Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mamm Genome ; 35(1): 68-76, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37979047

ABSTRACT

Animal models of diabetes, such as db/db mice, are a useful tool for deciphering the genetic background of molecular changes at the initial stages of disease development. Our goal was to find early transcriptomic changes in three tissues involved in metabolism regulation in db/db mice: adipose tissue, muscle tissue and liver tissue. Nine animals (three per time point) were studied. Tissues were collected at 8, 12 and 16 weeks of age. Transcriptome-wide analysis was performed using mRNA-seq. Libraries were sequenced on NextSeq (Illumina). Differential expression (DE) analysis was performed with edgeR. The analysis of the gene expression profile shared by all three tissues revealed eight upregulated genes (Irf7, Sp100, Neb, Stat2, Oas2, Rtp4, H2-T24 and Oasl2) as early as between 8 and 12 weeks of age. The most pronounced differences were found in liver tissue: nine DE genes between 8 and 12 weeks of age (Irf7, Ly6a, Ly6g6d, H2-Dma, Pld4, Ly86, Fcer1g, Ly6e and Idi1) and five between 12 and 16 weeks of age (Irf7, Plac8, Ifi44, Xaf1 and Ly6a) (adj. p-value < 0.05). The mitochondrial transcriptomic profile also changed with time: we found two downregulated genes in mice between 8 and 12 weeks old (Ckmt2 and Cox6a2) and five DE genes between 12 and 16 weeks of age (Mavs, Tomm40L, Mtfp1, Ckmt2 and Cox6a2). The KEGG pathway analysis showed significant enrichment in pathways related to the autoimmune response and cytosolic DNA sensing. Our results suggest an important involvement of the immunological response, mainly cytosolic nucleic acid sensing, and mitochondrial signalling in the early stages of diabetes and obesity.


Subject(s)
Diabetes Mellitus , Nucleic Acids , Mice , Animals , Transcriptome , Gene Expression Profiling , Mice, Inbred Strains , Antigens, Surface , Membrane Glycoproteins
2.
Int J Mol Sci ; 23(7)2022 Mar 27.
Article in English | MEDLINE | ID: mdl-35409027

ABSTRACT

Blood platelet dysfunctions are strongly involved in the development of the micro- and macrovascular complications in diabetes mellitus (DM). However, the molecular causes of abnormal platelet activation in DM remain unclear. Experimental data suggests that platelet mitochondria can regulate the prothrombotic phenotype of platelets, and changes in these organelles may influence platelet activation and modify platelet responses to stimulation. The present study evaluates the impact of DM on mitochondrial respiratory parameters and blood platelet activation/reactivity in a rat model of experimental diabetes following 1, 2.5 and 5 months of streptozotocin (STZ)-induced diabetes. Moreover, a mild inhibition of the mitochondrial respiratory chain with the use of metformin under in vitro and in vivo conditions was tested as a method to reduce platelet activation and reactivity. The platelets were studied with a combination of flow cytometry and advanced respirometry. Our results indicate that prolonged exposure of blood platelets to high concentrations of glucose, as in diabetes, can result in elevated blood platelet mitochondrial respiration; this may be an effect of cell adaptation to the high availability of energy substrates. However, as these alterations occur later than the changes in platelet activation/reactivity, they may not constitute the major reason for abnormal platelet functioning in DM. Moreover, metformin was not able to inhibit platelet activation and reactivity under in vitro conditions despite causing a decrease in mitochondrial respiration. This indicates that the beneficial effect of metformin on the coagulation system observed in vivo can be related to other mechanisms than via the inhibition of platelet activation.


Subject(s)
Diabetes Mellitus, Experimental , Metformin , Animals , Blood Platelets/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Energy Metabolism , Metformin/metabolism , Metformin/pharmacology , Mitochondria/metabolism , Platelet Activation , Rats
3.
Int J Mol Sci ; 23(5)2022 Feb 27.
Article in English | MEDLINE | ID: mdl-35269760

ABSTRACT

Hexokinase 2 (HK2), an enzyme of the sugar kinase family, plays a dual role in glucose metabolism and mediating cancer cell apoptosis, making it an attractive target for cancer therapy. While positive HK2 expression usually promotes cancer cells survival, silencing or inhibiting this enzyme has been found to improve the effectiveness of anti-cancer drugs and even result in cancer cell death. Previously, benitrobenrazide (BNBZ) was characterized as a potent HK2 inhibitor with good anti-cancer activity in mice, but the effect of its trihydroxy moiety (pyrogallol-like) on inhibitory activity and some cellular functions has not been fully understood. Therefore, the main goal of this study was to obtain the parent BNBZ (2a) and its three dihydroxy derivatives 2b-2d and to conduct additional physicochemical and biological investigations. The research hypothesis assumed that the HK2 inhibitory activity of the tested compounds depends on the number and location of hydroxyl groups in their chemical structure. Among many studies, the binding affinity to HK2 was determined and two human liver cancer cell lines, HepG2 and HUH7, were used and exposed to chemicals at various times: 24 h, 48 h and 72 h. The study showed that the modifications to the structures of the new BNBZ derivatives led to significant changes in their activities. It was also found that these compounds tend to aggregate and exhibit toxic effects. They were found to contribute to: (a) DNA damage, (b) increased ROS production, and (c) disruption of cell cycle progression. It was observed that, HepG2, occurred much more sensitive to the tested chemicals than the HUH7 cells; However, regardless of the used cell line it seems that the increase in the expression of HK2 in cancer cells compared to normal cells which have HK2 at a very low level, is a serious obstacle in anti-cancer therapy and efforts to find the effective inhibitors of this enzyme should be intensified.


Subject(s)
Antineoplastic Agents , Liver Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line , Cell Line, Tumor , Glycolysis , Hexokinase/metabolism , Liver Neoplasms/drug therapy , Mice
4.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502240

ABSTRACT

Blood platelets are considered as promising candidates as easily-accessible biomarkers of mitochondrial functioning. However, their high sensitivity to various stimulus types may potentially affect mitochondrial respiration and lead to artefactual outcomes. Therefore, it is crucial to identify the factors associated with platelet preparation that may lead to changes in mitochondrial respiration. A combination of flow cytometry and advanced respirometry was used to examine the effect of blood anticoagulants, the media used to suspend isolated platelets, respiration buffers, storage time and ADP stimulation on platelet activation and platelet mitochondria respiration. Our results clearly show that all the mentioned factors can affect platelet mitochondrial respiration. Briefly, (i) the use of EDTA as anticoagulant led to a significant increase in the dissipative component of respiration (LEAK), (ii) the use of plasma for the suspension of isolated platelets with MiR05 as a respiration buffer allows high electron transfer capacity and low platelet activation, and (iii) ADP stimulation increases physiological coupling respiration (ROUTINE). Significant associations were observed between platelet activation markers and mitochondrial respiration at different preparation steps; however, the fact that these relationships were not always apparent suggests that the method of platelet preparation may have a greater impact on mitochondrial respiration than the platelet activation itself.


Subject(s)
Anticoagulants/pharmacology , Blood Platelets/physiology , Cell Respiration/physiology , Culture Media/pharmacology , Mitochondria/physiology , Platelet Activation , Blood Platelets/drug effects , Flow Cytometry , Humans , Mitochondria/drug effects
5.
Int J Mol Sci ; 22(17)2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34502416

ABSTRACT

In spite of the continuous improvement in our knowledge of the nature of cancer, the causes of its formation and the development of new treatment methods, our knowledge is still incomplete. A key issue is the difference in metabolism between normal and cancer cells. The features that distinguish cancer cells from normal cells are the increased proliferation and abnormal differentiation and maturation of these cells, which are due to regulatory changes in the emerging tumour. Normal cells use oxidative phosphorylation (OXPHOS) in the mitochondrion as a major source of energy during division. During OXPHOS, there are 36 ATP molecules produced from one molecule of glucose, in contrast to glycolysis which provides an ATP supply of only two molecules. Although aerobic glucose metabolism is more efficient, metabolism based on intensive glycolysis provides intermediate metabolites necessary for the synthesis of nucleic acids, proteins and lipids, which are in constant high demand due to the intense cell division in cancer. This is the main reason why the cancer cell does not "give up" on glycolysis despite the high demand for energy in the form of ATP. One of the evolving trends in the development of anti-cancer therapies is to exploit differences in the metabolism of normal cells and cancer cells. Currently constructed therapies, based on cell metabolism, focus on the attempt to reprogram the metabolic pathways of the cell in such a manner that it becomes possible to stop unrestrained proliferation.


Subject(s)
Adenosine Triphosphate/metabolism , Glucose/metabolism , Glycolysis , Mitochondria/metabolism , Neoplasms/metabolism , Oxidative Phosphorylation , Animals , Humans , Mitochondria/pathology , Neoplasms/pathology
6.
Sci Rep ; 11(1): 4544, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33633284

ABSTRACT

Doxorubicin (DOX) is considered one of the most powerful chemotherapeutic agents but its clinical use has several limitations, including cardiomyopathy and cellular resistance to the drug. By using transferrin (Tf) as a drug carrier, however, the adverse effects of doxorubicin as well as drug resistance can be reduced. The main objective of this study was to determine the exact nature and extent to which mitochondrial function is influenced by DOX-Tf conjugate treatment, specifically in human breast adenocarcinoma cells. We assessed the potential of DOX-Tf conjugate as a drug delivery system, monitoring its cytotoxicity using the MTT assay and ATP measurements. Moreover, we measured the alterations of mitochondrial function and oxidative stress markers. The effect of DOX-Tf was the most pronounced in MDA-MB-231, triple-negative breast cancer cells, whereas non-cancer endothelial HUVEC-ST cells were more resistant to DOX-Tf conjugate than to free DOX treatment. A different sensitivity of two investigate breast cancer cell lines corresponded to the functionality of their cellular antioxidant systems and expression of estrogen receptors. Our data also revealed that conjugate treatment mediated free radical generation and altered the mitochondrial bioenergetics in breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Doxorubicin/pharmacology , Energy Metabolism/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Transferrin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Homeostasis , Humans , Matrix Metalloproteinases/metabolism , Mitochondria/genetics , Oxidation-Reduction , Oxygen Consumption
7.
J Clin Med ; 8(5)2019 May 16.
Article in English | MEDLINE | ID: mdl-31100888

ABSTRACT

The present study is centered on molecular mechanisms of the cytoprotective effect of geranylgeraniol (GGOH) in skeletal muscle harmed by statin-associated myopathy (SAM). GGOH via autophagy induction was purportedly assumed to prevent skeletal muscle viability impaired by statins, atorvastatin (ATR) or simvastatin (SIM). The C2C12 cell line was used as the 'in vitro' model of muscle cells at different stages of muscle formation, and the effect of ATR or SIM on the cell viability, protein expression and mitochondrial respiration were tested. Autophagy seems to be important for the differentiation of muscle cells; however, it did not participate in the observed GGOH cytoprotective effects. We showed that ATR- and SIM-dependent loss in cell viability was reversed by GGOH co-treatment, although GGOH did not reverse the ATR-induced drop in the cytochrome c oxidase protein expression level. It has been unambiguously revealed that the mitochondria of C2C12 cells are not sensitive to SIM, although ATR effectively inhibits mitochondrial respiration. GGOH restored proper mitochondria functioning. Apoptosis might, to some extent, explain the lower viability of statin-treated myotubes as the pan-caspase inhibitor, N-Benzyloxycarbonyl-Val-Ala-Asp(O-Me) fluoromethyl ketone (Z-VAD-FMK), partly reversed ATR- or SIM-induced cytotoxic effects; however, it does not do so in conjunction with caspase-3. It appears that the calpain inhibitor, N-Acetyl-L-leucyl-L-leucyl-L-norleucinal (ALLM), restored the viability that was reduced by ATR and SIM (p < 0.001). GGOH prevents SAM, in part, as a consequence of a caspase-3 independent pathway, probably by calpain system inactivation.

8.
Curr Pharm Biotechnol ; 18(2): 150-157, 2017.
Article in English | MEDLINE | ID: mdl-28034293

ABSTRACT

BACKGROUND: In this study, the effect of low concentrations of poly(amido)amine dendrimers (G2-G4) on human lymphocytes was studied. Some works revealed that PAMAMs can adversely affect the morphology of blood components and mitochondria functions. In this context, the present report aimed to investigate the in vitro cationic dendrimers' effect on mitochondrial respiration and cell morphology in lymphocytes isolated from human blood. METHODS: To monitor the mitochondrial changes, the high-resolution respirometer was used, whereas the cell morphology was analyzed using a flow cytometer and fluorescence microscopy. RESULTS: The concentration-dependent dendrimers' influence on lymphocytes morphology was shown. Changes in mitochondrial respiration revealed the concentration- and generation-dependent differences between dendrimer activity. There were no alterations in the routine respiration and in the state of the inner mitochondrial membrane (L/E), but decreased ADP- and FCCP-stimulated respirations were detected after treatment with G3 and G4 dendrimers. The markers of mitochondrial membrane integrity (RCR) and OXPHOS efficiency (P/E) significantly decreased regardless of the dendrimer generation used. CONCLUSION: Based on these in vitro evaluations, we state that cationic PAMAM dendrimers can impair both the morphology and the bioenergetics of human lymphocytes, even when used at low concentrations and in a short time (up to 1 h). However, these results do not imply that similar findings could be possible for in vivo observations.


Subject(s)
Cell Respiration/drug effects , Dendrimers/toxicity , Lymphocytes/drug effects , Mitochondria/drug effects , Oxygen/metabolism , Cations , Cells, Cultured , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Microscopy, Fluorescence , Mitochondria/metabolism
9.
Int J Pharm ; 500(1-2): 379-89, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26772535

ABSTRACT

We explored the hypothesis that zeta potential altered by polycations affects blood platelet activation and reactivity, the phenomena associated with membrane lipid fluidity and platelet mitochondrial bioenergetics. PAMAM dendrimers generation- and dose-dependently enhanced zeta potential of platelets (from -10.7 mV to -4.3 mV). Increased expressions of activation markers, P-selectin and the active complex αIIbß3, as well as significantly enhanced fibrinogen binding occurred upon the in vitro incubation of blood platelets in the presence of PAMAMs G3 and G4 (resp. 62.1% and 69.4% vs. 1.4% and 2.7% in control for P-selectin, P<0.0001). PAMAM dendrimers increased fluidity of platelet membrane lipid bilayer, while they did not affect platelet mitochondria respiration. Increased platelet activation and their responses to agonists in vitro were statistically associated with the revealed alterations in zeta potential. Our results support the hypothesis that polycation-mediated "neutralized" zeta potential may underlie the activating effects of PAMAMs on blood platelets.


Subject(s)
Blood Platelets/drug effects , Dendrimers/pharmacology , Platelet Activation/drug effects , Adult , Blood Platelets/physiology , Cell Membrane , Electron Spin Resonance Spectroscopy , Female , Humans , Male , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen/metabolism , Young Adult
10.
Int J Pharm ; 503(1-2): 247-61, 2016 Apr 30.
Article in English | MEDLINE | ID: mdl-26319628

ABSTRACT

Direct use of poly(amido)amine (PAMAM) dendrimers as drugs may be limited, due to uncertain (cyto)toxicity. Peripheral blood components, which constitute the first line of a contact with administered pharmaceuticals, may become vastly affected by PAMAM dendrimers. The aim of this study was to explore how PAMAMs' polycationicity might affect blood platelet activation and reactivity, and thus trigger various haemostatic events. We monitored blood platelet reactivity in rats with experimental diabetes upon a long-term administration of the unmodified PAMAM dendrimers. In parallel, the effects on blood flow in a systemic circulation was recorded intravitally in mice administered with PAMAM G2, G3 or G4. Compounding was the in vitro approach to monitor the impact of PAMAM dendrimers on blood platelet activation and reactivity and on selected haemostatic and protein conformation parameters. We demonstrated the activating effects of polycations on blood platelets. Some diversity of the revealed outcomes considerably depended on the used approach and the particular technique employed to monitor blood platelet function. We discovered undesirable impact of plain PAMAM dendrimers on primary haemostasis and their prothrombotic influence. We emphasize the need of a more profound verifying of all the promising findings collected for PAMAMs with the use of well-designed in vivo preclinical studies.


Subject(s)
Blood Platelets/drug effects , Dendrimers/pharmacology , Animals , Arachidonic Acid/pharmacology , Blood Platelets/physiology , Collagen/pharmacology , Diabetes Mellitus, Experimental/blood , Fibrinogen/metabolism , Humans , Male , Mice, Inbred C57BL , Platelet Activation/drug effects , Polylysine/pharmacology , Polymyxins/pharmacology , Rats, Wistar , Splanchnic Circulation/drug effects
11.
J Pharm Sci ; 104(1): 2-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25363074

ABSTRACT

PAMAM (polyamidoamine) dendrimers are commonly considered promising polymers that can be successfully used in various biomedical applications. Nevertheless, direct clinical adaptations of plain unmodified PAMAM dendrimers may be limited at present, mainly because of their toxicity, unpredictable behavior in living organisms, unknown bioavailability, biocompatibility or pharmacokinetic profile, problematic therapeutic dose selection, or high cost of production. On the basis of our studies concerning the possible use of unmodified PAMAM dendrimers as the scavengers of glucose and carbonyl stress in animal models of human pathology, as well as considering available literature on experimental data of other researchers, we have prepared the brief critical review of the biomedical activities of these unmodified compounds and their most alluring derivatives, especially in the context of possible future perspectives of PAMAMs. Thus, on the pages of this review, we made an attempt to briefly summarize obstacles, emerging from experimental, technical, and human limitations, that may, to some extent, restrain our belief in a brighter future of plain amine-terminated PAMAM dendrimers.


Subject(s)
Biocompatible Materials/adverse effects , Dendrimers/adverse effects , Polyamines/adverse effects , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/economics , Biocompatible Materials/therapeutic use , Dendrimers/chemistry , Dendrimers/economics , Dendrimers/therapeutic use , Drug Delivery Systems/adverse effects , Drug Delivery Systems/economics , Drug Delivery Systems/trends , Humans , Nanoparticles/adverse effects , Nanoparticles/chemistry , Nanoparticles/economics , Nanoparticles/therapeutic use , Nanotechnology/economics , Nanotechnology/trends , Polyamines/chemistry , Polyamines/economics , Polyamines/therapeutic use , Surface Properties
12.
Int J Pharm ; 464(1-2): 152-67, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24463003

ABSTRACT

Poly(amido)amine (PAMAM) G4 dendrimers, given intraperitoneally to diabetic rats, have been reported to scavenge excessive blood glucose and minimize the effects of hyperglycaemia, however, at the cost of reduced survival. This paper is the first to compare the effectiveness of three different routes of PAMAM G4 administration with regard to minimizing the adverse effects of hyperglycaemia in rats. Hence, the aim of the study is to identify the most effective and the least harmful method of dendrimer administration. Control and streptozotocin-diabetic Sprague-Dawley rats were exposed to PAMAM G4 (0.5 µmol/kg b.w.) for 60 days, administered intraperitoneally, intragastrically or subcutaneously. Intraperitoneal and subcutaneous administration of PAMAM G4 was found to be most effective in suppressing the long-term markers of hyperglycaemia, while the intragastric route appeared the least effective. Otherwise, the greatest incidence of adverse effects was associated with intraperitoneal and the lowest with subcutaneous delivery. Harmful effects of intragastrical administration were much lower compared to intraperitoneal route, but at the cost of reduced hypoglycaemizing potential. Otherwise, subcutaneous injection represents the best compromise of moderate PAMAM dendrimer toxicity and effective reduction in the markers of long-term severe hyperglycaemia in chronic experimental diabetes.


Subject(s)
Dendrimers/administration & dosage , Dendrimers/adverse effects , Diabetes Mellitus, Experimental/drug therapy , Nylons/adverse effects , Administration, Oral , Animals , Diabetes Mellitus, Experimental/pathology , Hyperglycemia/drug therapy , Hyperglycemia/pathology , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Rats , Rats, Sprague-Dawley , Treatment Outcome
13.
Molecules ; 18(11): 13769-811, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24213655

ABSTRACT

Diabetes mellitus, which is characterised by high blood glucose levels and the burden of various macrovascular and microvascular complications, is a cause of much human suffering across the globe. While the use of exogenous insulin and other medications can control and sometimes prevent various diabetes-associated sequelae, numerous diabetic complications are still commonly encountered in diabetic patients. Therefore, there is a strong need for safe and effective antihyperglycaemic agents that provide an alternative or compounding option for the treatment of diabetes. In recent years, amino-terminated poly(amido)amine (PAMAM) dendrimers (G2, G3 and G4) have attracted attention due to their protective value as anti-glycation and anti-carbonylation agents that can be used to limit the nonenzymatic modifications of biomacromolecules. The focus of this review is to present a detailed survey of our own data, as well as of the available literature regarding the toxicity, pharmacological properties and overall usefulness of PAMAM dendrimers. This presentation pays particular and primary attention to their therapeutic use in poorly controlled diabetes and its complications, but also in other conditions, such as Alzheimer's disease, in which such nonenzymatic modifications may underlie the pathophysiological mechanisms. The impact of dendrimer administration on the overall survival of diabetic animals and on glycosylation, glycoxidation, the brain-blood barrier and cellular bioenergetics are demonstrated. Finally, we critically discuss the potential advantages and disadvantages accompanying the use of PAMAM dendrimers in the treatment of metabolic impairments that occur under conditions of chronic hyperglycaemia.


Subject(s)
Dendrimers/therapeutic use , Animals , Dendrimers/adverse effects , Diabetes Mellitus/drug therapy , Humans , Hyperglycemia/drug therapy , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use
14.
Int J Pharm ; 436(1-2): 508-18, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22721855

ABSTRACT

We hypothesized that BBB is impaired in rat model of streptozotocin-induced diabetes and can be sealed by poly(amido)amine dendrimers G4.0 (PAMAM G4), which reveal anti-glycation activity. The BBB permeabilization was monitored in rats with the 60-day streptozotocin-diabetes and non-diabetic animals, using three fluorescent dyes (given intraperitoneally) differing in molecular weight: fluorescein, fluorescein isothiocyanate (FITC)-dextran and Evans blue. All animals were administered for 2 months with either PAMAM G4 dendrimer or placebo. The fluorescence intensities of the injected fluorescent markers were recorded in the homogenates of selected brain regions. The highest accumulations of the used fluorescent dyes were observed for fluorescein, predominantly in thalamus, hippocampus, frontal cortex, striatum and cerebellum. FITC-dextran leaked to much smaller extent, however, higher permeabilization for FITC-dextran was revealed in pons-medulla oblongata, frontal and parietal cortex of diabetic compared to control animals. Evans blue leaked very slowly into striatum and pons-medulla oblongata in diabetic rats. The treatment of diabetic animals with PAMAM G4 significantly reduced blood glucose concentration and hallmarks of late diabetic complications, compared to non-treated diabetic animals. PAMAM G4 significantly reduced diabetes-induced permeabilization of BBB, which remained in line with the reduced blood glucose and the amelioration of the biochemical hallmarks of severe hyperglycaemia.


Subject(s)
Brain/drug effects , Dendrimers/pharmacology , Diabetes Mellitus, Experimental/metabolism , Nylons/pharmacology , Animals , Blood Glucose/analysis , Brain/metabolism , Coumaric Acids/pharmacology , Dendrimers/therapeutic use , Dextrans/pharmacokinetics , Diabetes Mellitus, Experimental/drug therapy , Evans Blue/pharmacokinetics , Fluorescein/pharmacokinetics , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Symporters/antagonists & inhibitors , Symporters/metabolism
15.
Int J Pharm ; 430(1-2): 258-65, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22486957

ABSTRACT

Poly(amido)amine (PAMAM) dendrimer G3 was investigated for its ability to support the proper functioning of rat heart mitochondria exposed to hyperglycemia, in both the in vitro and in vivo experiments. The main aims of this study were to check whether PAMAM G3 dendrimer improves the efficiency of the impaired respiration of rat heart mitochondria. This study showed that mitochondria isolated from animals studied in different seasons respond to G3 (100 µM) exposure to a different extent. Probably, seasonal variations had the impact on rat metabolism and consequently on the received data. The used biological samples formed a heterogenous group and therefore the obtained results were not pooled together but treated separately. Nevertheless, the in vitro part of this study revealed that PAMAM G3 could be successfully used in the protection of heart mitochondria against MG-induced impaired respiratory activity. Despite these promising data, the protective effect of G3 was not confirmed in the in vivo experiment. This study revealed that dendrimer G3 (20 mg/kgbw) is toxic and very high mortality among the animals administered with G3 did not allow to perform a reliable data analysis.


Subject(s)
Dendrimers/toxicity , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Energy Metabolism/drug effects , Mitochondria, Heart/metabolism , Animals , Cell Respiration/drug effects , Cytoprotection , Dendrimers/administration & dosage , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/chemically induced , Glycation End Products, Advanced/metabolism , Injections, Intraperitoneal , Male , Mitochondria, Heart/drug effects , Pyruvaldehyde/metabolism , Rats , Rats, Wistar , Seasons
16.
Int J Mol Sci ; 12(11): 8013-26, 2011.
Article in English | MEDLINE | ID: mdl-22174647

ABSTRACT

Diabetes is associated with a mitochondrial dysfunction. Hyperglycaemia is also clearly recognized as the primary culprit in the pathogenesis of cardiac complications. In response to glycation and oxidative stress, cardiac mitochondria undergo cumulative alterations, often leading to heart deterioration. There is a continuous search for innovative treatment strategies for protecting the heart mitochondria from the destructive impact of diabetes. Aminoguanidine derivatives have been successfully used in animal model studies on the treatment of experimental diabetes, as well as the diabetes-driven dysfunctions of peripheral tissues and cells. Considerable attention has been paid particularly to ß-resorcylidene aminoguanidine (RAG), often shown as the efficient anti-glycation and anti-oxidant agent in both animal studies and in vitro experiments. The aim of the present study was to test the hypothesis that RAG improves oxidative phosphorylation and electron transport capacity in mitochondria impaired by hyperglycaemia. Diabetes mellitus was induced in Wistar rats by a single intraperitoneal injection of streptozotocin (70 mg/kg body weight). Heart mitochondria were isolated from healthy rats and rats with streptozotocin-diabetes. Mitochondrial respiratory capacity was measured by high resolution respirometry with the OROBOROS Oxygraph-2k according to experimental protocol including respiratory substrates and inhibitors. The results revealed that RAG protects the heart against diabetes-associated injury by improving the mitochondrial bioenergetics, thus suggesting a possible novel pharmacological strategy for cardioprotection.


Subject(s)
Energy Metabolism/drug effects , Guanidines/pharmacology , Heart/drug effects , Hyperglycemia/drug therapy , Mitochondria, Heart/drug effects , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Electron Transport , Enzyme Inhibitors/pharmacology , Heart/physiology , Male , Mitochondria, Heart/metabolism , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...