Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Drug Des Devel Ther ; 15: 2357-2373, 2021.
Article in English | MEDLINE | ID: mdl-34121838

ABSTRACT

PURPOSE: To prepare the levocarnitine thermosensitive in situ gel (LCTG) and evaluate its effect on dry eye disease (DED). METHODS: Draize eye irritation test and other examinations were used to evaluate the eye irritation after multiple administration of LCTG. The Schirmer test, fluorescein sodium staining, HE staining and TUNEL staining were used to detect the tear secretion, corneal injury, histopathological changes of the cornea and lacrimal gland, and the apoptosis rate of cornea epithelial cells after 3 days of the administration. The conjunctival goblet cell density was detected by PAS staining, and the expression levels of matrix metalloproteinase-3 (MMP-3) and matrix metalloproteinase-9 (MMP-9) of corneal epithelial cells were detected by immunofluorescence staining after 7 days of the administration. RESULTS: LCTG is non-irritating to rabbit eyes and has good biocompatibility. LCTG administration for 3 days can significantly increase the amount of tear secretion in mice with DED, promote corneal epithelial integrity and central corneal epithelium thickness recovery, and improve the pathological morphology and structure of corneal and lacrimal gland tissues, and reduce the apoptosis rate of the corneal epithelial cells. After 7 days of the administration, the preparation can promote the proliferation of conjunctival goblet cells and down-regulate the cornea expression levels of MMP-3 and MMP-9 in epithelial cells. CONCLUSION: The LCTG has a good curative effect on mice with DED, and the overall curative effect is better than that of levocarnitine solution.


Subject(s)
Carnitine/administration & dosage , Cornea/metabolism , Drug Delivery Systems , Dry Eye Syndromes/drug therapy , Administration, Ophthalmic , Animals , Carnitine/pharmacology , Carnitine/toxicity , Disease Models, Animal , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Female , Gene Expression Regulation/drug effects , Goblet Cells/drug effects , Goblet Cells/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred BALB C , Rabbits , Temperature , Treatment Outcome
2.
Mol Nutr Food Res ; 63(20): e1900514, 2019 10.
Article in English | MEDLINE | ID: mdl-31368236

ABSTRACT

SCOPE: A gut-microbiota-dependent metabolite of L-carnitine, trimethylamine-N-oxide (TMAO), has been recently discovered as an independent and dose-dependent risk factor for cardiovascular disease (CVD). This study aims to investigate the effects of pterostilbene on reducing TMAO formation and on decreasing vascular inflammation in carnitine-feeding mice. METHODS AND RESULTS: C57BL/6 mice are treated with 1.3% carnitine in drinking water with or without pterostilbene supplementation. Using LC-MS/MS, the result shows that mice treated with 1.3% carnitine only significantly increased the plasma TMAO and pterostilbene supplementation group can reverse it. Additionally, pterostilbene decreases hepatic flavin monooxygenase 3 (FMO3) mRNA levels compared to carnitine only group. It appears that pterostilbene can alter host physiology and create an intestinal microenvironment favorable for certain gut microbiota. Gut microbiota analysis reveals that pterostilbene increases the abundance of Bacteroides. Further, pterostilbene decreases mRNA levels of vascular inflammatory markers tumor necrosis factor-α (TNF-α), vascular cell adhesion molecule 1 (VCAM-1), and E-selectin). CONCLUSION: These data suggest that amelioration of carnitine-induced vascular inflammation after consumption of pterostilbene is partially mediated via modulation of gut microbiota composition and hepatic enzyme FMO3 gene expression.


Subject(s)
Gastrointestinal Microbiome/physiology , Methylamines/metabolism , Stilbenes/pharmacology , Vasculitis/prevention & control , Animals , Carnitine/toxicity , Female , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Oxygenases/genetics , Tumor Necrosis Factor-alpha/genetics
3.
Nutrients ; 9(5)2017 May 12.
Article in English | MEDLINE | ID: mdl-28498348

ABSTRACT

In chronic kidney disease (CKD), accumulation of uremic toxins is associated with an increased risk of death. Some uremic toxins are ingested with the diet, such as phosphate and star fruit-derived caramboxin. Others result from nutrient processing by gut microbiota, yielding precursors of uremic toxins or uremic toxins themselves. These nutrients include l-carnitine, choline/phosphatidylcholine, tryptophan and tyrosine, which are also sold over-the-counter as nutritional supplements. Physicians and patients alike should be aware that, in CKD patients, the use of these supplements may lead to potentially toxic effects. Unfortunately, most patients with CKD are not aware of their condition. Some of the dietary components may modify the gut microbiota, increasing the number of bacteria that process them to yield uremic toxins, such as trimethylamine N-Oxide (TMAO), p-cresyl sulfate, indoxyl sulfate and indole-3 acetic acid. Circulating levels of nutrient-derived uremic toxins are associated to increased risk of death and cardiovascular disease and there is evidence that this association may be causal. Future developments may include maneuvers to modify gut processing or absorption of these nutrients or derivatives to improve CKD patient outcomes.


Subject(s)
Gastrointestinal Microbiome , Micronutrients/toxicity , Renal Insufficiency, Chronic/microbiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/mortality , Carnitine/administration & dosage , Carnitine/toxicity , Choline/administration & dosage , Choline/toxicity , Diet , Humans , Methylamines/administration & dosage , Methylamines/toxicity , Micronutrients/administration & dosage , Oxalates/administration & dosage , Oxalates/toxicity , Phosphates/administration & dosage , Phosphates/toxicity , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/toxicity , Tryptophan/administration & dosage , Tryptophan/toxicity , Tyrosine/administration & dosage , Tyrosine/toxicity
4.
Reproduction ; 152(4): 283-91, 2016 10.
Article in English | MEDLINE | ID: mdl-27402869

ABSTRACT

l-Carnitine (l-Cn), despite the beneficial role as energy-generating substance delivering long-chain fatty acids to the ß-oxidation pathway in mitochondria, has been accused to cause an endometriosis-like state to BALB/c mice manifested by increased inflammatory cytokines in serum and peritoneal fluid, accumulation of immune cells in the peritoneal cavity and uterine walls and most importantly, correlating to infertility. Exploring this type of infertility, the effect of l-Cn on preimplantation embryo development, ovarian integrity and systemic maternal immunity was studied. Using nonlinear microscopy analysis, which was shown to be a powerful tool for determining embryo quality by quantitatively estimating the lipid body (LB) content of the cells, it was shown that in vitro and in vivo administration of l-Cn significantly decreased LB mean area in zygotes. Daily intraperitoneal administration of 2.5mg l-Cn for 3, 4 and 7days to mice significantly decreased the percent of normal zygotes. However, only the 7-day treatment persisted by affecting 2- and 8-cell stage embryos, while almost abolishing blastocyst development. Such effects were accompanied by abnormal ovarian histology, showing increased numbers of corpora luteus and elevated progesterone concentration in the serum. In addition, it was shown that the 7-day l-Cn treatment pushed maternal systemic immunity toward inflammation and immunosuppression by increasing CD11b-, CD25- and CD11bGr1-positive cells in spleen, which opposed the necessity for immunostimulation at these early stages of pregnancy. In conclusion, the results presented here demonstrated that elevated doses of l-Cn affect early stages of embryo development, leading to infertility.


Subject(s)
Blastocyst/drug effects , Carnitine/toxicity , Embryonic Development/drug effects , Infertility/etiology , Animals , Blastocyst/cytology , Cells, Cultured , Female , Infertility/pathology , Mice , Mice, Inbred BALB C , Pregnancy
5.
Nutr Clin Pract ; 30(4): 559-69, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26113560

ABSTRACT

The American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) started an intensive review of commercially available parenteral vitamin and trace element (TE) products in 2009. The chief findings were that adult multi-TE products currently available in the United States (U.S.) provide potentially toxic amounts of manganese, copper, and chromium, and neonatal/pediatric multi-TE products provide potentially toxic amounts of manganese and chromium. The multivitamin products appeared safe and effective; however, a separate parenteral vitamin D product is needed for those patients on standard therapy who continue to be vitamin D depleted and are unresponsive to oral supplements. The review process also extended to parenteral choline and carnitine. Although choline and carnitine are not technically vitamins or trace elements, choline is an essential nutrient in all age groups, and carnitine is an essential nutrient in infants, according to the Food and Nutrition Board of the Institute of Medicine. A parenteral choline product needs to be developed and available. Efforts are currently under way to engage the U.S. Food and Drug Administration (FDA) and the parenteral nutrient industry so A.S.P.E.N.'s recommendations can become a commercial reality.


Subject(s)
Dietary Supplements/standards , Micronutrients/standards , Parenteral Nutrition/standards , United States Food and Drug Administration/standards , Adult , Carnitine/standards , Carnitine/toxicity , Choline/standards , Choline/toxicity , Dietary Supplements/toxicity , Drug Approval , Humans , Infant , Lipotropic Agents/standards , Lipotropic Agents/toxicity , Micronutrients/toxicity , Trace Elements/standards , Trace Elements/toxicity , United States , Vitamin D/standards , Vitamin D/toxicity , Vitamins/standards , Vitamins/toxicity
6.
Int J Dev Neurosci ; 30(5): 383-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22472139

ABSTRACT

Medium-chain fatty acids and acylcarnitines accumulate in medium-chain acyl-CoA dehydrogenase deficiency (MCADD), the most frequent fatty acid oxidation defect clinically characterized by episodic crises with vomiting, seizures and coma. Considering that the pathophysiology of the neurological symptoms observed in MCADD is poorly known and, to our knowledge, there is no report on the involvement of acylcarnitines in the brain damage presented by the affected patients, the objective of the present study was to investigate the in vitro effects of hexanoylcarnitine (HC), octanoylcarnitine, decanoylcarnitine (DC) and cis-4-decenoylcarnitine (cDC) at concentrations varying from 0.01 to 1.0mM on important oxidative stress parameters in cerebral cortex of young rats. HC, DC and cDC significantly induced lipid peroxidation, as determined by increased thiobarbituric acid-reactive substances (TBA-RS) values. In addition, carbonyl formation was significantly augmented and sulfhydryl content diminished by DC, reflecting induction of protein oxidative damage. HC, DC and cDC also decreased glutathione (GSH) levels, the most important brain antioxidant defense. Furthermore, DC-induced elevation of TBA-RS values and decrease of GSH levels were prevented by the free radical scavengers melatonin and α-tocopherol, indicating the involvement of reactive oxygen species in these effects. We also found that l-carnitine itself did not induce lipid and protein oxidative damage, neither reduced the antioxidant defenses. Our present data show that the major medium-chain acylcarnitines accumulating in MCADD elicit oxidative stress in rat brain. It is therefore presumed that these compounds may be involved to a certain extent in the pathogenesis of the neurologic dysfunction of MCADD.


Subject(s)
Cerebral Cortex/physiopathology , Homeostasis/physiology , Lipid Metabolism, Inborn Errors/pathology , Acyl-CoA Dehydrogenase/deficiency , Animals , Carnitine/analogs & derivatives , Carnitine/toxicity , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Glutathione/metabolism , Homeostasis/drug effects , Homeostasis/ethics , Lipid Metabolism, Inborn Errors/chemically induced , Lipid Metabolism, Inborn Errors/prevention & control , Lipid Peroxidation/drug effects , Male , Melatonin/administration & dosage , Oxidative Stress/drug effects , Oxidative Stress/physiology , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Statistics, Nonparametric , Thiobarbituric Acid Reactive Substances/metabolism , alpha-Tocopherol/administration & dosage
7.
Pharmacology ; 88(1-2): 10-7, 2011.
Article in English | MEDLINE | ID: mdl-21709430

ABSTRACT

AIM: To evaluate the efficacy and safety of 2 analogs of L-carnitine on rats made insulin resistant by a high-fructose diet. METHODS: Using rats made insulin resistant by a high-fructose diet, we investigated the impact of 2 analogs of L-carnitine (25 mg/kg) and L-carnitine (250 mg/kg) on glucose, triglycerides and cholesterol blood levels, and liver glycogen. We also evaluated the safety of both analogs by the assessment of some biochemical and hematological parameters, a histological analysis and a study of embryotoxicity. RESULTS: Both analogs reduced the levels of triglycerides in the liver and plasma, but only analog 2 reduced the cholesterol levels in insulin-resistant rats. No changes were observed in glycogen content. Safety evaluations revealed alterations in blood lymphocytes and embryotoxicity data. CONCLUSION: This study demonstrated that the 2 analogs maintain the pharmacological properties of L-carnitine but have a different efficacy, potency and toxicity.


Subject(s)
Carnitine/pharmacology , Fructose/pharmacology , Insulin Resistance/physiology , Sweetening Agents/pharmacology , Vitamin B Complex/pharmacology , Animals , Blood Glucose/analysis , Body Weight , Carnitine/analogs & derivatives , Carnitine/therapeutic use , Carnitine/toxicity , Chick Embryo , Cholesterol/blood , Diet , Disease Models, Animal , Drug Evaluation, Preclinical , Embryo, Nonmammalian/drug effects , Glycogen/blood , Insulin/blood , Insulin/physiology , Liver/chemistry , Liver/metabolism , Male , Rats , Rats, Wistar , Sweetening Agents/analysis , Sweetening Agents/chemical synthesis , Sweetening Agents/toxicity , Teratogens/toxicity , Triglycerides/blood , Vitamin B Complex/therapeutic use , Vitamin B Complex/toxicity
8.
Regul Toxicol Pharmacol ; 59(2): 285-92, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21078356

ABSTRACT

Soy products are primarily composed of proteins, phytochemicals such as isoflavones, soy lipids, and carbohydrates. Recently, soy isoflavones with L-carnitine were reported to exhibit anti-obesity effects in mice. FCD, a combination of soybean extract and L-carnitine, is a newly developed food substance. As a part of its safety assessment, acute and 13-week subchronic toxicity studies were performed in a total of 100 Sprague-Dawley (SD) rats. In the acute study, a single limit dose of 2000 mg/kg was orally administered to five male and five female rats. No adverse effects or mortality was observed during a 14-day period or upon gross pathological examination. In the subchronic study, FCD was orally administered in daily doses of 500, 1000, and 2000 mg/kg for 13 weeks, resulting in no mortality, and no changes in hematological and serum biochemistry parameters, gross pathology or histopathology. However, body weights of females were significantly decreased 10 weeks after treatment at an average of 2000 mg/kg. In addition, a slight decrease in mean food and water consumption was observed at the same dose level for 13 weeks. Therefore, the no-observed-adverse-effect-level (NOAEL) of FCD was considered to be 2000 mg/kg for male and 1000 mg/kg for female SD rats.


Subject(s)
Carnitine/toxicity , Glycine max/chemistry , Isoflavones/toxicity , Plant Extracts/toxicity , beta-Glucans/toxicity , Animals , Body Weight/drug effects , Carnitine/administration & dosage , Female , Isoflavones/administration & dosage , Male , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Plant Extracts/administration & dosage , Rats , Rats, Sprague-Dawley , Toxicity Tests, Acute , Toxicity Tests, Chronic , beta-Glucans/administration & dosage
9.
FEMS Yeast Res ; 10(3): 270-81, 2010 May.
Article in English | MEDLINE | ID: mdl-20199579

ABSTRACT

l-Carnitine plays a well-documented role in eukaryotic energy homeostasis by acting as a shuttling molecule for activated acyl residues across intracellular membranes. This activity, supported by carnitine acyl-transferases and transporters, is referred to as the carnitine shuttle. However, several pleiotropic and often beneficial effects of carnitine in humans have been reported that appear to be unrelated to shuttling activity, but little conclusive evidence regarding molecular mechanisms exists. We have recently demonstrated a role of carnitine, independent of the carnitine shuttle, in yeast stress protection. Here, we show that carnitine specifically protects against oxidative stress caused by H(2)O(2) and the superoxide-generating agent menadione. Surprisingly, carnitine has a detrimental effect on survival when combined with thiol-modifying agents. Central elements of the oxidative stress response, specifically the transcription factors Yap1p and Skn7p, are shown to be required for carnitine's protective effect, but several downstream effectors are dispensable. A DNA microarray-based analysis identifies Cyc3p, a cytochrome c heme lyase, as being important for carnitine's impact during oxidative stress. These findings establish a direct genetic link to a carnitine-related phenotype that is independent of the shuttle system and suggests that Saccharomyces cerevisiae should provide a useful model for further elucidation of carnitine's physiological roles.


Subject(s)
Carnitine/pharmacology , Carnitine/toxicity , Microbial Viability/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Vitamin B Complex/pharmacology , Vitamin B Complex/toxicity , Antioxidants/pharmacology , Antioxidants/toxicity , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hydrogen Peroxide/toxicity , Lyases/genetics , Lyases/metabolism , Oxidants/toxicity , Oxidative Stress , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Stress, Physiological , Transcription Factors/genetics , Transcription Factors/metabolism , Vitamin K 3/toxicity
10.
J Med Chem ; 51(13): 3713-9, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18529042

ABSTRACT

We studied the antitumor properties of the dinuclear copper(II) complex of l-carnitine [Cu 2( l-carnitine) 2Cl 2(H 2O) 2]Cl 2, as well as those of l-carnitine and copper chloride dihydrate, in human leukemic cells. The complex was synthesized and characterized using EPR, (1)H NMR, (13)C NMR, IR, and UV-vis analyses. Its cytotoxic effect on the human leukemia cell lines HL-60 and K562 was studied by assessing the metabolic activity of cells (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, MTT method), the structural integrity of cell membrane using Trypan blue assay, and the proliferation capacity of cells studying growth curves. Both leukemia cell lines showed a concentration-specific increased cytotoxicity of the complex, compared to l-carnitine or copper chloride dihydrate, with distinct underlying mechanisms, which were decreased proliferation efficiency for HL-60 cells and increased necrotic phenomena for K562 cells. Our results are indicative of a concentration-specific enhanced antileukemic effect of the complex, implying its value as a tool in the implementation of leukemia.


Subject(s)
Carnitine/chemistry , Carnitine/toxicity , Chlorides/chemistry , Copper/chemistry , Water/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Electron Spin Resonance Spectroscopy , Humans , Molecular Structure
11.
Biofizika ; 53(6): 1025-32, 2008.
Article in Russian | MEDLINE | ID: mdl-19137688

ABSTRACT

It has been shown using the fluorescent microscopy technique that long-chain fatty acid derivatives, myristoylcarnitine and palmitoylcarnitine, exert the most toxic effect on rat ventricular cardiomyoctes. The addition of 20-50 microM acylcarnitines increases calcium concentration in cytoplasm ([Ca2+]i) and causes cell death after the 4-8 min lag-period. This effect is independent on extracellular calcium and L-type calcium channel inhibitors. Free acids (myristic and palmitic acids) at a concentration of 300-500 microM have a little effect on [Ca2+]i within 30 min. We suggest that the toxic effect is due to the activation of sarcoplasmic reticulum calcium channels by acylcarnitines and resulting acyl-CoA. Mitochondria play a role of calcium-buffer system in these conditions. The calcium capacity of this buffer determines the lag-period. Phosphate increases the calcium capacity of mitochondrial and the lag-period. In the presence of rotenone and oligomycin the elevation of [Ca2+]i after the addition of acylcarnitines occurs without the lag-period. The exhaustion of the mitochondrial calcium-buffer capacity or significant depolarization of mitochondrial leads to a rapid release of calcium from mitochondria and cell death. Thus, the activation of reticular calcium channels is the main reason of the toxicity of myristoylcarnitine and palmitoylcarnitine.


Subject(s)
Calcium/metabolism , Carnitine/analogs & derivatives , Cytosol/metabolism , Myocytes, Cardiac/drug effects , Myristic Acids/toxicity , Palmitoylcarnitine/toxicity , Animals , Calcium Channels/physiology , Carnitine/toxicity , Cell Death , In Vitro Techniques , Mitochondria, Heart/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology
12.
Biomed Khim ; 53(6): 672-82, 2007.
Article in Russian | MEDLINE | ID: mdl-18323152

ABSTRACT

We have investigated effects of Mildronate, gamma-butyrobetaine (GBB) and their combination ("Neomildronate") on the plasma levels of thyroid gland hormones and some intermediates of basal metabolism (free fatty acids, triglycerides, glucose) in rats with different dysfunctions of thyroid gland, including idiopathic hyperfunction and hypofunction induced by propylthiouracil or L-carnitin administration. Histological investigation of the thyroid gland was also performed. Intraperitoneal injections of Mildronate (150 mg/kg) during 20 days to Wistar male rats with elevated level of thyroid hormones and basal metabolism normalized thyroxine level and parameters of lipid metabolism. Mildronate, GBB and their combination did not affect the natural resurgence of rats with experimental hypofunction induced by propylthiouracil or L-carnitin administration. The possible biochemical role of given treatment in regulation of thyroid gland function is discussed.


Subject(s)
Betaine/analogs & derivatives , Carbohydrate Metabolism/drug effects , Cardiovascular Agents/pharmacology , Carnitine/pharmacology , Lipid Metabolism/drug effects , Methylhydrazines/pharmacology , Thyroid Diseases/blood , Thyroxine/blood , Animals , Antithyroid Agents/toxicity , Betaine/pharmacology , Carnitine/toxicity , Male , Propylthiouracil/toxicity , Rats , Rats, Wistar , Thyroid Diseases/chemically induced , Thyroid Diseases/pathology , Thyroid Gland/metabolism , Thyroid Gland/pathology , Vitamin B Complex/toxicity
13.
Biofactors ; 8(1-2): 73-8, 1998.
Article in English | MEDLINE | ID: mdl-9699012

ABSTRACT

The effects of L-carnitine on toxicity induced by paraquat (PQ) in mice were investigated. L-carnitine pretreatment surprisingly promoted the toxicity of PQ in a dose-dependent manner shortening the survival time. The maximum effect occurred when L-carnitine, at a dose of 16 mmol/kg, was intraperitoneally administered 30 min before an intraperitoneal injection of PQ (75 mg/kg). Lipid peroxidation in lung significantly increased 6 h after PQ administration. L-carnitine accelerated this effect since L-carnitine-pretreated mice already showed a significant increase of lung malondialdehyde 1 h after PQ administration. In liver, PQ administration did not produce lipid peroxidation; nevertheless L-carnitine-pretreated mice showed a significant increase of malondialdehyde 6 h after PQ administration. Lung and liver glutathione decreased in mice receiving only PQ but this effect was not significantly changed by L-carnitine pretreatment. These results indicate that L-carnitine accelerates PQ-induced mortality rate by facilitating lipid peroxidation.


Subject(s)
Carnitine/toxicity , Paraquat/toxicity , Analysis of Variance , Animals , Drug Synergism , Glutathione/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Malondialdehyde/analysis , Mice
15.
Ann Acad Med Stetin ; 39: 185-205, 1993.
Article in Polish | MEDLINE | ID: mdl-8154618

ABSTRACT

Investigations were carried out on 24 pregnant white New Zealand rabbits and their 210 fetuses. Eight pregnant rabbits received intravenous injections of carnitine on the 25th, 26th and 27th day of pregnancy, in doses of 80 mg/kg/24 hours. Eight other pregnant rabbits were given carnitine in the same manner and betamethasone intramuscularly on the 25th and 26th day of pregnancy in doses of 0.1 mg/kg/24 hours. Eight control pregnant rabbits received intravenous and intramuscular injections of normal saline. In all the groups caesarean section was performed on the 28th day of pregnancy. The contents of lecithin and sphingomyelin in lung homogenates of fetuses and in amniotic fluid were determined by thin-layer chromatography. The lungs of the fetuses were subjected to morphological studies in light and in electron microscopes. The studies proved that carnitine given during pregnancy accelerated biochemical and morphological maturation of fetal rabbit lungs. However, administration of carnitine in combination with betamethasone does not provide better effect than the administration of carnitine alone.


Subject(s)
Betamethasone/toxicity , Carnitine/toxicity , Fetus/drug effects , Lung/embryology , Animals , Female , Fetal Organ Maturity/drug effects , Lung/drug effects , Maternal-Fetal Exchange , Pregnancy , Rabbits
16.
Metabolism ; 36(12): 1192-6, 1987 Dec.
Article in English | MEDLINE | ID: mdl-3683188

ABSTRACT

A study was designed to examine the hypolipidemic effect of L-carnitine treatment (4 weeks, 170 mg/kg/d) in rabbits fed a high fat diet (5% corn oil/0.5% cholesterol, w/w). Eight weeks of exposure to the high fat diet significantly increased plasma total cholesterol and triglycerides. VLDL associated triglycerides, cholesterol, apo-B, and total protein were also significantly increased with the diet. There was no change in HDL-cholesterol levels. Plasma concentration of carnitine (free, acyl, and total) all increased significantly with the high fat diet. The content of free, short-chain, and total carnitine were decreased in the liver whereas the content of long-chain acylcarnitines was increased. The diet generated a significant steatosis within the livers of these animals. Four weeks of treatment of L-carnitine reduced the extent of the liver steatosis and significantly decreased plasma total cholesterol, triglycerides, VLDL associated triglycerides, cholesterol, and total protein. HDL-cholesterol levels were unaffected by the treatment. All plasma fractions of carnitine (free, acetyl, acyl, and total) were significantly increased above those levels seen after 8 weeks of the high fat diet alone. The content of liver carnitine and its esters was normalized following treatment. The high fat diet decreased liver HMG-CoA reductase activity and increased the activities of 7-alpha-hydroxylase and acylcholesterol acyltransferase (ACAT). L-Carnitine treatment blunted the magnitude of the diet induced increase in 7 alpha-hydroxylase activity, yet overall the activity still remained elevated relative to controls. ACAT activity increased (1.5 times) with the high fat diet and increased further (4.5 times) following carnitine treatment.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Carnitine/therapeutic use , Hyperlipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Lipids/blood , Animals , Body Weight/drug effects , Carnitine/toxicity , Dietary Fats/administration & dosage , Fatty Liver/chemically induced , Hyperlipidemias/blood , Hypolipidemic Agents/toxicity , Liver/enzymology , Male , Rabbits
17.
Teratology ; 32(2): 267-72, 1985 Oct.
Article in English | MEDLINE | ID: mdl-4049285

ABSTRACT

A single 1.0-ml volume of L- or D-carnitine solution, at several selected mmole concentrations, was applied to the extraembryonic membranes of 3- and 4-day chick embryos in ovo. Hamburger-Hamilton stages of chick development ranged from 17 to 23. During the 17-18th days of incubation, embryos were dissected, and both survival and intracardiac anomaly rates were determined. Only at extremely high doses, both stereoisomers of carnitine exhibited a statistically significant toxigenic effect (p less than 0.001) as measured by a sharp decrease in survival rate when compared to chick Ringer's saline controls. Furthermore, since the anomaly rates became significant only near the LD50's, this indicated that intracardiac anomalies were induced only at toxic doses. Therefore, it is suggested that cardiovascular teratogenicity may be the result of toxicity. Below the LD50, anomaly rates were not significantly different from those of control embryos. In comparison, L- and D-carnitine were significantly different from one another (p less than 0.001) both in survival rate and percent affected embryos at a dose of 0.5 mmole. In summary, exogenous carnitine administration to the chick embryo does not appear to be deleterious to the developing cardiovascular system.


Subject(s)
Cardiovascular System/embryology , Carnitine/toxicity , Teratogens , Animals , Cardiovascular Abnormalities , Cardiovascular System/drug effects , Chick Embryo , Dose-Response Relationship, Drug , Lethal Dose 50 , Stereoisomerism , Structure-Activity Relationship
18.
Arch Toxicol ; 55(4): 239-46, 1984 Oct.
Article in English | MEDLINE | ID: mdl-6517701

ABSTRACT

A primary rat hepatocyte culture system has been developed for the study of peroxisome proliferation. Maximal induction of peroxisomal activity requires supplementation of the culture medium with hydrocortisone. The addition of clofibric acid (0.01-1 mM), mono-(2-ethylhexyl)phthalate (0.01-0.5 mM) and trichloroacetic acid (0.1-5 mM) to cultured rat hepatocytes resulted in a time- and dose-related increase in CN- insensitive palmitoyl CoA oxidation (maximal increases: 27-, 15.5-, and 5-fold respectively) and mitochondrial alpha-glycerophosphate dehydrogenase activity (maximal increases: 7.3-, 5.8-, and 1.6-fold respectively). Electron microscopic examination revealed smooth endoplasmic reticulum proliferation and morphometric analysis indicated an increase in fractional peroxisomal volume of X 8 and X 4 for clofibric acid (1 mM) and trichloroacetic acid (2.5 mM), respectively. SDS-PAGE of cell homogenates revealed an intensified protein band of mol. wt. 76-78,000. The induction of peroxisomal beta-oxidation by clofibric acid was elevated from 9- to 12-fold by supplementation of the medium with L-carnitine (2 mM).


Subject(s)
Liver/drug effects , Microbodies/drug effects , Animals , Carnitine/toxicity , Cell Division/drug effects , Cells, Cultured , Clofibric Acid/toxicity , Diethylhexyl Phthalate/analogs & derivatives , Diethylhexyl Phthalate/toxicity , Dose-Response Relationship, Drug , Glucosephosphate Dehydrogenase/analysis , Hydrocortisone/pharmacology , Male , Microbodies/metabolism , Palmitoyl Coenzyme A/metabolism , Rats , Rats, Inbred Strains , Trichloroacetic Acid/toxicity
19.
Acta Neurol Scand Suppl ; 100: 39-47, 1984.
Article in English | MEDLINE | ID: mdl-6148832

ABSTRACT

More than 40 drugs are known to increase muscle relaxation, to aggravate myasthenia gravis, or to induce myasthenic syndromes in "normal" individuals with or without a known impaired safety factor. Many of these drugs have been known for a long time. In the last ten years some other drugs have been added to the long list: Penicillamine, beta-adrenergic blockers, carnitine, some antibiotics, lithium carbonate, catharactics containing magnesium salts, etc. An immunological basis for the penicillamine-induced myasthenia gravis is generally approved, but there may be other examples such as trimethadione and phenytoin. The knowledge of these possible side effects is important for the interpretation of unwanted effects, particularly diplopia and other mild myasthenic symptoms.


Subject(s)
Muscular Diseases/chemically induced , Adrenergic beta-Antagonists/adverse effects , Adult , Anti-Arrhythmia Agents/adverse effects , Anti-Bacterial Agents/adverse effects , Autoimmune Diseases/chemically induced , Baclofen/adverse effects , Carnitine/toxicity , Collagen Diseases/chemically induced , Diazepam/adverse effects , Female , Humans , Male , Middle Aged , Myasthenia Gravis/chemically induced , Neuromuscular Junction/drug effects , Penicillamine/adverse effects , Phenytoin/adverse effects , Streptomycin/adverse effects , Synaptic Transmission/drug effects , Syndrome
20.
J Submicrosc Cytol ; 14(2): 337-45, 1982 Apr.
Article in English | MEDLINE | ID: mdl-7077714

ABSTRACT

A quantitative ultrastructural analysis of leptomeres in cardiac muscle was performed using an adriamycin/carnitine chronic mouse model. Five animals from each of the following four groups were sacrificed for evaluation by electron microscopy: the control group received saline injections only; the carnitine group received carnitine orally; the adriamycin group received a chronic dose of adriamycin (10 mg/kg over 4 weeks); the adriamycin/carnitine group received both the adriamycin and carnitine dosage regimens. The leptomeres were quantitated using a double-blind method. The total number of leptomeres were scored for 20 random low-power electron micrographs from each of 5 animals within each group. The adriamycin group contained 3 of the 4 lowest leptomere scores. The relationships between myofibrillar disruption (MD) and the number of leptomeres found in an intramyofibrillar location(IML) were then determined for each of the four groups using linear regression analysis. In the control group, increased MD was associated with a decrease in the number of IML. In the carnitine group, increased MD was associated with an increase in IML. In the adriamycin group, no significant change in the number of IML occurred with increasing levels of MD. The adriamycin/carnitine group was similar to the control group. It was concluded from this study that 1) a relationship exists between MD and the number of IML in normal murine cardiac muscle; 2) carnitine produces an inverse effect on this normal relationship; 3) adriamycin disturbs the normal relationship between IML and MD; and 4) the administration of carnitine to adriamycin-treated animals restored the normal relationship. The physiological significance of the leptomere to the cardiac cell is then discussed.


Subject(s)
Carnitine/toxicity , Doxorubicin/toxicity , Myocardium/ultrastructure , Myofibrils/ultrastructure , Animals , Mice , Microscopy, Electron , Myocardium/pathology , Myofibrils/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...