Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 156: 113926, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36308917

ABSTRACT

Natalizumab is a recombinant, humanized form of a monoclonal antibody that binds to CD49d. The presented study was conducted to explore the suitability of Göttingen pigs as a pharmacokinetic/pharmacodynamic model in the preclinical phase of biosimilar natalizumab development. The minipigs were treated with 1.286 or 3.0 mg of natalizumab (Tysabri®) per kg of body weight by a single 1-hour intravenous infusion. Six days before (baseline) and 30 days after administration of the single dose of natalizumab, blood samples were taken for analysis. No signs of local or general intolerance were observed. The pharmacokinetics plot shows a biphasic profile dependent on anti-drug antibody (ADA) levels. A dose-related increase in the CD49d saturation was observed immediately after the end of the infusion. The soluble vascular cell adhesion molecule (sVCAM) concentrations of the female animals were moderately decreased immediately after the end of infusion compared to the predose levels. The soluble mucosal addressin cell adhesion molecule (sMAdCAM) concentrations were slightly decreased compared to the predose levels starting immediately after the end of infusion and lasting for the next 30 days. All animals treated appeared to produce ADA. The concentrations of the ADA ranged from 15.8 to 16,748 ng/mL Göttingen pigs represent a suitable model for pharmacokinetic analysis and mechanism of action evaluation related to saturation of CD49d.


Subject(s)
Antibodies, Monoclonal, Humanized , Integrin alpha4 , Female , Animals , Swine , Natalizumab/pharmacology , Natalizumab/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Swine, Miniature , Antibodies, Monoclonal/therapeutic use
2.
J Vet Sci ; 22(5): e47, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34423596

ABSTRACT

BACKGROUND: Due to multiple similarities in the structure and physiology of human and pig skin, the pig model is extremely useful for biological drug testing after subcutaneous administration. Knowledge of the differences between subcutaneous injection sites could have a significant impact on the absorption phase and pharmacokinetic profiles of biological drugs. OBJECTIVES: This study aimed to analyze the impact of administration site on pharmacokinetics and selected biochemical and hematological parameters after a single subcutaneous administration of ustekinumab in pigs. Drug concentrations in blood plasma were analyzed by enzyme-linked immunosorbent assay. Pharmacokinetic analyses were performed based on raw data using Phoenix WinNonlin 8.1 software and ThothPro v 4.1. METHODS: The study included 12 healthy, female, large white piglets. Each group received a single dose of ustekinumab given as a 1 mg/kg subcutaneous injection into the internal part of the inguinal fold or the external part of the inguinal fold. RESULTS: The differences in absorption rate between the internal and external parts of the inguinal fold were not significant. However, the time of maximal concentration, clearance, area under the curve calculated between zero and mean residence time and mean residence time between groups were substantially different (p > 0.05). The relative bioavailability after administration of ustekinumab into the external part of the inguinal fold was 40.36% lower than after administration of ustekinumab into the internal part of the inguinal fold. CONCLUSIONS: Healthy breeding pigs are a relevant model to study the pharmacokinetic profile of subcutaneously administered ustekinumab.


Subject(s)
Dermatologic Agents/pharmacokinetics , Sus scrofa/metabolism , Ustekinumab/pharmacokinetics , Animals , Biological Availability , Female , Humans , Injections, Subcutaneous , Models, Animal
4.
Stem Cell Res Ther ; 6: 61, 2015 Apr 12.
Article in English | MEDLINE | ID: mdl-25889676

ABSTRACT

INTRODUCTION: Diabetes is associated with reduced expression of heme oxygenase-1 (HO-1), a heme-degrading enzyme with cytoprotective and proangiogenic properties. In myoblasts and muscle satellite cells HO-1 improves survival, proliferation and production of proangiogenic growth factors. Induction of HO-1 in injured tissues facilitates neovascularization, the process impaired in diabetes. We aimed to examine whether conditioned media from the HO-1 overexpressing myoblast cell line can improve a blood-flow recovery in ischemic muscles of diabetic mice. METHODS: Analysis of myogenic markers was performed at the mRNA level in primary muscle satellite cells, isolated by a pre-plate technique from diabetic db/db and normoglycemic wild-type mice, and then cultured under growth or differentiation conditions. Hind limb ischemia was performed by femoral artery ligation in db/db mice and blood recovery was monitored by laser Doppler measurements. Mice were treated with a single intramuscular injection of conditioned media harvested from wild-type C2C12 myoblast cell line, C2C12 cells stably transduced with HO-1 cDNA, or with unconditioned media. RESULTS: Expression of HO-1 was lower in muscle satellite cells isolated from muscles of diabetic db/db mice when compared to their wild-type counterparts, what was accompanied by increased levels of Myf5 or CXCR4, and decreased Mef2 or Pax7. Such cells also displayed diminished differentiation potential when cultured in vitro, as shown by less effective formation of myotubes and reduced expression of myogenic markers (myogenic differentiation antigen - myoD, myogenin and myosin). Blood flow recovery after induction of severe hind limb ischemia was delayed in db/db mice compared to that in normoglycemic individuals. To improve muscle regeneration after ischemia, conditioned media collected from differentiating C2C12 cells (control and HO-1 overexpressing) were injected into hind limbs of diabetic mice. Analysis of blood flow revealed that media from HO-1 overexpressing cells accelerated blood-flow recovery, while immunohistochemical staining assessment of vessel density in injected muscle confirmed increased angiogenesis. The effect might be mediated by stromal-cell derived factor-1α proangiogenic factor, as its secretion is elevated in HO-1 overexpressing cells. CONCLUSIONS: In conclusion, paracrine stimulation of angiogenesis in ischemic skeletal muscle using conditioned media may be a safe approach exploiting protective and proangiogenic properties of HO-1 in diabetes.


Subject(s)
Culture Media, Conditioned/pharmacology , Diabetes Mellitus, Experimental/pathology , Muscle, Skeletal/physiology , Neovascularization, Physiologic/drug effects , Regeneration/drug effects , Animals , Cells, Cultured , Chemokine CXCL12/analysis , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Ischemia/complications , Ischemia/metabolism , Ischemia/pathology , MEF2 Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Myoblasts/cytology , Myoblasts/metabolism , Myogenic Regulatory Factor 5/metabolism , PAX7 Transcription Factor/metabolism , Receptors, CXCR4/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Vascular Endothelial Growth Factor A/analysis
5.
Cardiovasc Diabetol ; 13: 150, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25361524

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor-γ (PPARγ) agonists, which have been used as insulin sensitizers in diabetic patients, may improve functions of endothelial cells (ECs). We investigated the effect of PPARγ on angiogenic activities of murine ECs and bone marrow-derived proangiogenic cells (PACs). METHODS: PACs were isolated from bone marrow of 10-12 weeks old, wild type, db/db and PPARγ heterozygous animals. Cells were cultured on fibronectin and gelatin coated dishes in EGM-2MV medium. For in vitro stimulations, rosiglitazone (10 µmol/L) or GW9662 (10 µmol/L) were added to 80% confluent cell cultures for 24 hours. Angiogenic potential of PACs and ECs was tested in vitro and in vivo in wound healing assay and hind limb ischemia model. RESULTS: ECs and PACs isolated from diabetic db/db mice displayed a reduced angiogenic potential in ex vivo and in vitro assays, the effect partially rescued by incubation of cells with rosiglitazone (PPARγ activator). Correction of diabetes by administration of rosiglitazone in vivo did not improve angiogenic potential of isolated PACs or ECs. In a hind limb ischemia model we demonstrated that local injection of conditioned media harvested from wild type PACs improved the blood flow restoration in db/db mice, confirming the importance of paracrine action of the bone marrow-derived cells. CONCLUSIONS: In summary, activation of PPARγ by rosiglitazone improves angiogenic potential of diabetic ECs and PACs, but decreased expression of PPARγ in diabetes does not impair angiogenesis.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow/drug effects , Endothelial Cells/drug effects , PPAR gamma/metabolism , Stem Cells/metabolism , Animals , Bone Marrow Cells/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Hypoglycemic Agents/pharmacology , Ischemia/drug therapy , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , PPAR gamma/genetics , Rosiglitazone , Stem Cells/cytology , Stem Cells/drug effects , Thiazolidinediones/pharmacology , Wound Healing/drug effects
6.
Biochem Cell Biol ; 92(1): 1-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24471913

ABSTRACT

Effects of equimolar concentrations of proinsulin C-peptide and insulin on glucose synthesis were studied in primary cultures of rabbit kidney-cortex tubules grown in the presence of alanine, glycerol, and octanoate. The rhodamine-labeled C-peptide entered renal tubular cells and localized in nuclei, both in the presence and absence of insulin; preincubations with the unlabeled compound inhibited internalization. C-peptide did not affect glucose formation when added alone but potentiated the inhibitory action of insulin by about 20% due to a decrease in flux through glucose-6-phosphate isomerase (GPI) and (or) glucose-6-phosphatase (G6Pase). GPI inhibition was caused by: (i) increased intracellular contents of fructose-1,6-bisphosphate and fructose-1-phosphate, inhibitors of the enzyme and (ii) reduced level of the phosphorylated GPI, which exhibits higher enzymatic activity in the presence of casein kinase 2. A decrease in flux through G6Pase, due to diminished import of G6P by G6P-transporter from the cytoplasm into endoplasmic reticulum lumen, is also suggested. The data show for the first time that in the presence of insulin and C-peptide, both GPI and G6P-ase may act as regulatory enzymes of renal gluconeogenic pathway.


Subject(s)
C-Peptide/metabolism , Glucose/biosynthesis , Insulin/metabolism , Kidney Tubules/metabolism , Animals , C-Peptide/pharmacology , Cells, Cultured , Humans , Insulin/pharmacology , Kidney Tubules/cytology , Kidney Tubules/drug effects , Male , Rabbits
7.
PLoS One ; 8(5): e64805, 2013.
Article in English | MEDLINE | ID: mdl-23705015

ABSTRACT

Anserine (beta-alanyl-N(Pi)-methyl-L-histidine), a naturally occurring derivative of carnosine (beta-alanyl-L-histidine), is an abundant constituent of skeletal muscles and brain of many vertebrates. Although it has long been proposed to serve as a proton buffer, radicals scavenger and transglycating agent, its physiological function remains obscure. The formation of anserine is catalyzed by carnosine N-methyltransferase which exhibits unknown molecular identity. In the present investigation, we have purified carnosine N-methyltransferase from chicken pectoral muscle about 640-fold until three major polypeptides of about 23, 26 and 37 kDa coeluting with the enzyme were identified in the preparation. Mass spectrometry analysis of these polypeptides resulted in an identification of histamine N-methyltransferase-like (HNMT-like) protein as the only meaningful candidate. Analysis of GenBank database records indicated that the hnmt-like gene might be a paralogue of histamine N-methyltransferase gene, while comparison of their protein sequences suggested that HNMT-like protein might have acquired a new activity. Chicken HNMT-like protein was expressed in COS-7 cells, purified to homogeneity, and shown to catalyze the formation of anserine as confirmed by both chromatographic and mass spectrometry analysis. Both specificity and kinetic studies carried out on the native and recombinant enzyme were in agreement with published data. Particularly, several compounds structurally related to carnosine, including histamine and L-histidine, were tested as potential substrates for the enzyme, and carnosine was the only methyl group acceptor. The identification of the gene encoding carnosine N-methyltransferase might be beneficial for estimation of the biological functions of anserine.


Subject(s)
Carnosine/metabolism , Histamine N-Methyltransferase/metabolism , Methyltransferases/metabolism , Amino Acid Sequence , Animals , Anserine/biosynthesis , Blotting, Western , COS Cells , Chickens , Chlorocebus aethiops , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Histamine N-Methyltransferase/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Mass Spectrometry , Methylation , Molecular Sequence Data , Muscles/enzymology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Time Factors
8.
Antioxid Redox Signal ; 16(2): 113-27, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21827279

ABSTRACT

AIMS: Heme oxygenase-1 (HMOX1) is a cytoprotective enzyme degrading heme to biliverdin, iron ions, and carbon monoxide, whose expression is induced in response to oxidative stress. Its overexpression has been suggested as a strategy improving survival of transplanted muscle precursors. RESULTS: Here we demonstrated that HMOX1 inhibits differentiation of myoblasts and modulates miRNA processing: downregulates Lin28 and DGCR8, lowers the total pool of cellular miRNAs, and specifically blocks induction of myomirs. Genetic or pharmacological activation of HMOX1 in C2C12 cells reduces the abundance of miR-1, miR-133a, miR-133b, and miR-206, which is accompanied by augmented production of SDF-1 and miR-146a, decreased expression of MyoD, myogenin, and myosin, and disturbed formation of myotubes. Similar relationships between HMOX1 and myomirs were demonstrated in murine primary satellite cells isolated from skeletal muscles of HMOX1(+/+), HMOX1(+/-), and HMOX1(-/-) mice or in human rhabdomyosarcoma cell lines. Inhibition of myogenic development is independent of antioxidative properties of HMOX1. Instead it is mediated by CO-dependent inhibition of c/EBPδ binding to myoD promoter, can be imitated by SDF-1, and partially reversed by enforced expression of miR-133b and miR-206. Control C2C12 myoblasts injected to gastrocnemius muscles of NOD-SCID mice contribute to formation of muscle fibers. In contrast, HMOX1 overexpressing C2C12 myoblasts form fast growing, hyperplastic tumors, infiltrating the surrounding tissues, and disseminating to the lungs. INNOVATION: We evidenced for the first time that HMOX1 inhibits differentiation of myoblasts, affects the miRNA processing enzymes, and modulates the miRNA transcriptome. CONCLUSION: HMOX1 improves the survival of myoblasts, but concurrently through regulation of myomirs, may act similarly to oncogenes, increasing the risk of hyperplastic growth of myogenic precursors.


Subject(s)
Cell Differentiation/physiology , Heme Oxygenase-1/physiology , Myoblasts/cytology , Animals , Cell Line , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Transcriptome
9.
Postepy Biochem ; 57(2): 200-6, 2011.
Article in Polish | MEDLINE | ID: mdl-21913421

ABSTRACT

Chronic metabolic and cardiovascular diseases, described as the epidemics of XXI century, are connected to the resistance of peripheral tissues, such as liver, muscle and fat, to insulin. Insulin resistance, which precedes the development of type 2 diabetes by several years, is difficult to diagnose, mainly because of practical limitations to the use of "gold standard" hyperinsulinemic euglycemic clamp technique for screening. It is also begins a certain vicious circle, in which insulin resistant peripheral tissues force pancreatic beta cells to increased insulin release, and sustained high concentrations of insulin cause further development of insulin resistance. Currently, there are two major hypotheses describing the mechanism of insulin resistance: one relating to the "lipid overload" in liver and muscle cells as the key factor and another one emphasizing the role of lipid accumulation in adipocytes, which leads to the overgrowth of fatty tissue and chronic local inflammation.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Lipid Metabolism , Muscle, Skeletal/metabolism , Adipocytes/metabolism , Humans , Insulin Secretion , Liver/metabolism
10.
Postepy Biochem ; 57(2): 207-14, 2011.
Article in Polish | MEDLINE | ID: mdl-21913422

ABSTRACT

Nuclear receptors regulate many basic cellular processes and their malfunction can lead to serious consequences including metabolic disorders, obesity and type 2 diabetes. Among many nuclear receptor families, the best known for their therapeutic use are the PPARs. These are key transcription factors determining, proper cellular metabolism of glucose and lipids, tissue sensitivity to insulin, appropriate immune responses including inflammatory processes and finally cell division and differentiation. Currently two types of PPAR activators are in medical use: in the therapy of type 2 diabetes--thiazolidinediones (TZDs), which act via PPARgamma receptors and in the treatment of dyslipidemia-fibrates, which act via PPARalpha receptors. The search for new drugs acting through PPAR mechanism consists in the design of new molecules with tissue specific proprieties, which would selectively bind and modulate the activity of appropriate receptors, thus reducing the number of adverse events typically observed with the use of full agonists. These molecules have been named selective nuclear receptor modulators (SNuRMs).


Subject(s)
Metabolic Diseases/drug therapy , Peroxisome Proliferator-Activated Receptors/agonists , Diabetes Mellitus, Type 2/drug therapy , Drug Design , Humans , Metabolic Diseases/metabolism , PPAR alpha/agonists , PPAR gamma/agonists , Peroxisome Proliferator-Activated Receptors/metabolism , Thiazolidinediones/pharmacology
11.
Genet Vaccines Ther ; 8: 6, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20804557

ABSTRACT

BACKGROUND: Impaired wound healing in diabetes is related to decreased production of growth factors. Hence, gene therapy is considered as promising treatment modality. So far, efforts concentrated on single gene therapy with particular emphasis on vascular endothelial growth factor-A (VEGF-A). However, as multiple proteins are involved in this process it is rational to test new approaches. Therefore, the aim of this study was to investigate whether single AAV vector-mediated simultaneous transfer of VEGF-A and fibroblast growth factor 4 (FGF4) coding sequences will improve the wound healing over the effect of VEGF-A in diabetic (db/db) mice. METHODS: Leptin receptor-deficient db/db mice were randomized to receive intradermal injections of PBS or AAVs carrying ß-galactosidase gene (AAV-LacZ), VEGF-A (AAV-VEGF-A), FGF-4 (AAV-FGF4-IRES-GFP) or both therapeutic genes (AAV-FGF4-IRES-VEGF-A). Wound healing kinetics was analyzed until day 21 when all animals were sacrificed for biochemical and histological examination. RESULTS: Complete wound closure in animals treated with AAV-VEGF-A was achieved earlier (day 19) than in control mice or animals injected with AAV harboring FGF4 (both on day 21). However, the fastest healing was observed in mice injected with bicistronic AAV-FGF4-IRES-VEGF-A vector (day 17). This was paralleled by significantly increased granulation tissue formation, vascularity and dermal matrix deposition. Mechanistically, as shown in vitro, FGF4 stimulated matrix metalloproteinase-9 (MMP-9) and VEGF receptor-1 expression in mouse dermal fibroblasts and when delivered in combination with VEGF-A, enhanced their migration. CONCLUSION: Combined gene transfer of VEGF-A and FGF4 can improve reparative processes in the wounded skin of diabetic mice better than single agent treatment.

12.
Biochem Cell Biol ; 86(5): 396-404, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18923541

ABSTRACT

Therapeutic effect of rosiglitazone has been reported to result from an improvement of insulin sensitivity and inhibition of glucose synthesis. As the latter process occurs in both liver and kidney cortex the aim of this study was to elucidate the rosiglitazone action on glucose formation in both tissues. Primary cultured cells of both liver and kidney cortex grown in defined medium were use throughout. To identify the mechanism responsible for drug-induced changes, intracellular gluconeogenic intermediates and enzyme activities were determined. In contrast to hepatocytes, the administration of a 10 micromol/L concentration of rosiglitazone to renal tubules resulted in about a 70% decrease in the rate of gluconeogenesis, accompanied by an approximately 75% decrease in alanine utilization and a 35% increase in lactate synthesis. The effect of rosiglitazone was not abolished by GW9662, the PPAR-gamma irreversible antagonist, indicating that this action is not dependent on PPAR-gamma activation. In view of rosiglitazone-induced changes in gluconeogenic intermediates and a diminished incorporation of 14CO2 into pyruvate, it is likely that the drug causes a decline in flux through pyruvate carboxylase and (or) phosphoenolpyruvate carboxykinase. It is likely that the hypoglycemic action of rosiglitazone is PPAR-gamma independent and results mainly from its inhibitory effects on renal gluconeogenesis.


Subject(s)
Gluconeogenesis/drug effects , Hypoglycemic Agents/pharmacology , Kidney Cortex/anatomy & histology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , PPAR gamma/metabolism , Thiazolidinediones/pharmacology , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/metabolism , Cells, Cultured , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Kidney Tubules/cytology , Lactic Acid/metabolism , Male , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Rabbits , Rosiglitazone , Trimethoprim/metabolism
13.
Chem Biol Interact ; 170(3): 162-76, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17767924

ABSTRACT

The action of selegiline, a selective and irreversible inhibitor of monoamine oxidase B, commonly applied in the therapy of Parkinson's disease, on glucose formation was investigated in isolated rabbit hepatocytes and kidney-cortex tubules, maintaining the whole body glucose homeostasis via gluconeogenic pathway activity. An intensive hepatic metabolism of selegiline resulted in formation of selegiline-N-oxide, desmethylselegiline, methamphetamine and amphetamine, whereas during slow degradation of the drug in freshly isolated renal tubules selegiline-N-oxide was mainly produced. At 100 microM concentration selegiline markedly diminished glucose synthesis in isolated renal tubules incubated with dihydroxyacetone or alanine+glycerol+octanoate (by about 60 and 30%, respectively), while at 5 microM concentration a similar degree of inhibition was achieved in renal tubules grown in primary culture under the same conditions (about 40 and 60%, respectively). Moreover, desmethylselegiline and selegiline-N-oxide considerably diminished glucose production in renal tubules whereas selegiline and its metabolites did not affect gluconeogenesis in hepatocytes. Contrary to control animals, following selegiline administration to alloxan-diabetic rabbits for 8 days (10 mg kg(-1) body wt. daily) the blood glucose and serum creatinine levels were significantly diminished, suggesting a decrease in renal gluconeogenesis and improvement of kidney functions. Since in renal tubules selegiline induced a decline in the intracellular levels of gluconeogenic intermediates and ATP content accompanied by a decrease in oxygen consumption in both kidney-cortex and hepatic mitochondria it seems possible that its inhibitory action on renal gluconeogenesis might result from an impairment of mitochondrial function, while an intensive selegiline metabolism in hepatocytes causes decrease of its concentration and in consequence no inhibition of gluconeogenesis. In view of these observations it is likely that an increased risk of selegiline-induced hypoglycemia might be expected particularly in patients exhibiting an impairment of liver function and following transdermal administration of this drug, i.e. under conditions of increased serum selegiline concentrations.


Subject(s)
Glucose/biosynthesis , Hepatocytes/drug effects , Hepatocytes/metabolism , Kidney Cortex/drug effects , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Selegiline/pharmacology , Adenosine Triphosphate/metabolism , Animals , Body Weight/drug effects , Cell Separation , Cells, Cultured , Kidney Cortex/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Oxygen/metabolism , Rabbits , Selegiline/chemistry , Selegiline/metabolism , Superoxides/metabolism
14.
Yeast ; 24(10): 871-82, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17640084

ABSTRACT

Murine DNA methyltransferases Dnmt1 and Dnmt3a were expressed in the yeast Saccharomyces cerevisiae. Adjustment to yeast preferences of the nucleotide sequences upstream and downstream of the translation initiation sites of both cDNAs was needed to obtain significant levels of the methyltransferases. Both proteins were correctly localized to the nucleus and their presence had no measurable influence on the functioning of yeast cells. Both Dnmt1 and Dnmt3a expressed in yeast cells were enzymatically active in vitro, and in vivo in the genomic DNA of the transgenic S. cerevisiae ca. 0.06% and 0.4%, respectively, of cytosines became methylated. This level of DNA methylation is about 100- to 10-fold less than that observed in mammalian cells. The constructed system may be used to investigate the in vivo specificity of individual mammalian DNA methyltransferases and to search for additional factors needed to allow more efficient in vivo methylation of chromatin-contained DNA and to study their mechanism of action.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Saccharomyces cerevisiae/genetics , Animals , Base Sequence , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Mice , Molecular Sequence Data
15.
J Pineal Res ; 42(2): 203-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17286753

ABSTRACT

The antioxidative effects of melatonin (Mel), 5-hydroxytryptophan (5-HTP) and taurine (TAU) on hyperglycemia-induced oxidative stress was investigated in primary cultures of kidney-cortex tubule cells grown in metabolically and hormonally defined medium. In the presence of 30 mm glucose (hyperglycemic conditions), cell viability was decreased by about 35% in comparison with that estimated in the glucose-depleted medium probably as a result of induction of apoptosis, as concluded from: (i) chromatin condensation and DNA fragmentation assays, (ii) a significant enhancement of reactive oxygen species (ROS) production, (iii) 8-hydroxydeoxyguanosine (8-OHdG) generation, (iv) an increased protein peroxidation and (v) a decline of reduced glutathione (GSH) levels leading to a disturbed glutathione redox state. The addition of 100 microm Mel to the hyperglycemic medium resulted in a twofold decrease in both 8-OHdG accumulation and protein peroxidation as well as restoration of the control intracellular ROS levels accompanied by a substantial increase in GSH/oxidized glutathione (GSSG) ratio due to a decline in GSSG content. ROS elimination was also achieved in the presence of 1 mm TAU which diminished protein and DNA injuries by about 25% and 30%, respectively. On the contrary, the action of 100 microm 5-HTP on ROS level, 8-OHdG generation, protein peroxidation and GSH/GSSG ratio was negligible. Thus, in contrast to 5-HTP and TAU, Mel might be considered as beneficial for diabetes therapy, particularly in terms of reduction of hyperglycemia-induced kidney injury.


Subject(s)
5-Hydroxytryptophan/physiology , Hyperglycemia/metabolism , Kidney Cortex/metabolism , Kidney Tubules/metabolism , Melatonin/physiology , Taurine/physiology , Animals , Hyperglycemia/complications , Hyperglycemia/pathology , Kidney Cortex/pathology , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney Tubules/pathology , Male , Organ Culture Techniques , Rabbits
16.
J Inorg Biochem ; 101(3): 493-505, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17222910

ABSTRACT

Although selenium is taken with diet mainly as selenoamino acids, its hypoglycaemic action on hepatic gluconeogenesis has been studied with the use of inorganic selenium derivatives. The aim of the present investigation was to compare relative efficacies of inorganic and organic selenium compounds in reducing glucose synthesis in hepatocytes and renal tubules, significantly contributing to the glucose homeostasis. In contrast to hepatocytes, both selenite and methylselenocysteine inhibited renal gluconeogenesis by about 40-45% in control rabbits. Selenate did not affect this process, whereas selenomethionine inhibited gluconeogenesis by about 20% in both hepatocytes and renal tubules. In contrast to methylselenocysteine, selenite decreased intracellular ATP content, glutathione reduced/glutathione oxidized (GSH/GSSG) ratio and pyruvate carboxylase, PEPCK and FBPase activities, while methylselenocysteine diminished PEPCK activity due to elevation of intracellular 2-oxoglutarate and GSSG, inhibitors of this enzyme. Experiments in vivo indicate that in 3 of 9 alloxan-diabetic rabbits treated for 14 days with methylselenocysteine (0.182mg/kg body weight) blood glucose level was normalized, whereas in all diabetic rabbits plasma creatinine and urea levels decreased from 2.52+/-0.18 and 87.4+/-9.7 down to 1.63+/-0.11 and 39.0+/-2.8, respectively. In view of these data selenium supplementation might be beneficial for protection against diabetes-induced nephrotoxicity despite selenium accumulation in kidneys and liver.


Subject(s)
Blood Glucose/drug effects , Gluconeogenesis/drug effects , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Kidney Tubules/drug effects , Selenium Compounds/pharmacology , Alloxan , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Glucose-6-Phosphatase/metabolism , Hepatocytes/metabolism , Kidney Tubules/metabolism , Male , Pyruvate Carboxylase/metabolism , Rabbits
17.
J Pineal Res ; 38(3): 164-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15725337

ABSTRACT

The effect of melatonin on glucose metabolism in the presence and absence of insulin has been investigated in the primary cultures of renal tubules grown in a defined medium. In the absence of glucose in the medium containing 5 microg/mL of insulin and 2 mm alanine + 5 mm glycerol + 0.5 mm octanoate, 100 nm melatonin stimulated both glucose and lactate synthesis, while in the medium devoid of insulin melatonin action was negligible. Melatonin-induced increase in glucose and lactate synthesis was accompanied by an enhancement of alanine and glycerol consumption. In view of measurements of [U-14C]L-alanine and [U-14C]L-glycerol incorporation into glucose, it is likely that melatonin increased alanine utilization for glucose production, while accelerated lactate synthesis was because of an enhanced glycerol consumption. As (i) 10 nm luzindole attenuated the stimulatory action of melatonin on glucose formation and (ii) the indole induced a decrease in intracellular cAMP level, it seems likely that in renal tubules melatonin binds to ML1 membrane receptor subtype. In view of a decline of intracellular fructose-1,6-bisphosphate content accompanied by a significant rise in hexose-6-phosphate and glucose levels, melatonin might result in an acceleration of flux through fructose-1,6-bisphosphatase probably because of an increase in the active, dephosphorylated form of this enzyme. Thus, the administration of melatonin in combination with insulin might be beneficial for diabetic therapy because of protection against hypoglycemia.


Subject(s)
Glucose/metabolism , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Melatonin/pharmacology , Alanine/metabolism , Animals , Diabetes Mellitus/drug therapy , Fructose-Bisphosphatase/metabolism , Gluconeogenesis/drug effects , Glycerol/metabolism , Insulin/pharmacology , Kinetics , Lactic Acid/metabolism , Melatonin/metabolism , Rabbits , Receptor, Melatonin, MT1/drug effects , Receptor, Melatonin, MT1/metabolism , Tissue Culture Techniques , Tryptamines/pharmacology
18.
Arch Biochem Biophys ; 434(2): 282-8, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15639228

ABSTRACT

Effects of various cAMP analogues on gluconeogenesis in isolated rabbit kidney tubules have been investigated. In contrast to N(6),2'-O-dibutyryladenosine-3',5'-cyclic monophosphate (db-cAMP) and cAMP, which accelerate renal gluconeogenesis, 8-bromoadenosine-3',5'-cyclic monophosphate (Br-cAMP) and 8-(4-chlorophenylthio)-cAMP (pCPT-cAMP) inhibit glucose production. Stimulatory action of cAMP and db-cAMP may be evoked by butyrate and purinergic agonists generated during their extracellular and intracellular metabolism resulting in an increase in flux through fructose-1,6-bisphosphatase and in consequence acceleration of the rate of glucose formation. On the contrary, Br-cAMP is poorly metabolized in renal tubules and induces a fall of flux through glyceraldehyde-3-phosphate dehydrogenase. The contribution of putative extracellular cAMP receptors to the inhibitory Br-cAMP action is doubtful in view of a decline of glucose formation in renal tubules grown in the primary culture supplemented with forskolin. The presented data indicate that in contrast to hepatocytes, in kidney-cortex tubules an increased intracellular cAMP level results in an inhibition of glucose production.


Subject(s)
Cyclic AMP/metabolism , Gluconeogenesis/physiology , Kidney Tubules/metabolism , Kidney/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/metabolism , Animals , Colforsin/pharmacology , Dose-Response Relationship, Drug , Fructose-Bisphosphatase/metabolism , Glucose/metabolism , Hepatocytes/metabolism , Kidney Cortex/metabolism , Male , Rabbits , Time Factors
19.
Alcohol Alcohol ; 39(2): 93-100, 2004.
Article in English | MEDLINE | ID: mdl-14998823

ABSTRACT

AIMS: The effect of ethanol on glucose synthesis in kidney-cortex tubules of control and diabetic rabbits has been investigated. METHODS: Both freshly isolated and grown in primary cultures, kidney-cortex tubules were incubated with alanine or aspartate plus lactate or glycerol plus octanoate in the absence and presence of 100 mmol/l ethanol. RESULTS: In freshly isolated renal tubules incubated in the presence of alanine plus lactate or glycerol plus octanoate, and in tubules grown in primary culture in the medium containing alanine plus lactate plus octanoate alcohol, resulted in about 30% decrease in glucose formation. A diminished glucose production in freshly isolated tubules was accompanied by: (i) a decrease in alanine utilization, (ii) an increase in lactate or glycerol consumptions and (iii) a decline in GSH:GSSG ratio. The ethanol action was not abolished by 4-methylpyrazole, an inhibitor of alcohol dehydrogenase (ADH). In view of ethanol-induced changes in gluconeogenic intermediates it is likely that in the presence of alanine plus glycerol plus octanoate ethanol causes a decline in flux through phosphoenolpyruvate carboxykinase, probably due to either an increase in intracellular content of 2-oxoglutarate, inhibitor of this key gluconeogenic enzyme and/or an enhanced flux through pyruvate kinase, as concluded from an increased lactate formation in the presence of glycerol in the incubation medium. In renal tubules grown in primary cultures in the presence of alanine plus lactate plus octanoate a decrease in GSH:GSSG ratio was accompanied by elevated generation of reactive oxygen species (ROS). Upon replacement of alanine by aspartate ethanol affected neither glucose production, substrate uptake, ROS accumulation nor GSH:GSSG ratio. CONCLUSIONS: In the presence of alanine ethanol-induced decrease in glucose production and elevation of ROS might cause a limited NADPH generation resulting in a decrease in the intracellular GSH:GSSG ratio. On the contrary, aspartate might protect against ROS generation, so intensive gluconeogenesis supports NADPH generation and in consequence high values of the intracellular GSH:GSSG ratio are maintained.


Subject(s)
Amino Acids/pharmacology , Ethanol/pharmacology , Glucose/biosynthesis , Kidney Cortex/drug effects , Kidney Tubules/drug effects , Animals , Dose-Response Relationship, Drug , Kidney Cortex/metabolism , Kidney Tubules/metabolism , Male , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL