensor. FASEB J 2010; 25:765-74. [PMID: 20959516 DOI: 10.1096/fj.10-166595] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This study was undertaken to investigate the link between dietary fat content and intestinal levels of anorectic N-acylethanolamines (NAEs), including oleoylethanolamide (OEA), palmitoylethanolamide (PEA), and linoleoylethanolamide (LEA). Male rats were fed high-fat diets (HFDs) with variable percentages of fat [20-45% of total energy (E%)] for 1-7 d; afterward, the jejunums were isolated, and jejunal NAE levels were measured by liquid-chromatography mass spectrometry. Enzyme activities and mRNA expression levels were measured for two synthesizing enzymes, N-acylphosphatidylethanolamine-specific phospholipase D (NAPE-PLD) and glycerophosphodiesterase (GDE1), and one degrading enzyme, fatty acid amide hydrolase (FAAH). We found a dose-response relation between the quantity/percentage of dietary fat, irrespective of the energy density, and the reduction of intestinal levels of OEA, PEA, and LEA. The reductions were present after 1 d of 45E% HFD. LEA, the major NAE species, was shown to have an anorectic potency slightly less than that of OEA but higher than PEA. Regulation at the enzyme level seems not to explain the changes in NAE levels. The results suggest the presence of a fat sensor, mediating the reduced intestinal NAE levels. The intestinal NAE levels are reduced in a dose- and time-dependent manner in response to dietary fat intake, and this may contribute to the well-known hyperphagic effect of HFDs.
Collapse
Affiliation(s)
- Thi Ai Diep
- Department of Pharmacology and Pharmacotheraphy, Faculty of Pharmaceutical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
49
|
Barrera epidérmica y nutrición lipídica. La conexión PPAR e inmunopatología inflamatoria como nuevas dianas de tratamiento en dermatitis atópica y psoriasis. ACTAS DERMO-SIFILIOGRAFICAS 2010; 101:585-599. [DOI: 10.1016/j.ad.2010.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Revised: 03/07/2010] [Accepted: 03/30/2010] [Indexed: 12/20/2022] Open
|
50
|
Villarrubia V, Vidal-Asensi S, Pérez-Bañasco V, Cuevas-Santos J, Cisterna-Cáncer R. Lipid Nutrition and the Epidermal Barrier: The Connection Between Immune-Mediated Inflammatory Diseases and Peroxisome Proliferator-Activated Receptors, a New Therapeutic Target in Psoriasis and Atopic Dermatitis. ACTAS DERMO-SIFILIOGRAFICAS 2010. [DOI: 10.1016/s1578-2190(10)70681-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|