1
|
Yang H, Su M, Liu M, Sheng Y, Zhu L, Yang L, Mu R, Zou J, Liu X, Liu L. Hepatic retinaldehyde deficiency is involved in diabetes deterioration by enhancing PCK1- and G6PC-mediated gluconeogenesis. Acta Pharm Sin B 2023; 13:3728-3743. [PMID: 37719384 PMCID: PMC10501888 DOI: 10.1016/j.apsb.2023.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/14/2023] [Accepted: 05/06/2023] [Indexed: 09/19/2023] Open
Abstract
Type 2 diabetes (T2D) is often accompanied with an induction of retinaldehyde dehydrogenase 1 (RALDH1 or ALDH1A1) expression and a consequent decrease in hepatic retinaldehyde (Rald) levels. However, the role of hepatic Rald deficiency in T2D progression remains unclear. In this study, we demonstrated that reversing T2D-mediated hepatic Rald deficiency by Rald or citral treatments, or liver-specific Raldh1 silencing substantially lowered fasting glycemia levels, inhibited hepatic glucogenesis, and downregulated phosphoenolpyruvate carboxykinase 1 (PCK1) and glucose-6-phosphatase (G6PC) expression in diabetic db/db mice. Fasting glycemia and Pck1/G6pc mRNA expression levels were strongly negatively correlated with hepatic Rald levels, indicating the involvement of hepatic Rald depletion in T2D deterioration. A similar result that liver-specific Raldh1 silencing improved glucose metabolism was also observed in high-fat diet-fed mice. In primary human hepatocytes and oleic acid-treated HepG2 cells, Rald or Rald + RALDH1 silencing resulted in decreased glucose production and downregulated PCK1/G6PC mRNA and protein expression. Mechanistically, Rald downregulated direct repeat 1-mediated PCK1 and G6PC expression by antagonizing retinoid X receptor α, as confirmed by luciferase reporter assays and molecular docking. These results highlight the link between hepatic Rald deficiency, glucose dyshomeostasis, and the progression of T2D, whilst also suggesting RALDH1 as a potential therapeutic target for T2D.
Collapse
Affiliation(s)
- Hanyu Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mengxiang Su
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ming Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yun Sheng
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liang Zhu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ruijing Mu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jianjun Zou
- Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaodong Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Li Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
2
|
Peng DQ, Smith SB, Lee HG. Vitamin A regulates intramuscular adipose tissue and muscle development: promoting high-quality beef production. J Anim Sci Biotechnol 2021; 12:34. [PMID: 33663602 PMCID: PMC7934359 DOI: 10.1186/s40104-021-00558-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/18/2021] [Indexed: 01/07/2023] Open
Abstract
During growth in cattle, the development of intramuscular adipose tissue and muscle is dependent upon cell hyperplasia (increased number of adipocytes) and hypertrophy (increased size of adipocytes). Based on the results of previous studies, other adipose tissue depots (e.g., perirenal and subcutaneous) develop from the fetal stage primarily as brown adipose tissue. The hyperplastic stage of intramuscular adipose is considered to develop from late pregnancy, but there is no evidence indicating that intramuscular adipose tissue develops initially as brown adipose tissue. Hyperplastic growth of intramuscular adipose continues well into postweaning and is dependent on the timing of the transition to grain-based diets; thereafter, the late-stage development of intramuscular adipose tissue is dominated by hypertrophy. For muscle development, hyperplasia of myoblasts lasts from early (following development of somites in the embryo) to middle pregnancy, after which growth of muscle is the result of hypertrophy of myofibers. Vitamin A is a fat-soluble compound that is required for the normal immunologic function, vision, cellular proliferation, and differentiation. Here we review the roles of vitamin A in intramuscular adipose tissue and muscle development in cattle. Vitamin A regulates both hyperplasia and hypertrophy in in vitro experiments. Vitamin A supplementation at the early stage and restriction at fattening stage generate opposite effects in the beef cattle. Appropriate vitamin A supplementation and restriction strategy increase intramuscular adipose tissue development (i.e., marbling or intramuscular fat) in some in vivo trials. Besides, hyperplasia and hypertrophy of myoblasts/myotubes were affected by vitamin A treatment in in vitro trials. Additionally, some studies reported an interaction between the alcohol dehydrogenase-1C (ADH1C) genotype and vitamin A feed restriction for the development of marbling and/or intramuscular adipose tissue, which was dependent on the timing and level of vitamin A restriction. Therefore, the feed strategy of vitamin A has the visible impact on the marbling and muscle development in the cattle, which will be helpful to promote the quality of the beef.
Collapse
Affiliation(s)
- Dong Qiao Peng
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Stephen B Smith
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Hong Gu Lee
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea.
| |
Collapse
|
3
|
Abstract
Hepatic stellate cells (HSCs) are resident non-parenchymal liver pericytes whose plasticity enables them to regulate a remarkable range of physiologic and pathologic responses. To support their functions in health and disease, HSCs engage pathways regulating carbohydrate, mitochondrial, lipid, and retinoid homeostasis. In chronic liver injury, HSCs drive hepatic fibrosis and are implicated in inflammation and cancer. To do so, the cells activate, or transdifferentiate, from a quiescent state into proliferative, motile myofibroblasts that secrete extracellular matrix, which demands rapid adaptation to meet a heightened energy need. Adaptations include reprogramming of central carbon metabolism, enhanced mitochondrial number and activity, endoplasmic reticulum stress, and liberation of free fatty acids through autophagy-dependent hydrolysis of retinyl esters that are stored in cytoplasmic droplets. As an archetype for pericytes in other tissues, recognition of the HSC's metabolic drivers and vulnerabilities offer the potential to target these pathways therapeutically to enhance parenchymal growth and modulate repair.
Collapse
Affiliation(s)
- Parth Trivedi
- Division of Liver Diseases, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shuang Wang
- Division of Liver Diseases, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Scott L Friedman
- Division of Liver Diseases, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
4
|
Wellmann KB, Kim J, Urso PM, Smith ZK, Johnson BJ. Evaluation of the dietary vitamin A requirement of finishing steers via systematic depletion and repletion, and its effects on performance and carcass characteristics. J Anim Sci 2020; 98:5894116. [PMID: 32812033 DOI: 10.1093/jas/skaa266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/14/2020] [Indexed: 11/14/2022] Open
Abstract
A randomized complete block design experiment with 30 yearling crossbred steers (initial average body weight [BW] = 297.6 ± 32.8 kg) fed a steam-flaked corn-based diet was used to evaluate finishing performance and carcass characteristics when provided with different concentrations of vitamin A (Rovimix A 1000; DSM Nutritional Products Ltd., Sisseln, Switzerland) subsequent to a depletion phase. Steers were blocked by BW (n = 5 blocks; 6 steers per block), assigned to pens (n = 2 steers per pen), and randomly assigned to one of the following dietary treatments: no added vitamin A (0IU; 0.0 IU/kg dry matter [DM] basis of additional vitamin A), vitamin A supplemented at the estimated National Academies of Sciences, Engineering, and Medicine (NASEM) requirement (2,200IU; 2,200 IU/kg of dietary DM of additional vitamin A), and vitamin A supplemented at 5× the estimated requirement (11,000IU; 11,000 IU/kg of dietary DM of additional vitamin A). The basal diet included minimal vitamin A activity (<200 IU of vitamin A activity/kg of dietary DM) via the provitamin A, beta-carotene. After all animals underwent a 91-d vitamin A depletion period, additional vitamin A was top-dressed at feeding via a ground corn carrier. Liver biopsy samples, BW, and blood were obtained on days -91, -35, 0, 28, 56, 84, and 112. Final BW was collected prior to shipping on day 112. Carcass data were collected by trained personnel upon harvest. Sera and liver samples were used to monitor circulating vitamin A and evaluate true vitamin A status of the cattle. Vitamin A status did not affect interim average daily gain or feed efficiency (G:F; P > 0.05). Throughout the duration of the study, dry matter intake for the 0IU cattle was depressed (P = 0.01). Differences were not observed across treatments for hot carcass weight, rib eye area, back fat thickness, kidney-pelvic-heart fat %, marbling score, or dressing percent (P ≥ 0.10). A treatment × day interaction occurred for both (P < 0.01) sera retinol and liver retinol during phase 2 of the trial. The treatments and sera retinol concentrations were incorporated into a repletion model, resulting in an estimation of liver retinol changes (P < 0.01; R2 = 0.682). However, models used to evaluate depleted animals were less effective. The current NASEM recommended that vitamin A requirement of 2,200 IU/kg is adequate for repletion of vitamin A status of feedlot steers.
Collapse
Affiliation(s)
| | - Jongkyoo Kim
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| | - Phil M Urso
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| | - Zachary K Smith
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| | - Bradley J Johnson
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| |
Collapse
|
5
|
Attrill E, Ramsay C, Ross R, Richards S, Sutherland BA, Keske MA, Eringa E, Premilovac D. Metabolic-vascular coupling in skeletal muscle: A potential role for capillary pericytes? Clin Exp Pharmacol Physiol 2019; 47:520-528. [PMID: 31702069 DOI: 10.1111/1440-1681.13208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/22/2019] [Accepted: 11/05/2019] [Indexed: 01/23/2023]
Abstract
The matching of capillary blood flow to metabolic rate of the cells within organs and tissues is a critical microvascular function which ensures appropriate delivery of hormones and nutrients, and the removal of waste products. This relationship is particularly important in tissues where local metabolism, and hence capillary blood flow, must be regulated to avoid a mismatch between nutrient demand and supply that would compromise normal function. The consequences of a mismatch in microvascular blood flow and metabolism are acutely apparent in the brain and heart, where a sudden cessation of blood flow, for example following an embolism, acutely manifests as stroke or myocardial infarction. Even in more resilient tissues such as skeletal muscle, a short-term mismatch reduces muscle performance and exercise tolerance, and can cause intermittent claudication. In the longer-term, a microvascular-metabolic mismatch in skeletal muscle reduces insulin-mediated muscle glucose uptake, leading to disturbances in whole-body metabolic homeostasis. While the notion that capillary blood flow is fine-tuned to meet cellular metabolism is well accepted, the mechanisms that control this function and where and how different parts of the vascular tree contribute to capillary blood flow regulation remain poorly understood. Here, we discuss the emerging evidence implicating pericytes, mural cells that surround capillaries, as key mediators that match tissue metabolic demand with adequate capillary blood flow in a number of organs, including skeletal muscle.
Collapse
Affiliation(s)
- Emily Attrill
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tas, Australia
| | - Ciaran Ramsay
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tas, Australia
| | - Renee Ross
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tas, Australia
| | - Stephen Richards
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tas, Australia
| | - Brad A Sutherland
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tas, Australia
| | - Michelle A Keske
- The Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Vic., Australia
| | - Etto Eringa
- Laboratory for Physiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Dino Premilovac
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tas, Australia
| |
Collapse
|
6
|
Wang Z, Yang H, Xu J, Zhao K, Chen Y, Liang L, Li P, Chen N, Geng D, Zhang X, Liu X, Liu L. Prediction of Atorvastatin Pharmacokinetics in High-Fat Diet and Low-Dose Streptozotocin-Induced Diabetic Rats Using a Semiphysiologically Based Pharmacokinetic Model Involving Both Enzymes and Transporters. Drug Metab Dispos 2019; 47:1066-1079. [PMID: 31399507 DOI: 10.1124/dmd.118.085902] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 07/01/2019] [Indexed: 12/16/2022] Open
Abstract
Atorvastatin is a substrate of cytochrome P450 3a (CYP3a), organic anion-transporting polypeptides (OATPs), breast cancer-resistance protein (BCRP), and P-glycoprotein (P-gp). We aimed to develop a semiphysiologically based pharmacokinetic (semi-PBPK) model involving both enzyme and transporters for predicting the contributions of altered function and expression of CYP3a and transporters to atorvastatin transport in diabetic rats by combining high-fat diet feeding and low-dose streptozotocin injection. Atorvastatin metabolism and transport parameters comes from in situ intestinal perfusion, primary hepatocytes, and intestinal or hepatic microsomes. We estimated the expressions and functions of these proteins and their contributions. Diabetes increased the expression of hepatic CYP3a, OATP1b2, and P-gp but decreased the expression of intestinal CYP3a, OATP1a5, and P-gp. The expression and function of intestinal BCRP were significantly decreased in 10-day diabetic rats but increased in 22-day diabetic rats. Based on alterations in CYP3a and transporters by diabetes, the developed semi-PBPK model was successfully used to predict atorvastatin pharmacokinetics after oral and intravenous doses to rats. Contributions to oral atorvastatin PK were intestinal OATP1a5 < intestinal P-gp < intestinal CYP3a < hepatic CYP3a < hepatic OATP1b2 < intestinal BRCP. Contributions of decreased expression and function of intestinal CYP3a and P-gp by diabetes to oral atorvastatin plasma exposure were almost attenuated by increased expression and function of hepatic CYP3a and OATP1b2. Opposite alterations in oral plasma atorvastatin exposure in 10- and 22-day diabetic rats may be explained by altered intestinal BCRP. In conclusion, the altered atorvastatin pharmacokinetics by diabetes was the synergistic effects of altered intestinal or hepatic CYP3a and transporters and could be predicted using the developed semi-PBPK.
Collapse
Affiliation(s)
- Zhongjian Wang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hanyu Yang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiong Xu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kaijing Zhao
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yang Chen
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Limin Liang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ping Li
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Nan Chen
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Donghao Geng
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiangping Zhang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
7
|
Abstract
Much evidence has accumulated in the literature over the last fifteen years that indicates vitamin A has a role in metabolic disease prevention and causation. This literature proposes that vitamin A can affect obesity development and the development of obesity-related diseases including insulin resistance, type 2 diabetes, hepatic steatosis and steatohepatitis, and cardiovascular disease. Retinoic acid, the transcriptionally active form of vitamin A, accounts for many of the reported associations. However, a number of proteins involved in vitamin A metabolism, including retinol-binding protein 4 (RBP4) and aldehyde dehydrogenase 1A1 (ALDH1A1, alternatively known as retinaldehyde dehydrogenase 1 or RALDH1), have also been identified as being associated with metabolic disease. Some of the reported effects of these vitamin A-related proteins are proposed to be independent of their roles in assuring normal retinoic acid homeostasis. This review will consider both human observational data as well as published data from molecular studies undertaken in rodent models and in cells in culture. The primary focus of the review will be on the effects that vitamin A per se and proteins involved in vitamin A metabolism have on adipocytes, adipose tissue biology, and adipose-related disease, as well as on early stage liver disease, including non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH).
Collapse
Affiliation(s)
- William S Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032.
| |
Collapse
|
8
|
Reddy MRG, Asha GV, Manchiryala SK, Putcha UK, Vajreswari A, Jeyakumar SM. High-Fat Diet Elevates Liver Docosahexaenoic Acid Possibly through Over-Expression of Very Long-Chain Fatty Acid Elongase 2 in C57BL/6J Mice. INT J VITAM NUTR RES 2019; 89:62-72. [PMID: 30957704 DOI: 10.1024/0300-9831/a000432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The liver is the main site of lipid metabolism and vitamin A storage. Dietary factors are known to affect liver function, thereby leading to metabolic abnormalities. Here, we assessed the impact of long-term feeding of a high-fat diet on hepatic vitamin A status and lipid metabolism. For this purpose, 14 male and 14 female 35-day-old mice (strain C57BL/6J) were each divided into 2 groups of 7 animals and fed either a stock diet or a high-fat (HF) diet for 26 weeks. In addition to increased body weight/weight gain, the HF diet induced hypertriglyceridemia in both (p < 0.01). However, liver triglyceride levels were comparable among groups, which could be partly explained by unaltered expression of various lipogenic pathway proteins such as sterol regulatory element binding protein 1 (SREBP1), fatty acid synthase (FAS), microsomal triglyceride transfer protein (MTTP), and glycerol 3-phosphate acyl transferase (GPAT). On the other hand, hepatic retinol stores increased significantly in both sexes, whereas males displayed elevated circulatory retinol levels. Notably, long-term feeding of a HF diet elevated n-3 polyunsaturated fatty acid (PUFA) and docosahexaenoic acid (DHA, C22:6) levels in the liver (p ≤ 0.001), which is in line with the over-expression of very long-chain fatty acid elongase 2 (ELOVL2) protein in both sexes of mice (p < 0.01). In conclusion, very long-term feeding of a HF diet increased hepatic retinol stores and induced hypertriglyceridemia. However, it had no effect on hepatic triglyceride accumulation, possibly due to increased DHA levels arising from the ELOVL2-mediated elongation pathway.
Collapse
Affiliation(s)
- Mooli Raja Gopal Reddy
- 1Lipid Biochemistry Division, National Institute of Nutrition, Jamai Osmania, Hyderabad, India
| | - Gundluri Venkata Asha
- 1Lipid Biochemistry Division, National Institute of Nutrition, Jamai Osmania, Hyderabad, India
| | | | - Uday Kumar Putcha
- 2Pathology Division, National Institute of Nutrition, Jamai Osmania, Hyderabad, India
| | | | - Shanmugam M Jeyakumar
- 1Lipid Biochemistry Division, National Institute of Nutrition, Jamai Osmania, Hyderabad, India
| |
Collapse
|
9
|
Abstract
Stellate cells are resident lipid-storing cells of the pancreas and liver that transdifferentiate to a myofibroblastic state in the context of tissue injury. Beyond having roles in tissue homeostasis, stellate cells are increasingly implicated in pathological fibrogenic and inflammatory programs that contribute to tissue fibrosis and that constitute a growth-permissive tumor microenvironment. Although the capacity of stellate cells for extracellular matrix production and remodeling has long been appreciated, recent research efforts have demonstrated diverse roles for stellate cells in regulation of epithelial cell fate, immune modulation, and tissue health. Our present understanding of stellate cell biology in health and disease is discussed here, as are emerging means to target these multifaceted cells for therapeutic benefit.
Collapse
Affiliation(s)
- Mara H Sherman
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon 97201, USA;
| |
Collapse
|
10
|
The stellate cell system (vitamin A-storing cell system). Anat Sci Int 2017; 92:387-455. [PMID: 28299597 DOI: 10.1007/s12565-017-0395-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/15/2017] [Indexed: 01/18/2023]
Abstract
Past, present, and future research into hepatic stellate cells (HSCs, also called vitamin A-storing cells, lipocytes, interstitial cells, fat-storing cells, or Ito cells) are summarized and discussed in this review. Kupffer discovered black-stained cells in the liver using the gold chloride method and named them stellate cells (Sternzellen in German) in 1876. Wake rediscovered the cells in 1971 using the same gold chloride method and various modern histological techniques including electron microscopy. Between their discovery and rediscovery, HSCs disappeared from the research history. Their identification, the establishment of cell isolation and culture methods, and the development of cellular and molecular biological techniques promoted HSC research after their rediscovery. In mammals, HSCs exist in the space between liver parenchymal cells (PCs) or hepatocytes and liver sinusoidal endothelial cells (LSECs) of the hepatic lobule, and store 50-80% of all vitamin A in the body as retinyl ester in lipid droplets in the cytoplasm. SCs also exist in extrahepatic organs such as pancreas, lung, and kidney. Hepatic (HSCs) and extrahepatic stellate cells (EHSCs) form the stellate cell (SC) system or SC family; the main storage site of vitamin A in the body is HSCs in the liver. In pathological conditions such as liver fibrosis, HSCs lose vitamin A, and synthesize a large amount of extracellular matrix (ECM) components including collagen, proteoglycan, glycosaminoglycan, and adhesive glycoproteins. The morphology of these cells also changes from the star-shaped HSCs to that of fibroblasts or myofibroblasts.
Collapse
|
11
|
Saeed A, Hoekstra M, Hoeke MO, Heegsma J, Faber KN. The interrelationship between bile acid and vitamin A homeostasis. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:496-512. [PMID: 28111285 DOI: 10.1016/j.bbalip.2017.01.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/04/2017] [Accepted: 01/18/2017] [Indexed: 12/12/2022]
Abstract
Vitamin A is a fat-soluble vitamin important for vision, reproduction, embryonic development, cell differentiation, epithelial barrier function and adequate immune responses. Efficient absorption of dietary vitamin A depends on the fat-solubilizing properties of bile acids. Bile acids are synthesized in the liver and maintained in an enterohepatic circulation. The liver is also the main storage site for vitamin A in the mammalian body, where an intimate collaboration between hepatocytes and hepatic stellate cells leads to the accumulation of retinyl esters in large cytoplasmic lipid droplet hepatic stellate cells. Chronic liver diseases are often characterized by disturbed bile acid and vitamin A homeostasis, where bile production is impaired and hepatic stellate cells lose their vitamin A in a transdifferentiation process to myofibroblasts, cells that produce excessive extracellular matrix proteins leading to fibrosis. Chronic liver diseases thus may lead to vitamin A deficiency. Recent data reveal an intricate crosstalk between vitamin A metabolites and bile acids, in part via the Retinoic Acid Receptor (RAR), Retinoid X Receptor (RXR) and the Farnesoid X Receptor (FXR), in maintaining vitamin A and bile acid homeostasis. Here, we provide an overview of the various levels of "communication" between vitamin A metabolites and bile acids and its relevance for the treatment of chronic liver diseases.
Collapse
Affiliation(s)
- Ali Saeed
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Institute of Molecular biology & Bio-technology, Bahauddin Zakariya University, Multan, Pakistan.
| | - Mark Hoekstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Martijn Oscar Hoeke
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Janette Heegsma
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
12
|
Grumet L, Taschler U, Lass A. Hepatic Retinyl Ester Hydrolases and the Mobilization of Retinyl Ester Stores. Nutrients 2016; 9:nu9010013. [PMID: 28035980 PMCID: PMC5295057 DOI: 10.3390/nu9010013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/12/2016] [Accepted: 12/21/2016] [Indexed: 12/26/2022] Open
Abstract
For mammals, vitamin A (retinol and metabolites) is an essential micronutrient that is required for the maintenance of life. Mammals cannot synthesize vitamin A but have to obtain it from their diet. Resorbed dietary vitamin A is stored in large quantities in the form of retinyl esters (REs) in cytosolic lipid droplets of cells to ensure a constant supply of the body. The largest quantities of REs are stored in the liver, comprising around 80% of the body’s total vitamin A content. These hepatic vitamin A stores are known to be mobilized under times of insufficient dietary vitamin A intake but also under pathological conditions such as chronic alcohol consumption and different forms of liver diseases. The mobilization of REs requires the activity of RE hydrolases. It is astounding that despite their physiological significance little is known about their identities as well as about factors or stimuli which lead to their activation and consequently to the mobilization of hepatic RE stores. In this review, we focus on the recent advances for the understanding of hepatic RE hydrolases and discuss pathological conditions which lead to the mobilization of hepatic RE stores.
Collapse
Affiliation(s)
- Lukas Grumet
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31, 8010 Graz, Austria.
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31, 8010 Graz, Austria.
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31, 8010 Graz, Austria.
| |
Collapse
|
13
|
Abstract
Large differences in time and dose needed to induce hypervitaminosis A have been observed. High doses of vitamin A in food and oily solutions are well tolerated, whereas emulsified preparations have higher toxicity. Chronic hypervitaminosis seems to be induced following daily doses of 300,000 to 600,000 IU of vitamin A (90–180 mg of retinol) in oily preparations for many months or years, whereas teratogenicity may be induced by daily doses as low as 40,000 IU of vitamin A (12 mg of retinol) in oil during the first trimester. for the provitamin A, β-carotene, serious adverse effects have been reported in large-scale prospective randomized trials: four years of supplementation with 20 to 30 mg β-carotene per day was associated with increased risk of lung cancer and cardiovascular disease among smokers and workers exposed to asbestos. These results strongly suggest that high doses of β-carotene should not be recommended for any group until the safety of such doses can be established.
Collapse
|
14
|
Harvey WA, Jurgensen K, Pu X, Lamb CL, Cornell KA, Clark RJ, Klocke C, Mitchell KA. Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) increases human hepatic stellate cell activation. Toxicology 2016; 344-346:26-33. [PMID: 26860701 DOI: 10.1016/j.tox.2016.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 12/02/2015] [Accepted: 02/04/2016] [Indexed: 01/18/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a halogenated aromatic hydrocarbon that elicits toxicity through the aryl hydrocarbon receptor (AhR). In the liver, gross markers of TCDD toxicity are attributed to AhR activation in parenchymal hepatocytes. However, less is known regarding the consequences of TCDD treatment on non-parenchymal cells in the liver. Hepatic stellate cells (HSCs) are non-parenchymal cells that store vitamin A when quiescent. Upon liver injury, activated HSCs lose this storage ability and instead function in the development and maintenance of inflammation and fibrosis through the production of pro-inflammatory mediators and collagen type I. Reports that TCDD exposure disrupts hepatic retinoid homeostasis and dysregulates extracellular matrix remodeling in the liver led us to speculate that TCDD treatment may disrupt HSC activity. The human HSC line LX-2 was used to test the hypothesis that TCDD treatment directly activates HSCs. Results indicate that exposure to 10nM TCDD almost completely inhibited lipid droplet storage in LX-2 cells cultured with retinol and palmitic acid. TCDD treatment also increased LX-2 cell proliferation, expression of α-smooth muscle actin, and production of monocyte chemoattractant protein-1 (MCP-1), all of which are characteristics of activated HSCs. However, TCDD treatment had no effect on Col1a1 mRNA levels in LX-2 cells stimulated with the potent profibrogenic mediator, transforming growth factor-β. The TCDD-mediated increase in LX-2 cell proliferation, but not MCP-1 production, was abolished when phosphoinositide 3-kinase was inhibited. These results indicate that HSCs are susceptible to direct modulation by TCDD and that TCDD likely increases HSC activation through a multi-faceted mechanism.
Collapse
Affiliation(s)
- Wendy A Harvey
- Department of Biological Sciences, Boise State University, Boise, ID 83725, United States
| | - Kimberly Jurgensen
- Department of Biological Sciences, Boise State University, Boise, ID 83725, United States
| | - Xinzhu Pu
- Biomolecular Research Center, Boise State University, Boise, ID 83725, United States
| | - Cheri L Lamb
- Biomolecular Sciences Ph.D. Program, Boise State University, Boise, ID 83725, United States
| | - Kenneth A Cornell
- Biomolecular Research Center, Boise State University, Boise, ID 83725, United States; Biomolecular Sciences Ph.D. Program, Boise State University, Boise, ID 83725, United States; Department of Chemistry and Biochemistry, Boise State University, Boise, ID 83725, United States
| | - Reilly J Clark
- Department of Biological Sciences, Boise State University, Boise, ID 83725, United States
| | - Carolyn Klocke
- Department of Biological Sciences, Boise State University, Boise, ID 83725, United States
| | - Kristen A Mitchell
- Department of Biological Sciences, Boise State University, Boise, ID 83725, United States; Biomolecular Sciences Ph.D. Program, Boise State University, Boise, ID 83725, United States.
| |
Collapse
|
15
|
Abstract
It is well established that chylomicron remnant (dietary) vitamin A is taken up from the circulation by hepatocytes, but more than 80 % of the vitamin A in the liver is stored in hepatic stellate cells (HSC). It presently is not known how vitamin A is transferred from hepatocytes to HSCs for storage. Since retinol-binding protein 4 (RBP4), a protein that is required for mobilizing stored vitamin A, is synthesized solely by hepatocytes and not HSCs, it similarly is not known how vitamin A is transferred from HSCs to hepatocytes. Although it has long been thought that RBP4 is absolutely essential for delivering vitamin A to tissues, recent research has proven that this notion is incorrect since total RBP4-deficiency is not lethal. In addition to RBP4, vitamin A is also found in the circulation bound to lipoproteins and as retinoic acid bound to albumin. It is not known how these different circulating pools of vitamin A contribute to the vitamin A needs of different tissues. In our view, better insight into these three issues is required to better understand vitamin A absorption, storage and mobilization. Here, we provide an up to date synthesis of current knowledge regarding the intestinal uptake of dietary vitamin A, the storage of vitamin A within the liver, and the mobilization of hepatic vitamin A stores, and summarize areas where our understanding of these processes is incomplete.
Collapse
|
16
|
Mondul A, Mancina RM, Merlo A, Dongiovanni P, Rametta R, Montalcini T, Valenti L, Albanes D, Romeo S. PNPLA3 I148M Variant Influences Circulating Retinol in Adults with Nonalcoholic Fatty Liver Disease or Obesity. J Nutr 2015; 145:1687-91. [PMID: 26136587 PMCID: PMC4516767 DOI: 10.3945/jn.115.210633] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 06/04/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Retinol is a lipid-soluble essential nutrient that is stored as retinyl esters in lipid droplets of hepatic stellate cells. Patatin-like phospholipase domain-containing 3 (PNPLA3), through its retinyl-palmitate lipase activity, releases retinol from lipid droplets in hepatic stellate cells in vitro and ex vivo. We have shown that the genetic variant I148M (rs738409) reduces the PNPLA3 retinyl-palmitate lipase activity. OBJECTIVE The aim of the present genetic association study was to test whether overweight/obese carriers of the PNPLA3 148M mutant allele had lower circulating concentrations of retinol than individuals who are homozygous for the 148I allele. METHODS PNPLA3 I148M (rs738409) was genotyped by Taqman assay in 76 overweight/obese individuals [BMI (kg/m(2)) ≥25; mean ± SD age: 59.7 ± 11.4 y; male gender: 70%] with a histologic diagnosis of nonalcoholic fatty liver disease (NAFLD; namely the Milan NAFLD cohort) and in 413 obese men (BMI ≥30; mean ± SD age: 57.1 ± 4.9 y) from the α-Tocopherol, β-Carotene Cancer Prevention (ATBC) Study. Serum concentrations of retinol and α-tocopherol were measured by HPLC in both cohorts. β-Carotene concentrations in the ATBC study were measured by using HPLC. RESULTS The PNPLA3 148M mutant allele was associated with lower fasting circulating concentrations of retinol (β = -0.289, P = 0.03) in adults with NAFLD (Milan NAFLD cohort). The PNPLA3 148M mutant allele was also associated with lower fasting circulating concentrations of retinol in adults with a BMI ≥30 (ATBC study; β = -0.043, P = 0.04). CONCLUSION We showed for the first time, to our knowledge, that carriers of the PNPLA3 148M allele with either fatty liver plus obesity or obesity alone have lower fasting circulating retinol concentrations.
Collapse
Affiliation(s)
- Alison Mondul
- Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD
| | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Andrea Merlo
- Internal Medicine, Institution of Scientific Inpatient Care (istituto di ricovero e cura a carattere scientifico, IRCCS) Ca'-Granda Polyclinic Hospital, Milan, Italy;,Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Paola Dongiovanni
- Internal Medicine, Institution of Scientific Inpatient Care (istituto di ricovero e cura a carattere scientifico, IRCCS) Ca'-Granda Polyclinic Hospital, Milan, Italy;,Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Raffaela Rametta
- Internal Medicine, Institution of Scientific Inpatient Care (istituto di ricovero e cura a carattere scientifico, IRCCS) Ca'-Granda Polyclinic Hospital, Milan, Italy;,Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Tiziana Montalcini
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, University Magna Graecia of Catanzaro, Catanzaro, Italy; and
| | - Luca Valenti
- Internal Medicine, Institution of Scientific Inpatient Care (istituto di ricovero e cura a carattere scientifico, IRCCS) Ca'-Granda Polyclinic Hospital, Milan, Italy;,Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Demetrius Albanes
- Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Medical and Surgical Sciences, Clinical Nutrition Unit, University Magna Graecia of Catanzaro, Catanzaro, Italy; and Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
17
|
Pirazzi C, Valenti L, Motta BM, Pingitore P, Hedfalk K, Mancina RM, Burza MA, Indiveri C, Ferro Y, Montalcini T, Maglio C, Dongiovanni P, Fargion S, Rametta R, Pujia A, Andersson L, Ghosal S, Levin M, Wiklund O, Iacovino M, Borén J, Romeo S. PNPLA3 has retinyl-palmitate lipase activity in human hepatic stellate cells. Hum Mol Genet 2014; 23:4077-85. [PMID: 24670599 PMCID: PMC4082369 DOI: 10.1093/hmg/ddu121] [Citation(s) in RCA: 264] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Retinoids are micronutrients that are stored as retinyl esters in the retina and hepatic stellate cells (HSCs). HSCs are key players in fibrogenesis in chronic liver diseases. The enzyme responsible for hydrolysis and release of retinyl esters from HSCs is unknown and the relationship between retinoid metabolism and liver disease remains unclear. We hypothesize that the patatin-like phospholipase domain-containing 3 (PNPLA3) protein is involved in retinol metabolism in HSCs. We tested our hypothesis both in primary human HSCs and in a human cohort of subjects with non-alcoholic fatty liver disease (N = 146). Here we show that PNPLA3 is highly expressed in human HSCs. Its expression is regulated by retinol availability and insulin, and increased PNPLA3 expression results in reduced lipid droplet content. PNPLA3 promotes extracellular release of retinol from HSCs in response to insulin. We also show that purified wild-type PNPLA3 hydrolyzes retinyl palmitate into retinol and palmitic acid. Conversely, this enzymatic activity is markedly reduced with purified PNPLA3 148M, a common mutation robustly associated with liver fibrosis and hepatocellular carcinoma development. We also find the PNPLA3 I148M genotype to be an independent (P = 0.009 in a multivariate analysis) determinant of circulating retinol-binding protein 4, a reliable proxy for retinol levels in humans. This study identifies PNPLA3 as a lipase responsible for retinyl-palmitate hydrolysis in HSCs in humans. Importantly, this indicates a potential novel link between HSCs, retinoid metabolism and PNPLA3 in determining the susceptibility to chronic liver disease.
Collapse
Affiliation(s)
- Carlo Pirazzi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Benedetta Maria Motta
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Piero Pingitore
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden Department BEST (Biologia, Ecologia, Scienze Della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Kristina Hedfalk
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Rosellina Margherita Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and Department of Medical and Surgical Sciences, Clinical Nutrition Unit, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Maria Antonella Burza
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and
| | - Cesare Indiveri
- Department BEST (Biologia, Ecologia, Scienze Della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Yvelise Ferro
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Tiziana Montalcini
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Cristina Maglio
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and
| | - Paola Dongiovanni
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Silvia Fargion
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Raffaela Rametta
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Arturo Pujia
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Linda Andersson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and
| | - Saswati Ghosal
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and
| | - Malin Levin
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and
| | - Michelina Iacovino
- Department of Pediatrics, LA Biomedical Research Institute at Harbor-UCLA, 1124 W. Carson Street, HH1, Torrance, CA 90502, USA
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory and Department of Medical and Surgical Sciences, Clinical Nutrition Unit, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
18
|
Nagatsuma K, Hano H, Murakami K, Shindo D, Matsumoto Y, Mitobe J, Tanaka K, Saito M, Maehashi H, Owada M, Ikegami M, Tsubota A, Ohkusa T, Aizawa Y, Takagi I, Tajiri H, Matsuura T. Hepatic stellate cells that coexpress LRAT and CRBP-1 partially contribute to portal fibrogenesis in patients with human viral hepatitis. Liver Int 2014; 34:243-52. [PMID: 23890161 DOI: 10.1111/liv.12255] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 06/12/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Precisely what type of cells mainly contributes to portal fibrosis, especially in chronic viral hepatitis, such as hepatic stellate cells (HSCs) in the parenchyma or myofibroblasts in the portal area, still remains unclear. It is necessary to clarify the characteristics of cells that contribute to portal fibrosis in order to determine the mechanism of portal fibrogenesis and to develop a therapeutic target for portal fibrosis. This study was undertaken to examine whether LRAT+/CRBP-1+ HSCs contribute to portal fibrosis on viral hepatitis. METHODS Antibodies to lecithin:retinol acyltransferase (LRAT), cellular retinol-binding protein-1 (CRBP-1) and widely ascertained antibodies to HSCs (alpha-smooth muscle actin, neurotrophin-3) and endothelial cells (CD31) were used for immunohistochemical studies to assess the distribution of cells that contribute to the development of portal fibrosis with the aid of fluorescence microscopy. A quantitative analysis of LRAT+/CRBP-1+ HSCs was performed. RESULTS The number of LRAT+/CRBP-1+ HSCs was increased in fibrotic liver in comparison with normal liver in the portal area and fibrous septa. The number of double positive cells was less than 20% of all cells/field in maximum. CONCLUSION This study provides evidence that functional HSCs coexpressing both LRAT and CRBP-1 that continue to maintain the ability to store vitamin A contribute in part to the development of portal fibrogenesis in addition to parenchymal fibrogenesis in patients with viral hepatitis.
Collapse
Affiliation(s)
- Keisuke Nagatsuma
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan; Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Senoo H, Mezaki Y, Morii M, Hebiguchi T, Miura M, Imai K. Uptake and storage of vitamin A as lipid droplets in the cytoplasm of cells in the lamina propria mucosae of the rat intestine. Cell Biol Int 2013; 37:1171-80. [PMID: 23765517 DOI: 10.1002/cbin.10140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 06/06/2013] [Indexed: 12/30/2022]
Abstract
Vitamin A (retinyl palmitate) was injected subcutaneously or administered to rats by tube feeding. After subcutaneous injection, vitamin A was taken up and stored in cells of the lamina propria mucosae of the rat intestine. After oral administration, vitamin A was absorbed by the intestinal absorptive epithelial cells and transferred to cells of the lamina propria mucosae, where cells took up and stored the transferred vitamin A. The morphology of these cells was similar to that of hepatic stellate cells (also called vitamin A-storing cells, lipocytes, interstitial cells, fat-storing cells or Ito cells). Thus, these cells in the intestine could take up vitamin A from the systemic circulation and as well as by intestinal absorption, and store the vitamin in the lipid droplets in their cytoplasm. The data suggest that these cells are extrahepatic stellate cells of the digestive tract that may play roles in both the absorption and homeostasis of vitamin A.
Collapse
Affiliation(s)
- Haruki Senoo
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | | | | | | | | | | |
Collapse
|
20
|
Senoo H, Imai K, Mezaki Y, Miura M, Morii M, Fujiwara M, Blomhoff R. Accumulation of Vitamin A in the Hepatic Stellate Cell of Arctic Top Predators. Anat Rec (Hoboken) 2012; 295:1660-8. [DOI: 10.1002/ar.22555] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 04/11/2012] [Accepted: 06/14/2012] [Indexed: 11/12/2022]
|
21
|
Tacke F, Weiskirchen R. Update on hepatic stellate cells: pathogenic role in liver fibrosis and novel isolation techniques. Expert Rev Gastroenterol Hepatol 2012; 6:67-80. [PMID: 22149583 DOI: 10.1586/egh.11.92] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Hepatic stellate cells (HSCs), also called Ito cells or lipocytes, are vitamin A-storing cells located in the Dissé space between hepatocytes and sinusoidal endothelial cells. Upon liver injury, these cells transdifferentiate into extracellular matrix-producing, highly proliferative myofibroblasts that promote hepatic fibrogenesis. Other possible collagen-producing cells in liver fibrosis include portal fibroblasts, bone marrow-derived cells (mesenchymal stem cells, fibrocytes and hematopoietic cells) and parenchymal cells undergoing epithelial-to-mesenchymal transition. Important factors and signaling pathways for HSC activation, as well as different functions of HSC during homeostasis and fibrosis, such as collagen production, secretion of cytokines and chemokines, immune modulation and changes in contractile features, as well as vitamin A storage capacity, have been identified in vitro and in vivo. Novel isolation techniques, specifically HSC sorting by FACS via autofluorescence and antibodies, will provide us with further opportunities to advance our understanding of HSC biology in health and disease.
Collapse
Affiliation(s)
- Frank Tacke
- Department of Medicine III RWTH, University Hospital Aachen, Aachen, Germany.
| | | |
Collapse
|
22
|
Schreiber R, Taschler U, Preiss-Landl K, Wongsiriroj N, Zimmermann R, Lass A. Retinyl ester hydrolases and their roles in vitamin A homeostasis. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:113-23. [PMID: 21586336 PMCID: PMC3242165 DOI: 10.1016/j.bbalip.2011.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 04/28/2011] [Accepted: 05/02/2011] [Indexed: 12/16/2022]
Abstract
In mammals, dietary vitamin A intake is essential for the maintenance of adequate retinoid (vitamin A and metabolites) supply of tissues and organs. Retinoids are taken up from animal or plant sources and subsequently stored in form of hydrophobic, biologically inactive retinyl esters (REs). Accessibility of these REs in the intestine, the circulation, and their mobilization from intracellular lipid droplets depends on the hydrolytic action of RE hydrolases (REHs). In particular, the mobilization of hepatic RE stores requires REHs to maintain steady plasma retinol levels thereby assuring constant vitamin A supply in times of food deprivation or inadequate vitamin A intake. In this review, we focus on the roles of extracellular and intracellular REHs in vitamin A metabolism. Furthermore, we will discuss the tissue-specific function of REHs and highlight major gaps in the understanding of RE catabolism. This article is part of a Special Issue entitled Retinoid and Lipid Metabolism.
Collapse
|
23
|
|
24
|
Shmarakov I, Fleshman MK, D'Ambrosio DN, Piantedosi R, Riedl KM, Schwartz SJ, Curley RW, von Lintig J, Rubin LP, Harrison EH, Blaner WS. Hepatic stellate cells are an important cellular site for β-carotene conversion to retinoid. Arch Biochem Biophys 2010; 504:3-10. [PMID: 20470748 DOI: 10.1016/j.abb.2010.05.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/07/2010] [Accepted: 05/10/2010] [Indexed: 11/19/2022]
Abstract
Hepatic stellate cells (HSCs) are responsible for storing 90-95% of the retinoid present in the liver. These cells have been reported in the literature also to accumulate dietary β-carotene, but the ability of HSCs to metabolize β-carotene in situ has not been explored. To gain understanding of this, we investigated whether β-carotene-15,15'-monooxygenase (Bcmo1) and β-carotene-9',10'-monooxygenase (Bcmo2) are expressed in HSCs. Using primary HSCs and hepatocytes purified from wild type and Bcmo1-deficient mice, we establish that Bcmo1 is highly expressed in HSCs; whereas Bcmo2 is expressed primarily in hepatocytes. We also confirmed that HSCs are an important cellular site within the liver for accumulation of dietary β-carotene. Bcmo2 expression was found to be significantly elevated for livers and hepatocytes isolated from Bcmo1-deficient compared to wild type mice. This elevation in Bcmo2 expression was accompanied by a statistically significant increase in hepatic apo-12'-carotenal levels of Bcmo1-deficient mice. Although apo-10'-carotenal, like apo-12'-carotenal, was readily detectable in livers and serum from both wild type and Bcmo1-deficient mice, we were unable to detect either apo-8'- or apo-14'-carotenals in livers or serum from the two strains. We further observed that hepatic triglyceride levels were significantly elevated in livers of Bcmo1-deficient mice fed a β-carotene-containing diet compared to mice receiving no β-carotene. Collectively, our data establish that HSCs are an important cellular site for β-carotene accumulation and metabolism within the liver.
Collapse
Affiliation(s)
- Igor Shmarakov
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Affiliation(s)
- Theo J C van Berkel
- Division of Biopharmaceutics, LACDR, Leiden University, Gorlaeus Laboratory, P.O. Box 9502, Einsteinweg 55, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
26
|
Vinculin and cellular retinol-binding protein-1 are markers for quiescent and activated hepatic stellate cells in formalin-fixed paraffin embedded human liver. Histochem Cell Biol 2008; 131:313-25. [PMID: 19052772 DOI: 10.1007/s00418-008-0544-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2008] [Indexed: 12/15/2022]
Abstract
Hepatic stellate cells (HSCs) have important roles in the pathogenesis of liver fibrosis and cirrhosis. As response to chronic injury HSCs are activated and change from quiescent into myofibroblast-like cells. Several HSC-specific markers have been described in rat or mouse models. The aim of our work was to identify the best marker(s) for human HSCs. To this end we used the automated high throughput NexES IHC staining device (Ventana Medical Systems) to incubate sections under standardized conditions. Formalin fixed paraffin embedded (FFPE) normal and diseased human livers were studied. With immunohistochemistry we examined the expression of synemin, desmin, vimentin, vinculin, neurotrophin-3 (NT-3), alpha-smooth muscle actin (alpha-SMA), cellular retinol-binding protein-1 (CRBP-1), glial fibrillary acidic protein (GFAP), cysteine- and glycine-rich protein 2 (CRP2), and cytoglobin/stellate cell activation-associated protein (cygb/STAP). This is the first study in which a series of HSC markers is compared on serial FFPE human tissues. CRBP-1 clearly stains lobular HSCs without reacting with smooth muscle cells (SMCs) and shows variable cholangiocyte positivity. Vinculin has a similar staining pattern as CRBP-1 but additionally stains SMCs, and (myo)fibroblasts. In conclusion, we therefore propose to use CRBP-1 and/or vinculin to stain HSCs in human liver tissues.
Collapse
|
27
|
Blaner WS, O'Byrne SM, Wongsiriroj N, Kluwe J, D'Ambrosio DM, Jiang H, Schwabe RF, Hillman EMC, Piantedosi R, Libien J. Hepatic stellate cell lipid droplets: a specialized lipid droplet for retinoid storage. Biochim Biophys Acta Mol Cell Biol Lipids 2008; 1791:467-73. [PMID: 19071229 DOI: 10.1016/j.bbalip.2008.11.001] [Citation(s) in RCA: 309] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Revised: 11/05/2008] [Accepted: 11/12/2008] [Indexed: 01/08/2023]
Abstract
The majority of retinoid (vitamin A and its metabolites) present in the body of a healthy vertebrate is contained within lipid droplets present in the cytoplasm of hepatic stellate cells (HSCs). Two types of lipid droplets have been identified through histological analysis of HSCs within the liver: smaller droplets bounded by a unit membrane and larger membrane-free droplets. Dietary retinoid intake but not triglyceride intake markedly influences the number and size of HSC lipid droplets. The lipids present in rat HSC lipid droplets include retinyl ester, triglyceride, cholesteryl ester, cholesterol, phospholipids and free fatty acids. Retinyl ester and triglyceride are present at similar concentrations, and together these two classes of lipid account for approximately three-quarters of the total lipid in HSC lipid droplets. Both adipocyte-differentiation related protein and TIP47 have been identified by immunohistochemical analysis to be present in HSC lipid droplets. Lecithin:retinol acyltransferase (LRAT), an enzyme responsible for all retinyl ester synthesis within the liver, is required for HSC lipid droplet formation, since Lrat-deficient mice completely lack HSC lipid droplets. When HSCs become activated in response to hepatic injury, the lipid droplets and their retinoid contents are rapidly lost. Although loss of HSC lipid droplets is a hallmark of developing liver disease, it is not known whether this contributes to disease development or occurs simply as a consequence of disease progression. Collectively, the available information suggests that HSC lipid droplets are specialized organelles for hepatic retinoid storage and that loss of HSC lipid droplets may contribute to the development of hepatic disease.
Collapse
Affiliation(s)
- William S Blaner
- Department of Medicine, Columbia University, 630 W. 168th St., New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Croce AC, De Simone U, Vairetti M, Ferrigno A, Boncompagni E, Freitas I, Bottiroli G. Liver autofluorescence properties in animal model under altered nutritional conditions. Photochem Photobiol Sci 2008; 7:1046-53. [PMID: 18754051 DOI: 10.1039/b804836c] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Autofluorescence spectroscopy is a promising and powerful approach for an in vivo, real time characterization of liver functional properties. In this work, preliminary results on the dependence of liver autofluorescence parameters on the nutritional status are reported, with particular attention to vitamin A and lipid accumulation in liver tissue. Normally fed and 24 h starving rats were used as animal models. Histochemical and autofluorescence analysis showed that lipids and vitamin A colocalize in the liver parenchyma. Fasting condition results in a parallel increase in both lipids and vitamin A. Autofluorescence imaging and microspectrofluorometric analysis carried out on unfixed, unstained tissue sections under 366 nm excitation, evidenced differences in both spectral shape and response to continuous irradiation between liver biopsies from fed and starving rats. As to photobleaching, in particular, fitting analysis evidenced a reduction of about 85% of the signal attributable solely to vitamin A during the first 10 s of irradiation. The tissue whole emission measured in fed and starving rat livers exhibited reductions of about 35% and 52%, respectively, that are closely related to vitamin A contents. The findings open interesting perspectives for the set up of an in situ, real time diagnostic procedure for the assessment of liver lipid accumulation, exploiting the photophysical properties of vitamin A.
Collapse
Affiliation(s)
- Anna Cleta Croce
- IGM-CNR Histochemistry and Cytometry Section and Department of Animal Biology, University of Pavia, Piazza Botta, 10, 27100, Pavia, Italy
| | | | | | | | | | | | | |
Collapse
|
29
|
Mello T, Nakatsuka A, Fears S, Davis W, Tsukamoto H, Bosron WF, Sanghani SP. Expression of carboxylesterase and lipase genes in rat liver cell-types. Biochem Biophys Res Commun 2008; 374:460-4. [PMID: 18639528 DOI: 10.1016/j.bbrc.2008.07.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 07/08/2008] [Indexed: 11/24/2022]
Abstract
Approximately 80% of the body vitamin A is stored in liver stellate cells with in the lipid droplets as retinyl esters. In low vitamin A status or after liver injury, stellate cells are depleted of the stored retinyl esters by their hydrolysis to retinol. However, the identity of retinyl ester hydrolase(s) expressed in stellate cells is unknown. The expression of carboxylesterase and lipase genes in purified liver cell-types was investigated by real-time PCR. We found that six carboxylesterase and hepatic lipase genes were expressed in hepatocytes. Adipose triglyceride lipase was expressed in Kupffer cells, stellate cells and endothelial cells. Lipoprotein lipase expression was detected in Kupffer cells and stellate cells. As a function of stellate cell activation, expression of adipose triglyceride lipase decreased by twofold and lipoprotein lipase increased by 32-fold suggesting that it may play a role in retinol ester hydrolysis during stellate cell activation.
Collapse
Affiliation(s)
- Tommaso Mello
- Gastroenterology Unit, Department of Clinical Pathophysiology, University of Florence, Florence, Italy
| | | | | | | | | | | | | |
Collapse
|
30
|
Cifelli CJ, Green JB, Green MH. Use of model-based compartmental analysis to study vitamin A kinetics and metabolism. VITAMINS AND HORMONES 2008; 75:161-95. [PMID: 17368316 DOI: 10.1016/s0083-6729(06)75007-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We discuss the use of mathematical modeling, and specifically model-based compartmental analysis, to analyze vitamin A kinetic data obtained in rat and human studies over the past 25 years. Following an overview of whole-body vitamin A metabolism, a review of early kinetic studies, and an introduction to the approach and terminology of compartmental analysis, we summarize studies done in this laboratory to develop models of whole-body vitamin A metabolism in rats at varying levels of vitamin A status. Highlights of the results of these studies include the extensive recycling of vitamin A among plasma and tissues before irreversible utilization and the existence of significant extrahepatic pools of the vitamin. Our studies also document important differences in vitamin A kinetics as a function of vitamin A status and the importance of plasma retinol pool size in vitamin A utilization rate. Later we describe vitamin A kinetics and models developed for specific organs including the liver, eyes, kidneys, small intestine, lungs, testes, adrenals, and remaining carcass, and we discuss the effects of various exogenous factors (e.g., 4-HPR, dioxin, iron deficiency, dietary retinoic acid, and inflammation) on vitamin A dynamics. We also briefly review the retrospective application of model-based compartmental analysis to human vitamin A kinetic data. Overall, we conclude that the application of model-based compartmental analysis to vitamin A kinetic data provides unique insights into both quantitative and descriptive aspects of vitamin A metabolism and homeostasis in the intact animal.
Collapse
Affiliation(s)
- Christopher J Cifelli
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16801, USA
| | | | | |
Collapse
|
31
|
Liu L, Tang XH, Gudas LJ. Homeostasis of retinol in lecithin: retinol acyltransferase gene knockout mice fed a high retinol diet. Biochem Pharmacol 2008; 75:2316-24. [PMID: 18455147 DOI: 10.1016/j.bcp.2008.03.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 03/18/2008] [Accepted: 03/20/2008] [Indexed: 11/25/2022]
Abstract
We analyzed the retinoid levels and gene expression in various tissues after wild-type (Wt) and lecithin:retinol acyltransferase (LRAT-/-) knockout mice were fed a high retinol diet (250 IU/g). As compared to Wt, LRAT-/- mice exhibited a greater and faster increase in serum retinol concentration (mean+/-S.D., Wt, 1.3 +/- 0.2 microM to 1.5 +/- 0.3 microM in 48 h, p > 0.05; LRAT-/-, 1.3 +/- 0.2 microM to 2.2+/-0.3 microM in 48 h, p < 0.01) and a higher level of retinol in adipose tissue (17.2 +/- 2.4 pmol/mg in Wt vs. 34.6 +/- 8.0 pmol/mg in LRAT-/-). In the small intestines of Wt mice higher levels of retinol (96.4 +/- 13.0 pmol/mg in Wt vs. 13.7 +/- 7.6 pmol/mg in LRAT-/- and retinyl esters (2493.4 +/- 544.8 pmol/mg in Wt vs. 8.2 +/- 2.6 pmol/mg in LRAT-/- were detected. More retinol was detected in the feces of LRAT-/- mice (69.3 +/- 32.6 pmol/mg in LRAT-/- vs. 24.1 +/- 8.6 pmol/mg in Wt). LRAT mRNA levels increased in the lungs, small intestines, and livers of Wt mice on the high retinol diet, while CYP26A1 mRNA levels increased greatly only in the LRAT-/- mice. After 4 weeks, no significant differences between Wt mice and LRAT-/- mice were observed in either the serum retinol level or in the prevalence of Goblet cells in jejunal crypts. Our data indicate that the LRAT-/- mice maintain the homeostasis of retinol as the dietary retinol increases by increasing the excretion of retinol from the gastrointestinal tract, increasing the distribution of retinol to adipose tissue, and enhancing the catabolism by CYP26A1. We show that LRAT plays a role in maintaining a stable serum retinol concentration when dietary retinol concentration fluctuates.
Collapse
Affiliation(s)
- Limin Liu
- Department of Pharmacology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA
| | | | | |
Collapse
|
32
|
Yamaguchi K, Yang L, McCall S, Huang J, Yu XX, Pandey SK, Bhanot S, Monia BP, Li YX, Diehl AM. Diacylglycerol acyltranferase 1 anti-sense oligonucleotides reduce hepatic fibrosis in mice with nonalcoholic steatohepatitis. Hepatology 2008; 47:625-35. [PMID: 18000880 DOI: 10.1002/hep.21988] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
UNLABELLED Retinyl ester (RE) stores decrease during hepatic stellate cell (HSC) activation and liver fibrosis. Although retinol esterification is mostly catalyzed by lecithin:retinol acyltransferase (LRAT), diacylglycerol acyltransferase (DGAT)1 also does this. In previous reports, LRAT(-/-) mice had reduced hepatic RE but neither excessive HSC activation nor liver fibrosis, and DGAT1(-/-) mice had increased liver levels of RE and retinol. We sought to clarify the role of DGAT1 in liver fibrosis. Expression of DGAT1/2 was compared by real time PCR in freshly isolated, primary mouse HSCs and hepatocytes. To induce nonalcoholic steatohepatitis (NASH) and liver fibrosis, adult male db/db mice were fed methionine choline-deficient (MCD) diets. Half were treated with DGAT1 antisense oligonucleotide (ASO); the rest were injected with saline. Results were compared with chow-fed controls. Inhibition of DGAT1 in liver had no effect on hepatic triglyceride content or liver necroinflammation but reduced HSC activation and liver fibrosis in mice with NASH. To evaluate the role of DGAT1 in HSC activation, HSC were isolated from healthy rats treated with DGAT1 ASO or saline. DGAT1 was expressed at relatively high levels in HSCs. HSC isolated from DGAT1 ASO-treated rats had reduced DGAT1 expression and increased messenger RNA (mRNA) levels of LRAT and cellular retinol binding protein-1. During culture, they retained more vitamin A, had repressed collagen a2 (I) transcriptional activity, and expressed less collagen a1 (I) and a2 (I) mRNA. CONCLUSION DGAT1 may be a therapeutic target in NASH because inhibiting DGAT1 favorably altered. HSC retinoid homeostasis and inhibited hepatic fibrosis in mice with NASH.
Collapse
Affiliation(s)
- Kanji Yamaguchi
- Division of Gastroenterology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Guimarães ELM, Franceschi MFS, Andrade CMB, Guaragna RM, Borojevic R, Margis R, Bernard EA, Guma FCR. Hepatic stellate cell line modulates lipogenic transcription factors. Liver Int 2007; 27:1255-64. [PMID: 17919238 DOI: 10.1111/j.1478-3231.2007.01578.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND/AIMS Pre-adipocyte differentiation into adipocyte is a terminal differentiation process triggered by a cascade of transcription factors. Conversely, hepatic stellate cells (HSC) can switch between lipid storing and the myofibroblast phenotype in association with liver fibrotic processes. Here, adipogenic/lipogenic-related transcription factors and downstream-regulated genes were evaluated in a murine HSC cell line. GRX-HSC cells are transitional myofibroblasts that differentiate into lipocytes following retinol or indomethacin treatment. METHODS/RESULTS Specific mRNAs were quantified by a real-time polymerase chain reaction after 24 h or 7 days of cell culture with indomethacin or retinol. Proliferator-activated receptorgamma and Pex16 transcripts were increased either by retinol or indomethacin. Retinol induced a minor increase in C/enhancer binding proteinalpha transcripts, while only indomethacin increased adipsin transcripts. CONCLUSIONS Our results showed that the myofibroblast to lipocyte phenotype switch follows partially different transcriptional pathways, according to the effector. Retinol induces lipid synthesis and storage without affecting characteristic adipocytic genes, while indomethacin treatment restores the lipocytic phenotype with increased adipisin expression.
Collapse
|
34
|
Abstract
Vitamins A and E are essential, naturally occurring, fat-soluble nutrients that are involved in several important biological processes such as immunity, protection against tissue damage, reproduction, growth and development. They are extremely important during the early stages of life and must be transferred adequately to the young during gestation and lactation. The present article presents an overview of their biological functions, metabolism and dynamics of transfer to offspring in mammals. Among other topics, the review focuses on the biochemical aspects of their intestinal absorption, blood transport, tissue uptake, storage and catabolism. It also describes their different roles as well as their use as preventive and therapeutic agents. Finally, the mechanisms involved in their transfer during gestation and lactation are discussed.
Collapse
Affiliation(s)
- C Debier
- Institut des Sciences de la Vie, Unité de Biochimie de la Nutrition, Université catholique de Louvain, Croix du Sud 2/8, B-1348 Louvain-la-Neuve, Belgium.
| | | |
Collapse
|
35
|
Abstract
Retinoids (vitamin A) are crucial for most forms of life. In chordates, they have important roles in the developing nervous system and notochord and many other embryonic structures, as well as in maintenance of epithelial surfaces, immune competence, and reproduction. The ability of all-trans retinoic acid to regulate expression of several hundred genes through binding to nuclear transcription factors is believed to mediate most of these functions. The role of all-trans retinoic may extend beyond the regulation of gene transcription because a large number of noncoding RNAs also are regulated by retinoic acid. Additionally, extra-nuclear mechanisms of action of retinoids are also being identified. In organisms ranging from prokaryotes to humans, retinal is covalently linked to G protein-coupled transmembrane receptors called opsins. These receptors function as light-driven ion pumps, mediators of phototaxis, or photosensory pigments. In vertebrates phototransduction is initiated by a photochemical reaction where opsin-bound 11-cis-retinal is isomerized to all-trans-retinal. The photosensitive receptor is restored via the retinoid visual cycle. Multiple genes encoding components of this cycle have been identified and linked to many human retinal diseases. Central aspects of vitamin A absorption, enzymatic oxidation of all-trans retinol to all-trans retinal and all-trans retinoic acid, and esterification of all-trans retinol have been clarified. Furthermore, specific binding proteins are involved in several of these enzymatic processes as well as in delivery of all-trans retinoic acid to nuclear receptors. Thus, substantial progress has been made in our understanding of retinoid metabolism and function. This insight has improved our view of retinoids as critical molecules in vision, normal embryonic development, and in control of cellular growth, differentiation, and death throughout life.
Collapse
Affiliation(s)
- Rune Blomhoff
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | | |
Collapse
|
36
|
Sarem M, Znaidak R, Macías M, Rey R. [Hepatic stellate cells: it's role in normal and pathological conditions]. GASTROENTEROLOGIA Y HEPATOLOGIA 2006; 29:93-101. [PMID: 16448612 DOI: 10.1157/13083906] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hepatic fibrosis is a dynamic and sophisticatedly regulated wound healing response to chronic hepatocellular injury. This fibrotic process results from the accumulation of extracellular matrix (ECM) including collagen, proteoglycan, and adhesive glycoproteins which are principally produced by hepatic stellate cells (HSC), a mesenchymal cell type located between parenchymal cell plates and sinusoidal endothelial cells in the space of Disse. In physiological conditions, quiescent HSCs play important roles in the regulation of retinoid homeostasis and ECM remodeling by producing ECM components as well as metalloproteases and its inhibitor. However during hepatic fibrogenesis, HSCs are known to be activated or "transdifferentiated" to myofibroblast-like cells which play a pivotal role in ECM remodeling and hepatic blood flow regulation. Activation of HSC is now well established as the key process involved in the development of hepatic fibrosis. Both basic morphology and functions of HSCs in normal conditions and its role in pathological fibrosis will be discussed in this review.
Collapse
Affiliation(s)
- M Sarem
- Facultad de Medicina, Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
37
|
Schindler R, Fielenbach T, Rave G. A comparative study on the effects of oral amiodarone and trimeprazine, two in vitroretinyl ester hydrolase inhibitors, on the metabolic availability of vitamin A in rats. Br J Nutr 2005; 94:675-83. [PMID: 16277768 DOI: 10.1079/bjn20051495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Amiodarone, an antiarrhythmic drug, and trimeprazine, an antipsychotic drug, are bothin vitroinhibitors of retinyl ester hydrolase. To determine whether these agents have deleterious effects on aspects of vitamin A metabolism, Brown Norway rats (n18) were treated at clinically equivalent doses once daily for 26d with either oral drug. On day 27, a tolerance test was used to determine whether these agents interfered with vitamin absorption. During the first 8d, the plasma retinol level declined in all animals. Between days 12 and 27, it rose to near pre-treatment concentrations in the control and trimeprazine groups and remained relatively constant at low levels (P<0·001) in the amiodarone group. The intestinal absorption of vitamin A was reduced (P<0·05) in the amiodarone group compared with the placebo and trimeprazine groups, which did not differ significantly from each other. At the end of the 4-week treatment period, hepatic retinyl ester hydrolase activity was lower in the drug-dosed rats (P=0·06 for amiodarone) than in the controls. With regard to effects on liver reserves, drug treatment resulted in vitamin A depletion (P<0·019), and distinctive patterns of retinol and its esters were seen in response to dosing. In conclusion, amiodarone and trimeprazine have been shown to influence different aspects of retinoid metabolism, namely absorption, storage and transport. In clinical practice, the routine unmonitored use of these drugs and the suggestion that these agents be taken with meals are not recommended.
Collapse
Affiliation(s)
- Rainer Schindler
- Department of Human Nutrition and Food Science, Christian-Albrechts-University zu Kiel, D-24 116, Germany.
| | | | | |
Collapse
|
38
|
Natarajan SK, Thomas S, Ramachandran A, Pulimood AB, Balasubramanian KA. Retinoid metabolism during development of liver cirrhosis. Arch Biochem Biophys 2005; 443:93-100. [PMID: 16248980 DOI: 10.1016/j.abb.2005.09.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2005] [Revised: 08/30/2005] [Accepted: 09/03/2005] [Indexed: 11/29/2022]
Abstract
The changes in retinoid metabolism have been documented in liver cirrhosis. However, the dynamic alterations in levels of this vitamin between circulation and liver during development of the liver cirrhosis are not well understood. The aim of this study was to measure retinoids in the liver and circulation in parallel, during and after development of cirrhosis induced by carbon tetrachloride and thioacetamide. Retinoid levels were measured by HPLC. A decrease in retinaldehyde and total retinol, together with an increase in retinoic acid was evident in liver from both carbon tetrachloride or thioacetamide treated rats within a month after initiation of treatment. Activity of enzymes involved in retinoid metabolism such as retinaldehyde oxidase, retinaldehyde dehydrogenase, and retinaldehyde reductase were decreased in the liver. In parallel, levels of retinol and retinaldehyde in the serum were increased while retinoic acid was decreased. This study indicates that during development of cirrhosis, there is reciprocal transfer of retinoid metabolites between the circulation and the liver.
Collapse
Affiliation(s)
- Sathish Kumar Natarajan
- The Wellcome Trust Research Laboratory, Department of Gastrointestinal Sciences, Christian Medical College, Ida Scudder Road, Vellore 632004, India
| | | | | | | | | |
Collapse
|
39
|
Abstract
Mechanisms involved in the digestion and absorption of dietary vitamin A require the participation of several proteins. Dietary retinyl esters are hydrolyzed in the intestine by the pancreatic enzyme, pancreatic triglyceride lipase, and intestinal brush border enzyme, phospholipase B. Unesterified retinol taken up by the enterocyte is complexed with cellular retinol-binding protein type 2 and the complex serves as a substrate for reesterification of the retinol by the enzyme lecithin:retinol acyltransferase (LRAT). The retinyl esters are then incorporated into chylomicrons, intestinal lipoproteins containing other dietary lipids, such as triglycerides, phospholipids, and free and esterified cholesterol, and apolipoprotein B. Chylomicrons containing newly absorbed retinyl esters are then secreted into the lymph. Although under normal dietary conditions much of the dietary vitamin A is absorbed via the chylomicron/lymphatic route, it is also clear that under some circumstances there is substantial absorption of unesterified retinol via the portal route. Evidence supports the idea that the cellular uptake and efflux of unesterified retinol by enterocytes is mediated by lipid transporters, but the exact number, identity, and role of these proteins is not known and is an active area of research.
Collapse
Affiliation(s)
- Earl H Harrison
- Human Nutrition Research Center, United States Department of Agriculture, Beltsville, Maryland 20705, USA.
| |
Collapse
|
40
|
Kaschula CH, Jin MH, Desmond-Smith NS, Travis GH. Acyl CoA:retinol acyltransferase (ARAT) activity is present in bovine retinal pigment epithelium. Exp Eye Res 2005; 82:111-21. [PMID: 16054134 DOI: 10.1016/j.exer.2005.05.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2005] [Accepted: 05/27/2005] [Indexed: 11/16/2022]
Abstract
Visual perception is mediated by a family of G protein-coupled receptors called the opsins. The light-absorbing chromophore in most opsins is 11-cis-retinaldehyde, which is isomerized to all-trans-retinaldehyde upon absorption of a photon. Restoration of light sensitivity to the photobleached opsin requires chemical re-isomerization of the chromophore. This is carried out by an enzymatic pathway called the visual cycle in retinal pigment epithelial cells. The isomerase in this pathway uses fatty-acyl esters of all-trans-retinol as substrate. A retinyl-ester synthase that produces these esters, called lecithin:retinol acyltransferase (LRAT), has been extensively characterized. Based on prior biochemical studies and the phenotype in lrat(-/-) knockout mice, it has been assumed that LRAT is the sole or dominant retinyl-ester synthase in the retinal pigment epithelium. Here we demonstrate the presence of a second ester synthase activity in these cells called acyl CoA:retinol acyltransferase (ARAT). We show that this activity uses palmitoyl coenzyme A as an acyl donor, unlike LRAT which uses phosphatidylcholine. Similar to LRAT, ARAT esterifies both all-trans-retinol and 11-cis-retinol. LRAT and ARAT are both potently inhibited by the retinyl-ester analog, all-trans-retinylbromoacetate, but only ARAT is inhibited by progesterone. Unexpectedly, the maximum turnover rate (V(max)) of ARAT was similar to that of LRAT. However, the Michaelis constant (K(M)) of ARAT was 10-fold higher than the K(M) of LRAT for all-trans-retinol. These observations suggest that ARAT may complement LRAT to provide additional retinyl-ester synthase activity under conditions of high all-trans-retinol. These conditions occur in the retina following exposure to bright light.
Collapse
|
41
|
Yen CLE, Monetti M, Burri BJ, Farese RV. The triacylglycerol synthesis enzyme DGAT1 also catalyzes the synthesis of diacylglycerols, waxes, and retinyl esters. J Lipid Res 2005; 46:1502-11. [PMID: 15834126 DOI: 10.1194/jlr.m500036-jlr200] [Citation(s) in RCA: 203] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The final step of triacylglycerol biosynthesis is catalyzed by acyl CoA:diacylglycerol acyltransferase (DGAT) enzymes. The two known DGATs, DGAT1 and DGAT2, are encoded by unrelated genes. Although both DGAT1 and DGAT2 knockout mice have reduced tissue triacylglycerol contents, they have disparate phenotypes, prompting us to investigate whether the two enzymes have unrecognized functional differences. We now report that DGAT1 exhibits additional acyltransferase activities in vitro, including those of acyl CoA:monoacylglycerol acyltransferase (MGAT), wax monoester and wax diester synthases, and acyl CoA:retinol acyltransferase (ARAT), which catalyze the synthesis of diacylglycerols, wax esters, and retinyl esters, respectively. These activities were demonstrated in in vitro assays with membranes from insect cells or homogenates from COS7 cells overexpressing DGAT1. Wax synthase and ARAT activities were also demonstrated in intact COS7 cells expressing DGAT1. Additionally, cells and tissues from DGAT1-deficient mice exhibited reduced ARAT activity, and the mice had increased levels of unesterified retinol in their livers on a high-retinol diet. Our findings indicate that DGAT1 can utilize a variety of acyl acceptors as substrates in vitro and suggest that these activities may be relevant to the in vivo functions of DGAT1.
Collapse
Affiliation(s)
- Chi-Liang Eric Yen
- Gladstone Institute of Cardiovascular Disease, University of California, San Francisco, CA 94158, USA.
| | | | | | | |
Collapse
|
42
|
Schindler R, Fielenbach T, Rave G. Flupenthixol and cefotiam: effects on vitamin A metabolism in rats. Br J Nutr 2004; 92:597-605. [PMID: 15522128 DOI: 10.1079/bjn20041236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We examined the alterations in vitamin A metabolism as a result of flupenthixol or cefotiam administration. The impact of these drugs on indices of vitamin A status was evaluated in Brown Norway and Long-Evans rats. Intramuscular drug administration for 28 d resulted in a decline in systemic retinol. Changes in circulating retinol with time for chronic dosing showed drug treatment (P<0.001) and time (P<0.03) to be significant factors, but rat strain (P=0.33) was not a significant factor. Flupenthixol was the most active retinol-lowering compound (P<0.005). At the end of the 28 d period, hepatic retinyl ester hydrolase activity was greater in drug-treated rats than in controls (P<0.05). With regard to effects on liver reserves: (1) flupenthixol treatment resulted in vitamin A depletion (P<0.05); (2) cefotiam treatment stimulated vitamin A accumulation; (3) distinctive patterns of retinol and its esters were seen in response to treatment. It is reasonable to assume that the drugs interfere with vitamin A in at least two ways: (1) lowering of plasma retinol, an early event in the interaction, may be caused by inhibition of hepatic holo-retinol-binding protein secretion or stimulation of clearance, or both; (2) when plasma retinol levels are persistently low, and as the hepatic deposits of the xenobiotics build up, there are changes in the vitamin A pool size and composition of the liver. Candidate enzymes are retinyl ester hydrolase and cytochrome P450. The relationship between these two events will be studied in further detail.
Collapse
Affiliation(s)
- Rainer Schindler
- Department of Human Nutrition and Food Science, Christian-Albrechts-University, Kiel, Germany.
| | | | | |
Collapse
|
43
|
Lepreux S, Bioulac-Sage P, Gabbiani G, Sapin V, Housset C, Rosenbaum J, Balabaud C, Desmoulière A. Cellular retinol-binding protein-1 expression in normal and fibrotic/cirrhotic human liver: different patterns of expression in hepatic stellate cells and (myo)fibroblast subpopulations. J Hepatol 2004; 40:774-80. [PMID: 15094224 DOI: 10.1016/j.jhep.2004.01.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2003] [Revised: 12/16/2003] [Accepted: 01/08/2004] [Indexed: 01/11/2023]
Abstract
BACKGROUND/AIMS Cellular retinol-binding protein-1 (CRBP-1) which is involved in vitamin A metabolism is highly expressed in liver cells, particularly in hepatic stellate cells (HSCs). In this work, the CRBP-1 expression was studied by immunohistochemistry in the different liver cell populations, including HSCs and portal fibroblasts, of normal liver and of fibrotic and cirrhotic liver. METHODS Normal liver, fibrotic liver in different stages and cirrhotic liver sections were studied. Immunohistochemistry was performed using antibodies against CRBP-1, alpha-smooth muscle actin (SMA), CD 68 and CD 34. RESULTS In normal liver, quiescent HSCs expressed CRBP-1, while portal fibroblasts did not. In fibrotic or cirrhotic liver, activated HSCs co-expressed CRBP-1 and alpha-SMA; a variable proportion of portal and septal (myo)fibroblasts, more important in cirrhosis, neo-expressed both CRBP-1 and alpha-SMA. Biliary epithelial cells both in normal and pathological situations expressed CRBP-1. Neither Kupffer cells, nor endothelial cells showed CRBP-1 expression. CONCLUSIONS Our study demonstrates that CRBP-1 is a good marker to identify HSC in normal human liver. Furthermore, in fibrotic or cirrhotic liver, the different patterns of expression for CRBP-1 and alpha-SMA allow the distinction of different subsets of fibroblastic cells involved in fibrogenesis and septa formation.
Collapse
Affiliation(s)
- Sébastien Lepreux
- Department of Pathology, Centre Médical Universitaire, Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Shibata N, Watanabe T, Okitsu T, Sakaguchi M, Takesue M, Kunieda T, Omoto K, Yamamoto S, Tanaka N, Kobayashi N. Establishment of an immortalized human hepatic stellate cell line to develop antifibrotic therapies. Cell Transplant 2003; 12:499-507. [PMID: 12953924 DOI: 10.3727/000000003108747064] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Because human hepatic stellate cells (HSCs) perform a crucial role in the progress of hepatic fibrosis, it is of great value to establish an immortalized human cell line that exhibits HSC characteristics and grows well in tissue cultures for the development of antifibrotic therapies. Thus, we engineered an immortalized human hepatic stellate cell (HSC) line TWNT-4 by retrovirally inducing human telomerase reverse transcriptase (hTERT) into LI 90 cells established from a human liver mesenchymal tumor. Parental LI 90 entered replicative senescence, whereas TWNT-4 showed telomerase activity and proliferated for more than population doubling level (PDL) 200 without any crisis. TWNT-4 expressed platelet-derived growth factor-beta receptor (PDGF-betaR), alpha-smooth muscle actin (alpha-SMA), and type I collagen (alpha1) and was considered to be an activated form of HSCs. Treatment of TWNT-4 cells with either 100 U/ml of IFN-gamma or 1 ng/ml of rapamycin (Rapa) for 14 days led to lower expression of type I collagen (alpha1) at RNA and protein levels. Exposure of TWNT-4 cells to both of IFN-gamma (10 U/ml) and Rapa (0.1 ng/ml) for 14 days effectively decreased the expression of type I collagen (alpha1), PDGF-betaR, and alpha-SMA expression and suppressed TGF-beta1 secretion of TWNT-4 cells. We successfully induced apoptosis by transducing TNF-related apoptosis-inducing ligand (TRAIL) into TWNT-4 cells using adenovirus vectors Ad/GT-TRAIL and Ad/PGK-GV-17. These findings suggested that immortalized activated HSC line TWNT-4 would be a useful means to develop antifibrotic therapies.
Collapse
Affiliation(s)
- Norikuni Shibata
- Division of Gastroenterology, Department of Internal Medicine, Kawasaki Medical School, 577 Matsushima, Kurashiki 701-0114, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Fortuna VA, Martucci RB, Trugo LC, Borojevic R. Hepatic stellate cells uptake of retinol associated with retinol-binding protein or with bovine serum albumin. J Cell Biochem 2003; 90:792-805. [PMID: 14587034 DOI: 10.1002/jcb.10703] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Retinol is stored in liver, and the dynamic balance between its accumulation and mobilization is regulated by hepatic stellate cells (HSC). Representing less than 1% total liver protein, HSC can reach a very high intracellular retinoid (vitamin-A and its metabolites) concentration, which elicits their conversion from the myofibroblast to the fat-storing lipocyte phenotype. Circulating retinol is associated with plasma retinol-binding protein (RBP) or bovine serum albumin (BSA). Here we have used the in vitro model of GRX cells to compare incorporation and metabolism of BSA versus RBP associated [(3)H]retinol in HSC. We have found that lipocytes, but not myofibroblasts, expressed a high-affinity membrane receptor for RBP-retinol complex (KD = 4.93 nM), and both cell types expressed a low-affinity one (KD = 234 nM). The RBP-retinol complex, but not the BSA-delivered retinol, could be dislodged from membranes by treatments that specifically disturb protein-protein interactions (high RBP concentrations). Under both conditions, treatments that disturb the membrane lipid layer (detergent, cyclodextrin) released the membrane-bound retinol. RBP-delivered retinol was found in cytosol, microsomal fraction and, as retinyl esters, in lipid droplets, while albumin-delivered retinol was mainly associated with membranes. Disturbing the clathrin-mediated endocytosis did not interfere with retinol uptake. Retinol derived from the holo-RBP complex was differentially incorporated in lipocytes and preferentially reached esterification sites close to lipid droplets through a specific intracellular traffic route. This direct influx pathway facilitates the retinol uptake into HSC against the concentration gradients, and possibly protects cell membranes from undesirable and potentially noxious high retinol concentrations.
Collapse
Affiliation(s)
- Vitor A Fortuna
- Departamento de Histologia e Embriologia, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-970 Cidade Universitária, Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
46
|
Higashi N, Senoo H. Distribution of vitamin A-storing lipid droplets in hepatic stellate cells in liver lobules--a comparative study. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2003; 271:240-8. [PMID: 12552640 DOI: 10.1002/ar.a.10036] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
To investigate the storage mechanisms of vitamin A, we examined the liver of adult polar bears and arctic foxes, which physiologically store a large amount of vitamin A, by high-performance liquid chromatography (HPLC), transmission electron microscopy (TEM) morphometry, gold chloride staining, fluorescence microscopy for the detection of autofluorescence of vitamin A, staining with hematoxylin-eosin (H&E), Masson's trichrome, and Ishii and Ishii's silver impregnation. HPLC revealed that the polar bears and arctic foxes contained 1.8-1.9 x 10(4) nmol total retinol (retinol plus retinyl esters) per gram liver. In the arctic foxes, the composition of the retinyl esters was found to be 51.1% palmitate, 26.6% oleate, 15.4% stearate, and 7% linoleate. The hepatic stellate cells of the arctic animals were demonstrated by TEM to contain the bulk of the vitamin A-lipid droplets in their cytoplasm. The liver lobules of the arctic animals showed a zonal gradient in the storage of vitamin A. The gradient was expressed as a symmetric crescendo-decrescendo profile starting at the periportal zone, peaking at the middle zone, and sloping down toward the central zone in the liver lobule. The density (i.e., cell number per area) of hepatic stellate cells was essentially the same among the zones. The gradient and the composition of the retinyl esters in storing vitamin A were not changed by differences in the vitamin A amount in the livers. These results indicate that the heterogeneity of vitamin A-storage capacity in hepatic stellate cells of arctic foxes and polar bears is genetically determined.
Collapse
Affiliation(s)
- Nobuyo Higashi
- Department of Anatomy, Akita University School of Medicine, Akita, Japan
| | | |
Collapse
|
47
|
Hellemans K, Rombouts K, Quartier E, Dittié AS, Knorr A, Michalik L, Rogiers V, Schuit F, Wahli W, Geerts A. PPARbeta regulates vitamin A metabolism-related gene expression in hepatic stellate cells undergoing activation. J Lipid Res 2003; 44:280-95. [PMID: 12576510 DOI: 10.1194/jlr.m200376-jlr200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Activation of cultured hepatic stellate cells correlated with an enhanced expression of proteins involved in uptake and storage of fatty acids (FA translocase CD36, Acyl-CoA synthetase 2) and retinol (cellular retinol binding protein type I, CRBP-I; lecithin:retinol acyltransferases, LRAT). The increased expression of CRBP-I and LRAT during hepatic stellate cells activation, both involved in retinol esterification, was in contrast with the simultaneous depletion of their typical lipid-vitamin A (vitA) reserves. Since hepatic stellate cells express high levels of peroxisome proliferator activated receptor beta (PPARbeta), which become further induced during transition into the activated phenotype, we investigated the potential role of PPARbeta in the regulation of these changes. Administration of L165041, a PPARbeta-specific agonist, further induced the expression of CD36, B-FABP, CRBP-I, and LRAT, whereas their expression was inhibited by antisense PPARbeta mRNA. PPARbeta-RXR dimers bound to CRBP-I promoter sequences. Our observations suggest that PPARbeta regulates the expression of these genes, and thus could play an important role in vitA storage. In vivo, we observed a striking association between the enhanced expression of PPARbeta and CRBP-I in activated myofibroblast-like hepatic stellate cells and the manifestation of vitA autofluorescent droplets in the fibrotic septa after injury with CCl4 or CCl4 in combination with retinol.
Collapse
Affiliation(s)
- Karine Hellemans
- Laboratory of Molecular Liver Cell Biology, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Casu A, Bassi AM, Canepa C, Maloberti G, Nanni G. Thioacetamide impairs retinol storage and dolichol content in rat liver cells in vivo. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1583:266-72. [PMID: 12176393 DOI: 10.1016/s1388-1981(02)00251-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The aim of this paper was to ascertain whether chronic pretreatment with thioacetamide (TAA) might alter the uptake of a load of retinol and dolichol distribution in hepatocytes (HC), hepatic stellate cells (HSC) (Ito-1 and Ito-2 subfractions), Kupffer (KC) and sinusoidal endothelial cells (SEC). The reason why retinol and dolichol content was studied is that their metabolism and transport might be interrelated and that the two isoprenoids might exert different functions in the cells of the hepatic sinusoid. Rats were treated for 2 and 4 months with TAA, a known fibrogenic hepatotoxin, at a low dosage, to produce an early stage of damage. Three days before sacrifice, the rats were given a load of vitamin A, and cells were isolated to investigate its uptake. In HC, the load of retinol was taken up and accumulated, while a decrease in dolichol preceded retinol increase. In HSC, much less of the retinol load was stored than in controls, and dolichol content also decreased. Various minor modifications were seen in KC and SEC.Collectively, the results show that the distribution of these two isoprenoids, which play important roles in cellular differentiation and proliferation, is differently altered in the multiple cell types that line the hepatic sinusoid, and that both isoprenoids seem to participate in the first steps of liver damage.
Collapse
Affiliation(s)
- A Casu
- Department of Experimental Medicine, Section of General Pathology, University of Genoa, Via L.B. Alberti 2, 16132 Genoa, Italy
| | | | | | | | | |
Collapse
|
49
|
Sauvant P, Sapin V, Abergel A, Schmidt CK, Blanchon L, Alexandre-Gouabau MC, Rosenbaum J, Bommelaer G, Rock E, Dastugue B, Nau H, Azaïs-Braesco V. PAV-1, a new rat hepatic stellate cell line converts retinol into retinoic acid, a process altered by ethanol. Int J Biochem Cell Biol 2002; 34:1017-29. [PMID: 12007639 DOI: 10.1016/s1357-2725(02)00023-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
During liver fibrogenesis or long term culture, hepatic stellate cells (HSCs) evolved from "quiescent" to activated phenotype called "myofibroblast-like", a transition prevented by retinoic acid (RA). Little is known about RA generation by HSCs. Our study aimed to check the ability of these cells to produce RA from retinol (Rol) and the alterations of this metabolic step by ethanol. To study this metabolic pathway, primary cultures of HSCs represent the most physiological model but technically suffer several drawbacks. To circumvent these problems, an immortalized rat HSC line (named PAV-1) has been established. We validated PAV-1 cell line as a convenient model to study retinoids metabolism by HSCs. Then, we showed that PAV-1 cells express Rol-binding proteins (RBPs), enzymes and nuclear receptors involved in RA signaling pathway. We also demonstrated in situ generation of functional all-trans-RA (ATRA), using transient transfections with a RA-sensitive reporter gene, in situ modulation of tissue transglutaminase (tTG) activity and HPLC experiments. This production was Rol dose-dependent; 4-methylpyrazole, citral, and ethanol-inhibited which argues in favor of an enzymatic process.In conclusion, we first demonstrate in situ RA generation from Rol in a newly immortalized rat HSC line, named PAV-1. Inhibition of RA production by ethanol in PAV-1 and recent data, suggesting fundamental role of RA to prevent fibrosis development in the liver, allow us to hypothesize that Rol metabolism could be a primary target for ethanol during development of hepatic fibrosis.
Collapse
Affiliation(s)
- Patrick Sauvant
- INRA-Unité des Maladies Métaboliques et Micronutriments; Equipe Vitamines, -Theix, 63122 Saint Genès Champanelle, Clermont-Ferrand, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tokairin T, Nishikawa Y, Doi Y, Watanabe H, Yoshioka T, Su M, Omori Y, Enomoto K. A highly specific isolation of rat sinusoidal endothelial cells by the immunomagnetic bead method using SE-1 monoclonal antibody. J Hepatol 2002; 36:725-33. [PMID: 12044521 DOI: 10.1016/s0168-8278(02)00048-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND/AIMS To develop a specific isolation method of hepatic sinusoidal endothelial cells (SEC), we applied the immunomagnetic method using a monoclonal antibody (SE-1) that recognizes a membranous antigen expressed only in rat SEC. METHODS Cells were isolated by incubating mixed non-parenchymal cells, which were obtained by collagenase digestion of the liver, with SE-1-conjugated superparamagnetic polystyrene beads. The conventional Percoll method was also performed in parallel to compare with the immunomagnetic method. The isolated cells were cultured on glass coverslips coated with type I collagen in the presence of various growth factors for 6 days. RESULTS Approximately 98% of the isolated cells were positive for SE-1 and the contamination of Kupffer cells or stellate cells was less than 1%. The purity was significantly better than that obtained by the Percoll method. The cultured cells showed typical SEC features, such as sieve plates and uptake of acetylated low-density lipoprotein. Although the cells continuously underwent apoptotic cell death after 2 days, they started robust cell growth after 3 days and were well maintained during the culture period. CONCLUSIONS Our simple and specific isolation method enables us to culture SEC with high purity and should be useful for the biological analysis of SEC.
Collapse
Affiliation(s)
- Takuo Tokairin
- Department of Pathology, Akita University School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | | | | | | | | | | | | | | |
Collapse
|