Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Lipids Health Dis ; 18(1): 172, 2019 Sep 14.
Article in English | MEDLINE | ID: mdl-31521175

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH) is a common disease and feeding mice a methionine-choline-deficient (MCD) diet is a frequently used model to study its pathophysiology. Genetic and environmental factors influence NASH development and liver lipid content, which was studied herein using C57BL/6 J mice bred in two different animal facilities. METHODS: Age-matched male C57BL/6 J mice bred in two different animal facilities (later on referred to as WT1 and WT2) at the University Hospital of Regensburg were fed identical MCD or control chows for 2 weeks. Hepatic gene and protein expression and lipid composition were determined. RESULTS: NASH was associated with increased hepatic triglycerides, which were actually higher in WT1 than WT2 liver in both dietary groups. Cholesterol contributes to hepatic injury but was only elevated in WT2 NASH liver. Ceramides account for insulin resistance and cell death, and ceramide species d18:1/16:0 and d18:1/18:0 were higher in the NASH liver of both groups. Saturated sphingomyelins only declined in WT1 NASH liver. Lysophosphatidylcholine concentrations were quite normal in NASH and only one of the 12 altered phosphatidylcholine species declined in NASH liver of both groups. Very few phosphatidylethanolamine, phosphatidylserine, and phosphatidylinositol species were comparably regulated in NASH liver of both animal groups. Seven of these lipid species declined and two increased in NASH. Notably, hepatic mRNA expression of proinflammatory (F4/80, CD68, IL-6, TNF and chemerin) and profibrotic genes (TGF beta and alpha SMA) was comparable in WT1 and WT2 mice. CONCLUSIONS: Mice housed and bred in different animal facilities had comparable disease severity of NASH whereas liver lipids varied among the groups. Thus, there was no specific lipid signature for NASH in the MCD model.


Subject(s)
Animal Experimentation/standards , Choline Deficiency/metabolism , Liver/metabolism , Methionine/deficiency , Non-alcoholic Fatty Liver Disease/metabolism , Actins/genetics , Actins/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Ceramides/metabolism , Cholesterol/metabolism , Choline Deficiency/etiology , Choline Deficiency/genetics , Diet/adverse effects , Disease Models, Animal , Gene Expression Regulation , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/pathology , Lysophosphatidylcholines/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Phosphatidylethanolamines/metabolism , Phosphatidylinositols/metabolism , Phosphatidylserines/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sphingomyelins/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Triglycerides/metabolism
2.
Biofactors ; 44(3): 289-298, 2018 May.
Article in English | MEDLINE | ID: mdl-29672963

ABSTRACT

One of the worldwide metabolic health dilemma is nonalcoholic fatty liver diseases (NAFLD). Researchers are searching effective drug to manage NAFLD patients. One of the best way to manage the metabolic imperfection is through natural principal isolated from different sources. Butein, a natural compound known to have numerous pharmacological application. In the current study we assessed the therapeutic effect of butein administration on liver function tests, oxidative stress, antioxidants, lipid abnormalities, serum inflammatory cytokines, and mitochondrial reactive oxygen species levels, in rats with methionine-choline deficient (MCD) diet induced NAFLD. Male Wistar rats were treated with MCD diet with/without butein (200 mg/kg body wt. orally) for 6 weeks. The protective effect of butein, were evident from decreased transaminase activities, restoration of albumin, globulin, albumin/globulin ratio, and oxidants in serum (P < 0.01), further it improved liver antioxidant status (P < 0.01). Butein significantly lowered lipid profile parameters (P < 0.01), suppressed inflammatory cytokines (P < 0.01), and improved liver histology. Further to understand the possible mechanism behind the hepatoprotective and lipid lowering effect of butein, the activities of heme oxygenase (HO1), myeloperoxidase (MPO), and mitochondrial reactive oxygen species (ROS) were measured. We found that butein supplementation significantly decreased the activity of HO1 (P < 0.001), and increased the activity of MPO (P < 0.001). Furthermore butein attenuated mitochondrial ROS produced in NAFLD condition. Present study shows that butein supplementation restore liver function by altering liver oxidative stress, inflammatory markers, vital defensive enzyme activities, and mitochondrial ROS. In summary, butein has remarkable potential to develop effective hepato-protective drug. © 2018 BioFactors, 44(3):289-298, 2018.


Subject(s)
Chalcones/pharmacology , Choline Deficiency/drug therapy , Diet/adverse effects , Hypolipidemic Agents/pharmacology , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Administration, Oral , Albumins/metabolism , Animals , Choline/metabolism , Choline Deficiency/etiology , Choline Deficiency/metabolism , Choline Deficiency/pathology , Globulins/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Liver/metabolism , Liver/pathology , Male , Methionine/deficiency , Mitochondria/drug effects , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Peroxidase/antagonists & inhibitors , Peroxidase/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Transaminases/antagonists & inhibitors , Transaminases/metabolism
3.
J Vis Exp ; (128)2017 10 21.
Article in English | MEDLINE | ID: mdl-29155718

ABSTRACT

Chronic liver diseases, such as viral hepatitis, alcoholic liver disease, or non-alcoholic fatty liver disease, are characterized by continual inflammation, progressive destruction and regeneration of the hepatic parenchyma, liver progenitor cell proliferation, and fibrosis. The end-stage of every chronic liver disease is cirrhosis, a major risk factor for the development of hepatocellular carcinoma. To study processes regulating disease initiation, establishment, and progression, several animal models are used in laboratories. Here we describe a six-week time course of the choline-deficient and ethionine-supplemented (CDE) mouse model, which involves feeding six-week old male C57BL/6J mice with choline-deficient chow and 0.15% DL-ethionine-supplemented drinking water. Monitoring of animal health and a typical body weight loss curve are explained. The protocol demonstrates the gross examination of a CDE-treated liver and blood collection by cardiac puncture for subsequent serum analyses. Next, the liver perfusion technique and collection of different hepatic lobes for standard evaluations are shown, including liver histology assessments by hematoxylin and eosin or Sirius Red stainings, immunofluorescent detection of hepatic cell populations as well as transcriptome profiling of the liver microenvironment. This mouse model is suitable for studying inflammatory, fibrogenic, and liver progenitor cell dynamics induced through chronic liver disease and can be used to test potential therapeutic agents that may modulate these processes.


Subject(s)
Choline Deficiency/etiology , Disease Models, Animal , Ethionine/administration & dosage , Lung Injury/etiology , Animals , Cell Proliferation/physiology , Choline Deficiency/metabolism , Diet , Dietary Supplements , Liver/pathology , Lung Injury/metabolism , Lung Injury/pathology , Male , Mice , Mice, Inbred C57BL
4.
Biochem Biophys Res Commun ; 486(2): 264-269, 2017 04 29.
Article in English | MEDLINE | ID: mdl-28286271

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is characterized by excess accumulation of lipids in liver, accompanied with hepatocyte injury, cell death and inflammation. Although p16 is known as tumor suppressor in multiple cancer types, it remains unclear whether p16 plays a critical role in NASH. To determine whether p16 could play a role in the pathogenesis of NASH, wild-type mice and p16-/- mice were fed on a methionine and choline-deficient (MCD) diet for 3 weeks, and liver steatosis, fibrosis, and inflammation were evaluated. Our data show that p16-/- mice fed with MCD diet displayed more significant hepatic steatosis, hepatocyte damage, increased oxidative stress and inflammatory cell infiltration compared to MCD-fed WT mice. It was also clear that the increased ROS and the accumulation of lipid in BEL-7402 cells occurred when p16 expression was depleted with siRNA. These findings indicate that p16 may play a critical role in the development of NASH by reining in ROS production and by inhabiting inflammatory response.


Subject(s)
Choline Deficiency/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Hepatocytes/metabolism , Liver Cirrhosis/genetics , Non-alcoholic Fatty Liver Disease/genetics , Animals , Cell Line, Tumor , Choline Deficiency/etiology , Choline Deficiency/metabolism , Choline Deficiency/pathology , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p18/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Disease Models, Animal , Food, Formulated/adverse effects , Gene Expression Regulation , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Methionine/deficiency , Mice , Mice, Inbred BALB C , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
5.
Biochem Biophys Res Commun ; 486(3): 632-638, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28322783

ABSTRACT

Endoplasmic reticulum (ER) stress caused by accumulation of misfolded proteins is observed in several kinds of diseases. Since ER stress is reported to be involved in the progression of non-alcoholic steatohepatitis (NASH), highly sensitive and simple measurement methods are required for research into developing novel therapy for NASH. To investigate the involvement of ER stress in NASH pathogenesis in a mouse model, an assay for liver ER stress was developed using ER stress activated indicator-luciferase (ERAI-Luc) mice. To establish the assay method for detection of ER stress in the liver, tunicamycin (TM) (0.3 mg/kg i. p.) was administered to ERAI-Luc mice, and the luciferase activity was measured in ex vivo and in vivo. To evaluate ER stress in the NASH model, ERAI-Luc mice were fed a modified choline-deficient l-amino acid-defined (mCDAA) diet for 14 weeks. After measurement of ER stress by luminescence imaging, levels of liver lipids and pro-fibrotic and pro-inflammatory gene expression were measured as NASH-related indexes. In non-invasive whole-body imaging, TM elevated luciferase activity in the liver, induced by activation of ER stress. The highest luminescence in the liver was confirmed by ex vivo imaging of isolated tissues. In parallel with progression of NASH, elevated luminescence induced by ER stress in liver was observed in mCDAA diet-fed ERAI-Luc mice. Luciferase activity was significantly and positively correlated to levels of triglyceride and free cholesterol in the liver, as well as to the mRNA expression of type 1 collagen α1 chain and tumor necrosis factor α. These data indicated that the use of ERAI-Luc mice was effective in the detection of ER stress in the liver. Moreover, the NASH model using ERAI-Luc mice can be a useful tool to clarify the role of ER stress in pathogenesis of NASH and to evaluate effects of drugs targeted against ER stress.


Subject(s)
Choline Deficiency/genetics , Collagen Type I/genetics , Endoplasmic Reticulum Stress/genetics , Food, Formulated/adverse effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Biological Assay , Cholesterol/metabolism , Choline Deficiency/etiology , Choline Deficiency/metabolism , Choline Deficiency/pathology , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Gene Expression Regulation , Genes, Reporter , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/pathology , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tunicamycin/pharmacology
6.
J Nutr Biochem ; 35: 81-86, 2016 09.
Article in English | MEDLINE | ID: mdl-27469995

ABSTRACT

Methionine is required for protein synthesis and provides a methyl group for >50 critical transmethylation reactions including creatine and phosphatidylcholine synthesis as well as DNA and protein methylation. However, the availability of methionine depends on dietary sources as well as remethylation of demethylated methionine (i.e., homocysteine) by the dietary methyl donors folate and choline (via betaine). By restricting dietary methyl supply, we aimed to determine the extent that dietary methyl donors contribute to methionine availability for protein synthesis and transmethylation reactions in neonatal piglets. Piglets 4-8 days of age were fed a diet deficient (MD-) (n=8) or sufficient (MS+) (n=7) in folate, choline and betaine. After 5 days, dietary methionine was reduced to 80% of requirement in both groups to elicit a response. On day 8, animals were fed [(3)H-methyl]methionine for 6h to measure methionine partitioning into hepatic protein, phosphatidylcholine, creatine and DNA. MD- feeding reduced plasma choline, betaine and folate (P<.05) and increased homocysteine ~3-fold (P<.05). With MD- feeding, hepatic phosphatidylcholine synthesis was 60% higher (P<.05) at the expense of creatine synthesis, which was 30% lower during MD- feeding (P<.05); protein synthesis as well as DNA and protein methylation were unchanged. In the liver, ~30% of dietary label was traced to phosphatidylcholine and creatine together, with ~50% traced to methylation of proteins and ~20% incorporated in synthesized protein. Dietary methyl donors are integral to neonatal methionine requirements and can affect methionine availability for transmethylation pathways.


Subject(s)
Creatine/metabolism , Diet/adverse effects , Hyperhomocysteinemia/etiology , Liver/metabolism , Methionine/metabolism , Phosphatidylcholines/metabolism , Animals , Animals, Newborn , Betaine/administration & dosage , Choline Deficiency/blood , Choline Deficiency/etiology , Choline Deficiency/metabolism , Choline Deficiency/physiopathology , Female , Folic Acid Deficiency/blood , Folic Acid Deficiency/etiology , Folic Acid Deficiency/metabolism , Folic Acid Deficiency/physiopathology , Homocysteine/blood , Homocysteine/metabolism , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/metabolism , Male , Methylation , Protein Biosynthesis , Protein Processing, Post-Translational , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Swine , Swine, Miniature , Tritium
7.
Nat Commun ; 7: 11869, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27323669

ABSTRACT

Receptor-interacting protein kinase 3 (RIPK3) mediates necroptosis, a form of programmed cell death that promotes inflammation in various pathological conditions, suggesting that it might be a privileged pharmacological target. However, its function in glucose homeostasis and obesity has been unknown. Here we show that RIPK3 is over expressed in the white adipose tissue (WAT) of obese mice fed with a choline-deficient high-fat diet. Genetic inactivation of Ripk3 promotes increased Caspase-8-dependent adipocyte apoptosis and WAT inflammation, associated with impaired insulin signalling in WAT as the basis for glucose intolerance. Similarly to mice, in visceral WAT of obese humans, RIPK3 is overexpressed and correlates with the body mass index and metabolic serum markers. Together, these findings provide evidence that RIPK3 in WAT maintains tissue homeostasis and suppresses inflammation and adipocyte apoptosis, suggesting that systemic targeting of necroptosis might be associated with the risk of promoting insulin resistance in obese patients.


Subject(s)
Adipose Tissue, White/enzymology , Choline Deficiency/genetics , Glucose Intolerance/genetics , Intra-Abdominal Fat/enzymology , Necrosis/enzymology , Obesity/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Adipocytes/enzymology , Adipocytes/pathology , Adipose Tissue, White/pathology , Animals , Apoptosis/genetics , Body Mass Index , Caspase 8/genetics , Caspase 8/metabolism , Choline/metabolism , Choline Deficiency/enzymology , Choline Deficiency/etiology , Choline Deficiency/pathology , Diet, High-Fat , Gene Expression Regulation , Glucose Intolerance/enzymology , Glucose Intolerance/etiology , Glucose Intolerance/pathology , Homeostasis , Humans , Inflammation , Insulin/metabolism , Insulin Resistance , Intra-Abdominal Fat/pathology , Male , Mice , Necrosis/genetics , Necrosis/pathology , Obesity/enzymology , Obesity/etiology , Obesity/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
8.
Toxicol Lett ; 258: 159-167, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27320964

ABSTRACT

Evidence indicates that hepatic fibrosis is the initial lesion of cirrhosis or hepatocellular carcinoma in diseases such as nonalcoholic steatohepatitis (NASH). To induce NASH, we fed rats a choline-deficient and iron-supplemented L-amino acid-defined (CDAA) diet. Histopathological examination revealed that fibrosis appeared from week 4 and progressed to bridging fibrosis from week 12. Using qRT-PCR assays, we detected increased expression of miR-21, Mmp-9, and Timp-1 in liver that peaked during week 4, when fibrosis was first detected. The expression pattern of miR-21 in plasma paralleled that in liver. Fibrosis tended to be resolved within 12 weeks of a recovery period after 12 weeks of feeding, and the expression of miR-21, Timp-1, and Mmp-9 decreased in liver. Comprehensive analyses of miRNA and mRNA expression in the liver using samples acquired at week 4 detected 16 miRNAs and 11 mRNAs that are mutually-interacting fibrosis-related factors. We therefore conclude that miR-21 was closely associated with fibrosis in a rat model of NASH and has potential as a plasma biomarker for hepatic fibrosis.


Subject(s)
Disease Models, Animal , Liver Cirrhosis/etiology , Liver/metabolism , Matrix Metalloproteinase 9/metabolism , MicroRNAs/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Biomarkers/blood , Choline/therapeutic use , Choline Deficiency/complications , Choline Deficiency/diet therapy , Choline Deficiency/etiology , Choline Deficiency/physiopathology , Diet/adverse effects , Dietary Supplements/poisoning , Disease Progression , Gene Expression Profiling , Gene Expression Regulation , Iron Overload/complications , Iron Overload/etiology , Iron Overload/physiopathology , Iron, Dietary/poisoning , Liver/immunology , Liver/pathology , Liver/physiopathology , Liver Cirrhosis/prevention & control , Matrix Metalloproteinase 9/genetics , MicroRNAs/blood , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , RNA, Messenger/metabolism , Rats, Wistar , Specific Pathogen-Free Organisms , Tissue Inhibitor of Metalloproteinase-1/genetics
9.
J Pediatr Gastroenterol Nutr ; 62(4): 618-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26465792

ABSTRACT

BACKGROUND: Choline depletion is seen in cystic fibrosis (CF) and pancreatic insufficiency in spite of enzyme treatment and may result in liver, fatty acid, and muscle abnormalities. This study evaluated the efficacy and safety of an easily absorbed choline-rich structured lipid (LYM-X-SORB™ [LXS]) to improve choline status. METHODS: Children with CF and pancreatic insufficiency were randomized to LXS or placebo in a 12-month double blind trial. Dietary choline intake, plasma cholines, plasma and fecal phospholipids, coefficient of fat absorption, pulmonary function, growth status, body composition, and safety measures were assessed. Magnetic resonance spectroscopy for calf muscle choline and liver fat were assessed in a subgroup and compared with a healthy comparison group matched for age, sex, and body size. RESULTS: A total of 110 subjects were enrolled (age 10.4 ±â€Š3.0 years). Baseline dietary choline, 88% recommended, increased 3-fold in the LXS group. Plasma choline, betaine, and dimethylglycine increased in the LXS but not placebo (P = 0.007). Plasma lysophosphatidylcholine and phosphatidylcholine increased, and fecal phosphatidylcholine/phosphatidylethanolamine ratio decreased (P ≤ 0.05) in LXS only, accompanied by a 6% coefficient of fat absorption increase (P = 0.001). Children with CF had higher liver fat than healthy children and depleted calf muscle choline at baseline. Muscle choline concentration increased in LXS and was associated with improvement in plasma choline status. No relevant changes in safety measures were evident. CONCLUSIONS: LXS had improved choline intake, plasma choline status, and muscle choline stores compared with placebo group. The choline-rich supplement was safe, accepted by participants, and improved choline status in children with CF.


Subject(s)
Adolescent Nutritional Physiological Phenomena , Child Nutritional Physiological Phenomena , Choline/therapeutic use , Cystic Fibrosis/diet therapy , Dietary Fats , Dietary Supplements , Lysophosphatidylcholines/therapeutic use , Nutritional Status , Adolescent , Child , Child, Preschool , Choline/adverse effects , Choline/analysis , Choline/blood , Choline Deficiency/etiology , Choline Deficiency/prevention & control , Cystic Fibrosis/blood , Cystic Fibrosis/metabolism , Dietary Fats/adverse effects , Dietary Fats/analysis , Dietary Fats/metabolism , Dietary Supplements/adverse effects , Dietary Supplements/analysis , Double-Blind Method , Female , Humans , Intestinal Absorption , Leg , Lipid Metabolism , Liver/metabolism , Lysophosphatidylcholines/adverse effects , Lysophosphatidylcholines/analysis , Lysophosphatidylcholines/metabolism , Male , Muscle, Skeletal/metabolism , Patient Acceptance of Health Care
10.
FASEB J ; 29(5): 1663-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25466896

ABSTRACT

Fibroblasts from a patient with postural orthostatic tachycardia syndrome (POTS), who presented with low plasma choline and betaine, were studied to determine the metabolic characteristics of the choline deficiency. Choline is required for the synthesis of the phospholipid phosphatidylcholine (PC) and for betaine, an important osmoregulator. Here, choline transport, lipid homeostasis, and mitochondria function were analyzed in skin fibroblasts from POTS and compared with control cells. The choline transporter-like protein 1/solute carrier 44A1 (CTL1/SLC44A1) and mRNA expression were 2-3 times lower in POTS fibroblasts, and choline uptake was reduced 60% (P < 0.05). Disturbances of membrane homeostasis were observed by reduced ratios between PC:phosphatidylethanolamine and sphingomyelin:cholesterol, as well as by modified phospholipid fatty acid composition. Choline deficiency also impaired mitochondria function, which was observed by a reduction in oxygen consumption, mitochondrial potential, and glycolytic activity. When POTS cells were treated with choline, transporter was up-regulated, and uptake of choline increased, offering an option for patient treatment. The characteristics of the POTS fibroblasts described here represent a first model of choline and CTL1/SLC44A1 deficiency, in which choline transport, membrane homeostasis, and mitochondrial function are impaired.


Subject(s)
Cell Membrane/pathology , Choline Deficiency/etiology , Choline/pharmacology , Fibroblasts/pathology , Mitochondria/pathology , Postural Orthostatic Tachycardia Syndrome/complications , Skin/pathology , Biological Transport , Blotting, Western , Case-Control Studies , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Choline Deficiency/metabolism , Choline Deficiency/pathology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Oxygen Consumption/drug effects , Phosphatidylcholines/metabolism , Phospholipids/metabolism , Postural Orthostatic Tachycardia Syndrome/physiopathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin/drug effects , Skin/metabolism
11.
Eur J Nutr ; 52(3): 1269-78, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22961562

ABSTRACT

BACKGROUND: Choline forms the head group of phosphatidylcholines, comprising 40-50 % of cellular membranes and 70-95 % of phospholipids in surfactant, bile, and lipoproteins. Moreover, choline serves as the precursor of acetylcholine and is important for brain differentiation and function. While accepted as essential for fetal and neonatal development, its role in preterm infant nutrition has not yet gained much attention. METHODS: The adequate intake of choline of preterm infants was estimated from international recommendations for infants, children, and adults. Choline intake relative to other nutrients was determined retrospectively in all inborn infants below 1,000 g (extremely low birth weight) or below 28 weeks gestational age, admitted to our department in 2006 and 2007 (N = 93). RESULTS: Estimation of adequate intake showed that children with 290 g body weight need more choline than those with 1,200 g (31.4 and 25.2 mg/kg/day, respectively). Day-by-day variability was high for all nutrient intakes including choline. In contrast to the continuous intrauterine choline delivery, median supply reached a plateau at d11 (21.7 mg/kg/day; 25th/75th percentile: 19.6; 23.9). Individual choline supply at d0-d1 and d2-d3 was <10 mg/kg/day in 100 and 69 % of infants, respectively. Furthermore, intakes <10 mg/kg/day were frequently observed beyond day 11. Median adequate intakes (27.4 mg/kg/day at 735 g body weight) were achieved in <2 %. CONCLUSIONS: Nutritional intake of choline in this cohort of preterm infants was frequently less than the estimated adequate intake, with particular shortage until postnatal d10. Because choline is important for brain development, future studies are needed to investigate the effects of adequate nutritional choline intake on long-term neurodevelopment in VLBW infants.


Subject(s)
Child Development , Choline Deficiency/etiology , Choline/administration & dosage , Diet/adverse effects , Infant Nutritional Physiological Phenomena , Infant, Premature, Diseases/etiology , Choline Deficiency/epidemiology , Choline Deficiency/physiopathology , Cohort Studies , Female , Germany/epidemiology , Guidelines as Topic , Hospitals, University , Humans , Incidence , Infant, Extremely Low Birth Weight , Infant, Extremely Premature , Infant, Newborn , Infant, Premature, Diseases/epidemiology , Infant, Premature, Diseases/physiopathology , Intensive Care Units, Neonatal , Male , Nutritional Requirements , Quality Assurance, Health Care , Retrospective Studies
12.
Br J Nutr ; 109(3): 511-9, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-22717142

ABSTRACT

Choline and betaine are nutrients involved in one-carbon metabolism. Choline is essential for neurodevelopment and brain function. We studied the associations between cognitive function and plasma concentrations of free choline and betaine. In a cross-sectional study, 2195 subjects (55 % women), aged 70-74 years, underwent extensive cognitive testing including the Kendrick Object Learning Test (KOLT), Trail Making Test (part A, TMT-A), modified versions of the Digit Symbol Test (m-DST), Block Design (m-BD), Mini-Mental State Examination (m-MMSE) and Controlled Oral Word Association Test (COWAT). Compared with low concentrations, high choline (>8·4 µmol/l) was associated with better test scores in the TMT-A (56·0 v. 61·5, P=0·004), m-DST (10·5 v. 9·8, P=0·005) and m-MMSE (11·5 v. 11·4, P=0·01). A generalised additive regression model showed a positive dose-response relationship between the m-MMSE and choline (P=0·012 from a corresponding linear regression model). Betaine was associated with the KOLT, TMT-A and COWAT, but after adjustments for potential confounders, the associations lost significance. Risk ratios (RR) for poor test performance roughly tripled when low choline was combined with either low plasma vitamin B12 (≤257 pmol/l) concentrations (RR(KOLT)=2·6, 95 % CI 1·1, 6·1; RR(m-MMSE)=2·7, 95 % CI 1·1, 6·6; RR(COWAT)=3·1, 95 % CI 1·4, 7·2) or high methylmalonic acid (MMA) (≥3·95 µmol/l) concentrations (RR(m-BD)=2·8, 95 % CI 1·3, 6·1). Low betaine (≤31·1 µmol/l) combined with high MMA was associated with elevated RR on KOLT (RR(KOLT)=2·5, 95 % CI 1·0, 6·2). Low plasma free choline concentrations are associated with poor cognitive performance. There were significant interactions between low choline or betaine and low vitamin B12 or high MMA on cognitive performance.


Subject(s)
Aging , Betaine/blood , Choline Deficiency/physiopathology , Choline/blood , Cognitive Dysfunction/etiology , Aged , Biomarkers/blood , Choline Deficiency/etiology , Cognitive Dysfunction/blood , Cognitive Dysfunction/epidemiology , Cohort Studies , Cross-Sectional Studies , Diet/adverse effects , Female , Follow-Up Studies , Geriatric Assessment , Humans , Male , Methylmalonic Acid/blood , Norway/epidemiology , Risk Factors , Statistics as Topic , Vitamin B 12/blood , Vitamin B 12 Deficiency/etiology , Vitamin B 12 Deficiency/physiopathology
13.
Am J Clin Nutr ; 96(6): 1465-72, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23134891

ABSTRACT

BACKGROUND: Choline is essential for fetal brain development, and it is not known whether a typical American diet contains enough choline to ensure optimal brain development. OBJECTIVE: The study was undertaken to determine whether supplementing pregnant women with phosphatidylcholine (the main dietary source of choline) improves the cognitive abilities of their offspring. DESIGN: In a double-blind, randomized controlled trial, 140 pregnant women were randomly assigned to receive supplemental phosphatidylcholine (750 mg) or a placebo (corn oil) from 18 wk gestation through 90 d postpartum. Their infants (n = 99) were tested for short-term visuospatial memory, long-term episodic memory, language development, and global development at 10 and 12 mo of age. RESULTS: The women studied ate diets that delivered ∼360 mg choline/d in foods (∼80% of the recommended intake for pregnant women, 65% of the recommended intake for lactating women). The phosphatidylcholine supplements were well tolerated. Groups did not differ significantly in global development, language development, short-term visuospatial memory, or long-term episodic memory. CONCLUSIONS: Phosphatidylcholine supplementation of pregnant women eating diets containing moderate amounts of choline did not enhance their infants' brain function. It is possible that a longer follow-up period would reveal late-emerging effects. Moreover, future studies should determine whether supplementing mothers eating diets much lower in choline content, such as those consumed in several low-income countries, would enhance infant brain development.


Subject(s)
Child Development , Choline Deficiency/diet therapy , Cognition , Dietary Supplements , Lactation , Maternal Nutritional Physiological Phenomena , Phosphatidylcholines/therapeutic use , Choline Deficiency/etiology , Choline Deficiency/physiopathology , Diet/adverse effects , Dietary Supplements/adverse effects , Double-Blind Method , Female , Humans , Infant, Newborn , Language Development , Male , Memory, Long-Term , Memory, Short-Term , North Carolina , Phosphatidylcholines/adverse effects , Pregnancy , Pregnancy Complications/diet therapy , Pregnancy Complications/etiology , Pregnancy Complications/physiopathology , Severity of Illness Index
14.
J Pediatr Gastroenterol Nutr ; 53(1): 115-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21694550

ABSTRACT

Choline deficiency leads to steatohepatitis, elevated transaminases, susceptibility to septic shock, and an increased risk of central catheter thrombosis. Children with intestinal failure (IF) are at risk for choline deficiency. In an unblinded, open-label study, we studied 7 children with IF on parenteral nutrition, measured their plasma free choline level, and, if low, supplemented enterally with adequate intake (AI) doses of choline. Four to 6 weeks later we remeasured their plasma free choline. Unlike adults, infants did not respond to oral choline supplementation at AI doses. Additionally, we have calculated plasma free choline percentiles versus age for normal children.


Subject(s)
Choline/therapeutic use , Dietary Supplements , Intestinal Diseases/diet therapy , Intestines/physiopathology , Administration, Oral , Adolescent , Age Factors , Child , Choline/blood , Choline Deficiency/etiology , Choline Deficiency/prevention & control , Female , Humans , Infant , Intestinal Diseases/blood , Intestinal Diseases/physiopathology , Male , Parenteral Nutrition , Pilot Projects , Short Bowel Syndrome/blood , Short Bowel Syndrome/diet therapy , Short Bowel Syndrome/physiopathology
15.
J Inherit Metab Dis ; 34(1): 3-15, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20446114

ABSTRACT

Choline is an essential nutrient, but is also formed by de novo synthesis. Choline and its derivatives serve as components of structural lipoproteins, blood and membrane lipids, and as a precursor of the neurotransmitter acetylcholine. Pre-and postnatal choline availability is important for neurodevelopment in rodents. Choline is oxidized to betaine that serves as an osmoregulator and is a substrate in the betaine-homocysteine methyltransferase reaction, which links choline and betaine to the folate-dependent one-carbon metabolism. Choline and betaine are important sources of one-carbon units, in particular, during folate deficiency. Choline or betaine supplementation in humans reduces concentration of total homocysteine (tHcy), and plasma betaine is a strong predictor of plasma tHcy in individuals with low plasma concentration of folate and other B vitamins (B2, B6, and B12) in combination TT genotype of the methylenetetrahydrofolate reductase 677 C->T polymorphism. The link to one-carbon metabolism and the recent availability of food composition data have motivated studies on choline and betaine as risk factors of chronic diseases previously studied in relation to folate and homocysteine status. High intake and plasma level of choline in the mother seems to afford reduced risk of neural tube defects. Intake of choline and betaine shows no consistent relation to cancer or cardiovascular risk or risk factors, whereas an unfavorable cardiovascular risk factor profile was associated with high choline and low betaine concentrations in plasma. Thus, choline and betaine showed opposite relations with key components of metabolic syndrome, suggesting a disruption of mitochondrial choline oxidation to betaine as part of the mitochondrial dysfunction in metabolic syndrome.


Subject(s)
Betaine/pharmacology , Choline/physiology , Disease/etiology , Health , Animals , Betaine/metabolism , Choline/metabolism , Choline Deficiency/complications , Choline Deficiency/etiology , Choline Deficiency/therapy , Eating/physiology , Humans , Models, Biological , Water-Electrolyte Balance/physiology
16.
Metab Brain Dis ; 25(3): 269-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20838865

ABSTRACT

Diabetic encephalopathy describes the moderate cognitive deficits, neurophysiological and structural central nervous system changes associated with untreated diabetes. It involves neurotoxic effects such as the generation of oxidative stress, the enhanced formation of advanced glycation end-products, as well as the disturbance of calcium homeostasis. Due to the direct connection of choline (Ch) with acetylcholine availability and signal transduction, a background of Ch-deficiency might be unfavorable for the pathology and subsequently for the treatment of several metabolic brain diseases, including that of diabetic encephalopathy. The aim of this study was to shed more light on the effects of adult-onset streptozotocin (STZ)-induced diabetes and/or Ch-deprivation on the activities of acetylcholinesterase (AChE) and two important adenosine triphosphatases, namely Na(+),K(+)-ATPase and Mg(2+)-ATPase. Male adult Wistar rats were divided into four main groups, as follows: control (C), diabetic (D), Ch-deprived (CD), and Ch-deprived diabetic (D+CD). Deprivation of Ch was provoked through the administration of Ch-deficient diet. Both the induction of diabetes and the beginning of dietary-mediated provoking of Ch-deprivation occurred at the same day, and rats were killed by decapitation after 30 days (1 month; groups C1, D1, CD1 and D1+CD1) and 60 days (2 months; groups C2, D2, CD2 and D2+CD2, respectively). The adult rat brain AChE activity was found to be significantly increased by both diabetes (+10%, p < 0.001 and +11%, p < 0.01) and Ch-deprivation (+19%, p < 0.001 and +14%, p < 0.001) when compared to the control group by the end of the first (C1) and the second month (C2), respectively. However, the Ch-deprived diabetic rats' brain AChE activity was significantly altered only after a 60-day period of exposure, resulting in a +27% increase (D2+CD2 vs. C2, p < 0.001). Although the only significant change recorded in the brain Na(+),K(+)-ATPase activity after the end of the first month is attributed to Ch-deprivation (+21%, p < 0.05, CD1 vs. C1), all groups of the second month exhibited a statistically significant decrease in brain Na(+),K(+)-ATPase activity (-24%, p < 0.01, D2 vs. C2; -21%, p < 0.01, CD2 vs. C2; -22%, p < 0.01, D2+CD2 vs. C2). As concerns Mg(2+)-ATPase, the enzyme's activity demonstrates no significant changes, with the sole exception of the D2+CD2 group (+21%, p < 0.05, D2+CD2 vs. C2). In addition, statistically significant time-dependent changes concerning the brain Mg(2+)-ATPase activity were recorded within the diabetic (p < 0.05, D2 vs. D1) and the Ch-deprived (p < 0.05, CD2 vs. CD1) rat groups. Our data indicate that Ch-deprivation seems to be an undesirable background for the above-mentioned enzymatic activities under untreated diabetes, in a time-evolving way. Further studies on the issue should focus on a region-specific reevaluation of these crucial enzymes' activities as well as on the possible oxidative mechanisms involved.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/etiology , Brain/enzymology , Choline Deficiency/enzymology , Choline Deficiency/etiology , Diabetes Complications/enzymology , Diabetes Mellitus, Experimental/enzymology , Rats, Wistar , Acetylcholinesterase/biosynthesis , Animals , Brain/metabolism , Brain Ischemia/metabolism , Choline Deficiency/metabolism , Diabetes Complications/complications , Diabetes Complications/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Enzyme Activation/physiology , Male , Rats , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Up-Regulation/physiology
17.
Liver Int ; 29(4): 575-84, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19323784

ABSTRACT

BACKGROUND: Hepatic oval cells, progenitor cells in the liver, can differentiate into hepatocytes and bile duct cells both in vitro and in vivo. Although hepatic stellate cells are another important cell component in the liver, less attention has been focused on the relationship between hepatic oval cells and hepatic stellate cells. METHODS: Hepatic oval cells were isolated from rats fed a choline-deficient diet supplemented with 0.1% ethionine for 6 weeks and characterized by electron microscopy, flow cytometry, reverse transcription polymerase chain reaction, Western blot and bi-direction differentiation. After treatment with transforming growth factor-beta1 (TGF-beta1), changes in cell viability, morphology, extracellular matrix (ECM) expression and immune phenotype were analysed in these cultured and adherent hepatic oval cells. RESULTS: The primary cultured hepatic oval cells were positive for the oval cell-specific markers OV-6, BD-1/BD-2 and M2PK as well as the hepatocyte markers albumin and alpha-foetoprotein. These hepatic oval cells differentiated bipotentially into hepatocytes or bile duct-like cells under appropriate conditions. It is noteworthy that these bipotential hepatic oval cells expressed ECM genes stably, including collagens, matrix metalloproteinases and tissue inhibitor of mellatoproteinase. Furthermore, except for growth inhibition and morphological changes in the hepatic oval cells after exposure to TGF-beta1, there was an increased expression of ECM genes, the onset expression of snail and loss expression of E-cadherin. During this process, TGF-beta1 treatment induced an upregulation of marker genes for hepatic stellate cells in hepatic oval cells, such as desmin and GFAP. CONCLUSION: Except for the expression of ECM, the cultured hepatic oval cells could induce an increased expression of hepatic stellate cell markers by TGF-beta1 through an epithelial-mesenchymal transition process, which might indicate the contribution of hepatic oval cells to liver fibrosis.


Subject(s)
Extracellular Matrix Proteins/metabolism , Gene Expression/drug effects , Liver/metabolism , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Albumins/metabolism , Animals , Antimetabolites/administration & dosage , Antimetabolites/adverse effects , Bile Ducts/drug effects , Bile Ducts/metabolism , Bile Ducts/pathology , Biomarkers/metabolism , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Choline Deficiency/etiology , Choline Deficiency/metabolism , Choline Deficiency/pathology , Desmin/genetics , Desmin/metabolism , Disease Models, Animal , Ethionine/administration & dosage , Ethionine/adverse effects , Extracellular Matrix Proteins/drug effects , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/pharmacology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/pathology , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Stem Cells/ultrastructure , Transforming Growth Factor beta/pharmacology , alpha-Fetoproteins/metabolism
18.
Neurology ; 71(1): 44-9, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18591504

ABSTRACT

OBJECTIVE: To investigate whether secondary impairment of the transmethylation pathway is a mechanism underlying the neurologic involvement in homocystinuria due to remethylation defects. METHODS: Twelve patients with neurologic disease due to remethylation defects were examined by brain magnetic resonance spectroscopic imaging ((1)H MRSI). Brain N-acetylaspartate, choline-containing compounds (Cho), and creatine (Cr) were quantified and compared to with controls. Metabolites of remethylation cycle and creatine biosynthesis pathway were measured in plasma and urine. RESULTS: MRSI revealed isolated Cho deficiency in all regions examined (mean concentration units +/- SD, patients vs controls): frontal white matter (0.051 +/- 0.010 vs 0.064 +/- 0.010; p = 0.001), lenticular nucleus (0.056 +/- 0.011 vs 0.069 +/- 0.009; p < 0.001), and thalamus (0.063 +/- 0.010 vs 0.071 +/- 0.007; p = 0.006). In contrast to controls, the Cho/Cr ratio decreased with age in patients in the three brain regions examined. Low creatine urinary excretion (p < 0.005), normal urine and plasma guanidinoacetate, and a paradoxical increase in plasma S-adenosylmethionine (p < 0.005) concentrations were observed. CONCLUSION: Patients with homocystinuria due to remethylation defects have an isolated brain choline deficiency, probably secondary to depletion of labile methyl groups produced by the transmethylation pathway. Although biochemical studies suggest mild peripheral creatine deficiency, brain creatine is in the reference range, indicating a possible compartmentation phenomenon. Paradoxical increase of S-adenosylmethionine suggests that secondary inhibition of methylases contributes to the transmethylation defect in these conditions.


Subject(s)
Brain/metabolism , Choline Deficiency/metabolism , Choline/metabolism , Homocysteine S-Methyltransferase/metabolism , Homocystinuria/blood , Homocystinuria/urine , Adolescent , Adult , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Brain/physiopathology , Brain Chemistry/physiology , Child , Child, Preschool , Choline Deficiency/etiology , Choline Deficiency/physiopathology , Creatine/blood , Creatine/urine , Female , Homocystinuria/physiopathology , Humans , Magnetic Resonance Spectroscopy , Male , Methylation , S-Adenosylmethionine/metabolism
19.
J Hepatol ; 47(6): 851-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17825453

ABSTRACT

BACKGROUND/AIMS: High serum ferritin and liver iron concentrations were found in some patients with NASH, suggesting a role for iron as a co-factor that aggravates liver injury. The aim of this study is to investigate the effects of parenteral iron in a rat model of NASH induced by a methionine choline deficient diet (MCDD). METHODS: Wistar rats were divided into 1 - Control, 2 - Iron (Fe), 3 - MCDD, 4 - MCDD&Fe groups. Iron dextran 100mg/kg was administered intra-muscularly in groups 2 and 4. All rats were fed MCDD, Groups 1 and 2 were supplied with choline and methionine. Blood and tissue samples were obtained after 4weeks. RESULTS: The iron injection alone did not affect the liver whereas MCDD led to steatohepatitis. Iron worsened steatosis without any obvious effect on accompanying inflammation. It aggravated tissue injury by increasing apoptosis. Liver fibrosis was observed only in 3 out of 10 rats in the MCDD&Fe group. CONCLUSIONS: Observation of liver fibrosis only in the MCDD&Fe group suggests that iron induced increase in apoptosis contributes to the development of fibrosis at an earlier time than expected.


Subject(s)
Apoptosis/drug effects , Choline Deficiency/complications , Fatty Liver/etiology , Iron, Dietary/adverse effects , Animals , Choline Deficiency/etiology , Diet , Iron-Dextran Complex/administration & dosage , Iron-Dextran Complex/adverse effects , Liver Cirrhosis/chemically induced , Nutrition Disorders , Rats , Rats, Wistar
20.
Liver Int ; 27(2): 227-34, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17311618

ABSTRACT

BACKGROUND: Oxidative stress plays a role in the pathogenesis of nonalcoholic steatohepatitis (NASH). Yo jyo hen shi ko (YHK) is a complex compound purported to reduce reactive oxygen species (ROS) by blocking the propagation of radical-induced reactions. The aim of this study was to evaluate the role of the effect of YHK in experimental NASH. METHODS: NASH was induced in male ob/ob mice by a high-fat (HF) diet or methionine/choline-deficient (MCD) diet for 4 weeks. YHK-treated animals received YHK solution orally (20 mg/kg/day) in both experimental diets (n=6; each group) while control animals received only vehicle. RESULTS: The MCD and HF groups developed moderate diffuse macrosteatosis, hepatocellular ballooning, and a diffuse inflammatory infiltrate. With the addition of YHK, there was a marked reduction in macrosteatosis in both groups. This was associated with decreased lipoperoxide and reduced glutathione-GSH concentrations as well as reduced serum aminotransferases and improved histological markers of inflammation. These changes were also associated with weight loss in the MCD+YHK group and diminished weight gain in the HF+YHK group. CONCLUSION: YHK therapy blunts the development of macrosteatosis in these models of NASH and significantly reduces markers of oxidative stress. YHK also diminishes weight gain in this obesity prone model. Our findings warrant further study on the mechanisms involved with these effects.


Subject(s)
Choline Deficiency , Dietary Fats/administration & dosage , Drugs, Chinese Herbal/pharmacology , Fatty Liver/prevention & control , Hepatitis/prevention & control , Methionine/deficiency , Animals , Biomarkers/metabolism , Body Weight , Choline Deficiency/etiology , Diet , Dose-Response Relationship, Drug , Lipids/blood , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Obese , Oxidative Stress , Transaminases/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...