1
|
Rifa RA, Lavado R. Cytotoxic impacts of seven alternative bisphenols on human in vitro cellular models. CHEMOSPHERE 2024; 366:143408. [PMID: 39326710 DOI: 10.1016/j.chemosphere.2024.143408] [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: 04/11/2024] [Revised: 07/27/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Bisphenols (BPs), common in plastics, coatings, and resins, are under scrutiny for potential endocrine disruption. Despite banning bisphenol A (BPA), its perceived safer alternatives may still pose health risks, urging thorough studies on their toxicity mechanisms. This study aimed to investigate the cellular toxicity of the top seven most commonly used BPs, bisphenol S (BPS), bisphenol F (BPF), bisphenol AF (BPAF), bisphenol P (BPP), bisphenol AP (BPAP), bisphenol B (BPB), bisphenol E (BPE) in eight different relevant human in vitro cell models: liver (HepaRG), intestinal (Caco-2), breast (T47D), brain (HMC-3), lungs (MRC-5), kidney (HEK293), endothelial (HMEC-1), and skin (HEK-001) cell lines. BPE manifested the highest cytotoxicity in Caco-2 cells, presenting an EC50 value of roughly 0.2 μM (95% confidence interval). In contrast, HEK293 and HepaRG cells demonstrated significant resilience to BPS (EC50 > 1000 μM). BPAF, BPP, and BPAP had consistently low EC50 values across cell lines (6-27.9 μM, 0.6-134.7 μM, and 3.6-178.8 μM), indicating elevated toxicity. After 24 h, all bisphenols adhered to nominal concentrations except BPAF, BPP, and BPS. BPP's concentration notably decreased (30.82 ± 5.53% of nominal value). The results revealed diverse effects of bisphenol analogs on different cell types. These findings emphasized the considerable cytotoxic potential of specific bisphenol analogs across various human cell models, underlining the necessity for a re-evaluation of their safety and regulatory standards.
Collapse
Affiliation(s)
- Rafia Afroze Rifa
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Ramon Lavado
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA.
| |
Collapse
|
2
|
Noguchi M, Shimizu M, Lu P, Takahashi Y, Yamauchi Y, Sato S, Kiyono H, Kishino S, Ogawa J, Nagata K, Sato R. Lactic acid bacteria-derived γ-linolenic acid metabolites are PPARδ ligands that reduce lipid accumulation in human intestinal organoids. J Biol Chem 2022; 298:102534. [PMID: 36162507 PMCID: PMC9636582 DOI: 10.1016/j.jbc.2022.102534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Gut microbiota regulate physiological functions in various hosts, such as energy metabolism and immunity. Lactic acid bacteria, including Lactobacillus plantarum, have a specific polyunsaturated fatty acid saturation metabolism that generates multiple fatty acid species, such as hydroxy fatty acids, oxo fatty acids, conjugated fatty acids, and trans-fatty acids. How these bacterial metabolites impact host physiology is not fully understood. Here, we investigated the ligand activity of lactic acid bacteria–produced fatty acids in relation to nuclear hormone receptors expressed in the small intestine. Our reporter assays revealed two bacterial metabolites of γ-linolenic acid (GLA), 13-hydroxy-cis-6,cis-9-octadecadienoic acid (γHYD), and 13-oxo-cis-6,cis-9-octadecadienoic acid (γKetoD) activated peroxisome proliferator-activated receptor delta (PPARδ) more potently than GLA. We demonstrate that both γHYD and γKetoD bound directly to the ligand-binding domain of human PPARδ. A docking simulation indicated that four polar residues (T289, H323, H449, and Y473) of PPARδ donate hydrogen bonds to these fatty acids. Interestingly, T289 does not donate a hydrogen bond to GLA, suggesting that bacterial modification of GLA introducing hydroxy and oxo group determines ligand selectivity. In human intestinal organoids, we determined γHYD and γKetoD increased the expression of PPARδ target genes, enhanced fatty acid β-oxidation, and reduced intracellular triglyceride accumulation. These findings suggest that γHYD and γKetoD, which gut lactic acid bacteria could generate, are naturally occurring PPARδ ligands in the intestinal tract and may improve lipid metabolism in the human intestine.
Collapse
Affiliation(s)
- Makoto Noguchi
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Makoto Shimizu
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo.
| | - Peng Lu
- Food Biotechnology and Structural Biology Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Yu Takahashi
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Yoshio Yamauchi
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo; Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Shintaro Sato
- Department of Microbiology and Immunology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama
| | - Hiroshi Kiyono
- Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Future Medicine Education and Research Organization, Chiba University, Chiba
| | - Shigenobu Kishino
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto
| | - Jun Ogawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto
| | - Koji Nagata
- Food Biotechnology and Structural Biology Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Ryuichiro Sato
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo.
| |
Collapse
|
3
|
Organoid-derived intestinal epithelial cells are a suitable model for preclinical toxicology and pharmacokinetic studies. iScience 2022; 25:104542. [PMID: 35754737 PMCID: PMC9218437 DOI: 10.1016/j.isci.2022.104542] [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: 11/15/2021] [Revised: 01/06/2022] [Accepted: 06/02/2022] [Indexed: 12/28/2022] Open
Abstract
Intestinal organoids are physiologically relevant tools used for cellular models. However, the suitability of organoids to examine biological functions over existing established cell lines lacks sufficient evidence. Cytochrome P450 3A4 (CYP3A4) induction by pregnane X receptor ligands, glucose uptake via sodium/glucose cotransporter 1, and microsomal triglyceride transfer protein-dependent ApoB-48 secretion, which are critical for human intestinal metabolism, were observed in organoid-derived two-dimensional cells but little in Caco-2 cells. CYP3A4 induction evaluation involved a simplified method of establishing organoids that constitutively expressed a reporter gene. Compound screening identified several anticancer drugs with selective activities toward Caco-2 cells, highlighting their characteristics as cancer cells. Another compound screening revealed a decline in N-(4-hydroxyphenyl)retinamide cytotoxicity upon rifampicin treatment in organoid-derived cells, under CYP3A4-induced conditions. This study shows that organoid-derived intestinal epithelial cells (IECs) possess similar physiological properties as intestinal epithelium and can serve as tools for enhancing the prediction of biological activity in humans. Comparison of mRNA expression between organoid-derived intestinal epithelial cells (IECs) and Caco-2 cells Evaluation of CYP3A4, SGLT1, and MTP protein function in organoid-derived IECs Identification of anti-cancer drugs as selective cytotoxicity against Caco-2 cells Reduction of N-(4-hydroxyphenyl)retinamide (4-HPR) cytotoxicity by rifampicin in organoid-derived IECs
Collapse
|
4
|
He B, Wang Z, Moreau R. Chylomicron production is repressed by RPTOR knockdown, R-α-lipoic acid and 4-phenylbutyric acid in human enterocyte-like Caco-2 cells. J Nutr Biochem 2022; 108:109087. [PMID: 35691593 DOI: 10.1016/j.jnutbio.2022.109087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 03/28/2022] [Accepted: 05/14/2022] [Indexed: 10/18/2022]
Abstract
Although the role of mechanistic target of rapamycin complex 1 (mTORC1) in lipid metabolism has been the subject of previous research, its function in chylomicron production is not known. In this study, we created three stable human colorectal adenocarcinoma Caco-2 cell lines exhibiting normal, low or high mTORC1 kinase activity, and used these cells to investigate the consequences of manipulating mTORC1 activity on enterocyte differentiation and chylomicron-like particle production. Constitutively active mTORC1 induced Caco-2 cell proliferation and differentiation (as judged by alkaline phosphatase activity) but weakened transepithelial electrical resistance (TEER). Repressed mTORC1 activity due to the knockdown of RPTOR significantly decreased the expression of lipogenic genes FASN, DGAT1 and DGAT2, lipoprotein assembly genes APOB and MTTP, reduced protein expression of APOB, MTTP and FASN, downregulated the gene expression of very long-chain fatty acyl-CoA ligase (FATP2), acyl-CoA binding protein (DBI), and prechylomicron transport vesicle-associated proteins VAMP7 (vesicle-associated membrane protein 7) and SAR1B (secretion associated Ras related GTPase 1B) resulting in the repression of apoB-containing triacylglycerol-rich lipoprotein secretion. Exposure of Caco-2 cells harboring a constitutively active mTORC1 to short-chain fatty acid derivatives, R-α-lipoic acid and 4-phenylbutyric acid, downregulated chylomicron-like particle secretion by interfering with the lipidation and assembly of the particles, and concomitantly repressed mTORC1 activity with no change to Raptor abundance or PRAS40 (Thr246) phosphorylation. R-α-lipoic acid and 4-phenylbutyric acid may be useful to mitigate intestinal lipoprotein overproduction and associated postprandial inflammation.
Collapse
Affiliation(s)
- Bo He
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Zhigang Wang
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Régis Moreau
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| |
Collapse
|
5
|
Velagacherla V, Suresh A, Mehta CH, Nayak UY. Advances and challenges in nintedanib drug delivery. Expert Opin Drug Deliv 2021; 18:1687-1706. [PMID: 34556001 DOI: 10.1080/17425247.2021.1985460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Nintedanib (N.T.B) is an orally administered tyrosine kinase inhibitor that has been approved recently by U.S.F.D.A for idiopathic pulmonary fibrosis (I.P.F) and systemic sclerosis-associated interstitial lung disease (S.Sc-I.L.D). N.T.B is also prescribed in COVID-19 patients associated with I.P.F. However, it has an extremely low bioavailability of around 4.7%, and hence, researchers are attempting to address this drawback by different approaches. AREAS COVERED This review article focuses on enlisting all the formulation attempts explored by researchers to increase the bioavailability of N.T.B while also providing meaningful insight into the unexplored areas in formulation development, such as targeting of the lymphatic system and transdermal delivery. All the patents on the formulation development of N.T.B have also been summarized. EXPERT OPINION N.T.B has the potential to act on multiple diseases that are still being discovered, but its extremely low bioavailability is a challenge that is to be dealt with for obtaining the full benefit. Few studies have been performed aiming at improving the bioavailability, but there are unexplored areas that can be used, a few of which are explained in this article. However, the ability to reproduce laboratory results when scaling up to the industry level is the only factor to be taken into consideration.
Collapse
Affiliation(s)
- Varalakshmi Velagacherla
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Akhil Suresh
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Chetan H Mehta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
6
|
Intestinal and Hepatic Uptake of Dietary Peroxidized Lipids and Their Decomposition Products, and Their Subsequent Effects on Apolipoprotein A1 and Paraoxonase1. Antioxidants (Basel) 2021; 10:antiox10081258. [PMID: 34439506 PMCID: PMC8389297 DOI: 10.3390/antiox10081258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/23/2022] Open
Abstract
Both pro- and antiatherosclerotic effects have been ascribed to dietary peroxidized lipids. Confusion on the role of peroxidized lipids in atherosclerotic cardiovascular disease is punctuated by a lack of understanding regarding the metabolic fate and potential physiological effects of dietary peroxidized lipids and their decomposition products. This study sought to determine the metabolic fate and physiological ramifications of 13-hydroperoxyoctadecadienoic acid (13-HPODE) and 13-HODE (13-hydroxyoctadecadienoic acid) supplementation in intestinal and hepatic cell lines, as well as any effects resulting from 13-HPODE or 13-HODE degradation products. In the presence of Caco-2 cells, 13-HPODE was rapidly reduced to 13-HODE. Upon entering the cell, 13-HODE appears to undergo decomposition, followed by esterification. Moreover, 13-HPODE undergoes autodecomposition to produce aldehydes such as 9-oxononanoic acid (9-ONA). Results indicate that 9-ONA was oxidized to azelaic acid (AzA) rapidly in cell culture media, but AzA was poorly absorbed by intestinal cells and remained detectable in cell culture media for up to 18 h. An increased apolipoprotein A1 (ApoA1) secretion was observed in Caco-2 cells in the presence of 13-HPODE, 9-ONA, and AzA, whereas such induction was not observed in HepG2 cells. However, 13-HPODE treatments suppressed paraoxonase 1 (PON1) activity, suggesting the induction of ApoA1 secretion by 13-HPODE may not represent functional high-density lipoprotein (HDL) capable of reducing oxidative stress. Alternatively, AzA induced both ApoA1 secretion and PON1 activity while suppressing ApoB secretion in differentiated Caco-2 cells but not in HepG2. These results suggest oxidation of 9-ONA to AzA might be an important phenomenon, resulting in the accumulation of potentially beneficial dietary peroxidized lipid-derived aldehydes.
Collapse
|
7
|
Koudoufio M, Feldman F, Ahmarani L, Delvin E, Spahis S, Desjardins Y, Levy E. Intestinal protection by proanthocyanidins involves anti-oxidative and anti-inflammatory actions in association with an improvement of insulin sensitivity, lipid and glucose homeostasis. Sci Rep 2021; 11:3878. [PMID: 33594093 PMCID: PMC7886900 DOI: 10.1038/s41598-020-80587-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/16/2020] [Indexed: 12/16/2022] Open
Abstract
Recent advances have added another dimension to the complexity of cardiometabolic disorders (CMD) by directly implicating the gastrointestinal tract as a key player. In fact, multiple factors could interfere with intestinal homeostasis and elicit extra-intestinal CMD. As oxidative stress (OxS), inflammation, insulin resistance and lipid abnormalities are among the most disruptive events, the aim of the present study is to explore whether proanthocyanidins (PACs) exert protective effects against these disorders. To this end, fully differentiated intestinal Caco-2/15 cells were pre-incubated with PACs with and without the pro-oxidant and pro-inflammatory iron/ascorbate (Fe/Asc). PACs significantly reduce malondialdehyde, a biomarker of lipid peroxidation, and raise antioxidant SOD2 and GPx via the increase of NRF2/Keap1 ratio. Likewise, PACs decrease the inflammatory agents TNFα and COX2 through abrogation of NF-κB. Moreover, according to crucial biomarkers, PACs result in lipid homeostasis improvement as reflected by enhanced fatty acid β-oxidation, diminished lipogenesis, and lowered gluconeogenesis as a result of PPARα, γ and SREBP1c modulation. Since these metabolic routes are mainly regulated by insulin sensitivity, we have examined the insulin signaling pathway and found an upregulation of phosphoPI3K/Akt and downregulation of p38-MAPK expressions, indicating beneficial effects in response to PACs. Taken together, PACs display the potential to counterbalance OxS and inflammation in Fe/Asc-exposed intestinal cells, in association with an improvement of insulin sensitivity, which ameliorates lipid and glucose homeostasis.
Collapse
Affiliation(s)
- Mireille Koudoufio
- Research Centre, CHU Sainte-Justine, Université de Montréal, 3175 Ste Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Nutrition, Université de Montréal, Montreal, QC, H3T 1A8, Canada.,Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Francis Feldman
- Research Centre, CHU Sainte-Justine, Université de Montréal, 3175 Ste Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Nutrition, Université de Montréal, Montreal, QC, H3T 1A8, Canada.,Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Lena Ahmarani
- Research Centre, CHU Sainte-Justine, Université de Montréal, 3175 Ste Catherine Road, Montreal, QC, H3T 1C5, Canada.,Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Edgard Delvin
- Research Centre, CHU Sainte-Justine, Université de Montréal, 3175 Ste Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Biochemistry, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Schohraya Spahis
- Research Centre, CHU Sainte-Justine, Université de Montréal, 3175 Ste Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Nutrition, Université de Montréal, Montreal, QC, H3T 1A8, Canada.,Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Yves Desjardins
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Emile Levy
- Research Centre, CHU Sainte-Justine, Université de Montréal, 3175 Ste Catherine Road, Montreal, QC, H3T 1C5, Canada. .,Department of Nutrition, Université de Montréal, Montreal, QC, H3T 1A8, Canada. .,Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC, G1V 0A6, Canada.
| |
Collapse
|
8
|
Jeon JW, Lee SH, Kim D, Sung JH. In vitro hepatic steatosis model based on gut-liver-on-a-chip. Biotechnol Prog 2021; 37:e3121. [PMID: 33393209 DOI: 10.1002/btpr.3121] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022]
Abstract
Hepatic steatosis, also known as fatty liver disease, occurs due to abnormal lipid accumulation in the liver. It has been known that gut absorption also plays an important role in the mechanism underlying hepatic steatosis. Conventional in vitro cell culture models have limitations in recapitulating the mechanisms of hepatic steatosis because it does not include the gut absorption process. Previously, we reported development of a microfluidic gut-liver chip that can recapitulate the gut absorption of fatty acids and subsequent lipid accumulation in liver cells. In this study, we performed a series of experiments to verify that our gut-liver chip reproduces various aspects of hepatic steatosis. The absorption of fatty acids was evaluated under various culture conditions. The anti-steatotic effect of turofexorate isopropyl (XL-335) and metformin was tested, and both drugs showed different action mechanisms. In addition, the oxidative stress induced by lipid absorption was evaluated. Our results demonstrate the potential of the gut-liver chip for use as a novel, physiologically realistic in vitro model to study fatty liver disease.
Collapse
Affiliation(s)
- Joong-Won Jeon
- Department of Chemical Engineering, Hongik University, Seoul, Republic of Korea
| | - Seung Hwan Lee
- Department of Bionano Engineering, Hanyang University, Ansan, Republic of Korea
| | - Donghyun Kim
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Vincent M, Ménard O, Etienne J, Ossemond J, Durand A, Buffin R, Loizon E, Meugnier E, Deglaire A, Dupont D, Picaud JC, Knibbe C, Michalski MC, Penhoat A. Human milk pasteurisation reduces pre-lipolysis but not digestive lipolysis and moderately decreases intestinal lipid uptake in a combination of preterm infant in vitro models. Food Chem 2020; 329:126927. [PMID: 32516717 DOI: 10.1016/j.foodchem.2020.126927] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 11/19/2022]
Abstract
Donor human milk, pasteurised for safety reasons, is the first alternative for feeding preterm infants when mothers' own milk is unavailable. Breastmilk pasteurisation impact on lipid digestion and absorption was evaluated by a static in vitro digestion model for preterm infants coupled with intestinal absorption using Caco-2/TC7 cells. Lipid absorption was quantified by digital image analysis of lipid droplets, by measurement of basolateral triglyceride concentration and by analysing the expression of major genes involved. After in vitro digestion, lipolysis extent was 13% lower in pasteurised human milk (PHM) than in raw human milk (RHM). In Caco-2/TC7 cells, the number of lipid droplets was identical for both milk types, while the mean droplet area was 17% smaller with PHM. Altogether, pasteurisation decreased the pre-lipolysis of human milk. This initial difference in free fatty acid amount was only partially buffered by the subsequent processes of in vitro digestion and cellular lipid absorption.
Collapse
Affiliation(s)
- Marine Vincent
- Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Pierre-Bénite, France; Neonatology Department, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France.
| | | | - Julie Etienne
- Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Pierre-Bénite, France; Project-Team BEAGLE, INRIA, Villeurbanne, France.
| | | | - Annie Durand
- Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Villeurbanne, France.
| | - Rachel Buffin
- Neonatology Department, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France.
| | - Emmanuelle Loizon
- Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Pierre-Bénite, France.
| | - Emmanuelle Meugnier
- Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Pierre-Bénite, France.
| | | | | | - Jean-Charles Picaud
- Neonatology Department, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France; Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Pierre-Bénite, France.
| | - Carole Knibbe
- Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Pierre-Bénite, France; Project-Team BEAGLE, INRIA, Villeurbanne, France.
| | - Marie-Caroline Michalski
- Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Pierre-Bénite, France.
| | - Armelle Penhoat
- Univ Lyon, CarMeN Laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon1, Pierre-Bénite, France.
| |
Collapse
|
10
|
Nano lipid based carriers for lymphatic voyage of anti-cancer drugs: An insight into the in-vitro, ex-vivo, in-situ and in-vivo study models. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101899] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
11
|
Probiotic from human breast milk, Lactobacillus fermentum, promotes growth in animal model of chronic malnutrition. Pediatr Res 2020; 88:374-381. [PMID: 32023624 DOI: 10.1038/s41390-020-0774-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/03/2020] [Accepted: 01/15/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND Chronic undernutrition leads to growth hormone resistance and poor growth in children, which has been shown to be modulated by microbiota. We studied whether Lactobacillus fermentum CECT5716 (Lf CECT5716), isolated from mother's breast milk, could promote juvenile growth through the modulation of lipid absorption in a model of starvation. METHODS Germ-free (GF) Drosophila melanogaster larvae were inoculated with Lf CECT5716 in conditions of undernutrition with and without infant formula. The impact of Lf CECT5716 on larval growth was assessed 7 days after egg laying (AED) by measuring the larval size and on maturation by measuring the emergence of pupae during 21 days AED. For lipid absorption test, Caco2/TC7 intestinal cells were incubated with Lf CECT5716 and challenged with mixed lipid micelles. RESULTS The mono-associated larvae with Lf CECT5716 were significantly longer than GF larvae (3.7 vs 2.5 mm; p < 0.0001). The effect was maintained when Lf CECT5716 was added to the infant formula. The maturation time of larvae was accelerated by Lf CECT5716 (12 vs 13.2 days; p = 0.01). Lf CECT5716 did not have significant impact on lipid absorption in Caco2/TC7 cells. CONCLUSIONS Lf CECT5716 is a growth-promoting strain upon undernutrition in Drosophila, with a maintained effect when added to an infant formula but without effect on lipid absorption in vitro.
Collapse
|
12
|
Glycomacropeptide Prevents Iron/Ascorbate-Induced Oxidative Stress, Inflammation and Insulin Sensitivity with an Impact on Lipoprotein Production in Intestinal Caco-2/15 Cells. Nutrients 2020; 12:nu12041175. [PMID: 32331475 PMCID: PMC7231176 DOI: 10.3390/nu12041175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/13/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Background. Metabolic Syndrome (MetS), a major worldwide concern for the public health system, refers to a cluster of key metabolic components, and represents a risk factor for diabetes and cardiovascular diseases. As oxidative stress (OxS) and inflammation are the major triggers of insulin sensitivity (IS), a cardinal MetS feature, the principal aim of the present work is to determine whether glycomacropeptide (GMP), a milk-derived bioactive peptide, exerts beneficial effects on their expression. Methods. Fully differentiated intestinal Caco-2/15 cells are used to evaluate the preventive action of 2 mg/mL GMP against OxS and inflammation induced by the mixture iron-ascorbate (Fe/Asc) (200 μM:2 mM). The potency of GMP of decreasing the production of lipoproteins, including chylomicrons (CM), very-low-density lipoproteins (VLDL) and low-density lipoproteins (LDL) is also assessed. Results. The administration of GMP significantly reduces malondialdehyde, a biomarker of lipid peroxidation, and raises superoxide dismutase 2 and glutathione peroxidase via the induction of the nuclear factor erythroid 2–related factor 2, a transcription factor, which orchestrates cellular antioxidant defenses. Similarly, GMP markedly lowers the inflammatory agents tumor necrosis factor-α and cyclooxygenase-2 via abrogation of the nuclear transcription factor-kB. Moreover, GMP-treated cells show a down-regulation of Fe/Asc-induced mitogen activated protein kinase pathway, suggesting greater IS. Finally, GMP decreases the production of CM, VLDL, and LDL. Conclusions. Our results highlight the effectiveness of GMP in attenuating OxS, inflammation and lipoprotein biogenesis, as well as improving IS, the key components of MetS. Further investigation is needed to elucidate the mechanisms mediating the preventive action of GMP.
Collapse
|
13
|
Hiebl V, Schachner D, Ladurner A, Heiss EH, Stangl H, Dirsch VM. Caco-2 Cells for Measuring Intestinal Cholesterol Transport - Possibilities and Limitations. Biol Proced Online 2020; 22:7. [PMID: 32308567 PMCID: PMC7149936 DOI: 10.1186/s12575-020-00120-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
Background The human Caco-2 cell line is a common in vitro model of the intestinal epithelial barrier. As the intestine is a major interface in cholesterol turnover and represents a non-biliary pathway for cholesterol excretion, Caco-2 cells are also a valuable model for studying cholesterol homeostasis, including cholesterol uptake and efflux. Currently available protocols are, however, either sketchy or not consistent among different laboratories. Our aim was therefore to generate a collection of optimized protocols, considering the different approaches of the different laboratories and to highlight possibilities and limitations of measuring cholesterol transport with this cell line. Results We developed comprehensive and quality-controlled protocols for the cultivation of Caco-2 cells on filter inserts in a single tight monolayer. A cholesterol uptake as well as a cholesterol efflux assay is described in detail, including suitable positive controls. We further show that Caco-2 cells can be efficiently transfected for luciferase reporter gene assays in order to determine nuclear receptor activation, main transcriptional regulators of cholesterol transporters (ABCA1, ABCB1, ABCG5/8, NPC1L1). Detection of protein and mRNA levels of cholesterol transporters in cells grown on filter inserts can pose challenges for which we highlight essential steps and alternative approaches for consideration. A protocol for viability assays with cells differentiated on filter inserts is provided for the first time. Conclusions The Caco-2 cell line is widely used in the scientific community as model for the intestinal epithelium, although with highly divergent protocols. The herein provided information and protocols can be a common basis for researchers intending to use Caco-2 cells in the context of cellular cholesterol homeostasis.
Collapse
Affiliation(s)
- Verena Hiebl
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Daniel Schachner
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Angela Ladurner
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Elke H Heiss
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Herbert Stangl
- 2Institute of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Verena M Dirsch
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
14
|
Increased Cellular Uptake of Polyunsaturated Fatty Acids and Phytosterols from Natural Micellar Oil. Nutrients 2020; 12:nu12010150. [PMID: 31948089 PMCID: PMC7019862 DOI: 10.3390/nu12010150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/16/2019] [Accepted: 01/03/2020] [Indexed: 12/15/2022] Open
Abstract
The transport of hydrophobic compounds to recipient cells is a critical step in nutrient supplementation. Here, we tested the effect of phospholipid-based emulsification on the uptake of hydrophobic compounds into various tissue culture cell lines. In particular, the uptake of ω-3 fatty acids from micellar or nonmicellar algae oil into cell models for enterocytes, epithelial cells, and adipocytes was tested. Micellization of algae oil did not result in adverse effects on cell viability in the target cells. In general, both micellar and nonmicellar oil increased intracellular docosahexaenoic acid (DHA) levels. However, micellar oil was more effective in terms of augmenting the intracellular levels of total polyunsaturated fatty acids (PUFAs) than nonmicellar oil. These effects were rather conserved throughout the cells tested, indicating that fatty acids from micellar oils are enriched by mechanisms independent of lipases or lipid transporters. Importantly, the positive effect of emulsification was not restricted to the uptake of fatty acids. Instead, the uptake of phytosterols from phytogenic oils into target cells also increased after micellization. Taken together, phospholipid-based emulsification is a straightforward, effective, and safe approach to delivering hydrophobic nutrients, such as fatty acids or phytosterols, to a variety of cell types in vitro. It is proposed that this method of emulsification is suitable for the effective supplementation of numerous hydrophobic nutrients.
Collapse
|
15
|
Román‐Carrasco P, Lieder B, Somoza V, Ponce M, Szépfalusi Z, Martin D, Hemmer W, Swoboda I. Only α-Gal bound to lipids, but not to proteins, is transported across enterocytes as an IgE-reactive molecule that can induce effector cell activation. Allergy 2019; 74:1956-1968. [PMID: 31102539 PMCID: PMC6852507 DOI: 10.1111/all.13873] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/09/2019] [Accepted: 04/18/2019] [Indexed: 01/11/2023]
Abstract
BACKGROUND The oligosaccharide galactose-α-1,3-galactose (α-Gal), present in mammalian proteins and lipids, causes an unusual delayed allergic reaction 3 to 6 hours after ingestion of mammalian meat in individuals with IgE antibodies against α-Gal. To better understand the delayed onset of allergic symptoms and investigate whether protein-bound or lipid-bound α-Gal causes these symptoms, we analyzed the capacity of α-Gal conjugated proteins and lipids to cross a monolayer of intestinal cells. METHODS Extracts of proteins and lipids from beef were prepared, subjected to in vitro digestions, and added to Caco-2 cells grown on permeable supports. The presence of α-Gal in the basolateral medium was investigated by immunoblotting, thin-layer chromatography with immunostaining and ELISA, and its allergenic activity was analyzed in a basophil activation test. RESULTS After addition of beef proteins to the apical side of Caco-2 cells, α-Gal containing peptides were not detected in the basolateral medium. Those peptides that crossed the Caco-2 monolayer did not activate basophils from an α-Gal allergic patient. Instead, when Caco-2 cells were incubated with lipids extracted from beef, α-Gal was detected in the basolateral medium. Furthermore, these α-Gal lipids were able to activate the basophils of an α-Gal allergic patient in a dose-dependent manner. CONCLUSION Only α-Gal bound to lipids, but not to proteins, is able to cross the intestinal monolayer and trigger an allergic reaction. This suggests that the slower digestion and absorption of lipids might be responsible for the unusual delayed allergic reactions in α-Gal allergic patients and identifies glycolipids as potential allergenic molecules.
Collapse
Affiliation(s)
| | - Barbara Lieder
- Department of Physiological Chemistry, Faculty of Chemistry University of Vienna Vienna Austria
| | - Veronika Somoza
- Department of Physiological Chemistry, Faculty of Chemistry University of Vienna Vienna Austria
| | - Marta Ponce
- Department of Pediatrics and Adolescent Medicine Medical University of Vienna Vienna Austria
| | - Zsolt Szépfalusi
- Department of Pediatrics and Adolescent Medicine Medical University of Vienna Vienna Austria
| | - Diana Martin
- Departamento de Producción y Caracterización de Nuevos Alimentos Instituto de Investigación en Ciencias de la Alimentación (CIAL) (CSIC‐UAM) Madrid Spain
| | | | - Ines Swoboda
- Molecular Biotechnology Section University of Applied Sciences Vienna Austria
| |
Collapse
|
16
|
Igarashi M, Watanabe K, Tsuduki T, Kimura I, Kubota N. NAPE-PLD controls OEA synthesis and fat absorption by regulating lipoprotein synthesis in an in vitro model of intestinal epithelial cells. FASEB J 2018; 33:3167-3179. [PMID: 30399323 DOI: 10.1096/fj.201801408r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oleoylethanolamide (OEA), a fatty acid ethanolamide (FAE), is a lipid mediator that controls food intake and lipid metabolism. Accumulating data imply the importance of intestinal OEA in controlling satiety in addition to gastrointestinal peptide hormones. Although the biochemical pathway of FAE production has been illustrated, the enzymes responsible for the cleavage of OEA from its precursor N-acyl-phosphatidylethanolamine (NAPE) must be identified among reported candidates in the gut. In this study, we assessed the involvement of NAPE-specific phospholipase D (NAPE-PLD), which can directly release FAEs from NAPE, in intestinal OEA synthesis and lipid metabolism. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPER-associated protein 9 (Cas9)-mediated deletion of the NAPE-PLD gene in intestinal epithelial-like Caco-2 cells reduced OEA levels, regardless of their differentiation states. Transcriptome analysis revealed that deletion of NAPE-PLD activates a transcriptional program for nutrient transportation, including lipids and lipoproteins, and inactivates cell-cycle or mitosis-related genes in Caco-2 cells. In addition, the basolateral secretion of lipoproteins was increased in NAPE-PLD-deleted cells although lipoprotein size was not affected. By contrast, cellular lipid levels were reduced in NAPE-PLD-deleted cells. Overall, these results indicate that NAPE-PLD plays important roles in OEA synthesis and fat absorption by regulating lipoprotein production in the intestinal epithelial cells.-Igarashi, M., Watanabe, K., Tsuduki, T., Kimura, I., Kubota, N. NAPE-PLD controls OEA synthesis and fat absorption by regulating lipoprotein synthesis in an in vitro model of intestinal epithelial cells.
Collapse
Affiliation(s)
- Miki Igarashi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan.,RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | | | - Tsuyoshi Tsuduki
- Department of Bioscience and Biotechnology for Future Bioindustries, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Naoto Kubota
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan.,Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Clinical Nutrition, National Institute of Health and Nutrition, Tokyo, Japan; and.,Department of Clinical Nutrition Therapy, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Saez-Tenorio M, Domenech J, García-Rodríguez A, Velázquez A, Hernández A, Marcos R, Cortés C. Assessing the relevance of exposure time in differentiated Caco-2/HT29 cocultures. Effects of silver nanoparticles. Food Chem Toxicol 2018; 123:258-267. [PMID: 30403969 DOI: 10.1016/j.fct.2018.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 10/30/2018] [Accepted: 11/03/2018] [Indexed: 12/24/2022]
Abstract
In vitro models of the intestinal barrier are being increasingly used to evaluate nanoparticles (NPs) exposure risk. Nevertheless, most of these studies have focused on short-term exposures lasting no more than 24 h of duration, which could underestimate the toxic effects of a given compound under a more realistic setting. Since the assessment of longer exposure time-points is crucial to evaluate the risk of cumulative exposure to NPs, we have analyzed the effects of AgNPs at different exposure time-points between 6 h and 4 days on the barrier model system constituted by Caco-2/HT29 cells. Our results indicate that i) the system is stable during this time frame; ii) AgNPs affect the barrier's integrity only at the highest concentration tested (100 μg/mL), and only after 96 h of exposure; iii) cellular uptake of AgNPs showed a time-dependent and concentration-dependent increase; iv) translocation through the barrier was only observed at the highest concentration and only after 96 h of exposure; v) the expression of genes involved in the barrier's structure differs depending on the exposure time analyzed. All these results reinforce our proposal of expanding exposure times beyond 24 h when performing assays for hazard assessment of NPs using in vitro models of the intestinal barrier.
Collapse
Affiliation(s)
- Miriam Saez-Tenorio
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Josefa Domenech
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Alba García-Rodríguez
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Antonia Velázquez
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain
| | - Alba Hernández
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain
| | - Ricard Marcos
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain.
| | - Constanza Cortés
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
18
|
Jafari F, Agh N, Noori F, Tokmachi A, Gisbert E. Effects of dietary soybean lecithin on growth performance, blood chemistry and immunity in juvenile stellate sturgeon (Acipenser stellatus). FISH & SHELLFISH IMMUNOLOGY 2018; 80:487-496. [PMID: 29906622 DOI: 10.1016/j.fsi.2018.06.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 06/08/2023]
Abstract
An eleven weeks feeding trial was conducted to determine the effects of different levels of dietary soybean lecithin (SBL) on growth performance, blood chemistry and immunity in juvenile stellate sturgeon (Acipenser stellatus). Fish were fed seven isoproteic (44% crude protein) and isolipidic (17% crude fat) diets containing graded levels of SBL: 0 (control), 1, 2, 4, 6, 8 and 10%. Results showed that dietary SBL supplementation significantly improved the final body weight (BW) and weight gain (WG). Fish fed 6% SBL showed the highest BW and WG values in comparison to fish fed the control diet (P < 0.05), whereas increasing SBL levels above 6% had little practical benefit in terms of somatic growth performance. The inclusion of SBL in diets significantly improved the immune response as data from lysozyme, total Ig levels, alternative complement, phagocytic and bactericidal activities indicated (P < 0.05). The broken-line regression analysis of immunological variable revealed that depending on the parameter considered, the optimal SBL levels in diets for stellate sturgeon juveniles varied. In particular, dietary SBL levels requirements in stellate sturgeon when considering the phagocytic activity rate were determined at 3.3%, whereas 4.1-4.2% were recommended when considering data from lysozyme, alternative complement and bactericidal activities. In contrast, the highest minimum dietary SBL content was estimated at 6.9% when data from total Ig levels were considered. These results indicated that dietary PLs are required for boosting innate immunity in stellate sturgeon, although their minimal level changed depending on the immunological parameter considered. Therefore, we assume that SBL levels comprised between 3.3 and 6.9% may be used as a prophylactic measure to improve the health status in stellate sturgeon. Red blood cell count, hemoglobin and hematocrit levels increased with increasing dietary SBL levels, especially in those sturgeons fed the diet with 6% SBL (P < 0.05). In addition, white blood cell counts significantly increased as dietary SBL levels increased from 4 to 8% in comparison to the control group. Blood biochemistry was also affected by different dietary SBL levels. In particular, significantly higher levels of glucose, cholesterol, HDL and triglycerides were detected in fish fed >6%, >4%, >2% and 2% SBL, respectively (P < 0.05). Based on somatic growth parameters, blood chemistry and systemic immunity parameters, diets containing ca. 6% SBL are recommended for juvenile stellate sturgeon.
Collapse
Affiliation(s)
- Fatemeh Jafari
- Artemia and Aquaculture Research Institute, Urmia University, Urmia, Iran
| | - Naser Agh
- Artemia and Aquaculture Research Institute, Urmia University, Urmia, Iran.
| | - Farzaneh Noori
- Artemia and Aquaculture Research Institute, Urmia University, Urmia, Iran
| | - Amir Tokmachi
- Faculty of Veterinary, Urmia University, Urmia, Iran
| | - Enric Gisbert
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Centre de Sant Carles de la Ràpita, Unitat de Cultius Aqüícoles, Crta. Poble Nou km 5.5, 43540, Sant Carles de la Rapita, Spain
| |
Collapse
|
19
|
García-Rodríguez A, Vila L, Cortés C, Hernández A, Marcos R. Effects of differently shaped TiO 2NPs (nanospheres, nanorods and nanowires) on the in vitro model (Caco-2/HT29) of the intestinal barrier. Part Fibre Toxicol 2018; 15:33. [PMID: 30086772 PMCID: PMC6081908 DOI: 10.1186/s12989-018-0269-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The biological effects of nanoparticles depend on several characteristics such as size and shape that must be taken into account in any type of assessment. The increased use of titanium dioxide nanoparticles (TiO2NPs) for industrial applications, and specifically as a food additive, demands a deep assessment of their potential risk for humans, including their abilities to cross biological barriers. METHODS We have investigated the interaction of three differently shaped TiO2NPs (nanospheres, nanorods and nanowires) in an in vitro model of the intestinal barrier, where the coculture of Caco-2/HT29 cells confers inherent intestinal epithelium characteristics to the model (i.e. mucus secretion, brush border, tight junctions, etc.). RESULTS Adverse effects in the intestinal epithelium were detected by studying the barrier's integrity (TEER), permeability (LY) and changes in the gene expression of selected specific markers. Using Laser Scanning Confocal Microscopy, we detected a different behaviour in the bio-adhesion and biodistribution of each of the TiO2NPs. Moreover, we were able to specifically localize each type of TiO2NPs inside the cells. Interestingly, general DNA damage, but not oxidative DNA damage effects, were detected by using the FPG version of the comet assay. CONCLUSIONS Results indicate different interactions and cellular responses related to differently shaped TiO2NPs, nanowires showing the most harmful effects.
Collapse
Affiliation(s)
- Alba García-Rodríguez
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici Cn, Campus de Bellaterra, 08193 Cerdanyola del Vallès, Barcelona Spain
| | - Laura Vila
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici Cn, Campus de Bellaterra, 08193 Cerdanyola del Vallès, Barcelona Spain
| | - Constanza Cortés
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici Cn, Campus de Bellaterra, 08193 Cerdanyola del Vallès, Barcelona Spain
| | - Alba Hernández
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici Cn, Campus de Bellaterra, 08193 Cerdanyola del Vallès, Barcelona Spain
- CIBER Epidemiología y Salud Pública, ISCIII, Madrid, Spain
| | - Ricard Marcos
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici Cn, Campus de Bellaterra, 08193 Cerdanyola del Vallès, Barcelona Spain
- CIBER Epidemiología y Salud Pública, ISCIII, Madrid, Spain
| |
Collapse
|
20
|
Wu H, Luo T, Li YM, Gao ZP, Zhang KQ, Song JY, Xiao JS, Cao YP. Granny Smith apple procyanidin extract upregulates tight junction protein expression and modulates oxidative stress and inflammation in lipopolysaccharide-induced Caco-2 cells. Food Funct 2018; 9:3321-3329. [DOI: 10.1039/c8fo00525g] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Granny Smith apple procyanidin extracts upregulate tight junction protein expression, probably acting via the modulation of oxidative stress and inflammation in lipopolysaccharide-induced Caco-2 cells.
Collapse
Affiliation(s)
- H. Wu
- Beijing Technology & Business University
- Beijing 100048
- China
- Beijing Key Lab of Plant Resource Research and Development
- Beijing 100048
| | - T. Luo
- Department of Pharmacology
- University of Texas Southwestern Medical Center
- Dallas
- USA
| | - Y. M. Li
- Beijing Technology & Business University
- Beijing 100048
- China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health
- Beijing 100048
| | - Z. P. Gao
- Beijing Technology & Business University
- Beijing 100048
- China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health
- Beijing 100048
| | - K. Q. Zhang
- Beijing Technology & Business University
- Beijing 100048
- China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health
- Beijing 100048
| | - J. Y. Song
- Beijing Technology & Business University
- Beijing 100048
- China
- Beijing Key Lab of Plant Resource Research and Development
- Beijing 100048
| | - J. S. Xiao
- Beijing Technology & Business University
- Beijing 100048
- China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health
- Beijing 100048
| | - Y. P. Cao
- Beijing Technology & Business University
- Beijing 100048
- China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health
- Beijing 100048
| |
Collapse
|
21
|
Poquet L, Wooster TJ. Infant digestion physiology and the relevance of in vitro biochemical models to test infant formula lipid digestion. Mol Nutr Food Res 2017; 60:1876-95. [PMID: 27279140 DOI: 10.1002/mnfr.201500883] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/05/2016] [Accepted: 05/31/2016] [Indexed: 01/30/2023]
Abstract
Lipids play an important role in the diet of preterm and term infants providing a key energy source and essential lipid components for development. While a lot is known about adult lipid digestion, our understanding of infant digestion physiology is still incomplete, the greatest gap being on the biochemistry of the small intestine, particularly the activity and relative importance of the various lipases active in the intestine. The literature has been reviewed to identify the characteristics of lipid digestion of preterm and term infants, but also to better understand the physiology of the infant gastrointestinal tract compared to adults that impacts the absorption of lipids. The main differences are a higher gastric pH, submicellar bile salt concentration, a far more important role of gastric lipases as well as differences at the level of the intestinal barrier. Importantly, the consequences of improper in vitro replication of gastric digestions conditions (pH and lipase specificity) are demonstrated using examples from the most recent of studies. It is true that some animal models could be adapted to study infant lipid digestion physiology, however the ethical relevance of such models is questionable, hence the development of accurate in vitro models is a must. In vitro models that combine up to date knowledge of digestion biochemistry with intestinal cells in culture are the best choice to replicate digestion and absorption in infant population, this would allow the adaptation of infant formula for a better digestion and absorption of dietary lipids by preterm and term infants.
Collapse
Affiliation(s)
- Laure Poquet
- Nestlé Research Center, Vers-Chez-Les-Blanc, Lausanne 26, Switzerland
| | - Tim J Wooster
- Nestlé Research Center, Vers-Chez-Les-Blanc, Lausanne 26, Switzerland
| |
Collapse
|
22
|
Sané AT, Seidman E, Peretti N, Kleme ML, Delvin E, Deslandres C, Garofalo C, Spahis S, Levy E. Understanding Chylomicron Retention Disease Through Sar1b Gtpase Gene Disruption: Insight From Cell Culture. Arterioscler Thromb Vasc Biol 2017; 37:2243-2251. [PMID: 28982670 DOI: 10.1161/atvbaha.117.310121] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/21/2017] [Indexed: 01/28/2023]
Abstract
BACKGROUND Understanding the specific mechanisms of rare autosomal disorders has greatly expanded insights into the complex processes regulating intestinal fat transport. Sar1B GTPase is one of the critical proteins governing chylomicron secretion by the small intestine, and its mutations lead to chylomicron retention disease, despite the presence of Sar1A paralog. OBJECTIVE The central aim of this work is to examine the cause-effect relationship between Sar1B expression and chylomicron output and to determine whether Sar1B is obligatory for normal high-density lipoprotein biogenesis. APPROACH AND RESULTS The SAR1B gene was totally silenced in Caco-2/15 cells using the zinc finger nuclease technique. SAR1B deletion resulted in significantly decreased secretion of triglycerides (≈40%), apolipoprotein B-48 (≈57%), and chylomicron (≈34.5%). The absence of expected chylomicron production collapse may be because of the compensatory SAR1A elevation observed in our experiments. Therefore, a double knockout of SAR1A and SAR1B was engineered in Caco-2/15 cells, which led to almost complete inhibition of triglycerides, apolipoprotein B-48, and chylomicron output. Further experiments with labeled cholesterol revealed the downregulation of high-density lipoprotein biogenesis in cells deficient in SAR1B or with the double knockout of the 2 SAR1 paralogs. Similarly, there was a fall in the movement of labeled cholesterol from cells to basolateral medium containing apolipoprotein A-I, thereby limiting newly synthesized high-density lipoprotein in genetically modified cells. The decreased cholesterol efflux was associated with impaired expression of ABCA1 (ATP-binding cassette subfamily A member 1). CONCLUSIONS These findings demonstrate that the deletion of the 2 SAR1 isoforms is required to fully eliminate the secretion of chylomicron in vitro. They also underscore the limited high-density lipoprotein production by the intestinal cells in response to SAR1 knockout.
Collapse
Affiliation(s)
- Alain Théophile Sané
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.)
| | - Ernest Seidman
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.)
| | - Noel Peretti
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.)
| | - Marie Laure Kleme
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.)
| | - Edgard Delvin
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.)
| | - Colette Deslandres
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.)
| | - Carole Garofalo
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.)
| | - Schohraya Spahis
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.)
| | - Emile Levy
- From the CHU Sainte-Justine Research Centre (A.T.S., M.L.K., E.D., C.D., C.G., S.S., E.L.), Department of Nutrition (M.L.K., S.S., E.L.), and Department of Pediatrics (C.D.), Université de Montréal, Quebec, Canada; Division of Gastroenterology, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada (E.S.); and Centre de recherche Rhône-Alpes en nutrition humaine, Hôpital Edouard-Herriot, Université de Lyon 1, France (N.P.).
| |
Collapse
|
23
|
Wu ZF, Meng FC, Cao LJ, Jiang CH, Zhao MG, Shang XL, Fang SZ, Ye WC, Zhang QW, Zhang J, Yin ZQ. Triterpenoids from Cyclocarya paliurus and their inhibitory effect on the secretion of apoliprotein B48 in Caco-2 cells. PHYTOCHEMISTRY 2017; 142:76-84. [PMID: 28688991 DOI: 10.1016/j.phytochem.2017.06.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/19/2017] [Accepted: 06/25/2017] [Indexed: 06/07/2023]
Abstract
Five previously undescribed compounds including two triterpenoid aglycones, 3β,23-dihydroxy-1,12-dioxo-olean-28-oic acid and 3β,23,27-trihydroxy-1-oxo-olean-12-ene-28-oic acid, and three triterpenoid glucosides cyclocarioside L-N, along with 17 known compounds were isolated from a CH3Cl-soluble extract of the leaves of Cyclocarya paliurus. Two 27-nor-triterpenoid glycosides were isolated from the genus for the first time. Furthermore, the characterized compounds were tested for the inhibitory effects on apoliprotein B48 secretion in Caco-2 cells. Seven triterpenoid aglycones together with four triterpenoid saponins significantly decreased the apoliprotein B48 oversecretion induced by oleic acid in Caco-2 cells.
Collapse
Affiliation(s)
- Zheng-Feng Wu
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China
| | - Fan-Cheng Meng
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao SAR, PR China
| | - Lan-Jie Cao
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China
| | - Cui-Hua Jiang
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China
| | - Meng-Ge Zhao
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China
| | - Xu-Lan Shang
- College of Forestry, Nanjing Forestry University, Nanjing, 210037, PR China
| | - Sheng-Zuo Fang
- College of Forestry, Nanjing Forestry University, Nanjing, 210037, PR China
| | - Wen-Cai Ye
- Institute of Traditional Chinese Medicine and Natural Products & Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, PR China
| | - Qing-Wen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao SAR, PR China
| | - Jian Zhang
- Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China.
| | - Zhi-Qi Yin
- Department of Natural Medicinal Chemistry & State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
24
|
Rodriguez Sawicki L, Bottasso Arias NM, Scaglia N, Falomir Lockhart LJ, Franchini GR, Storch J, Córsico B. FABP1 knockdown in human enterocytes impairs proliferation and alters lipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1587-1594. [PMID: 28919479 DOI: 10.1016/j.bbalip.2017.09.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 09/06/2017] [Accepted: 09/10/2017] [Indexed: 12/17/2022]
Abstract
Fatty Acid-Binding Proteins (FABPs) are abundant intracellular proteins that bind long chain fatty acids (FA) and have been related with inmunometabolic diseases. Intestinal epithelial cells express two isoforms of FABPs: liver FABP (LFABP or FABP1) and intestinal FABP (IFABP or FABP2). They are thought to be associated with intracellular dietary lipid transport and trafficking towards diverse cell fates. But still their specific functions are not well understood. To study FABP1's functions, we generated an FABP1 knockdown model in Caco-2 cell line by stable antisense cDNA transfection (FABP1as). In these cells FABP1 expression was reduced up to 87%. No compensatory increase in FABP2 was observed, strengthening the idea of differential functions of both isoforms. In differentiated FABP1as cells, apical administration of oleate showed a decrease in its initial uptake rate and in long term incorporation compared with control cells. FABP1 depletion also reduced basolateral oleate secretion. The secreted oleate distribution showed an increase in FA/triacylglyceride ratio compared to control cells, probably due to FABP1's role in chylomicron assembly. Interestingly, FABP1as cells exhibited a dramatic decrease in proliferation rate. A reduction in oleate uptake as well as a decrease in its incorporation into the phospholipid fraction was observed in proliferating cells. Overall, our studies indicate that FABP1 is essential for proper lipid metabolism in differentiated enterocytes, particularly concerning fatty acids uptake and its basolateral secretion. Moreover, we show that FABP1 is required for enterocyte proliferation, suggesting that it may contribute to intestinal homeostasis.
Collapse
Affiliation(s)
- Luciana Rodriguez Sawicki
- Instituto de Investigaciones Bioquímicas de La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Natalia María Bottasso Arias
- Instituto de Investigaciones Bioquímicas de La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Natalia Scaglia
- Instituto de Investigaciones Bioquímicas de La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Lisandro Jorge Falomir Lockhart
- Instituto de Investigaciones Bioquímicas de La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Gisela Raquel Franchini
- Instituto de Investigaciones Bioquímicas de La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Judith Storch
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ, USA
| | - Betina Córsico
- Instituto de Investigaciones Bioquímicas de La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
25
|
Aeschimann W, Staats S, Kammer S, Olieric N, Jeckelmann JM, Fotiadis D, Netscher T, Rimbach G, Cascella M, Stocker A. Self-assembled α-Tocopherol Transfer Protein Nanoparticles Promote Vitamin E Delivery Across an Endothelial Barrier. Sci Rep 2017; 7:4970. [PMID: 28694484 PMCID: PMC5504013 DOI: 10.1038/s41598-017-05148-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/24/2017] [Indexed: 01/16/2023] Open
Abstract
Vitamin E is one of the most important natural antioxidants, protecting polyunsaturated fatty acids in the membranes of cells. Among different chemical isoforms assimilated from dietary regimes, RRR-α-tocopherol is the only one retained in higher animals. This is possible thanks to α-Tocopherol Transfer Protein (α-TTP), which extracts α-tocopherol from endosomal compartments in liver cells, facilitating its distribution into the body. Here we show that, upon binding to its substrate, α-TTP acquires tendency to aggregation into thermodynamically stable high molecular weight oligomers. Determination of the structure of such aggregates by X-ray crystallography revealed a spheroidal particle formed by 24 protein monomers. Oligomerization is triggered by refolding of the N-terminus. Experiments with cultured cell monolayers demonstrate that the same oligomers are efficiently transported through an endothelial barrier (HUVEC) and not through an epithelial one (Caco-2). Discovery of a human endogenous transport protein with intrinsic capability of crossing endothelial tissues opens to new ways of drug delivery into the brain or other tissues protected by endothelial barriers.
Collapse
Affiliation(s)
- Walter Aeschimann
- University of Bern, Department of Chemistry and Biochemistry, Bern, Switzerland
| | - Stefanie Staats
- University of Kiel, Institute of Human Nutrition and Food Science, Kiel, Germany
| | - Stephan Kammer
- University of Bern, Department of Chemistry and Biochemistry, Bern, Switzerland
| | | | - Jean-Marc Jeckelmann
- University of Bern, Institute of Biochemistry and Molecular Medicine, Bern, Switzerland
| | - Dimitrios Fotiadis
- University of Bern, Institute of Biochemistry and Molecular Medicine, Bern, Switzerland
| | | | - Gerald Rimbach
- University of Kiel, Institute of Human Nutrition and Food Science, Kiel, Germany
| | - Michele Cascella
- University of Oslo, Department of Chemistry and Centre for Theoretical and Computational Chemistry (CTCC), Oslo, Norway.
| | - Achim Stocker
- University of Bern, Department of Chemistry and Biochemistry, Bern, Switzerland.
| |
Collapse
|
26
|
Ikeda I, Tamakuni K, Sakuma T, Ozawa R, Inoue N, Kishimoto Y. Resistant Maltodextrin Decreases Micellar Solubility of Lipids and Diffusion of Bile Salt Micelles and Suppresses Incorporation of Micellar Fatty Acids into Caco-2 Cells. J Nutr Sci Vitaminol (Tokyo) 2017; 62:335-340. [PMID: 27928121 DOI: 10.3177/jnsv.62.335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Several studies have suggested that resistant maltodextrin (RMD) suppresses intestinal lipid absorption in experimental animals and humans. However, possible mechanisms underlying this effect are not known. In this study, effects of RMD on processes of the absorption of various lipids were investigated in vitro. RMD dose-dependently suppressed the solubility of various lipid components, including 1-mono-oleoylglycerol, oleic acid, and phosphatidylcholine in bile salt micelles in vitro. When the diffusion rate of bile salt micelles through a filter membrane was investigated in vitro, bile salt micelles containing RMD diffused more slowly than those without RMD. Incorporation of [1-14C] oleic acid into Caco-2 cells from the RMD-containing bile salt micelles was significantly smaller than that from the control micelles (without RMD). These results show that RMD suppresses intestinal absorption of lipids by decreasing their micellar solubility and the diffusion rate of bile salt micelles.
Collapse
Affiliation(s)
- Ikuo Ikeda
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University
| | | | | | | | | | | |
Collapse
|
27
|
Yang Y, Xiao H, McClements DJ. Impact of Lipid Phase on the Bioavailability of Vitamin E in Emulsion-Based Delivery Systems: Relative Importance of Bioaccessibility, Absorption, and Transformation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3946-3955. [PMID: 28447464 DOI: 10.1021/acs.jafc.7b00955] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A simulated gastrointestinal tract/Caco-2 cell culture model was used to investigate the effects of lipid phase type on vitamin E (VE) bioavailability. Oil-in-water emulsions fortified with α-tocopherol acetate were fabricated using a natural emulsifier (quillaja saponin) and long or medium chain triglycerides (LCTs or MCTs) as lipids. The impact of lipid type on VE bioaccessibility, absorption, and transformation was determined. VE bioaccessibility was greater for LCT (46%) than MCT (19%) due to greater solubilization in mixed micelles assembled from longer fatty acids. VE absorption by Caco-2 cells was similar for LCT (28%) and MCT (30%). The transformation of α-tocopherol acetate to α-tocopherol was higher for LCT (90%) than MCT (75%) due to differences in esterase accessibility to VE. Emulsion-based delivery systems formulated using LCT are therefore more suitable for encapsulating and delivering vitamin E than those formulated using MCT.
Collapse
Affiliation(s)
- Ying Yang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Ministry of Agriculture , Beijing, 100193, China
- Biopolymers and Colloids Research Laboratory, Department of Food Science, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - Hang Xiao
- Biopolymers and Colloids Research Laboratory, Department of Food Science, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - David Julian McClements
- Biopolymers and Colloids Research Laboratory, Department of Food Science, University of Massachusetts , Amherst, Massachusetts 01003, United States
| |
Collapse
|
28
|
Guo Z, Martucci NJ, Moreno-Olivas F, Tako E, Mahler GJ. Titanium Dioxide Nanoparticle Ingestion Alters Nutrient Absorption in an In Vitro Model of the Small Intestine. NANOIMPACT 2017; 5:70-82. [PMID: 28944308 PMCID: PMC5604471 DOI: 10.1016/j.impact.2017.01.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Ingestion of titanium dioxide (TiO2) nanoparticles from products such as agricultural chemicals, processed food, and nutritional supplements is nearly unavoidable. The gastrointestinal tract serves as a critical interface between the body and the external environment, and is the site of essential nutrient absorption. The goal of this study was to examine the effects of ingesting the 30 nm TiO2 nanoparticles with an in vitro cell culture model of the small intestinal epithelium, and to determine how acute or chronic exposure to nano-TiO2 influences intestinal barrier function, reactive oxygen species generation, proinflammatory signaling, nutrient absorption (iron, zinc, fatty acids), and brush border membrane enzyme function (intestinal alkaline phosphatase). A Caco-2/HT29-MTX cell culture model was exposed to physiologically relevant doses of TiO2 nanoparticles for acute (four hours) or chronic (five days) time periods. Exposure to TiO2 nanoparticles significantly decreased intestinal barrier function following chronic exposure. Reactive oxygen species (ROS) generation, proinflammatory signaling, and intestinal alkaline phosphatase activity all showed increases in response to nano-TiO2. Iron, zinc, and fatty acid transport were significantly decreased following exposure to TiO2 nanoparticles. This is because nanoparticle exposure induced a decrease in absorptive microvilli in the intestinal epithelial cells. Nutrient transporter protein gene expression was also altered, suggesting that cells are working to regulate the transport mechanisms disturbed by nanoparticle ingestion. Overall, these results show that intestinal epithelial cells are affected at a functional level by physiologically relevant exposure to nanoparticles commonly ingested from food.
Collapse
Affiliation(s)
- Zhongyuan Guo
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902
| | - Nicole J. Martucci
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902
| | | | - Elad Tako
- Plant, Soil and Nutrition Laboratory, Agricultural Research Services, U.S. Department of Agriculture, Ithaca, NY
| | - Gretchen J. Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902
- Correspondence to Gretchen Mahler, PhD, Binghamton University, Department of Biomedical Engineering, 2608 Biotechnology Building, Binghamton, NY 13902, Phone: 607-777-5238, Fax: 607-777-5780,
| |
Collapse
|
29
|
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: 92] [Impact Index Per Article: 10.2] [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
|
30
|
Yuan X, Lu P, Xue X, Qin H, Fan C, Wang Y, Zhang Q. Discovery of 2-azetidinone and 1 H -pyrrole-2,5-dione derivatives containing sulfonamide group at the side chain as potential cholesterol absorption inhibitors. Bioorg Med Chem Lett 2016; 26:849-853. [DOI: 10.1016/j.bmcl.2015.12.077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 12/08/2015] [Accepted: 12/22/2015] [Indexed: 10/22/2022]
|
31
|
Han S, Hu L, Quach T, Simpson JS, Trevaskis NL, Porter CJH. Constitutive Triglyceride Turnover into the Mesenteric Lymph Is Unable to Support Efficient Lymphatic Transport of a Biomimetic Triglyceride Prodrug. J Pharm Sci 2016; 105:786-796. [PMID: 26540595 DOI: 10.1002/jps.24670] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/07/2015] [Accepted: 09/11/2015] [Indexed: 12/17/2022]
Abstract
The triglyceride (TG) mimetic prodrug (1,3-dipalmitoyl-2-mycophenoloyl glycerol, 2-MPA-TG) biochemically integrates into intestinal lipid transport and lipoprotein assembly pathways and thereby promotes the delivery of mycophenolic acid (MPA) into the lymphatic system. As lipoprotein (LP) formation occurs constitutively, even in the fasted state, the current study aimed to determine whether lymphatic transport of 2-MPA-TG was dependent on coadministered exogenous lipid. In vitro incubation of the prodrug with rat digestive fluid and in situ intestinal perfusion experiments revealed that hydrolysis and absorption of the prodrug were relatively unaffected by the quantity of lipid in formulations. In vivo studies in rats, however, showed that the lymphatic transport of TG and 2-MPA-TG was significantly higher following administration with higher quantities of lipid and that oleic acid (C18:1) was more effective in promoting prodrug transport than lipids with higher degrees of unsaturation. The recovery of 2-MPA-TG and TG in lymph correlated strongly (R(2) = 0.99) and more than 97% of the prodrug was associated with chylomicrons. Inhibition of LP assembly by Pluronic L81 simultaneously inhibited the lymphatic transport of 2-MPA-TG and TG. In conclusion, although the TG mimetic prodrug effectively incorporates into TG resynthetic pathways, lipid coadministration is still required to support efficient lymphatic transport.
Collapse
Affiliation(s)
- Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Tim Quach
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia; Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Jamie S Simpson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| |
Collapse
|
32
|
Crites KSM, Morin G, Orlando V, Patey N, Cantin C, Martel J, Brochiero E, Mailhot G. CFTR Knockdown induces proinflammatory changes in intestinal epithelial cells. JOURNAL OF INFLAMMATION-LONDON 2015; 12:62. [PMID: 26549988 PMCID: PMC4636765 DOI: 10.1186/s12950-015-0107-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 11/05/2015] [Indexed: 12/20/2022]
Abstract
Background Hyperinflammation is a hallmark feature of cystic fibrosis (CF) airways. However, inflammation has also been documented systemically and, more recently, in extrapulmonary CF-affected tissues such as the pancreas and intestine. The pathogenesis of CF-related inflammation and more specifically the role of the cystic fibrosis transmembrane conductance regulator (CFTR) in that respect are not entirely understood. We have tested the hypothesis that genetic depletion of CFTR will affect the inflammatory status of human intestinal epithelial cell lines. Methods CFTR expression was genetically depleted from Caco-2/15 and HT-29 cells using short hairpin RNA interference (shRNAi). Inflammatory conditions were induced by the addition of human recombinant tumor necrosis factor (TNF) or Interleukin-1β (IL-1β) for various periods of time. Gene expression, mRNA stability and secreted levels of interleukin (IL)-6, −8 and 10 were assessed. Analysis of pro- and anti-inflammatory signaling pathways including mitogen-activated protein kinases (p38, ERK 1/2 and JNK), nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (IκBα), and nuclear factor-kappa B (NF-κB) was also performed. Eosinophils were counted in the jejunal mucosa of Cftr−/− and Cftr+/+ mice. Results CFTR gene and protein knockdown caused a significant increase in basal secretion of IL-8 as well as in IL-1β-induced secretion of IL-6 and −8. Release of the anti-inflammatory cytokine, IL-10, remained unaffected by CFTR depletion. The enhanced secretion of IL-8 stems in part from increased IL8 mRNA levels and greater activation of ERK1/2 MAPK, IκBα and NF-κB in the CFTR knockdown cells. By contrast, phosphorylation levels of p38 and JNK MAPK did not differ between control and knockdown cells. We also found a higher number of infiltrating eosinophils in the jejunal mucosa of Cftr −/− females, but not males, compared to Cftr +/+ mice, thus providing in vivo support to our in vitro findings. Conclusion Collectively, these data underscore the role played by CFTR in regulating the intestinal inflammatory responses. Such findings lend support to the theory that CFTR exerts functions that may go beyond its role as a chloride channel whereby its disruption may prevent cells to optimally respond to exogenous or endogenous challenges. These observations are of particular interest to CF patients who were found to display alterations in their intestinal microbiota, thus predisposing them to pathogens that may elicit exaggerated inflammatory responses. Electronic supplementary material The online version of this article (doi:10.1186/s12950-015-0107-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Geneviève Morin
- Research Centre, CHU Sainte-Justine, 3175 Cote Sainte-Catherine Rd, Montreal, Quebec H3T 1C5 Canada
| | - Valérie Orlando
- Research Centre, CHU Sainte-Justine, 3175 Cote Sainte-Catherine Rd, Montreal, Quebec H3T 1C5 Canada
| | - Natacha Patey
- Research Centre, CHU Sainte-Justine, 3175 Cote Sainte-Catherine Rd, Montreal, Quebec H3T 1C5 Canada
| | - Catherine Cantin
- Research Centre, CHU Sainte-Justine, 3175 Cote Sainte-Catherine Rd, Montreal, Quebec H3T 1C5 Canada
| | - Judith Martel
- Research Centre, CHU Sainte-Justine, 3175 Cote Sainte-Catherine Rd, Montreal, Quebec H3T 1C5 Canada
| | - Emmanuelle Brochiero
- Research Center, CHUM, 900 Saint-Denis Street, Montreal, Quebec H2X 0A9 Canada ; Department of Medicine, Université de Montreal, 2900, Édouard-Montpetit Blvd, Montreal, Quebec H3T 1J4 Canada
| | - Geneviève Mailhot
- Research Centre, CHU Sainte-Justine, 3175 Cote Sainte-Catherine Rd, Montreal, Quebec H3T 1C5 Canada ; Department of Nutrition, Université de Montreal, 2405 Cote Sainte-Catherine Rd, Montreal, Quebec H3T 1A8 Canada
| |
Collapse
|
33
|
Daimiel-Ruiz L, Klett-Mingo M, Konstantinidou V, Micó V, Aranda JF, García B, Martínez-Botas J, Dávalos A, Fernández-Hernando C, Ordovás JM. Dietary lipids modulate the expression of miR-107, a miRNA that regulates the circadian system. Mol Nutr Food Res 2015; 59:1865-78. [DOI: 10.1002/mnfr.201570094] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Lidia Daimiel-Ruiz
- Nutritional Genomics of Cardiovascular Disease and Obesity; IMDEA-Food Institute, CEI UAM+CSIC; Madrid Spain
| | - Mercedes Klett-Mingo
- Nutritional Genomics of Cardiovascular Disease and Obesity; IMDEA-Food Institute, CEI UAM+CSIC; Madrid Spain
| | - Valentini Konstantinidou
- Nutritional Genomics of Cardiovascular Disease and Obesity; IMDEA-Food Institute, CEI UAM+CSIC; Madrid Spain
| | - Victor Micó
- Nutritional Genomics of Cardiovascular Disease and Obesity; IMDEA-Food Institute, CEI UAM+CSIC; Madrid Spain
| | - Juan Francisco Aranda
- Vascular Biology and Therapeutics Program; Yale University School of Medicine; New Haven CT USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program; Section of Comparative Medicine, Yale University School of Medicine; New Haven CT USA
| | - Belén García
- Nutritional Genomics of Cardiovascular Disease and Obesity; IMDEA-Food Institute, CEI UAM+CSIC; Madrid Spain
| | - Javier Martínez-Botas
- Servicio de Bioquímica-Investigación; Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS); Madrid Spain
| | - Alberto Dávalos
- Nutritional Genomics of Cardiovascular Disease and Obesity; IMDEA-Food Institute, CEI UAM+CSIC; Madrid Spain
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program; Yale University School of Medicine; New Haven CT USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program; Section of Comparative Medicine, Yale University School of Medicine; New Haven CT USA
| | - José Maria Ordovás
- Nutritional Genomics of Cardiovascular Disease and Obesity; IMDEA-Food Institute, CEI UAM+CSIC; Madrid Spain
- Nutrition and Genomics Laboratory; JM-USDA Human Nutrition Research Center on Aging at Tufts University; Boston MA USA
| |
Collapse
|
34
|
Daimiel L, Ordovás JM, Dávalos A. miRNAs modified by dietary lipids in Caco-2 cells. A microarray screening. GENOMICS DATA 2015; 5:171-2. [PMID: 26484250 PMCID: PMC4584012 DOI: 10.1016/j.gdata.2015.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/21/2015] [Indexed: 12/31/2022]
Abstract
We performed a screening of miRNAs regulated by dietary lipids in a cellular model of enterocytes, Caco-2 cells. Our aim was to describe new lipid-modified miRNAs with an implication in lipid homeostasis and cardiovascular disease [1,2]. For that purpose, we treated differentiated Caco-2 cells with micelles containing the assayed lipids (cholesterol, conjugated linoleic acid and docosahexaenoic acid) and the screening of miRNAs was carried out by microarray using the μParaflo®Microfluidic Biochip Technology of LC Sciences (Huston, TX, USA). Experimental design, microarray description and raw data have been made available in the GEO database with the reference number of GSE59153. Here we described in detail the experimental design and methods used to obtain the relative expression data.
Collapse
Affiliation(s)
- Lidia Daimiel
- Nutritional Genomics of Cardiovascular Disease and Obesity, IMDEA-Food Institute, CEI UAM+CSIC, Madrid, E-28049, Spain
| | - Jose M Ordovás
- Nutritional Genomics of Cardiovascular Disease and Obesity, IMDEA-Food Institute, CEI UAM+CSIC, Madrid, E-28049, Spain ; Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| | - Alberto Dávalos
- Nutritional Genomics of Cardiovascular Disease and Obesity, IMDEA-Food Institute, CEI UAM+CSIC, Madrid, E-28049, Spain
| |
Collapse
|
35
|
Tenore GC, Ritieni A, Campiglia P, Stiuso P, Di Maro S, Sommella E, Pepe G, D'Urso E, Novellino E. Antioxidant peptides from “Mozzarella di Bufala Campana DOP” after simulated gastrointestinal digestion: In vitro intestinal protection, bioavailability, and anti-haemolytic capacity. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.03.048] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
36
|
Gaibelet G, Allart S, Tercé F, Azalbert V, Bertrand-Michel J, Hamdi S, Collet X, Orlowski S. Specific cellular incorporation of a pyrene-labelled cholesterol: lipoprotein-mediated delivery toward ordered intracellular membranes. PLoS One 2015; 10:e0121563. [PMID: 25875769 PMCID: PMC4398402 DOI: 10.1371/journal.pone.0121563] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/25/2015] [Indexed: 11/18/2022] Open
Abstract
In the aim of testing tools for tracing cell trafficking of exogenous cholesterol, two fluorescent derivatives of cholesterol, 22-nitrobenzoxadiazole-cholesterol (NBD-Chol) and 21-methylpyrenyl-cholesterol (Pyr-met-Chol), with distinctive chemico-physical characteristics, have been compared for their cell incorporation properties, using two cell models differently handling cholesterol, with two incorporation routes. In the Caco-2 cell model, the cholesterol probes were delivered in bile salt micelles, as a model of intestinal absorption. The two probes displayed contrasting behaviors for cell uptake characteristics, cell staining, and efflux kinetics. In particular, Pyr-met-Chol cell incorporation involved SR-BI, while that of NBD-Chol appeared purely passive. In the PC-3 cell model, which overexpresses lipoprotein receptors, the cholesterol probes were delivered via the serum components, as a model of systemic delivery. We showed that Pyr-met-Chol-labelled purified LDL or HDL were able to specifically deliver Pyr-met-Chol to the PC-3 cells, while NBD-Chol incorporation was independent of lipoproteins. Observations by fluorescence microscopy evidenced that, while NBD-Chol readily stained the cytosolic lipid droplets, Pyr-met-Chol labelling led to the intense staining of intracellular structures of membranous nature, in agreement with the absence of detectable esterification of Pyr-met-Chol. A 48 h incubation of PC-3 cells with either Pyr-met-Chol-labelled LDL or HDL gave same staining patterns, mainly colocalizing with Lamp1, caveolin-1 and CD63. These data indicated convergent trafficking downwards their respective receptors, LDL-R and SR-BI, toward the cholesterol-rich internal membrane compartments, late endosomes and multivesicular bodies. Interestingly, Pyr-met-Chol staining of these structures exhibited a high excimer fluorescence emission, revealing their ordered membrane environment, and indicating that Pyr-met-Chol behaves as a fair cholesterol tracer regarding its preferential incorporation into cholesterol-rich domains. We conclude that, while NBD-Chol is a valuable marker of cholesterol esterification, Pyr-met-Chol is a reliable new lipoprotein fluorescent marker which allows to probe specific intracellular trafficking of cholesterol-rich membranes.
Collapse
Affiliation(s)
- Gérald Gaibelet
- INSERM U563/1048, CHU Purpan, 31024, Toulouse, cedex 3, France
- CEA, SB2SM and UMR8221/UMR9198 CNRS, I2BC, IBiTec-Saclay, 91191, Gif-sur-Yvette, cedex, France
- Université Toulouse III, UMR 1048, F-31000, Toulouse, France
| | - Sophie Allart
- Université Toulouse III, UMR 1048, F-31000, Toulouse, France
- Plateau technique d’Imagerie Cellulaire, INSERM U1043, F-31300, Toulouse, France
| | - François Tercé
- Université Toulouse III, UMR 1048, F-31000, Toulouse, France
- INSERM U1048, F-31400, Toulouse, France
| | - Vincent Azalbert
- Université Toulouse III, UMR 1048, F-31000, Toulouse, France
- INSERM U1048, F-31400, Toulouse, France
| | - Justine Bertrand-Michel
- Université Toulouse III, UMR 1048, F-31000, Toulouse, France
- INSERM U1048, Lipidomic Platform Metatoul, F-31400, Toulouse, France
| | - Safouane Hamdi
- INSERM U563/1048, CHU Purpan, 31024, Toulouse, cedex 3, France
| | - Xavier Collet
- Université Toulouse III, UMR 1048, F-31000, Toulouse, France
- INSERM U1048, F-31400, Toulouse, France
| | - Stéphane Orlowski
- INSERM U563/1048, CHU Purpan, 31024, Toulouse, cedex 3, France
- CEA, SB2SM and UMR8221/UMR9198 CNRS, I2BC, IBiTec-Saclay, 91191, Gif-sur-Yvette, cedex, France
- * E-mail:
| |
Collapse
|
37
|
TNF-alpha and IL-6 inhibit apolipoprotein A-IV production induced by linoleic acid in human intestinal Caco2 cells. JOURNAL OF INFLAMMATION-LONDON 2015; 12:22. [PMID: 25861245 PMCID: PMC4389805 DOI: 10.1186/s12950-015-0069-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 03/11/2015] [Indexed: 11/29/2022]
Abstract
Background Apolipoprotein A-IV (apoA-IV) is a protein mainly synthesized by enterocytes in the intestine. Its gene expression is suppressed during fasting and stimulated during active fat absorption. Chronic feeding of a high-fat (HF) diet abolishes the differential expression between fasting and fat-feeding and therefore may contribute to diet-induced obesity since apoA-IV is a potent satiety factor. It is well established that the circulating pro-inflammatory cytokines TNF-α and IL-6 are increased by HF feeding. Methods To determine whether pro-inflammatory cytokines are involved in the diminished response of apoA-IV gene expression to fat-feeding, different concentrations of linoleic acid (LA), an important dietary fatty acid, was used to stimulate apoA-IV expression in human intestinal Caco2 cells. Cells were pre-treated with or without human recombinant TNF-α, IL-6 or their combination before the addition of LA. Real-time PCR and ELISA were used to detect and quantify RNA transcripts and proteins of apoA-IV and the cytokines. Results LA stimulated gene and protein expression of apoA-IV in a dose and time dependent manner. Pre-treatment with the cytokines for 72 h significantly inhibited the increased expression of apoA-IV gene and protein induced by LA. Furthermore, the cytokines, especially TNF-α, also positively up-regulate the cytokine themselves in Caco2 cells. Conclusions Our data indicate that the pro-inflammatory cytokines may be responsible for the reduced apoA-IV production in response to fat feeding. Because of apoA-IV’s role in satiety, we propose the inhibitory effect of circulating pro-inflammatory cytokines on apoA-IV production contributes to diet-induced obesity.
Collapse
|
38
|
Trevaskis NL, Hu L, Caliph SM, Han S, Porter CJH. The mesenteric lymph duct cannulated rat model: application to the assessment of intestinal lymphatic drug transport. J Vis Exp 2015:52389. [PMID: 25866901 PMCID: PMC4401200 DOI: 10.3791/52389] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The intestinal lymphatic system plays key roles in fluid transport, lipid absorption and immune function. Lymph flows directly from the small intestine via a series of lymphatic vessels and nodes that converge at the superior mesenteric lymph duct. Cannulation of the mesenteric lymph duct thus enables the collection of mesenteric lymph flowing from the intestine. Mesenteric lymph consists of a cellular fraction of immune cells (99% lymphocytes), aqueous fraction (fluid, peptides and proteins such as cytokines and gut hormones) and lipoprotein fraction (lipids, lipophilic molecules and apo-proteins). The mesenteric lymph duct cannulation model can therefore be used to measure the concentration and rate of transport of a range of factors from the intestine via the lymphatic system. Changes to these factors in response to different challenges (e.g., diets, antigens, drugs) and in disease (e.g., inflammatory bowel disease, HIV, diabetes) can also be determined. An area of expanding interest is the role of lymphatic transport in the absorption of orally administered lipophilic drugs and prodrugs that associate with intestinal lipid absorption pathways. Here we describe, in detail, a mesenteric lymph duct cannulated rat model which enables evaluation of the rate and extent of lipid and drug transport via the lymphatic system for several hours following intestinal delivery. The method is easily adaptable to the measurement of other parameters in lymph. We provide detailed descriptions of the difficulties that may be encountered when establishing this complex surgical method, as well as representative data from failed and successful experiments to provide instruction on how to confirm experimental success and interpret the data obtained.
Collapse
Affiliation(s)
- Natalie L Trevaskis
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus);
| | - Luojuan Hu
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus)
| | - Suzanne M Caliph
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus)
| | - Sifei Han
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus)
| | - Christopher J H Porter
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus);
| |
Collapse
|
39
|
Ting Y, Zhao Q, Xia C, Huang Q. Using in vitro and in vivo models to evaluate the oral bioavailability of nutraceuticals. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:1332-1338. [PMID: 25615514 DOI: 10.1021/jf5047464] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Nutraceuticals are the bioactive compounds found in many dietary sources. Numerous publications have reported their ability to prevent the development of degenerative diseases through modulation of physiological and physiochemical processes in living systems. Having sufficient concentration at the target site of action is the most critical factor for nutraceuticals to deliver the health benefits. For consumers, it is commonly accepted to ingest these bioactive components through oral delivery route because it is convenient and cost-efficient and allows flexible dosing schedule. Thus, it is important to understand the oral bioavailability of nutraceuticals to evaluate their qualifications as disease preventive agents and to calculate the required ingestion dosages. To predict the oral bioavailability of nutraceuticals, many in vitro and in vivo models have been developed to reduce the need for frequent human pharmacokinetic studies, which are costly and time-consuming and involve ethical complications. These models evaluate one or more of the influential factors that contribute to the oral bioavailability and are efficient screening techniques with the potential of predicting the pharmacokinetic process in humans. To accurately predict human oral bioavailability, further research is required to develop not only a better correlation between the in vitro and in vivo models but also an accurate scaling factor that takes into account interspecies variations.
Collapse
Affiliation(s)
- Yuwen Ting
- Department of Food Science, Rutgers University , 65 Dudley Road, New Brunswick, New Jersey 08901, United States
| | | | | | | |
Collapse
|
40
|
Daimiel-Ruiz L, Klett-Mingo M, Konstantinidou V, Micó V, Aranda JF, García B, Martínez-Botas J, Dávalos A, Fernández-Hernando C, Ordovás JM. Dietary lipids modulate the expression of miR-107, an miRNA that regulates the circadian system. Mol Nutr Food Res 2015; 59:552-65. [PMID: 25522185 DOI: 10.1002/mnfr.201400616] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/01/2014] [Accepted: 12/07/2014] [Indexed: 02/06/2023]
Abstract
SCOPE The increased prevalence of cardiovascular diseases (CVDs) has been hypothesized to be the result of an increased exposure to a host of atherogenic environmental factors, paramount among them being unhealthy dietary habits. Long-chain n-3 polyunsaturated fatty acids have been shown to have cardio protective effects, partially due to their ability to regulate gene expression. In this regard, increasing attention has been devoted to the role of miRNAs as regulators of multiple metabolic pathways whose deregulation has been associated with CVD risk. METHODS AND RESULTS In this work, we investigated whether miRNA expression was regulated by docosahexanoic acid, conjugated linoleic acid, and cholesterol in Caco-2 cells. The modulated miRNAs, miR-107 was differentially expressed by all treatments and this modulation was independent of its hosting gene, PANK1, possibly through its own promoter, which contains binding sites for metabolically relevant transcription factors. Among the putative target genes of miR-107, we found some genes with key roles in circadian rhythm. Specifically, we demonstrated that binding of miR-107 to the CLOCK gene results in the deregulation of the circadian rhythm of the cells. CONCLUSION Since chronodisruption has been linked to metabolic disorders such as type 2 diabetes, atherosclerosis, obesity, and CVD, our findings suggests that miR-107 could represent a new approach for pharmacological treatment of these diseases.
Collapse
Affiliation(s)
- Lidia Daimiel-Ruiz
- Nutritional Genomics of Cardiovascular Disease and Obesity, IMDEA-Food Institute, CEI UAM+CSIC, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Prevention of oxidative stress, inflammation and mitochondrial dysfunction in the intestine by different cranberry phenolic fractions. Clin Sci (Lond) 2014; 128:197-212. [PMID: 25069567 DOI: 10.1042/cs20140210] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cranberry fruit has been reported to have high antioxidant effectiveness that is potentially linked to its richness in diversified polyphenolic content. The aim of the present study was to determine the role of cranberry polyphenolic fractions in oxidative stress (OxS), inflammation and mitochondrial functions using intestinal Caco-2/15 cells. The combination of HPLC and UltraPerformance LC®-tandem quadrupole (UPLC-TQD) techniques allowed us to characterize the profile of low, medium and high molecular mass polyphenolic compounds in cranberry extracts. The medium molecular mass fraction was enriched with flavonoids and procyanidin dimers whereas procyanidin oligomers (DP > 4) were the dominant class of polyphenols in the high molecular mass fraction. Pre-incubation of Caco-2/15 cells with these cranberry extracts prevented iron/ascorbate-mediated lipid peroxidation and counteracted lipopolysaccharide-mediated inflammation as evidenced by the decrease in pro-inflammatory cytokines (TNF-α and interleukin-6), cyclo-oxygenase-2 and prostaglandin E2. Cranberry polyphenols (CP) fractions limited both nuclear factor κB activation and Nrf2 down-regulation. Consistently, cranberry procyanidins alleviated OxS-dependent mitochondrial dysfunctions as shown by the rise in ATP production and the up-regulation of Bcl-2, as well as the decline of protein expression of cytochrome c and apoptotic-inducing factor. These mitochondrial effects were associated with a significant stimulation of peroxisome-proliferator-activated receptor γ co-activator-1-α, a central inducing factor of mitochondrial biogenesis and transcriptional co-activator of numerous downstream mediators. Finally, cranberry procyanidins forestalled the effect of iron/ascorbate on the protein expression of mitochondrial transcription factors (mtTFA, mtTFB1, mtTFB2). Our findings provide evidence for the capacity of CP to reduce intestinal OxS and inflammation while improving mitochondrial dysfunction.
Collapse
|
42
|
Jin Y, McFie PJ, Banman SL, Brandt C, Stone SJ. Diacylglycerol acyltransferase-2 (DGAT2) and monoacylglycerol acyltransferase-2 (MGAT2) interact to promote triacylglycerol synthesis. J Biol Chem 2014; 289:28237-48. [PMID: 25164810 DOI: 10.1074/jbc.m114.571190] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Acyl CoA:1,2-diacylglycerol acyltransferase (DGAT)-2 is an integral membrane protein that catalyzes triacylglycerol (TG) synthesis using diacylglycerol and fatty acyl CoA as substrates. DGAT2 resides in the endoplasmic reticulum (ER), but when cells are incubated with fatty acids, DGAT2 interacts with lipid droplets presumably to catalyze localized TG synthesis for lipid droplet expansion. Previous studies have shown that DGAT2 interacts with proteins that synthesize its fatty acyl CoA substrates. In this study, we provide additional evidence that DGAT2 is present in a protein complex. Using a chemical cross-linker, disuccinimidyl suberate (DSS), we demonstrated that DGAT2 formed a dimer and was also part of a protein complex of ∼ 650 kDa, both in membranes and on lipid droplets. Using co-immunoprecipitation experiments and an in situ proximity ligation assay, we found that DGAT2 interacted with monoacylglycerol acyltransferase (MGAT)-2, an enzyme that catalyzes the synthesis of diacylglycerol. Deletion mutagenesis showed that the interaction with MGAT2 was dependent on the two transmembrane domains of DGAT2. No significant interaction of DGAT2 with lipin1, another enzyme that synthesizes diacylglycerol, could be detected. When co-expressed in cells, DGAT2 and MGAT2 co-localized in the ER and on lipid droplets. Co-expression also resulted in increased TG storage compared with expression of DGAT2 or MGAT2 alone. Incubating McArdle rat hepatoma RH7777 cells with 2-monoacylglycerol caused DGAT2 to translocate to lipid droplets. This also led to the formation of large cytosolic lipid droplets, characteristic of DGAT2, but not DGAT1, and indicated that DGAT2 can utilize monoacylglycerol-derived diacylglycerol. These findings suggest that the interaction of DGAT2 and MGAT2 serves to channel lipid substrates efficiently for TG biosynthesis.
Collapse
Affiliation(s)
- Youzhi Jin
- From the Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada and
| | - Pamela J McFie
- From the Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada and
| | - Shanna L Banman
- Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B4, Canada
| | - Curtis Brandt
- From the Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada and
| | - Scot J Stone
- From the Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada and
| |
Collapse
|
43
|
Effects of eicosapentaenoic acid and docosahexaenoic acid on chylomicron and VLDL synthesis and secretion in Caco-2 cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:684325. [PMID: 24987699 PMCID: PMC4058467 DOI: 10.1155/2014/684325] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/19/2014] [Indexed: 11/18/2022]
Abstract
The present research was undertaken to determine the effects of EPA (20 : 5 n-3) and DHA (22 : 6 n-3) on chylomicron and VLDL synthesis and secretion by Caco-2 cells. Cells were incubated for 12 to 36 h with 400 μM OA, EPA, and DHA; then 36 h was chosen for further study because EPA and DHA decreased de novo triglycerides synthesis in a longer incubation compared with OA (P < 0.01). Neither the uptake nor oxidation was different in response to the respective fatty acids (P > 0.05). Compared with OA, intercellular and secreted nascent apolipoprotein B48 and B100 were decreased by EPA and DHA (P < 0.01). Both DHA and EPA resulted in a lower secretion of chylomicron and VLDL (P < 0.01). In contrast to OA, EPA and DHA were preferentially incorporated into phospholipids instead of triacylglycerols (P < 0.01). These discoveries demonstrated that exposure of DHA and EPA reduced the secretion of chylomicron and VLDL partly by regulating the synthesis of TG and apoB.
Collapse
|
44
|
Zhang LJ, Wang C, Yuan Y, Wang H, Wu J, Liu F, Li L, Gao X, Zhao YL, Hu PZ, Li P, Ye J. Cideb facilitates the lipidation of chylomicrons in the small intestine. J Lipid Res 2014; 55:1279-87. [PMID: 24831470 DOI: 10.1194/jlr.m046482] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Indexed: 02/01/2023] Open
Abstract
Cell death-inducing DFF45-like effector b (Cideb), an endoplasmic reticulum (ER)- and lipid droplet (LD)-associated protein, has been shown to play a critical role in maintaining hepatic lipid homeostasis by promoting the lipidation and maturation of VLDL particles. Here, we observed that Cideb is expressed in the jejunum and ileum sections of the small intestine, and its expression was induced by high-fat diet. Intragastric gavage with lipids resulted in the retention of lipids in the intestine in Cideb-deficient mice. In addition, we observed that mice with Cideb deficiency exhibited reduced intestinal chylomicron-TG secretion and increased lipid accumulation in the enterocytes. The sizes of chylomicrons secreted from the small intestine of Cideb-deficient mice were also smaller than those from wild-type mice. Furthermore, the overexpression of Cideb increased TG secretion and reduced lipid accumulation in the enterocyte-like Caco-2 cells. In addition, we proved that Cideb was localized to the ER and LDs and could interact with ApoB48 in Caco-2 cells. Overall, these data revealed that Cideb plays an important role in controlling intestinal chylomicron lipidation.
Collapse
Affiliation(s)
- Li-Jun Zhang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Chao Wang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Yuan Yuan
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Hui Wang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jie Wu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Fang Liu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Le Li
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xing Gao
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan-Lin Zhao
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Pei-Zhen Hu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Peng Li
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 10084, China
| | - Jing Ye
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
45
|
Wang X, Ai T, Meng X, Zhou J, Mao X. In vitro iron absorption of α-lactalbumin hydrolysate-iron and β-lactoglobulin hydrolysate-iron complexes. J Dairy Sci 2014; 97:2559-66. [DOI: 10.3168/jds.2013-7461] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/07/2014] [Indexed: 02/01/2023]
|
46
|
Zhou L, Yang H, Okoro EU, Guo Z. Up-regulation of cholesterol absorption is a mechanism for cholecystokinin-induced hypercholesterolemia. J Biol Chem 2014; 289:12989-99. [PMID: 24692543 DOI: 10.1074/jbc.m113.534388] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Excessive absorption of intestinal cholesterol is a risk factor for atherosclerosis. This report examines the effect of cholecystokinin (CCK) on plasma cholesterol level and intestinal cholesterol absorption using the in vivo models of C57BL/6 wild-type and low density lipoprotein receptor knock-out (LDLR(-/-)) mice. These data were supported by in vitro studies involving mouse primary intestinal epithelial cells and human Caco-2 cells; both express CCK receptor 1 and 2 (CCK1R and CCK2R). We found that intravenous injection of [Thr(28),Nle(31)]CCK increased plasma cholesterol levels and intestinal cholesterol absorption in both wild-type and LDLR(-/-) mice. Treatment of mouse primary intestinal epithelial cells with [Thr(28),Nle(31)]CCK increased cholesterol absorption, whereas selective inhibition of CCK1R and CCK2R with antagonists attenuated CCK-induced cholesterol absorption. In Caco-2 cells, CCK enhanced CCK1R/CCK2R heterodimerization. Knockdown of both CCK1R and CCK2 or either one of them diminished CCK-induced cholesterol absorption to the same extent. CCK also increased cell surface-associated NPC1L1 (Niemann-Pick C1-like 1) transporters but did not alter their total protein expression. Inhibition or knockdown of NPC1L1 attenuated CCK-induced cholesterol absorption. CCK enhanced phosphatidylinositide 3-kinase (PI3K) and Akt phosphorylation and augmented the interaction between NPC1L1 and Rab11a (Rab-GTPase-11a), whereas knockdown of CCK receptors or inhibition of G protein βγ dimer (Gβγ) diminished CCK-induced PI3K and Akt phosphorylation. Inhibition of PI3K and Akt or knockdown of PI3K diminished CCK-induced NPC1L1-Rab11a interaction and cholesterol absorption. Knockdown of Rab11a suppressed CCK-induced NPC1L1 translocation and cholesterol absorption. These data imply that CCK enhances cholesterol absorption by activation of a pathway involving CCK1R/CCK2R, Gβγ, PI3K, Akt, Rab11a, and NPC1L.
Collapse
Affiliation(s)
- LiChun Zhou
- From the Department of Physiology, Meharry Medical College, Nashville, Tennessee 37208
| | | | | | | |
Collapse
|
47
|
Failla ML, Chitchumronchokchai C, Ferruzzi MG, Goltz SR, Campbell WW. Unsaturated fatty acids promote bioaccessibility and basolateral secretion of carotenoids and α-tocopherol by Caco-2 cells. Food Funct 2014; 5:1101-12. [PMID: 24710065 DOI: 10.1039/c3fo60599j] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Bioavailability of carotenoids and tocopherols from foods is determined by the efficiency of transfer from food/meal to mixed micelles during digestion, incorporation into chylomicrons for trans-epithelial transport to lymphatic/blood system, and distribution to target tissues.
Collapse
Affiliation(s)
- Mark L. Failla
- Department of Human Sciences
- Human Nutrition Program
- The Ohio State University
- Columbus, USA
| | | | - Mario G. Ferruzzi
- Department of Food Science
- Purdue University
- West Lafayette, USA
- Department of Nutrition Science
- Purdue University
| | | | | |
Collapse
|
48
|
Grenier E, Mailhot G, Dion D, Ravid Z, Spahis S, Bendayan M, Levy E. Role of the apical and basolateral domains of the enterocyte in the regulation of cholesterol transport by a high glucose concentration. Biochem Cell Biol 2013; 91:476-86. [PMID: 24219290 DOI: 10.1139/bcb-2013-0053] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We have recently shown that a high glucose (HG) concentration raised intestinal cholesterol (CHOL) transport and metabolism in intestinal epithelial cells. The objective of the present work is to determine whether the stimulus for increased CHOL absorption by glucose originates from the apical site (corresponding to the intestinal lumen) or from the basolateral site (related to blood circulation). We tackled this issue by using differentiated Caco-2/15 cells. Only basolateral medium, supplemented with 25 mmol/L glucose, stimulated [(14)C]-CHOL uptake via the up-regulation of the critical CHOL transporter NPC1L1 protein, as confirmed by its specific ezetimibe inhibitor that abolished the rise in glucose-mediated CHOL capture. No significant changes were noted in SR-BI and CD36. Elevated CHOL uptake was associated with an increase in the transcription factors SREBP-2, LXR-β, and ChREBP, along with a fall in RXR-α. Interestingly, although the HG concentration in the apical medium caused modest changes in CHOL processing, its impact was synergetic with that of the basolateral medium. Our results suggest that HG concentration influences positively intestinal CHOL uptake when present in the basolateral medium. In addition, excessive consumption of diets containing high levels of carbohydrates may strengthen intestinal CHOL uptake in metabolic syndrome, thereby contributing to elevated levels of circulating CHOL and, consequently, the risk of developing type 2 diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Emilie Grenier
- a Research Centre, CHU Ste-Justine, 3175 Ste-Catherine Road, Montreal, QC H3T 1C5, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Pelaseyed T, Gustafsson JK, Gustafsson IJ, Ermund A, Hansson GC. Carbachol-induced MUC17 endocytosis is concomitant with NHE3 internalization and CFTR membrane recruitment in enterocytes. Am J Physiol Cell Physiol 2013; 305:C457-67. [PMID: 23784542 DOI: 10.1152/ajpcell.00141.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We have reported that transmembrane mucin MUC17 binds PDZ protein PDZK1, which retains MUC17 apically in enterocytes. MUC17 and transmembrane mucins MUC3 and MUC12 are suggested to build the enterocyte apical glycocalyx. Carbachol (CCh) stimulation of the small intestine results in gel-forming mucin secretion from goblet cells, something that requires adjacent enterocytes to secrete chloride and bicarbonate for proper mucin formation. Surface labeling and confocal imaging demonstrated that apically expressed MUC17 in Caco-2 cells and Muc3(17) in murine enterocytes were endocytosed upon stimulation with CCh. Relocation of MUC17 in response to CCh was specific as MUC3 and MUC12 did not relocate following CCh stimulation. MUC17 colocalized with PDZK1 under basal conditions, while MUC17 relocated to the terminal web and into early endosomes after CCh stimulation. CCh stimulation concomitantly internalized the Na(+/)H(+) exchanger 3 (NHE3) and recruited cystic fibrosis transmembrane conductance regulator (CFTR) to the apical membranes, a process that was important for CFTR-mediated bicarbonate secretion necessary for proper gel-forming mucin unfolding. The reason for the specific internalization of MUC17 is not understood, but it could limit the diffusion barrier for ion secretion caused by the apical enterocyte glycocalyx or alternatively act to sample luminal bacteria. Our results reveal well-orchestrated mucus secretion and trafficking of ion channels and the MUC17 mucin.
Collapse
Affiliation(s)
- Thaher Pelaseyed
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
50
|
Pan X, Munshi MK, Iqbal J, Queiroz J, Sirwi AA, Shah S, Younus A, Hussain MM. Circadian regulation of intestinal lipid absorption by apolipoprotein AIV involves forkhead transcription factors A2 and O1 and microsomal triglyceride transfer protein. J Biol Chem 2013; 288:20464-76. [PMID: 23729668 DOI: 10.1074/jbc.m113.473454] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have shown previously that Clock, microsomal triglyceride transfer protein (MTP), and nocturnin are involved in the circadian regulation of intestinal lipid absorption. Here, we clarified the role of apolipoprotein AIV (apoAIV) in the diurnal regulation of plasma lipids and intestinal lipid absorption in mice. Plasma triglyceride in apoAIV(-/-) mice showed diurnal variations similar to apoAIV(+/+) mice; however, the increases in plasma triglyceride at night were significantly lower in these mice. ApoAIV(-/-) mice absorbed fewer lipids at night and showed blunted response to daytime feeding. To explain reasons for these lower responses, we measured MTP expression; intestinal MTP was low at night, and its induction after food entrainment was less in apoAIV(-/-) mice. Conversely, apoAIV overexpression increased MTP mRNA in hepatoma cells, indicating transcriptional regulation. Mechanistic studies revealed that sequences between -204/-775 bp in the MTP promoter respond to apoAIV and that apoAIV enhances expression of FoxA2 and FoxO1 transcription factors and their binding to the identified cis elements in the MTP promoter at night. Knockdown of FoxA2 and FoxO1 abolished apoAIV-mediated MTP induction. Similarly, knockdown of apoAIV in differentiated Caco-2 cells reduced MTP, FoxA2, and FoxO1 mRNA levels, cellular MTP activity, and media apoB. Moreover, FoxA2 and FoxO1 expression showed diurnal variations, and their expression was significantly lower in apoAIV(-/-) mice. These data indicate that apoAIV modulates diurnal changes in lipid absorption by regulating forkhead transcription factors and MTP and that inhibition of apoAIV expression might reduce plasma lipids.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203, USA.
| | | | | | | | | | | | | | | |
Collapse
|