1
|
Somabattini RA, Sherin S, Siva B, Chowdhury N, Nanjappan SK. Unravelling the complexities of non-alcoholic steatohepatitis: The role of metabolism, transporters, and herb-drug interactions. Life Sci 2024; 351:122806. [PMID: 38852799 DOI: 10.1016/j.lfs.2024.122806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a mainstream halting liver disease with high prevalence in North America, Europe, and other world regions. It is an advanced form of NAFLD caused by the amassing of fat in the liver and can progress to the more severe form known as non-alcoholic steatohepatitis (NASH). Until recently, there was no authorized pharmacotherapy reported for NASH, and to improve the patient's metabolic syndrome, the focus is mainly on lifestyle modification, weight loss, ensuring a healthy diet, and increased physical activity; however, the recent approval of Rezdiffra (Resmetirom) by the US FDA may change this narrative. As per the reported studies, there is an increased articulation of uptake and efflux transporters of the liver, including OATP and MRP, in NASH, leading to changes in the drug's pharmacokinetic properties. This increase leads to alterations in the pharmacokinetic properties of drugs. Furthermore, modifications in Cytochrome P450 (CYP) enzymes can have a significant impact on these properties. Xenobiotics are metabolized primarily in the liver and constitute liver enzymes and transporters. This review aims to delve into the role of metabolism, transport, and potential herb-drug interactions in the context of NASH.
Collapse
Affiliation(s)
- Ravi Adinarayan Somabattini
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Sahla Sherin
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Bhukya Siva
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Neelanjan Chowdhury
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Satheesh Kumar Nanjappan
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India.
| |
Collapse
|
2
|
Osuna-Ramos JF, Farfan-Morales CN, Cordero-Rivera CD, De Jesús-González LA, Reyes-Ruiz JM, Hurtado-Monzón AM, Palacios-Rápalo SN, Jiménez-Camacho R, Meraz-Ríos MA, Del Ángel RM. Cholesterol-Lowering Drugs as Potential Antivirals: A Repurposing Approach against Flavivirus Infections. Viruses 2023; 15:1465. [PMID: 37515153 PMCID: PMC10383882 DOI: 10.3390/v15071465] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Flaviviruses, including Dengue (DENV), Zika (ZIKV), and Yellow Fever (YFV) viruses, represent a significant global health burden. The development of effective antiviral therapies against these viruses is crucial to mitigate their impact. This study investigated the antiviral potential of the cholesterol-lowering drugs atorvastatin and ezetimibe in monotherapy and combination against DENV, ZIKV, and YFV. In vitro results demonstrated a dose-dependent reduction in the percentage of infected cells for both drugs. The combination of atorvastatin and ezetimibe showed a synergistic effect against DENV 2, an additive effect against DENV 4 and ZIKV, and an antagonistic effect against YFV. In AG129 mice infected with DENV 2, monotherapy with atorvastatin or ezetimibe significantly reduced clinical signs and increased survival. However, the combination of both drugs did not significantly affect survival. This study provides valuable insights into the potential of atorvastatin and ezetimibe as antiviral agents against flaviviruses and highlights the need for further investigations into their combined therapeutic effects.
Collapse
Affiliation(s)
- Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
- Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán 80019, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana (UAM), Unidad Cuajimalpa, Mexico City 05348, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico
| | - José Manuel Reyes-Ruiz
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional "Adolfo Ruiz Cortines", Instituto Mexicano del Seguro Social (IMSS), Veracruz Norte, Veracruz 91810, Mexico
- Facultad de Medicina, Región Veracruz, Universidad Veracruzana (UV), Veracruz 91090, Mexico
| | - Arianna M Hurtado-Monzón
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Selvin Noé Palacios-Rápalo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Ricardo Jiménez-Camacho
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Marco Antonio Meraz-Ríos
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Rosa María Del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| |
Collapse
|
3
|
Okada BY, Kuroiwa S, Noi A, Tanaka A, Nishikawa J, Kondo Y, Ishitsuka Y, Irie T, Higaki K, Matsuo M, Ichikawa A. Effects of 6-O-α-maltosyl-β cyclodextrin on lipid metabolism in Npc1-deficient Chinese hamster ovary cells. Mol Genet Metab 2022; 137:239-248. [PMID: 36182715 DOI: 10.1016/j.ymgme.2022.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/02/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022]
Abstract
Niemann-Pick disease Type C (NPC) is a lysosomal storage disorder caused by mutation of the NPC1/NPC2 genes, which ultimately results in the accumulation of unesterified cholesterol (UEC) in lysosomes, thereby inducing symptoms such as progressive neurodegeneration and hepatosplenomegaly. This study determines the effects of 6-O-α-maltosyl-β cyclodextrin (Mal-βCD) on lipid levels and synthesis in Npc1-deficient (Npc1-KO cells) and vehicle CHO cells. Compared to vehicle cells, Npc1-KO cells exhibited high level of UEC, and low levels of esterified cholesterols (ECs) and long-chain fatty acids (LCFAs). The difference in lipid levels between Npc1-KO and CHO cells was largely ameliorated by Mal-βCD administration. Moreover, the effects of Mal-βCD were reproduced in the lysosomes prepared from Npc1-KO cells. Stable isotope tracer analysis with extracellular addition of D4-deuterated palmitic acid (D4-PA) to Npc1-KO cells increased the synthesis of D4-deuterated LCFAs (D4-LCFAs) and D4-deuterated ECs (D4-ECs) in a Mal-βCD-dependent manner. Simultaneous addition of D6-deuterated UEC (D6-UEC) and D4-PA promoted the Mal-βCD-dependent synthesis of D6-/D4-ECs, consisting of D6-UEC and D4-PA, D4-deuterated stearic acid, or D4-deuterated myristic acid, in Npc1-KO cells. These results suggest that Mal-βCD helps to maintain normal lipid metabolism by restoring balance among UEC, ECs, and LCFAs through acting on behalf of NPC1 in Npc1-KO cells and may therefore be useful in designing effective therapies for NPC.
Collapse
Affiliation(s)
- By Yasuyo Okada
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Kyuban-cho, Koshien, Nishinomiya, Hyogo 663-8179, Japan.
| | - Sayako Kuroiwa
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Kyuban-cho, Koshien, Nishinomiya, Hyogo 663-8179, Japan
| | - Ayaka Noi
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Kyuban-cho, Koshien, Nishinomiya, Hyogo 663-8179, Japan
| | - Ayaka Tanaka
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Kyuban-cho, Koshien, Nishinomiya, Hyogo 663-8179, Japan
| | - Junichi Nishikawa
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Kyuban-cho, Koshien, Nishinomiya, Hyogo 663-8179, Japan
| | - Yuki Kondo
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yoichi Ishitsuka
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Tetsumi Irie
- Department of Pharmaceutical Packaging Technology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Katsumi Higaki
- Research Initiative Center, Organization for Research Initiative and Promotion, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Muneaki Matsuo
- Department of Pediatrics, Faculty of Medicine, Saga University, 5-1-1, Nabeshima, Saga 849-8501, Japan
| | - Atsushi Ichikawa
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Kyuban-cho, Koshien, Nishinomiya, Hyogo 663-8179, Japan; Bio-Education Laboratory, Tawara Building #702, 1-21-33 Higashinakajima, Osaka 533-0033, Japan.
| |
Collapse
|
4
|
Mimicking natural cholesterol assimilation to elevate the oral delivery of liraglutide for type Ⅱ diabetes therapy. Asian J Pharm Sci 2022; 17:653-665. [DOI: 10.1016/j.ajps.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/15/2022] [Accepted: 08/15/2022] [Indexed: 11/21/2022] Open
|
5
|
Xu C, Li H, Tang CK. Sterol carrier protein 2 in lipid metabolism and non-alcoholic fatty liver disease: Pathophysiology, molecular biology, and potential clinical implications. Metabolism 2022; 131:155180. [PMID: 35311663 DOI: 10.1016/j.metabol.2022.155180] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/03/2022] [Accepted: 03/13/2022] [Indexed: 11/29/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered as the most common chronic liver disease and has become a rapidly global public health problem. Sterol carrier protein 2 (SCP-2), also called non-specific lipid-transfer protein, is predominantly expressed by the liver. SCP-2 plays a key role in intracellular lipid transport and metabolism. SCP-2 has been closely implicated in the development of NAFLD-related metabolic disorders, such as obesity, atherosclerosis, Type 2 diabetes mellitus (T2DM), and gallstones. Recent studies indicate that SCP-2 plays a beneficial role in NAFLD by regulating cholesterol-, endocannabinoid-, and fatty acid-related aspects of lipid metabolism. Hence, in this paper, we summarize the latest findings about the roles of SCP-2 in hepatic steatosis and further describe its molecular function in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Can Xu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital of University of South China, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Heng Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital of University of South China, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, The First Affiliated Hospital of University of South China, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
6
|
Kulathunga K, Wakimoto A, Hiraishi Y, Yadav MK, Gentleman K, Warabi E, Sakasai T, Miwa Y, Mizuno S, Takahashi S, Hamada M. Albino mice with the point mutation at the tyrosinase locus show high cholesterol diet-induced NASH susceptibility. Sci Rep 2021; 11:21827. [PMID: 34750345 PMCID: PMC8576022 DOI: 10.1038/s41598-021-00501-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/06/2021] [Indexed: 11/26/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) constitutes a metabolic disorder with high worldwide prevalence and increasing incidence. The inflammatory progressive state, non-alcoholic steatohepatitis (NASH), leads to liver fibrosis and carcinogenesis. Here, we evaluated whether tyrosinase mutation underlies NASH pathophysiology. Tyrosinase point-mutated B6 (Cg)-Tyrc-2J/J mice (B6 albino) and C57BL/6J black mice (B6 black) were fed with high cholesterol diet (HCD) for 10 weeks. Normal diet-fed mice served as controls. HCD-fed B6 albino exhibited high NASH susceptibility compared to B6 black, a phenotype not previously reported. Liver injury occurred in approximately 50% of B6 albino from one post HCD feeding, with elevated serum alanine aminotransferase and aspartate aminotransferase levels. NASH was induced following 2 weeks in severe-phenotypic B6 albino (sB6), but B6 black exhibited no symptoms, even after 10 weeks. HCD-fed sB6 albino showed significantly higher mortality rate. Histological analysis of the liver revealed significant inflammatory cell and lipid infiltration and severe fibrosis. Serum lipoprotein analysis revealed significantly higher chylomicron and very low-density lipoprotein levels in sB6 albino. Moreover, significantly higher small intestinal lipid absorption and lower fecal lipid excretion occurred together with elevated intestinal NPC1L1 expression. As the tyrosinase point mutation represents the only genetic difference between B6 albino and B6 black, our work will facilitate the identification of susceptible genetic factors for NASH development and expand the understanding of NASH pathophysiology.
Collapse
Affiliation(s)
- Kaushalya Kulathunga
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.,Department of Physiology, Faculty of Medicine, Sabaragamuwa University of Sri Lanka, Hidellana, P.O. Box 01, Ratnapura, Sri Lanka
| | - Arata Wakimoto
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yukiko Hiraishi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Manoj Kumar Yadav
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kyle Gentleman
- Integrated Master of Science Natural Sciences, University of Southampton, Highfield, Southampton, Hampshire, SO17 1BJ, UK
| | - Eiji Warabi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Tomoki Sakasai
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yoshihiro Miwa
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.,Gene Engineering Division, BioResource Research Center, RIKEN, 3-1-1, Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan. .,Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan. .,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan. .,Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
7
|
Efficacy of Polyphenols in the Management of Dyslipidemia: A Focus on Clinical Studies. Nutrients 2021; 13:nu13020672. [PMID: 33669729 PMCID: PMC7922034 DOI: 10.3390/nu13020672] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Polyphenols (PLPs), phytochemicals found in a wide range of plant-based foods, have gained extensive attention in view of their antioxidant, anti-inflammatory, immunomodulatory and several additional beneficial activities. The health-promoting effects noted in animal models of various non-communicable diseases explain the growing interest in these molecules. In particular, in vitro and animal studies reported an attenuation of lipid disorders in response to PLPs. However, despite promising preclinical investigations, the effectiveness of PLPs in human dyslipidemia (DLP) is less clear and necessitates revision of available literature. Therefore, the present review analyzes the role of PLPs in managing clinical DLP, notably by dissecting their potential in ameliorating lipid/lipoprotein metabolism and alleviating hyperlipidemia, both postprandially and in long-term interventions. To this end, PubMed was used for article search. The search terms included polyphenols, lipids, triglycerides, cholesterol, LDL-cholesterol and /or HDL-cholesterol. The critical examination of the trials published to date illustrates certain benefits on blood lipids along with co-morbidities in participant’s health status. However, inconsistent results document significant research gaps, potentially owing to study heterogeneity and lack of rigor in establishing PLP bioavailability during supplementation. This underlines the need for further efforts in order to elucidate and support a potential role of PLPs in fighting DLP.
Collapse
|
8
|
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: 3.3] [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
|
9
|
Levy E, Beaulieu JF, Spahis S. From Congenital Disorders of Fat Malabsorption to Understanding Intra-Enterocyte Mechanisms Behind Chylomicron Assembly and Secretion. Front Physiol 2021; 12:629222. [PMID: 33584351 PMCID: PMC7873531 DOI: 10.3389/fphys.2021.629222] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/06/2021] [Indexed: 11/13/2022] Open
Abstract
During the last two decades, a large body of information on the events responsible for intestinal fat digestion and absorption has been accumulated. In particular, many groups have extensively focused on the absorptive phase in order to highlight the critical "players" and the main mechanisms orchestrating the assembly and secretion of chylomicrons (CM) as essential vehicles of alimentary lipids. The major aim of this article is to review understanding derived from basic science and clinical conditions associated with impaired packaging and export of CM. We have particularly insisted on inborn metabolic pathways in humans as well as on genetically modified animal models (recapitulating pathological features). The ultimate goal of this approach is that "experiments of nature" and in vivo model strategy collectively allow gaining novel mechanistic insight and filling the gap between the underlying genetic defect and the apparent clinical phenotype. Thus, uncovering the cause of disease contributes not only to understanding normal physiologic pathway, but also to capturing disorder onset, progression, treatment and prognosis.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU Ste-Justine, Université de Montréal, Montreal, QC, Canada
- Department of Nutrition, Université de Montréal, Montreal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
| | - Jean François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Schohraya Spahis
- Research Centre, CHU Ste-Justine, Université de Montréal, Montreal, QC, Canada
- Department of Nutrition, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
10
|
Lo CC, Coschigano KT. ApoB48 as an Efficient Regulator of Intestinal Lipid Transport. Front Physiol 2020; 11:796. [PMID: 32733283 PMCID: PMC7360825 DOI: 10.3389/fphys.2020.00796] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/15/2020] [Indexed: 12/30/2022] Open
Abstract
Fatty meals induce intestinal secretion of chylomicrons (CMs) containing apolipoprotein (Apo) B48. These CMs travel via the lymphatic system before entering the circulation. ApoB48 is produced after post-transcriptional RNA modification by Apobec-1 editing enzyme, exclusively in the small intestine of humans and most other mammals. In contrast, in the liver where Apobec-1 editing enzyme is not expressed (except in rats and mice), the unedited transcript encodes a larger protein, ApoB100, which is used in the formation of very low-density lipoproteins (VLDL) to transport liver-synthesized fat to peripheral tissues. Apobec-1 knockout (KO) mice lack the ability to perform ApoB RNA editing, and thus, express ApoB100 in the intestine. These mice, maintained on either a chow diet or high fat diet, have body weight gain and food intake comparable to their wildtype (WT) counterparts on the respective diet; however, they secrete larger triglyceride (TG)-rich lipoprotein particles and at a slower rate than the WT mice. Using a lymph fistula model, we demonstrated that Apobec-1 KO mice also produced fewer CMs and exhibited reduced lymphatic transport of TG in response to duodenal infusion of TG at a moderate dose; in contrast, the Apobec-1 KO and WT mice had similar lymphatic transport of TG when they received a high dose of TG. Thus, the smaller, energy-saving ApoB48 appears to play a superior role in comparison with ApoB100 in the control of intestinal lipid transport in response to dietary lipid intake, at least at low to moderate lipid levels.
Collapse
Affiliation(s)
- Chunmin C Lo
- The Diabetes Institute, Interdisciplinary Program in Molecular and Cellular Biology, and Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Karen T Coschigano
- The Diabetes Institute, Interdisciplinary Program in Molecular and Cellular Biology, and Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
11
|
Huang CS, Yu X, Fordstrom P, Choi K, Chung BC, Roh SH, Chiu W, Zhou M, Min X, Wang Z. Cryo-EM structures of NPC1L1 reveal mechanisms of cholesterol transport and ezetimibe inhibition. SCIENCE ADVANCES 2020; 6:eabb1989. [PMID: 32596471 PMCID: PMC7304964 DOI: 10.1126/sciadv.abb1989] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/11/2020] [Indexed: 05/21/2023]
Abstract
The intestinal absorption of cholesterol is mediated by a multipass membrane protein, Niemann-Pick C1-Like 1 (NPC1L1), the molecular target of a cholesterol lowering therapy ezetimibe. While ezetimibe gained Food and Drug Administration approval in 2002, its mechanism of action has remained unclear. Here, we present two cryo-electron microscopy structures of NPC1L1, one in its apo form and the other complexed with ezetimibe. The apo form represents an open state in which the N-terminal domain (NTD) interacts loosely with the rest of NPC1L1, leaving the NTD central cavity accessible for cholesterol loading. The ezetimibe-bound form signifies a closed state in which the NTD rotates ~60°, creating a continuous tunnel enabling cholesterol movement into the plasma membrane. Ezetimibe blocks cholesterol transport by occluding the tunnel instead of competing with cholesterol binding. These findings provide insight into the molecular mechanisms of NPC1L1-mediated cholesterol transport and ezetimibe inhibition, paving the way for more effective therapeutic development.
Collapse
Affiliation(s)
- Ching-Shin Huang
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Xinchao Yu
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Preston Fordstrom
- Department of Cardiometabolic Disorders, Amgen Research, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Kaylee Choi
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Ben C. Chung
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Soung-Hun Roh
- Department of Bioengineering and James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Wah Chiu
- Department of Bioengineering and James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- Division of Cryo-EM and Bioimaging, SLAC National Accelerator Laboratory, Menlo Park, CA 94025, USA
| | - Mingyue Zhou
- Department of Cardiometabolic Disorders, Amgen Research, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Xiaoshan Min
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Zhulun Wang
- Department of Therapeutic Discovery, Amgen Research, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080, USA
| |
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: 1.8] [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
|
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: 1.3] [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
|
14
|
Thang SK, Chen PY, Gao WY, Wu MJ, Pan MH, Yen JH. Xanthohumol Suppresses NPC1L1 Gene Expression through Downregulation of HNF-4α and Inhibits Cholesterol Uptake in Caco-2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:11119-11128. [PMID: 31525874 DOI: 10.1021/acs.jafc.9b05221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Xanthohumol (Xan) is a prenylated chalcone mainly found in hops; it has been demonstrated to function against hypercholesterolemia, hyperlipidemia, and atherosclerosis. In this study, we focused on the hypocholesterolemic effect of Xan on cholesterol uptake and the underlying molecular mechanisms of Xan in human intestinal Caco-2 cells. The microarray data showed that Niemann-Pick C1-like 1 (NPC1L1), an essential transporter for dietary cholesterol absorption, was significantly downregulated in Xan-treated Caco-2 cells. We demonstrated that Xan (10 and 20 μM) suppressed the mRNA and protein expression of NPC1L1 by 0.65 ± 0.12-fold and 0.54 ± 0.15-fold and 0.72 ± 0.04-fold and 0.44 ± 0.12-fold, respectively, compared to that of the vehicle-treated Caco-2 cells. Moreover, Xan (10 and 20 μM) significantly inhibited cholesterol uptake by approximately 12 and 32% in Caco-2 cells. NPC1L1 promoter activity was significantly suppressed by Xan, and a DNA element within the NPC1L1 promoter involved in Xan-mediated NPC1L1 reduction located between the -120 and -20 positions was identified. Moreover, Xan markedly decreased the mRNA and protein levels of hepatocyte nuclear factor 4α (HNF-4α), a critical activator of NPC1L1 transcription, and subsequently attenuated HNF-4α/NPC1L1 promoter complex formation, resulting in the suppression of NPC1L1 gene expression. Finally, we demonstrated that Xan markedly abolished lovastatin-induced NPC1L1 overexpression in Caco-2 cells. These findings reveal that Xan suppresses NPC1L1 expression via downregulation of HNF-4α and exerts inhibitory effects on cholesterol uptake in the intestinal Caco-2 cells. Our findings suggest Xan could serve as a potential cholesterol-lowering agent and supplement for statin therapy.
Collapse
Affiliation(s)
- Sang Kim Thang
- Institute of Medical Sciences , Tzu Chi University , Hualien 970 , Taiwan
| | - Pei-Yi Chen
- Center of Medical Genetics , Hualien Tzu Chi Hospital, Buddhist Tzu Chi Foundation , Hualien 970 , Taiwan
| | - Wan-Yun Gao
- Department of Molecular Biology and Human Genetics , Tzu Chi University , Hualien 970 , Taiwan
| | - Ming-Jiuan Wu
- Department of Biotechnology , Chia-Nan University of Pharmacy and Science , Tainan 717 , Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology , National Taiwan University , Taipei 10617 , Taiwan
| | - Jui-Hung Yen
- Institute of Medical Sciences , Tzu Chi University , Hualien 970 , Taiwan
- Department of Molecular Biology and Human Genetics , Tzu Chi University , Hualien 970 , Taiwan
| |
Collapse
|
15
|
Kim YJ, Ryu R, Choi JY, Choi MS. Platycodon grandiflorus Root Ethanol Extract Induces Lipid Excretion, Lipolysis, and Thermogenesis in Diet-Induced Obese Mice. J Med Food 2019; 22:1100-1109. [PMID: 31566484 DOI: 10.1089/jmf.2019.4443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adipocytes regulate lipid metabolism according to physiological energy requirements. A dysfunctional lipid metabolism can lead to obesity and its complications such as hepatic steatosis, diabetes, and hyperlipidemia. In our study, the impact of Platycodon grandiflorus root ethanol extract (PGH) on lipid excretion and thermogenesis-related markers in diet-induced obesity mice was analyzed. Our data show that PGH elevated fatty acid uptake in epididymal adipose tissue by increasing Cd36, Slc27a1, Ffar2, and Ffar4 expression, which led to decreased blood free fatty acid concentrations. Moreover, PGH normalized body weight and fat mass in diet-induced obese mice by increasing lipolysis (Plin1, Atgl, and Hsl) and fatty acid oxidation. Changes in the levels of browning-related genes, enzyme activity of carnitine palmitoyltransferase, and the overall transcriptome (Bmp4, Cidec, Ucp3, Sirt3, and Cox4i1) led to promote brown adipose tissue-like features (browning) in epididymal white adipose tissue and enhanced energy expenditure. Our results suggest that PGH promotes lipid excretion and thermogenic function in high-fat diet-induced obese mice, which are mediated by regulation of fat metabolism.
Collapse
Affiliation(s)
- Ye Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyungpook National University, Daegu, Korea
| | - Ri Ryu
- Research Institute of Applied Animal Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Korea
| | - Ji-Young Choi
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Myung-Sook Choi
- Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu, Korea.,Department of Food Science and Nutrition, Kyungpook National University, Daegu, Korea
| |
Collapse
|
16
|
Sané A, Ahmarani L, Delvin E, Auclair N, Spahis S, Levy E. SAR1B GTPase is necessary to protect intestinal cells from disorders of lipid homeostasis, oxidative stress, and inflammation. J Lipid Res 2019; 60:1755-1764. [PMID: 31409740 PMCID: PMC6795079 DOI: 10.1194/jlr.ra119000119] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/29/2019] [Indexed: 12/12/2022] Open
Abstract
Genetic defects in SAR1B GTPase inhibit chylomicron (CM) trafficking to the Golgi and result in a huge intraenterocyte lipid accumulation with a failure to release CMs and liposoluble vitamins into the blood circulation. The central aim of this study is to test the hypothesis that SAR1B deletion (SAR1B−/−) disturbs enterocyte lipid homeostasis (e.g., FA β-oxidation and lipogenesis) while promoting oxidative stress and inflammation. Another issue is to compare the impact of SAR1B−/− to that of its paralogue SAR1A−/− and combined SAR1A−/−/B−/−. To address these critical issues, we have generated Caco-2/15 cells with a knockout of SAR1A, SAR1B, or SAR1A/B genes. SAR1B−/− results in lipid homeostasis disruption, reflected by enhanced mitochondrial FA β-oxidation and diminished lipogenesis in intestinal absorptive cells via the implication of PPARα and PGC1α transcription factors. Additionally, SAR1B−/−cells, which mimicked enterocytes of CM retention disease, spontaneously disclosed inflammatory and oxidative characteristics via the implication of NF-κB and NRF2. In most conditions, SAR1A−/− cells showed a similar trend, albeit less dramatic, but synergetic effects were observed with the combined defects of the two SAR1 paralogues. In conclusion, SAR1B and its paralogue are needed not only for CM trafficking but also for lipid homeostasis, prooxidant/antioxidant balance, and protection against inflammatory processes.
Collapse
Affiliation(s)
- Alain Sané
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Lena Ahmarani
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Edgard Delvin
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Nikolas Auclair
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Departments of Pharmacology, Université de Montréal, Montreal, Quebec, Canada
| | - Schohraya Spahis
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Emile Levy
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada .,Departments of Pharmacology, Université de Montréal, Montreal, Quebec, Canada.,Nutrition, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Liu Y, Chen D, Li J, Xia D, Yu M, Tao J, Zhang X, Li L, Gan Y. NPC1L1-Targeted Cholesterol-Grafted Poly(β-Amino Ester)/pDNA Complexes for Oral Gene Delivery. Adv Healthc Mater 2019; 8:e1800934. [PMID: 30773830 DOI: 10.1002/adhm.201800934] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/15/2019] [Indexed: 12/12/2022]
Abstract
Gene vectors for oral delivery encounter harsh conditions throughout the gastrointestinal tract, and the continuous peristaltic activity can quickly remove the vectors, leading to inefficient intestinal permeation. Therefore, vectors have demanding property requirements, such as stability under various pH and, more importantly, efficient uptake in different intestinal segments. In this study, a functional polymer, cholesterol-grafted poly(β-amino ester) (poly[hexamethylene diacrylate-β-(5-amino-1-pentanol)] (CH-PHP)), is synthesized and electrostatically interacted with plasmid DNA to form a CH-PHP/DNA complex (CPNC). This complex is designed to target the Niemann-Pick C1-like receptor, a cholesterol receptor, to improve oral gene delivery efficacy. With the presence of cholesterol, CH-PHP shows mitigated cytotoxicity, enhanced enzyme resistance, and improved gene condensing ability. CPNC further contributes to ≈43.1- and 2.3-fold increases in luciferase expression in Caco-2 cells compared with PNC and Lipo 2000/DNA complexes, respectively. In addition, the in vivo transfection efficacy of CPNC is ≈4.1-, 2.1-, and 1.6-fold higher than that of Lipo 2000/DNA complexes in rat duodenum, jejunum, and ileum, respectively. Therefore, CPNC may be a promising delivery vector for gene delivery, and using a cholesterol-specific endocytic pathway can be a novel approach to achieve efficient oral gene transfection.
Collapse
Affiliation(s)
- Yuan Liu
- Department of ChemistryShanghai University Shanghai 200444 P. R. China
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Dan Chen
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Jialin Li
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
- School of PharmacyShanghai University of Traditional Chinese Medicine Shanghai 201203 P. R. China
| | - Dengning Xia
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Miaorong Yu
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Jinsong Tao
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Xinxin Zhang
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Li Li
- Department of ChemistryShanghai University Shanghai 200444 P. R. China
| | - Yong Gan
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| |
Collapse
|
18
|
Han S, Zhang W, Zhang R, Jiao J, Fu C, Tong X, Zhang W, Qin L. Cereal fiber improves blood cholesterol profiles and modulates intestinal cholesterol metabolism in C57BL/6 mice fed a high-fat, high-cholesterol diet. Food Nutr Res 2019; 63:1591. [PMID: 30863273 PMCID: PMC6403461 DOI: 10.29219/fnr.v63.1591] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/24/2018] [Accepted: 12/29/2018] [Indexed: 01/06/2023] Open
Abstract
Background Dietary intake of cereal fiber has been reported to benefit lipid metabolism through multiple mechanisms. The present study aimed to discover the potential mechanisms by which cereal fiber could modify the intestinal cholesterol metabolism. Design Male C57BL/6 mice were fed a reference chow (RC) diet; high-fat, high-cholesterol (HFC) diet; HFC plus oat fiber diet; or HFC plus wheat bran fiber diet for 24 weeks. Serum lipids were measured by enzymatic methods. Western blot was used to determine the protein expressions involved in intestinal cholesterol metabolism. Results Our results showed that HFC-induced elevations of serum triglycerides, total cholesterol, and low-density lipoprotein cholesterol were normalized in both groups that received cereal fiber. At the protein level, compared with the HFC diet group, the two cereal fibers, especially the oat fiber, significantly increased the protein expression of peroxisome proliferator-activated receptor alpha, liver X receptor alpha, sterol regulatory element-binding protein (SREBP) 2, low-density lipoprotein receptor, adenosine triphosphate (ATP)-binding cassette A1, and ATP-binding cassette G1, while decreasing the protein expression of Niemann-Pick C1-like protein 1, SREBP-1, fatty acid synthase, and acetyl-coenzyme A carboxylase, which were involved in intestinal cholesterol metabolism. Conclusion Taken together, increased intake of cereal fiber improved blood cholesterol profiles and increased the intestinal cholesterol efflux and cholesterol clearance in C57BL/6 mice fed a HFC diet. Oat fiber had a stronger effect than wheat bran fiber on cholesterol metabolism by modulating the PPARα, LXRα, and SREBP signaling pathways.
Collapse
Affiliation(s)
- Shufen Han
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Wei Zhang
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China.,Suzhou Maternal and Child Health Care and Family Planning Service Center, Suzhou, China
| | - Ru Zhang
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Jun Jiao
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Chunling Fu
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Xing Tong
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | | | - Liqiang Qin
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| |
Collapse
|
19
|
Bonnefond-Ortega M, Goudable J, Chambrier C, Bétry C. L’absorption intestinale des vitamines hydrosolubles et liposolubles en pratique clinique. NUTR CLIN METAB 2018. [DOI: 10.1016/j.nupar.2017.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Auclair N, Melbouci L, St-Pierre D, Levy E. Gastrointestinal factors regulating lipid droplet formation in the intestine. Exp Cell Res 2018; 363:1-14. [PMID: 29305172 DOI: 10.1016/j.yexcr.2017.12.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022]
Abstract
Cytoplasmic lipid droplets (CLD) are considered as neutral lipid reservoirs, which protect cells from lipotoxicity. It became clear that these fascinating dynamic organelles play a role not only in energy storage and metabolism, but also in cellular lipid and protein handling, inter-organelle communication, and signaling among diverse functions. Their dysregulation is associated with multiple disorders, including obesity, liver steatosis and cardiovascular diseases. The central aim of this review is to highlight the link between intra-enterocyte CLD dynamics and the formation of chylomicrons, the main intestinal dietary lipid vehicle, after overviewing the morphology, molecular composition, biogenesis and functions of CLD.
Collapse
Affiliation(s)
- N Auclair
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Nutrition, Université de Montréal, Montreal, Quebec, Canada H3T 1C5
| | - L Melbouci
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Department of Sciences and Physical Activities, UQAM, Quebec, Canada H2X 1Y4
| | - D St-Pierre
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Department of Sciences and Physical Activities, UQAM, Quebec, Canada H2X 1Y4
| | - E Levy
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Nutrition, Université de Montréal, Montreal, Quebec, Canada H3T 1C5; Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada G1V 0A6.
| |
Collapse
|
21
|
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: 27] [Impact Index Per Article: 3.9] [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
|
22
|
Isorhamnetin derivatives and piscidic acid for hypercholesterolemia: cholesterol permeability, HMG-CoA reductase inhibition, and docking studies. Arch Pharm Res 2017; 40:1278-1286. [PMID: 28936788 DOI: 10.1007/s12272-017-0959-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/13/2017] [Indexed: 10/18/2022]
Abstract
Bioactive compounds, such as isorhamnetin and piscidic acid, were obtained from decoctions of cladodes (stem pads from Opuntia ficus-indica). The effect of these phenolic compounds, in a fiber-free extract, were evaluated as inhibitors of cholesterol permeation through a Caco-2 cell monolayer and as 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor. A reduction of 38% in cholesterol permeation through the Caco-2 cell monolayer was obtained, and the phenolic compounds all permeated between 6 and 9%. A mixture of these compounds showed an IC50 of 20.3 μg/mL as an enzyme inhibitor, whereas piscidic acid alone showed an IC50 of 149.6 μg/mL; this was slightly outperformed by the isorhamnetin derivatives. Docking studies confirmed that both piscidic acid and isorhamnetin derivatives, present in the decoction, could adequately bind to the enzyme active site. These results reveal that O. ficus-indica, and cladodes derived there from, is a promising plant for use in the development of new functional foods and pharmaceutical products.
Collapse
|
23
|
Kamishikiryo J, Haraguchi M, Nakashima S, Tasaka Y, Narahara H, Sugihara N, Nakamura T, Morita T. N-terminal domain of the cholesterol transporter Niemann–Pick C1-like 1 (NPC1L1) is essential for α-tocopherol transport. Biochem Biophys Res Commun 2017; 486:476-480. [DOI: 10.1016/j.bbrc.2017.03.065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 10/20/2022]
|
24
|
Sané A, Seidman E, Spahis S, Lamantia V, Garofalo C, Montoudis A, Marcil V, Levy E. New Insights In Intestinal Sar1B GTPase Regulation and Role in Cholesterol Homeostasis. J Cell Biochem 2016; 116:2270-82. [PMID: 25826777 DOI: 10.1002/jcb.25177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/25/2015] [Indexed: 12/12/2022]
Abstract
Sar1B GTPase is a key component of Coat protein complex II (COPII)-coated vesicles that bud from the endoplasmic reticulum to export newly synthesized proteins. The aims of this study were to determine whether Sar1B responds to lipid regulation and to evaluate its role in cholesterol (CHOL) homeostasis. The influence of lipids on Sar1B protein expression was analyzed in Caco-2/15 cells by Western blot. Our results showed that the presence of CHOL (200 μM) and oleic acid (0.5 mM), bound to albumin, increases Sar1B protein expression. Similarly, supplementation of the medium with micelles composed of taurocholate with monooleylglycerol or oleic acid also stimulated Sar1B expression, but the addition of CHOL (200 μM) to micelle content did not modify its regulation. On the other hand, overexpression of Sar1B impacted on CHOL transport and metabolism in view of the reduced cellular CHOL content along with elevated secretion when incubated with oleic acid-containing micelles for 24 h, thereby disclosing induced CHOL transport. This was accompanied with higher secretion of free- and esterified-CHOL within chylomicrons, which was not the case when oleic acid was replaced with monooleylglycerol or when albumin-bound CHOL was given alone. The aforementioned cellular CHOL depletion was accompanied with a low phosphorylated/non phosphorylated HMG-CoA reductase ratio, indicating elevated enzymatic activity. Combination of Sar1B overexpression with micelle incubation led to reduction in intestinal CHOL transporters (NPC1L1, SR-BI) and metabolic regulators (PCSK9 and LDLR). The present work showed that Sar1B is regulated in a time- and concentration-dependent manner by dietary lipids, suggesting an adaptation to alimentary lipid flux. Our data also suggest that Sar1B overexpression contributes to regulation of CHOL transport and metabolism by facilitating rapid uptake and transport of CHOL.
Collapse
Affiliation(s)
- Alain Sané
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada, H3T 1C5
| | - Ernest Seidman
- Research Institute, McGill University, Campus MGH, C10.148.6, Montreal, Quebec, Canada, H3G 1A4
| | - Schohraya Spahis
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada, H3T 1C5.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada, H3T 1A8
| | - Valérie Lamantia
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada, H3T 1C5
| | - Carole Garofalo
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada, H3T 1C5
| | - Alain Montoudis
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada, H3T 1C5
| | - Valérie Marcil
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada, H3T 1C5.,Research Institute, McGill University, Campus MGH, C10.148.6, Montreal, Quebec, Canada, H3G 1A4
| | - Emile Levy
- Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada, H3T 1C5.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada, H3T 1A8
| |
Collapse
|
25
|
Morgan A, Mooney K, Wilkinson S, Pickles N, Mc Auley M. Cholesterol metabolism: A review of how ageing disrupts the biological mechanisms responsible for its regulation. Ageing Res Rev 2016; 27:108-124. [PMID: 27045039 DOI: 10.1016/j.arr.2016.03.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/22/2016] [Accepted: 03/30/2016] [Indexed: 02/06/2023]
Abstract
Cholesterol plays a vital role in the human body as a precursor of steroid hormones and bile acids, in addition to providing structure to cell membranes. Whole body cholesterol metabolism is maintained by a highly coordinated balancing act between cholesterol ingestion, synthesis, absorption, and excretion. The aim of this review is to discuss how ageing interacts with these processes. Firstly, we will present an overview of cholesterol metabolism. Following this, we discuss how the biological mechanisms which underpin cholesterol metabolism are effected by ageing. Included in this discussion are lipoprotein dynamics, cholesterol absorption/synthesis and the enterohepatic circulation/synthesis of bile acids. Moreover, we discuss the role of oxidative stress in the pathological progression of atherosclerosis and also discuss how cholesterol biosynthesis is effected by both the mammalian target of rapamycin and sirtuin pathways. Next, we examine how diet and alterations to the gut microbiome can be used to mitigate the impact ageing has on cholesterol metabolism. We conclude by discussing how mathematical models of cholesterol metabolism can be used to identify therapeutic interventions.
Collapse
|
26
|
Kleme ML, Sané AT, Garofalo C, Levy E. Targeted CFTR gene disruption with zinc-finger nucleases in human intestinal epithelial cells induces oxidative stress and inflammation. Int J Biochem Cell Biol 2016; 74:84-94. [PMID: 26923293 DOI: 10.1016/j.biocel.2016.02.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 02/14/2016] [Accepted: 02/23/2016] [Indexed: 12/18/2022]
Abstract
UNLABELLED Cystic fibrosis (CF) is a multisystemic pathology caused by mutations of the CF transmembrane conductance regulator (CFTR) gene. OBJECTIVES As the intestine harbors the greatest number of CFTR transcripts after birth and since CFTR plays a role in glutathione transport, we hypothesized that CFTR deletion might produce oxidative stress (OxS) and inflammation in CF intestinal epithelial cell. METHODS CFTR gene was abrogated in Caco-2/15 enterocytes through the zinc-finger nuclease system. Their oxidative and inflammatory characteristics were appreciated under basal conditions and after the treatment with the pro-oxidant iron-ascorbate (Fe/Asc) complex and pro-inflammatory lipopolysaccharide (LPS). RESULTS Intestinal epithelial cells with CFTR knockout spontaneously exhibited an increased lipid peroxidation level, reflected by malondialdehyde overproduction and reduced antioxidant defense characterized by low enzymatic activities of glutathione peroxidase and catalase. CFTR silencing also resulted in elevated protein expression of pro-inflammatory tumor necrosis Factor-α, interleukin-6, cyclooxygenase-2, and the transcription factor nuclear factor-κB. Moreover, exaggerated OxS and inflammation processes occurred in CFTR(-/-) cells in response to the addition of Fe/Asc and LPS, respectively. CONCLUSIONS Intestinal Caco-2/15 cells with CFTR deletion, display innate oxidative and inflammatory features while being more sensitive to pro-oxidant and pro-inflammatory stimuli. These two pathophysiological processes could be implicated in CF-related intestinal disorders.
Collapse
Affiliation(s)
- Marie-Laure Kleme
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, Quebec H3T 1C4, Canada; Department of Nutrition, Université de Montréal, Montreal, Quebec H3T 1C5, Canada
| | - Alain Théophile Sané
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, Quebec H3T 1C4, Canada
| | - Carole Garofalo
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, Quebec H3T 1C4, Canada
| | - Emile Levy
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, Quebec H3T 1C4, Canada; Department of Nutrition, Université de Montréal, Montreal, Quebec H3T 1C5, Canada.
| |
Collapse
|
27
|
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: 0.8] [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]
|
28
|
Takekawa Y, Sato Y, Yamaki Y, Imai M, Noto K, Sumi M, Takekuma Y, Iseki K, Sugawara M. An Approach to Improve Intestinal Absorption of Poorly Absorbed Water-Insoluble Components via Niemann–Pick C1-Like 1. Biol Pharm Bull 2016; 39:301-7. [DOI: 10.1248/bpb.b15-00359] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuto Takekawa
- Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Yuki Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University
| | | | - Mei Imai
- Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Kazuma Noto
- Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Masato Sumi
- Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Yoh Takekuma
- Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Ken Iseki
- Faculty of Pharmaceutical Sciences, Hokkaido University
| | | |
Collapse
|
29
|
Abstract
Dietary lipids are efficiently absorbed by the small intestine, incorporated into triglyceride-rich lipoproteins (chylomicrons), and transported in the circulation to various tissues. Intestinal lipid absorption and mobilization and chylomicron synthesis and secretion are highly regulated processes. Elevated chylomicron production rate contributes to the dyslipidemia seen in common metabolic disorders such as insulin-resistant states and type 2 diabetes and likely increases the risk for atherosclerosis seen in these conditions. An in-depth understanding of the regulation of chylomicron production may provide leads for the development of drugs that could be of therapeutic utility in the prevention of dyslipidemia and atherosclerosis. Chylomicron secretion is subject to regulation by various factors, including diet, body weight, genetic variants, hormones, nutraceuticals, medications, and emerging interventions such as bariatric surgical procedures. In this review we discuss the regulation of chylomicron production, mechanisms that underlie chylomicron dysregulation, and potential avenues for future research.
Collapse
Affiliation(s)
- Satya Dash
- Departments of Medicine and Physiology and the Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, M5G 2C4 Canada;
| | | | | | | |
Collapse
|
30
|
Xu C, Liu Y, Gong Y, Duan X, Tang X, Zhang M, Pang D, Yu L, Wei H, Ouyang H. Overexpression of NPC1L1 in the livers of transgenic Bama miniature pigs accelerates lipid peroxidation. Genes Genomics 2015. [DOI: 10.1007/s13258-014-0235-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
Fomiroid A, a novel compound from the mushroom Fomitopsis nigra, inhibits NPC1L1-mediated cholesterol uptake via a mode of action distinct from that of ezetimibe. PLoS One 2014; 9:e116162. [PMID: 25551765 PMCID: PMC4281142 DOI: 10.1371/journal.pone.0116162] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/03/2014] [Indexed: 12/11/2022] Open
Abstract
Hypercholesterolemia is one of the key risk factors for coronary heart disease, a major cause of death in developed countries. Suppression of NPC1L1-mediated dietary and biliary cholesterol absorption is predicted to be one of the most effective ways to reduce the risk of hypercholesterolemia. In a screen for natural products that inhibit ezetimibe glucuronide binding to NPC1L1, we found a novel compound, fomiroid A, in extracts of the mushroom Fomitopsis nigra. Fomiroid A is a lanosterone derivative with molecular formula C30H48O3. Fomiroid A inhibited ezetimibe glucuronide binding to NPC1L1, and dose-dependently prevented NPC1L1-mediated cholesterol uptake and formation of esterified cholesterol in NPC1L1-expressing Caco2 cells. Fomiroid A exhibited a pharmacological chaperone activity that corrected trafficking defects of the L1072T/L1168I mutant of NPC1L1. Because ezetimibe does not have such an activity, the binding site and mode of action of fomiroid A are likely to be distinct from those of ezetimibe.
Collapse
|
32
|
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: 7.6] [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
|
33
|
Wang J, Mitsche MA, Lütjohann D, Cohen JC, Xie XS, Hobbs HH. Relative roles of ABCG5/ABCG8 in liver and intestine. J Lipid Res 2014; 56:319-30. [PMID: 25378657 PMCID: PMC4306686 DOI: 10.1194/jlr.m054544] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ABCG5 (G5) and ABCG8 (G8) form a sterol transporter that acts in liver and intestine to prevent accumulation of dietary sterols. Mutations in either G5 or G8 cause sitosterolemia, a recessive disorder characterized by sterol accumulation and premature coronary atherosclerosis. Hepatic G5G8 mediates cholesterol excretion into bile, but the function and relative importance of intestinal G5G8 has not been defined. To determine the role of intestinal G5G8, we developed liver-specific (L-G5G8(-/-)), intestine-specific (I-G5G8(-/-)), and total (G5G8(-/-)) KO mice. Tissue levels of sitosterol, the most abundant plant sterol, were >90-fold higher in G5G8(-/-) mice than in WT animals. Expression of G5G8 only in intestine or only in liver decreased tissue sterol levels by 90% when compared with G5G8(-/-) animals. Biliary sterol secretion was reduced in L-G5G8(-/-) and G5G8(-/-) mice, but not in I-G5G8(-/-) mice. Conversely, absorption of plant sterols was increased in I-G5G8(-/-) and G5G8(-/-) mice, but not in L-G5G8(-/-) mice. Reverse cholesterol transport, as assessed from the fraction of intravenously administered (3)H-cholesterol that appeared in feces, was reduced in G5G8(-/-), I-G5G8(-/-), and L-G5G8(-/-) mice. Thus, G5G8 expression in both the liver and intestine protects animals from sterol accumulation, and intestinal G5G8 contributes to extrahepatic cholesterol efflux in mice.
Collapse
Affiliation(s)
- Jin Wang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Matthew A Mitsche
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Dieter Lütjohann
- Department of Clinical Pharmacology, University of Bonn, D-53105 Bonn, Germany
| | - Jonathan C Cohen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Xiao-Song Xie
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Helen H Hobbs
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390 Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390 Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
34
|
Niesor EJ, Chaput E, Mary JL, Staempfli A, Topp A, Stauffer A, Wang H, Durrwell A. Effect of Compounds Affecting ABCA1 Expression and CETP Activity on the HDL Pathway Involved in Intestinal Absorption of Lutein and Zeaxanthin. Lipids 2014; 49:1233-43. [DOI: 10.1007/s11745-014-3958-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/12/2014] [Indexed: 10/24/2022]
|
35
|
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: 1.1] [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
|
36
|
Falé PL, Ferreira C, Maruzzella F, Helena Florêncio M, Frazão FN, Serralheiro MLM. Evaluation of cholesterol absorption and biosynthesis by decoctions of Annona cherimola leaves. JOURNAL OF ETHNOPHARMACOLOGY 2013; 150:718-723. [PMID: 24095697 DOI: 10.1016/j.jep.2013.09.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 09/16/2013] [Accepted: 09/20/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Decoctions of the leaves of Annona cherimola Mill. are traditionally used in Azores to treat hypercholesterolemia. Although they are sold and consumed by people in order to improve their health, these are products that cannot be sold with claims for health benefits as they have never been studied scientifically. MATERIALS AND METHODS The activities of decoctions from Annona cherimola leaves were analysed for the two therapeutic approaches currently used to reduce plasma cholesterol: inhibition of dietary cholesterol uptake and inhibition of HMG-CoA reductase activity. Furthermore, the composition of the decoction was elucidated by LC-MS and the permeability of the active components was analysed using Caco-2 cell monolayers as a model of the intestinal barrier (dietary cholesterol uptake). RESULTS The chemical composition of the Annona cherimola leaves' extract revealed that rutin was its main component. The in vitro gastrointestinal digestion did not modify the chemical composition of the extract. This extract was able to originate a slight reduction in cholesterol absorption through Caco-2 cells lines and to reduce the HMG-CoA reductase activity in 50% when using 137.3 μg of the extract/mL. Rutin, when used in the same concentration as that found in the extract, was able to reduce cholesterol absorption through Caco-2 cells monolayer in approximately 47%. This flavonoid had an IC50 of 17.85 μM relatively to the HMG-CoA reductase activity. CONCLUSIONS The traditional use of decoctions from the leaves of Annona cherimola may be justified, at least by the inhibition of HMG-CoA reductase activity.
Collapse
Affiliation(s)
- Pedro L Falé
- Centro Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Edifício C8, Campo Grande, 1749-016 Lisboa, Portugal
| | | | | | | | | | | |
Collapse
|
37
|
Hayashi H, Kawamura M. Lowering LDL cholesterol, but not raising LDL receptor activity, by ezetimibe. J Clin Lipidol 2013; 7:632-6. [DOI: 10.1016/j.jacl.2013.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/06/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022]
|
38
|
Targeting lipid biosynthesis and salvage in apicomplexan parasites for improved chemotherapies. Nat Rev Microbiol 2013; 11:823-35. [DOI: 10.1038/nrmicro3139] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
39
|
Reboul E. Absorption of vitamin A and carotenoids by the enterocyte: focus on transport proteins. Nutrients 2013; 5:3563-81. [PMID: 24036530 PMCID: PMC3798921 DOI: 10.3390/nu5093563] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/19/2013] [Accepted: 08/26/2013] [Indexed: 12/15/2022] Open
Abstract
Vitamin A deficiency is a public health problem in most developing countries, especially in children and pregnant women. It is thus a priority in health policy to improve preformed vitamin A and/or provitamin A carotenoid status in these individuals. A more accurate understanding of the molecular mechanisms of intestinal vitamin A absorption is a key step in this direction. It was long thought that β-carotene (the main provitamin A carotenoid in human diet), and thus all carotenoids, were absorbed by a passive diffusion process, and that preformed vitamin A (retinol) absorption occurred via an unidentified energy-dependent transporter. The discovery of proteins able to facilitate carotenoid uptake and secretion by the enterocyte during the past decade has challenged established assumptions, and the elucidation of the mechanisms of retinol intestinal absorption is in progress. After an overview of vitamin A and carotenoid fate during gastro-duodenal digestion, our focus will be directed to the putative or identified proteins participating in the intestinal membrane and cellular transport of vitamin A and carotenoids across the enterocyte (i.e., Scavenger Receptors or Cellular Retinol Binding Proteins, among others). Further progress in the identification of the proteins involved in intestinal transport of vitamin A and carotenoids across the enterocyte is of major importance for optimizing their bioavailability.
Collapse
Affiliation(s)
- Emmanuelle Reboul
- INRA, UMR1260, Nutrition, Obesity and Risk of Thrombosis, Marseille F-13385, France.
| |
Collapse
|
40
|
Abstract
Polytopic transmembrane protein, Niemann-Pick C1-Like 1 (NPC1L1) is localized at the apical membrane of enterocytes and the canalicular membrane of hepatocytes. It mediates intestinal cholesterol absorption and prevents extensive loss of cholesterol by transporting biliary cholesterol into hepatocytes. NPC1L1 is a molecular target of ezetimibe, an agent for hypercholesterolemia. Recently, NPC1L1 inhibition has been shown to prevent metabolic disorders such as fatty liver disease, obesity, diabetes, and atherosclerosis. In this review, the identification and characterization of NPC1L1, NPC1L1-dependent cholesterol transport, the relationship with pathogenesis of metabolic disease and its newly introduced function for virus entry are discussed.
Collapse
Affiliation(s)
- Sung-Woo Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
41
|
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: 1.3] [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
|
42
|
Yara S, Lavoie JC, Beaulieu JF, Delvin E, Amre D, Marcil V, Seidman E, Levy E. Iron-ascorbate-mediated lipid peroxidation causes epigenetic changes in the antioxidant defense in intestinal epithelial cells: impact on inflammation. PLoS One 2013; 8:e63456. [PMID: 23717425 PMCID: PMC3661745 DOI: 10.1371/journal.pone.0063456] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 04/03/2013] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION The gastrointestinal tract is frequently exposed to noxious stimuli that may cause oxidative stress, inflammation and injury. Intraluminal pro-oxidants from ingested nutrients especially iron salts and ascorbic acid frequently consumed together, can lead to catalytic formation of oxygen-derived free radicals that ultimately overwhelm the cellular antioxidant defense and lead to cell damage. HYPOTHESIS Since the mechanisms remain sketchy, efforts have been exerted to evaluate the role of epigenetics in modulating components of endogenous enzymatic antioxidants in the intestine. To this end, Caco-2/15 cells were exposed to the iron-ascorbate oxygen radical-generating system. RESULTS Fe/Asc induced a significant increase in lipid peroxidation as reflected by the elevated formation of malondialdehyde along with the alteration of antioxidant defense as evidenced by raised superoxide dismutase 2 (SOD2) and diminished glutathione peroxidase (GPx) activities and genes. Consequently, there was an up-regulation of inflammatory processes illustrated by the activation of NF-κB transcription factor, the higher production of interleukin-6 and cycloxygenase-2 as well as the decrease of IκB. Assessment of promoter's methylation revealed decreased levels for SOD2 and increased degree for GPx2. On the other hand, pre-incubation of Caco-2/15 cells with 5-Aza-2'-deoxycytidine, a demethylating agent, or Trolox antioxidant normalized the activities of SOD2 and GPx, reduced lipid peroxidation and prevented inflammation. CONCLUSION Redox and inflammatory modifications in response to Fe/Asc -mediated lipid peroxidation may implicate epigenetic methylation.
Collapse
Affiliation(s)
- Sabrina Yara
- Department of Nutrition, Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Claude Lavoie
- Department of Pediatrics, Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Jean-François Beaulieu
- Canadian Institutes for Health Research Team on the Digestive Epithelium, Department of Anatomy and Cellular Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Edgard Delvin
- Department of Biochemistry, Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Devendra Amre
- Department of Pediatrics, Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Valerie Marcil
- Research Institute, McGill University, Campus MGH, C10.148.6, Montreal, Quebec, Canada
| | - Ernest Seidman
- Canadian Institutes for Health Research Team on the Digestive Epithelium, Department of Anatomy and Cellular Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Research Institute, McGill University, Campus MGH, C10.148.6, Montreal, Quebec, Canada
| | - Emile Levy
- Department of Nutrition, Research Centre, CHU-Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
- Canadian Institutes for Health Research Team on the Digestive Epithelium, Department of Anatomy and Cellular Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
43
|
Alqahtani S, Mohamed LA, Kaddoumi A. Experimental models for predicting drug absorption and metabolism. Expert Opin Drug Metab Toxicol 2013; 9:1241-54. [DOI: 10.1517/17425255.2013.802772] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Abstract
BACKGROUND Although genome-wide association studies (GWAS) and subsequent meta-analyses have confirmed associations between the PTPN2 (protein tyrosine phosphatase, nonreceptor type 2) gene and Crohn's disease (CD), the potential causal variants remain unidentified. We aimed to dissect potential causal CD-associated PTPN2 variants, assess their functional significance, and relate PTPN2 protein expression with inflammation in CD. METHODS A 3-stage study was carried out. In stage 1, we genotyped tagging single nucleotide polymorphisms (tag-SNPs) in the PTPN2 gene in a sample of patients with CD (<20 years, n = 556) and controls (n = 602). In stage 2, we resequenced the putative promoter, target exons and introns in the PTPN2 gene, and examined associations with high-frequency variants with CD in the stage 1 cohort. In stage 3 we studied the relationship between PTPN2 protein expression and mucosal inflammation and carried out in silico analyses to study the functional characteristics of the PTPN2 CD-associated SNPs. RESULTS In stage 1, we observed associations between 5 intronic SNPs and CD including rs1893217 (P = 2 × 10⁻⁴), the SNP that is in perfect linkage disequilibrium with the lead genome-wide association studies SNP rs2542151. Resequencing revealed 2 known promoter polymorphisms. No associations between these promoter SNPs and CD were evident. In silico analyses revealed that the 5 associated intronic SNPs influenced PTPN2 expression and binding to important transcription factors. PTPN2 protein was overexpressed in inflamed intestinal tissues of patients with CD. CONCLUSIONS Our findings suggest that noncoding variation in the PTPN2 gene may represent the causal variations influencing susceptibility for CD.
Collapse
|
45
|
Terunuma S, Kumata N, Osada K. Ezetimibe impairs uptake of dietary cholesterol oxidation products and reduces alterations in hepatic cholesterol metabolism and antioxidant function in rats. Lipids 2013; 48:587-95. [PMID: 23588779 DOI: 10.1007/s11745-013-3790-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/22/2013] [Indexed: 12/27/2022]
Abstract
Dietary cholesterol oxidation products (COP) induce various adverse effects, including development of atherosclerosis, modulation of lipid metabolism, and unfavorable changes in the antioxidant system. Therefore, we examined the effects of ezetimibe, a cholesterol absorption inhibitor on hepatic cholesterol metabolism and down-regulation of the antioxidant system in rats fed COP. Rats were fed a purified diet containing 0.3 % COP with or without ezetimibe (0.07 mg/100 g body weight) for 27 days. Levels of COP in both the plasma and liver were lowered by ezetimibe through promotion of COP excretion into the feces. Reflecting this effect, an increase in the arteriosclerotic index and a reduction in the mRNA expression of hepatic cholesterol biosynthesis transcripts by dietary COP were observed. Moreover, the ferric reducing ability of the plasma also was significantly higher in rats fed COP plus ezetimibe than in those fed COP alone. Finally, we also observed that ezetimibe enhanced the down-regulation of hepatic fatty acid synthesis in rats fed COP. Thus, ezetimibe, which inhibits the absorption of dietary COP from the small intestine, may exert preventive effects on dietary COP-induced disruption of cholesterol and fatty acid metabolism in the liver and down-regulation of the antioxidant system.
Collapse
Affiliation(s)
- Shoichiro Terunuma
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | | | | |
Collapse
|
46
|
Lee CL, Wen JY, Hsu YW, Pan TM. Monascus-fermented yellow pigments monascin and ankaflavin showed antiobesity effect via the suppression of differentiation and lipogenesis in obese rats fed a high-fat diet. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:1493-1500. [PMID: 23360447 DOI: 10.1021/jf304015z] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Monascus-fermented monascin and ankaflavin are found to strongly inhibit differentiation and lipogenesis and stimulate lipolysis effects in a 3T3-L1 preadipocyte model, but the in vivo regulation mechanism is unclear. This study uses obese rats caused by a high-fat diet to examine the effects of daily monascin and ankaflavin feeding (8 weeks) on antiobesity effects and modulation of differentiation, lipogenesis, and lipid absorption. The results show that monascin and ankaflavin had a significant antiobesity effect, which should result from the modulation of monascin and ankaflavin on the inhibition of differentiation by inhibiting CCAT/enhancer-binding protein β (C/EBPβ) expression (36.4% and 48.3%) and its downstream peroxisome proliferator-activated receptor γ (PPARγ) (55.6% and 64.5%) and CCAT/enhancer-binding protein α (C/EBPα) expressions (25.2% and 33.2%) and the inhibition of lipogenesis by increasing lipase activity (14.0% and 10.7%) and decreasing heparin releasable lipoprotein lipase (HR-LPL) activity (34.8% and 30.5%). Furthermore, monascin and ankaflavin are the first agents found to suppress Niemann-Pick C1 Like 1 (NPC1L1) protein expression (73.6% and 26.1%) associated with small intestine tissue lipid absorption. Importantly, monascin and ankaflavin are not like monacolin K, which increases creatine phosphokinase (CPK) activity, known as a rhabdomyolysis indicator.
Collapse
Affiliation(s)
- Chun-Lin Lee
- Department of Life Science, National Taitung University, Taitung, Taiwan, Republic of China
| | | | | | | |
Collapse
|
47
|
Renner O, Lütjohann D, Richter D, Strohmeyer A, Schimmel S, Müller O, Stange EF, Harsch S. Role of the ABCG8 19H risk allele in cholesterol absorption and gallstone disease. BMC Gastroenterol 2013; 13:30. [PMID: 23406058 PMCID: PMC3598676 DOI: 10.1186/1471-230x-13-30] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 02/12/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gallstone disease is associated with p.D19H of ABCG8 as well as alterations of cholesterol and bile acid metabolism. However, molecular mechanisms have not been fully elucidated. It is important to understand the link between the sterol transporters ABCG5/8 and NPC1L1 and intestinal cholesterol absorption as well as de novo synthesis in gallstone patients stratified according to 19H risk allele. Moreover, the functional importance of the 19H variant on intestinal ABCG8 feature remains to be clarified. METHODS Measurements of serum surrogate markers of cholesterol absorption (plant sterols: sitosterol, campesterol) and synthesis (cholesterol precursor: lathosterol) were carried out by gas chromatography/mass spectrometry (GC/MS). For expression studies, total RNA was isolated from 168 ileal biopsies of study participants with (34) and without gallstone disease (134). Messenger RNA was measured by LightCycler real-time PCR. Genomic DNA was obtained from blood leukocytes. Genotype frequencies of p.D19H were established using MALDI-TOF mass spectrometry. RESULTS Compared to controls, cholesterol absorption but not synthesis in gallstone carriers was diminished by about 21% based on low serum sitosterol (P = 0.0269) and campesterol (P = 0.0231) to cholesterol ratios. D19H was found to be significantly associated with gallstones (odds ratio [OR] = 2.9, P = 0.0220, 95% confidence interval [CI]:1.22-6.89), particularly in the overweight cohort (OR = 3.2, P = 0.0430, 95% CI:1.07-9.26). Cholesterol absorption was about 24% lower in individuals carrying p.D19H compared to wild type (Psitosterol = 0.0080, Pcampesterol = 0.0206). Moreover, irrespective of phenotype, carriers of p.D19H displayed a significant lower absorption than carriers of the major allele. The most pronounced effect on cholesterol absorption ratio was observed for serum campesterol levels (wild type controls to mutated controls 28%, P = 0.0347 and wild type controls to gallstone carriers with 19H allele 37%, P = 0.0030). Notably, ABCG5/8 and NPC1L1 expression was similar in gallstone carriers and controls regardless of p.D19H presence. CONCLUSIONS Both gallstone disease and p.D19H of ABCG8 are associated with diminished cholesterol absorption. However, p.D19H is not responsible for the differences in small intestinal sterol transporter expression.
Collapse
Affiliation(s)
- Olga Renner
- Dr, Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tuebingen, 70376, Stuttgart, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Ehrenman K, Wanyiri JW, Bhat N, Ward HD, Coppens I. Cryptosporidium parvum scavenges LDL-derived cholesterol and micellar cholesterol internalized into enterocytes. Cell Microbiol 2013; 15:1182-97. [PMID: 23311949 DOI: 10.1111/cmi.12107] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/13/2012] [Accepted: 12/27/2012] [Indexed: 11/26/2022]
Abstract
Cryptosporidium spp. are responsible for devastating diarrhoea in immunodeficient individuals. In the intestinal tract, the developmental stages of the parasite are confined to the apical surfaces of epithelial cells. Upon invasion, Cryptosporidium incorporates the microvillous membrane of the enterocyte to form the parasitophorous vacuole (PV) and sequesters itself from the host cytoplasm by rearranging the host cytoskeleton. Cryptosporidium parvum has minimal anabolic capabilities and relies on transporters and salvage pathways to meet its basic metabolic requirements. The cholesterol salvage pathway is crucial for the development of protozoan parasites. In this study, we have examined the sources of cholesterol from C. parvum infecting enterocytes. We illustrated that the intracellular stages of Cryptosporidium as well as the oocysts shed by the host, contain cholesterol. Incubation of infected enterocytes in lipoprotein-free medium impairs parasite development and results in substantial decrease in cholesterol content associated with the PV. Among lipoproteins, LDL constitutes an important source of cholesterol for Cryptosporidium. Dietary cholesterol incorporated into micelles is internalized into enterocytes by the NPC1L1 transporter. We showed that C. parvum also obtains cholesterol from micelles in enterocytes.Pharmacological blockade of NPC1L1 function by ezetimibe or moderate downregulation of NPC1L1 expression decreases parasite infectivity. These observations indicate that, despite its dual sequestration from the intestinal lumen and the host cytoplasm, C. parvum can, in fact, obtain cholesterol both from the gut's lumen and the host cell. This study highlights the evolutionary advantages for epicellular pathogens to access to nutrients from the outside and inside of the host cell.
Collapse
Affiliation(s)
- Karen Ehrenman
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
49
|
Xie 谢畅 C, Zhou 周章森 ZS, Li 李钠 N, Bian 卞艳 Y, Wang 王永建 YJ, Wang 王丽娟 LJ, Li 李伯良 BL, Song 宋保亮 BL. Ezetimibe blocks the internalization of NPC1L1 and cholesterol in mouse small intestine. J Lipid Res 2012; 53:2092-2101. [PMID: 22811412 PMCID: PMC3435542 DOI: 10.1194/jlr.m027359] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 07/05/2012] [Indexed: 11/20/2022] Open
Abstract
The multiple transmembrane protein Niemann-Pick C1 like1 (NPC1L1) is essential for intestinal cholesterol absorption. Ezetimibe binds to NPC1L1 and is a clinically used cholesterol absorption inhibitor. Recent studies in cultured cells have shown that NPC1L1 mediates cholesterol uptake through vesicular endocytosis that can be blocked by ezetimibe. However, how NPC1L1 and ezetimibe work in the small intestine is unknown. In this study, we found that NPC1L1 distributed in enterocytes of villi and transit-amplifying cells of crypts. Acyl-CoA cholesterol acyltransferase 2 (ACAT2), another important protein for cholesterol absorption by providing cholesteryl esters to chylomicrons, was mainly presented in the apical cytoplasm of enterocytes. NPC1L1 and ACAT2 were highly expressed in jejunum and ileum. ACAT1 presented in the Paneth cells of crypts and mesenchymal cells of villi. In the absence of cholesterol, NPC1L1 was localized on the brush border of enterocytes. Dietary cholesterol induced the internalization of NPC1L1 to the subapical layer beneath the brush border and became partially colocalized with the endosome marker Rab11. Ezetimibe blocked the internalization of NPC1L1 and cholesterol and caused their retention in the plasma membrane. This study demonstrates that NPC1L1 mediates cholesterol entering enterocytes through vesicular endocytosis and that ezetimibe blocks this step in vivo.
Collapse
Affiliation(s)
- Chang Xie 谢畅
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhang-Sen Zhou 周章森
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Na Li 李钠
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Bian 卞艳
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yong-Jian Wang 王永建
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li-Juan Wang 王丽娟
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bo-Liang Li 李伯良
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Bao-Liang Song 宋保亮
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
50
|
Grenier E, Garofalo C, Delvin E, Levy E. Modulatory role of PYY in transport and metabolism of cholesterol in intestinal epithelial cells. PLoS One 2012; 7:e40992. [PMID: 22844422 PMCID: PMC3402548 DOI: 10.1371/journal.pone.0040992] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 06/19/2012] [Indexed: 12/17/2022] Open
Abstract
Background Gastrointestinal peptides are involved in modulating appetite. Other biological functions attributed to them include the regulation of lipid homeostasis. However, data concerning PYY remain fragmentary. The objectives of the study were: (i) To determine the effect of PYY on intestinal transport and synthesis of cholesterol, the biogenesis of apolipoproteins (apos) and assembly of lipoproteins and (ii) To analyze whether the effects of PYY are similar according to whether cells are exposed to PYY on apical or basolateral surface. Methodology/Principal Findings Caco-2/15 cells were incubated with PYY (1–36) administered either to the apical or basolateral medium, at concentrations of 50 or 200 nM for 24 hours. De novo synthesis of cholesterol, cholesterol uptake, and assembly of lipoproteins were evaluated through the incorporation of [14C]-acetate, [14C]-cholesterol, and [14C]-oleate, respectively. Biogenesis of apos (A-I, A-IV, E, B-48 and B-100) was examined by the incorporation of [35S]-methionine. The influence of PYY on protein and mRNA levels of many key mediators of lipid metabolism was analyzed by Western blot and PCR, respectively. Our results show that PYY influenced cholesterol metabolism in Caco-2/15 cells depending on the site of PYY delivery. Apical addition of PYY significantly lowered the incorporation of [14C]-cholesterol likely via the reduction of NPC1L1, stimulated intracellular cholesterol synthesis probably through an increase in SREBP-2 expression, whereas it concomitantly increased apo A-I synthesis and decreased LDL secretion. In contrast, basolateral PYY reduced the production of chylomicrons (CM) as well as the biogenesis of apos B-48 and B-100, while lowering the expression of the transcription factors RXRα and PPAR(α,β). Conclusions/Significance PYY is capable of influencing cholesterol homeostasis in intestinal Caco-2/15 cells depending on the site delivery. Apical PYY was able to decrease cholesterol uptake via NPC1L1 downregulation, whereas basolateral PYY diminished CM output through the biogenesis decline of apos B-48 and B-100.
Collapse
Affiliation(s)
- Emilie Grenier
- Research Centre, Centre Hospitalier Universitaire (CHU) Ste-Justine, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Carole Garofalo
- Research Centre, Centre Hospitalier Universitaire (CHU) Ste-Justine, Montreal, Quebec, Canada
| | - Edgard Delvin
- Research Centre, Centre Hospitalier Universitaire (CHU) Ste-Justine, Montreal, Quebec, Canada
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada
| | - Emile Levy
- Research Centre, Centre Hospitalier Universitaire (CHU) Ste-Justine, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|