Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Biomolecules ; 14(3)2024 Mar 07.
Article En | MEDLINE | ID: mdl-38540736

Duchenne muscular dystrophy is caused by loss of the dystrophin protein. This pathology is accompanied by mitochondrial dysfunction contributing to muscle fiber instability. It is known that mitochondria-targeted in vivo therapy mitigates pathology and improves the quality of life of model animals. In the present work, we applied mitochondrial transplantation therapy (MTT) to correct the pathology in dystrophin-deficient mdx mice. Intramuscular injections of allogeneic mitochondria obtained from healthy animals into the hind limbs of mdx mice alleviated skeletal muscle injury, reduced calcium deposits in muscles and serum creatine kinase levels, and improved the grip strength of the hind limbs and motor activity of recipient mdx mice. We noted normalization of the mitochondrial ultrastructure and sarcoplasmic reticulum/mitochondria interactions in mdx muscles. At the same time, we revealed a decrease in the efficiency of oxidative phosphorylation in the skeletal muscle mitochondria of recipient mdx mice accompanied by a reduction in lipid peroxidation products (MDA products) and reduced calcium overloading. We found no effect of MTT on the expression of mitochondrial signature genes (Drp1, Mfn2, Ppargc1a, Pink1, Parkin) and on the level of mtDNA. Our results show that systemic MTT mitigates the development of destructive processes in the quadriceps muscle of mdx mice.


Dystrophin , Muscular Dystrophy, Duchenne , Animals , Mice , Mice, Inbred mdx , Dystrophin/genetics , Calcium/metabolism , Quality of Life , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/pathology , Muscle, Skeletal/metabolism , Mitochondria/metabolism
2.
Int J Mol Sci ; 24(24)2023 Dec 09.
Article En | MEDLINE | ID: mdl-38139129

The pyrimidine nucleoside uridine and its phosphorylated derivates have been shown to be involved in the systemic regulation of energy and redox balance and promote the regeneration of many tissues, including the myocardium, although the underlying mechanisms are not fully understood. Moreover, rearrangements in mitochondrial structure and function within cardiomyocytes are the predominant signs of myocardial injury. Accordingly, this study aimed to investigate whether uridine could alleviate acute myocardial injury induced by isoprenaline (ISO) exposure, a rat model of stress-induced cardiomyopathy, and to elucidate the mechanisms of its action related to mitochondrial dysfunction. For this purpose, a biochemical analysis of the relevant serum biomarkers and ECG monitoring were performed in combination with transmission electron microscopy and a comprehensive study of cardiac mitochondrial functions. The administration of ISO (150 mg/kg, twice with an interval of 24 h, s.c.) to rats caused myocardial degenerative changes, a sharp increase in the serum cardiospecific markers troponin I and the AST/ALT ratio, and a decline in the ATP level in the left ventricular myocardium. In parallel, alterations in the organization of sarcomeres with focal disorganization of myofibrils, and ultrastructural and morphological defects in mitochondria, including disturbances in the orientation and packing density of crista membranes, were detected. These malfunctions were improved by pretreatment with uridine (30 mg/kg, twice with an interval of 24 h, i.p.). Uridine also led to the normalization of the QT interval. Moreover, uridine effectively inhibited ISO-induced ROS overproduction and lipid peroxidation in rat heart mitochondria. The administration of uridine partially recovered the protein level of the respiratory chain complex V, along with the rates of ATP synthesis and mitochondrial potassium transport, suggesting the activation of the potassium cycle through the mitoKATP channel. Taken together, these results indicate that uridine ameliorates acute ISO-induced myocardial injury and mitochondrial malfunction, which may be due to the activation of mitochondrial potassium recycling and a mild uncoupling leading to decreased ROS generation and oxidative damage.


Cardiomyopathies , Mitochondria, Heart , Rats , Animals , Isoproterenol/adverse effects , Mitochondria, Heart/metabolism , Uridine/pharmacology , Uridine/metabolism , Reactive Oxygen Species/metabolism , Cardiomyopathies/metabolism , Potassium/metabolism , Adenosine Triphosphate/metabolism
3.
Brain Struct Funct ; 228(9): 2041-2049, 2023 Dec.
Article En | MEDLINE | ID: mdl-37688593

The cellular mechanisms of neuroplastic changes in the structure of motoneurons and neuropils of the oculomotor (III) nuclei in mice after a 30-day space flight and 7 days after landing were studied. The results showed that microgravity caused degenerative phenomena in neurons: a decrease in the number of terminal dendritic branches was found both after flight and after readaptation to Earth's gravity. In mice after the flight, the number of axodendritic synapses was less than in the control, and their number was not restored after the readaptation. The number of mitochondria in the motoneurons of animals after the flight also decreased and after the readaptation reached only the control value. In addition, a significant number of dark motorneurons were found in mice after readaptation, which indicates that degeneration was caused not only by microgravity, but also by a reaction to the landing of the biosatellite. On the contrary, in the trochlear nucleus, as we showed earlier (Mikheeva et al. in Brain Res 15(1795):148077. https://doi.org/10.1016/j.brainres.2022.148077 , 2022), after readaptation, the dendrites and synaptic contacts were restored, and mitogenesis is significantly enhanced. It has been suggested that morphological changes in the oculomotor nucleus may be the main cause of microgravity-induced nystagmus.


Oculomotor Nuclear Complex , Space Flight , Weightlessness , Mice , Animals , Motor Neurons , Neuropil
4.
Biochemistry (Mosc) ; 88(2): 189-201, 2023 Feb.
Article En | MEDLINE | ID: mdl-37072326

Dystrophin-deficient muscular dystrophy (Duchenne dystrophy) is characterized by impaired ion homeostasis, in which mitochondria play an important role. In the present work, using a model of dystrophin-deficient mdx mice, we revealed decrease in the efficiency of potassium ion transport and total content of this ion in the heart mitochondria. We evaluated the effect of chronic administration of the benzimidazole derivative NS1619, which is an activator of the large-conductance Ca2+-dependent K+ channel (mitoBKCa), on the structure and function of organelles and the state of the heart muscle. It was shown that NS1619 improves K+ transport and increases content of the ion in the heart mitochondria of mdx mice, but this is not associated with the changes in the level of mitoBKCa protein and expression of the gene encoding this protein. The effect of NS1619 was accompanied by the decrease in the intensity of oxidative stress, assessed by the level of lipid peroxidation products (MDA products), and normalization of the mitochondrial ultrastructure in the heart of mdx mice. In addition, we found positive changes in the tissue manifested by the decrease in the level of fibrosis in the heart of dystrophin-deficient animals treated with NS1619. It was noted that NS1619 had no significant effect on the structure and function of heart mitochondria in the wild-type animals. The paper discusses mechanisms of influence of NS1619 on the function of mouse heart mitochondria in Duchenne muscular dystrophy and prospects for applying this approach to correct pathology.


Calcium , Dystrophin , Mice , Animals , Dystrophin/genetics , Dystrophin/metabolism , Calcium/metabolism , Mice, Inbred mdx , Benzimidazoles/pharmacology , Benzimidazoles/metabolism , Mitochondria, Heart/metabolism
5.
Int J Mol Sci ; 23(22)2022 Nov 17.
Article En | MEDLINE | ID: mdl-36430733

The effect of a single one-hour exposure to three modes of hypobaric hypoxia (HBH) differed in the content of O2 in inhaled air (FiO2-14%, 10%, 8%) in the development of mitochondrial-dependent adaptive processes in the myocardium was studied in vivo. The following parameters have been examined: (a) an urgent reaction of catalytic subunits of mitochondrial enzymes (NDUFV2, SDHA, Cyt b, COX2, ATP5A) in the myocardium as an indicator of the state of the respiratory chain electron transport function; (b) an urgent activation of signaling pathways dependent on GPR91, HIF-1α and VEGF, allowing us to assess their role in the formation of urgent mechanisms of adaptation to hypoxia in the myocardium; (c) changes in the ultrastructure of three subpopulations of myocardial mitochondria under these conditions. The studies were conducted on two rat phenotypes: rats with low resistance (LR) and high resistance (HR) to hypoxia. The adaptive and compensatory role of the mitochondrial complex II (MC II) in maintaining the electron transport and energy function of the myocardium in a wide range of reduced O2 concentrations in the initial period of hypoxic exposure has been established. The features of urgent reciprocal regulatory interaction of NAD- and FAD-dependent oxidation pathways in myocardial mitochondria under these conditions have been revealed. The data indicating the participation of GPR91, HIF-1a and VEGF in this process have been obtained. The ultrastructure of the mitochondrial subpopulations in the myocardium of LR and HR rats differed in normoxic conditions and reacted differently to hypoxia of varying severity. The parameters studied together are highly informative indicators of the quality of cardiac activity and metabolic biomarkers of urgent adaptation in various hypoxic conditions.


Succinic Acid , Vascular Endothelial Growth Factor A , Rats , Animals , Succinic Acid/metabolism , Vascular Endothelial Growth Factor A/metabolism , Hypoxia/metabolism , Myocardium/metabolism , Signal Transduction , Mitochondria, Heart/metabolism
6.
Pharmaceutics ; 14(11)2022 Oct 29.
Article En | MEDLINE | ID: mdl-36365155

Duchenne muscular dystrophy (DMD) is a progressive hereditary disease caused by the absence of the dystrophin protein. This is secondarily accompanied by a dysregulation of ion homeostasis, in which mitochondria play an important role. In the present work, we show that mitochondrial dysfunction in the skeletal muscles of dystrophin-deficient mdx mice is accompanied by a reduction in K+ transport and a decrease in its content in the matrix. This is associated with a decrease in the expression of the mitochondrial large-conductance calcium-activated potassium channel (mitoBKCa) in the muscles of mdx mice, which play an important role in cytoprotection. We observed that the BKCa activator NS1619 caused a normalization of mitoBKCa expression and potassium homeostasis in the muscle mitochondria of these animals, which was accompanied by an increase in the calcium retention capacity, mitigation of oxidative stress, and improvement in mitochondrial ultrastructure. This effect of NS1619 contributed to the reduction of degeneration/regeneration cycles and fibrosis in the skeletal muscles of mdx mice as well as a normalization of sarcomere size, but had no effect on the leakage of muscle enzymes and muscle strength loss. In the case of wild-type mice, we noted the negative effect of NS1619 manifested in the inhibition of the functional activity of mitochondria and disruption of their structure, which, however, did not significantly affect the state of the skeletal muscles of the animals. This article discusses the role of mitoBKCa in the development of DMD and the prospects of the approach associated with the correction of its function in treatments of this secondary channelopathy.

7.
Brain Res ; 1795: 148077, 2022 11 15.
Article En | MEDLINE | ID: mdl-36096199

The negative effect of hypogravity on the human organism is manifested to a greater extent after the astronauts return to the conditions of habitual gravity. In this work, to elucidate the causes underlying atypical nystagmus, arising after the flight, we studied structural changes in the motoneurons of the trochlear nerve after a 7-day readaptation of mice to the conditions of Earth's gravity. It is known, that motoneurons of the trochlear nerve innervate the muscle that controls the movement of the eyes in the vertical direction. We showed that the number of axodendritic synapses and some other morphological parameters of motoneurons changed by microgravity can return to their original state in 7 days. However, according to some parameters, motoneurons retain a "memory" of the action of microgravity and do not completely restore the structure. The volume of the soma, the shape of the nuclei, the number and orientation of dendrites do not return to pre-flight parameters. The number of dendrites after 7 days of adaptation remained increased, and the proportion of dendrites in the ventrolateral direction became 2.5 times greater than in motoneurons after space flight. The increased number of mitochondria after space flight became even more significant after readaptation. Microgravity-induced plastic changes retain to some extent "memory traces" after readaptation to Earth's gravity. It can be assumed that the restoration of the function of the trochlear nuclei (overcoming nystagmus) is carried out not only by reversible restoration of the structure of neurons, but partially using those mechanisms that are formed in weightlessness.


Space Flight , Weightlessness , Animals , Astronauts , Humans , Mice , Plastics , Trochlear Nerve , Weightlessness/adverse effects
8.
Int J Mol Sci ; 23(18)2022 Sep 13.
Article En | MEDLINE | ID: mdl-36142532

Long-term hyperglycemia in diabetes mellitus is associated with complex damage to cardiomyocytes and the development of mitochondrial dysfunction in the myocardium. Uridine, a pyrimidine nucleoside, plays an important role in cellular metabolism and is used to improve cardiac function. Herein, the antidiabetic potential of uridine (30 mg/kg/day for 21 days, i.p.) and its effect on mitochondrial homeostasis in the heart tissue were examined in a high-fat diet-streptozotocin-induced model of diabetes in C57BL/6 mice. We found that chronic administration of uridine to diabetic mice normalized plasma glucose and triglyceride levels and the heart weight/body weight ratio and increased the rate of glucose utilization during the intraperitoneal glucose tolerance test. Analysis of TEM revealed that uridine prevented diabetes-induced ultrastructural abnormalities in mitochondria and sarcomeres in ventricular cardiomyocytes. In diabetic heart tissue, the mRNA level of Ppargc1a decreased and Drp1 and Parkin gene expression increased, suggesting the disturbances of mitochondrial biogenesis, fission, and mitophagy, respectively. Uridine treatment of diabetic mice restored the mRNA level of Ppargc1a and enhanced Pink1 gene expression, which may indicate an increase in the intensity of mitochondrial biogenesis and mitophagy, and as a consequence, mitochondrial turnover. Uridine also reduced oxidative phosphorylation dysfunction and suppressed lipid peroxidation, but it had no significant effect on the impaired calcium retention capacity and potassium transport in the heart mitochondria of diabetic mice. Altogether, these findings suggest that, along with its hypoglycemic effect, uridine has a protective action against diabetes-mediated functional and structural damage to cardiac mitochondria and disruption of mitochondrial quality-control systems in the diabetic heart.


Diabetes Mellitus, Experimental , Animals , Blood Glucose/metabolism , Calcium/metabolism , Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat/adverse effects , Hypoglycemic Agents/adverse effects , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Potassium/metabolism , Protein Kinases/metabolism , RNA, Messenger/metabolism , Streptozocin/adverse effects , Triglycerides/metabolism , Ubiquitin-Protein Ligases/metabolism , Uridine/pharmacology , Uridine/therapeutic use
9.
Int J Mol Sci ; 23(18)2022 Sep 13.
Article En | MEDLINE | ID: mdl-36142572

Duchenne muscular dystrophy is caused by the loss of functional dystrophin that secondarily causes systemic metabolic impairment in skeletal muscles and cardiomyocytes. The nutraceutical approach is considered as a possible complementary therapy for this pathology. In this work, we have studied the effect of pyrimidine nucleoside uridine (30 mg/kg/day for 28 days, i.p.), which plays an important role in cellular metabolism, on the development of DMD in the skeletal muscles of dystrophin deficient mdx mice, as well as its effect on the mitochondrial dysfunction that accompanies this pathology. We found that chronic uridine administration reduced fibrosis in the skeletal muscles of mdx mice, but it had no effect on the intensity of degeneration/regeneration cycles and inflammation, pseudohypetrophy, and muscle strength of the animals. Analysis of TEM micrographs showed that uridine also had no effect on the impaired mitochondrial ultrastructure of mdx mouse skeletal muscle. The administration of uridine was found to lead to an increase in the expression of the Drp1 and Parkin genes, which may indicate an increase in the intensity of organelle fission and the normalization of mitophagy. Uridine had little effect on OXPHOS dysfunction in mdx mouse mitochondria, and moreover, it was suppressed in the mitochondria of wild type animals. At the same time, uridine restored the transport of potassium ions and reduced the production of reactive oxygen species; however, this had no effect on the impaired calcium retention capacity of mdx mouse mitochondria. The obtained results demonstrate that the used dose of uridine only partially prevents mitochondrial dysfunction in skeletal muscles during Duchenne dystrophy, though it mitigates the development of destructive processes in skeletal muscles.


Muscular Dystrophy, Duchenne , Animals , Calcium/metabolism , Disease Models, Animal , Dystrophin/metabolism , Mice , Mice, Inbred mdx , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Potassium/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases/metabolism , Uridine/metabolism , Uridine/pharmacology
10.
Biochim Biophys Acta Gen Subj ; 1866(9): 130184, 2022 09.
Article En | MEDLINE | ID: mdl-35660414

BACKGROUND: Dithiocarbamates and derivatives (including disulfiram, DSF) are currently investigated as antineoplastic agents. We have revealed earlier the ability of hydroxocobalamin (vitamin В12b) combined with diethyldithiocarbamate (DDC) to catalyze the formation of highly cytotoxic oxidized derivatives of DSF (DSFoxy, sulfones and sulfoxides). METHODS: Electron and fluorescent confocal microscopy, molecular biology and conventional biochemical techniques were used to study the morphological and functional responses of MCF-7 human breast cancer cells to treatment with DDC and B12b alone or in combination. RESULTS: DDC induces unfolded protein response in MCF-7 cells. The combined use of DDC and B12b causes MCF-7 cell death. Electron microscopy revealed the separation of ER and nuclear membranes, leading to the formation of both cytoplasmic and perinuclear vacuoles, with many fibers inside. The process of vacuolization coincided with the appearance of ER stress markers, a marked damage to mitochondria, a significant inhibition of 20S proteasome, and actin depolimerization at later stages. Specific inhibitors of apoptosis, necroptosis, autophagy, and ferroptosis did not prevent cell death. A short- time (6-h) exposure to DSFoxy caused a significant increase in the number of entotic cells. CONCLUSIONS: These observations indicate that MCF-7 cells treated with a mixture of DDC and B12b die by the mechanism of paraptosis. A short- time exposure to DSFoxy caused, along with paraptosis, a significant activation of the entosis and its final stage, lysosomal cell death. GENERAL SIGNIFICANCE: The results obtained open up opportunities for the development of new approaches to induce non-apoptotic death of cancer cells by dithiocarbamates.


Antineoplastic Agents , Breast Neoplasms , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Disulfiram/pharmacology , Ditiocarb/chemistry , Ditiocarb/pharmacology , Duration of Therapy , Entosis , Female , Humans , MCF-7 Cells
11.
Int J Mol Sci ; 22(18)2021 Sep 10.
Article En | MEDLINE | ID: mdl-34575944

Mitigation of calcium-dependent destruction of skeletal muscle mitochondria is considered as a promising adjunctive therapy in Duchenne muscular dystrophy (DMD). In this work, we study the effect of intraperitoneal administration of a non-immunosuppressive inhibitor of calcium-dependent mitochondrial permeability transition (MPT) pore alisporivir on the state of skeletal muscles and the functioning of mitochondria in dystrophin-deficient mdx mice. We show that treatment with alisporivir reduces inflammation and improves muscle function in mdx mice. These effects of alisporivir were associated with an improvement in the ultrastructure of mitochondria, normalization of respiration and oxidative phosphorylation, and a decrease in lipid peroxidation, due to suppression of MPT pore opening and an improvement in calcium homeostasis. The action of alisporivir was associated with suppression of the activity of cyclophilin D and a decrease in its expression in skeletal muscles. This was observed in both mdx mice and wild-type animals. At the same time, alisporivir suppressed mitochondrial biogenesis, assessed by the expression of Ppargc1a, and altered the dynamics of organelles, inhibiting both DRP1-mediated fission and MFN2-associated fusion of mitochondria. The article discusses the effects of alisporivir administration and cyclophilin D inhibition on mitochondrial reprogramming and networking in DMD and the consequences of this therapy on skeletal muscle health.


Dynamins/genetics , Dystrophin/genetics , GTP Phosphohydrolases/genetics , Muscular Dystrophy, Duchenne/drug therapy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Cyclophilins/genetics , Cyclosporine/pharmacology , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred mdx , Mitochondria/drug effects , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/genetics , Mitochondrial Dynamics/drug effects , Mitochondrial Permeability Transition Pore/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology
12.
Biology (Basel) ; 10(9)2021 Aug 28.
Article En | MEDLINE | ID: mdl-34571715

Diabetes mellitus is a systemic metabolic disorder associated with mitochondrial dysfunction, with the mitochondrial permeability transition (MPT) pore opening being considered as one of its possible mechanisms. The effect of alisporivir, a non-immunosuppressive cyclosporin derivative and a selective inhibitor of the MPT pore opening, on the ultrastructure and functions of the heart mitochondria of mice with diabetes mellitus induced by a high-fat diet combined with streptozotocin injections was studied. The treatment of diabetic animals with alisporivir (2.5 mg/kg ip for 20 days) increased the rate of glucose clearance during the glucose tolerance test. The blood glucose level and the indicator of heart rate in alisporivir-treated diabetic mice tended to restore. An electron microscopy analysis showed that alisporivir prevented mitochondrial swelling and ultrastructural alterations in cardiomyocytes of diabetic mice. Alisporivir canceled the diabetes-induced increases in the susceptibility of heart mitochondria to the MPT pore opening and the level of lipid peroxidation products, but it did not affect the decline in mitochondrial oxidative phosphorylation capacity. The mRNA expression levels of Pink1 and Parkin in the heart tissue of alisporivir-treated diabetic mice were elevated, suggesting the stimulation of mitophagy. In parallel, alisporivir decreased the level of mtDNA in the heart tissue. These findings suggest that targeting the MPT pore opening by alisporivir alleviates the development of mitochondrial dysfunction in the diabetic heart. The cardioprotective effect of the drug in diabetes can be mediated by the induction of mitophagy and the inhibition of lipid peroxidation in the organelles.

13.
Biomedicines ; 9(9)2021 Sep 16.
Article En | MEDLINE | ID: mdl-34572419

Supporting mitochondrial function is one of the therapeutic strategies that improve the functioning of skeletal muscle in Duchenne muscular dystrophy (DMD). In this work, we studied the effect of a non-immunosuppressive inhibitor of mitochondrial permeability transition pore (MPTP) alisporivir (5 mg/kg/day), reducing the intensity of the necrotic process and inflammation in skeletal muscles on the cardiac phenotype of dystrophin-deficient mdx mice. We found that the heart mitochondria of mdx mice show an increase in the intensity of oxidative phosphorylation and an increase in the resistance of organelles to the MPT pore opening. Alisporivir had no significant effect on the hyperfunctionalization of the heart mitochondria of mdx mice, and the state of the heart mitochondria of wild-type animals did not affect the dynamics of organelles but significantly suppressed mitochondrial biogenesis and reduced the amount of mtDNA in the heart muscle. Moreover, alisporivir suppressed mitochondrial biogenesis in the heart of wild-type mice. Alisporivir treatment resulted in a decrease in heart weight in mdx mice, which was associated with a significant modification of the transmission of excitation in the heart. The latter was also noted in the case of WT mice treated with alisporivir. The paper discusses the prospects for using alisporivir to correct the function of heart mitochondria in DMD.

14.
Int J Mol Sci ; 22(17)2021 Sep 02.
Article En | MEDLINE | ID: mdl-34502433

Diabetes mellitus is a systemic metabolic disorder associated with mitochondrial dysfunction, with mitochondrial permeability transition (MPT) pore opening being recognized as one of its pathogenic mechanisms. Alisporivir has been recently identified as a non-immunosuppressive analogue of the MPT pore blocker cyclosporin A and has broad therapeutic potential. The purpose of the present work was to study the effect of alisporivir (2.5 mg/kg/day i.p.) on the ultrastructure and functions of the skeletal muscle mitochondria of mice with diabetes mellitus induced by a high-fat diet combined with streptozotocin injections. The glucose tolerance tests indicated that alisporivir increased the rate of glucose utilization in diabetic mice. An electron microscopy analysis showed that alisporivir prevented diabetes-induced changes in the ultrastructure and content of the mitochondria in myocytes. In diabetes, the ADP-stimulated respiration, respiratory control, and ADP/O ratios and the level of ATP synthase in the mitochondria decreased, whereas alisporivir treatment restored these indicators. Alisporivir eliminated diabetes-induced increases in mitochondrial lipid peroxidation products. Diabetic mice showed decreased mRNA levels of Atp5f1a, Ant1, and Ppif and increased levels of Ant2 in the skeletal muscles. The skeletal muscle mitochondria of diabetic animals were sensitized to the MPT pore opening. Alisporivir normalized the expression level of Ant2 and mitochondrial susceptibility to the MPT pore opening. In parallel, the levels of Mfn2 and Drp1 also returned to control values, suggesting a normalization of mitochondrial dynamics. These findings suggest that the targeting of the MPT pore opening by alisporivir is a therapeutic approach to prevent the development of mitochondrial dysfunction and associated oxidative stress in the skeletal muscles in diabetes.


Cyclosporine/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Mitochondria, Muscle/drug effects , Animals , Cyclosporine/pharmacology , Diet, High-Fat , Drug Evaluation, Preclinical , Male , Mice, Inbred C57BL , Mitochondria, Muscle/ultrastructure , Mitochondrial Dynamics/drug effects , Mitochondrial Permeability Transition Pore
15.
Int J Mol Sci ; 22(16)2021 Aug 11.
Article En | MEDLINE | ID: mdl-34445340

This study was the first comprehensive investigation of the dependence of mitochondrial enzyme response (catalytic subunits of mitochondrial complexes (MC) I-V, including NDUFV2, SDHA, Cyt b, COX1 and ATP5A) and mitochondrial ultrastructure in the rat cerebral cortex (CC) on the severity and duration of in vivo hypoxic exposures. The role of individual animal's resistance to hypoxia was also studied. The respiratory chain (RC) was shown to respond to changes in environmental [O2] as follows: (a) differential reaction of mitochondrial enzymes, which depends on the severity of the hypoxic exposure and which indicates changes in the content and catalytic properties of mitochondrial enzymes, both during acute and multiple exposures; and (b) ultrastructural changes in mitochondria, which reflect various degrees of mitochondrial energization. Within a specific range of reduced O2 concentrations, activation of the MC II is a compensatory response supporting the RC electron transport function. In this process, MC I develops new kinetic properties, and its function recovers in hypoxia by reprograming the RC substrate site. Therefore, the mitochondrial RC performs as an in vivo molecular oxygen sensor. Substantial differences between responses of rats with high and low resistance to hypoxia were determined.


Adaptation, Physiological/physiology , Hypoxia/physiopathology , Mitochondria/enzymology , Mitochondria/ultrastructure , Animals , Animals, Outbred Strains , Cell Respiration/physiology , Cerebral Cortex/chemistry , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Electron Transport/physiology , Hypoxia/metabolism , Hypoxia/pathology , Mitochondria/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/physiology , Protein Conformation , Rats
16.
Mitochondrion ; 59: 246-254, 2021 07.
Article En | MEDLINE | ID: mdl-34144205

Dapagliflozin (DAPA), a selective inhibitor of sodium/glucose cotransporter SGLT2, is currently used as a hypoglycemic agent in the treatment of diabetes mellitus. In this work, we have assessed the effect of DAPA treatment (1 mg/kg/day) on the ultrastructure and functions of the liver mitochondria of C57BL/6NCrl mice with type 2 diabetes mellitus (T2DM) induced by a high-fat diet combined with low-dose streptozotocin injections. An electron microscopy study showed that DAPA prevented the mitochondrial swelling and normalized the average mitochondrial size in hepatocytes of diabetic animals. The treatment with DAPA reversed the decline in the mtDNA copy number in the liver of diabetic mice. DAPA-treated T2DM mice showed increased expression of the Ppargc1a, Mfn2 and Drp1 in the liver tissue. The treatment of diabetic animals with DAPA normalized the mitochondrial respiratory control ratio, significantly decreased the level of lipid peroxidation products in liver mitochondria, and decreased their resistance to the opening of the mitochondrial permeability transition pore. At the same time, DAPA had no effects on the studied parameters of control animals. The paper discusses the possible mechanisms of the effect of dapagliflozin on mitochondrial dysfunction in the liver of diabetic animals.


Benzhydryl Compounds/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diet, High-Fat/adverse effects , Glucosides/administration & dosage , Mitochondria, Liver/genetics , Obesity/complications , Animals , Benzhydryl Compounds/pharmacology , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Dynamins/genetics , GTP Phosphohydrolases/genetics , Gene Dosage/drug effects , Glucosides/pharmacology , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Obesity/chemically induced , Obesity/metabolism , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Random Allocation , Sodium-Glucose Transporter 2 Inhibitors , Streptozocin
17.
Brain Res ; 1758: 147331, 2021 05 01.
Article En | MEDLINE | ID: mdl-33539796

During spaceflight and immediately after it, adaptive neuroplastic changes occur in the sensorimotor structures of the central nervous system, which are associated with changes of mainly vestibular and visual signals. It is known that the movement of the eyeball in the vertical direction is carried out by muscles that are innervated by the trochlear nerve (CN IV) and the oculomotor nerve (CN III). To elucidate the cellular processes underlying the atypical vertical nystagmus that occurs under microgravity conditions, it seems necessary to study the state of these nuclei in animals in more detail after prolonged space flights. We carried out a qualitative and quantitative light-optical and ultrastructural analysis of the nuclei of the trochlear nerve in mice after a 30-day flight on the Bion-M1 biosatellite. As a result, it was shown that the dendrites of motoneurons in the nucleus of the trochlear nerve significantly reorganized their geometry and orientation under microgravity conditions. The number of dendritic branches was increased, possibly in order to amplify the reduced signal flow. To ensure such plastic changes, the number and size of mitochondria in the soma of motoneurons and in axons coming from the vestibular structures increased. Thus, the main role in the adaptation of the trochlear nucleus to microgravity conditions, apparently, belongs to the dendrites of motoneurons, which rearrange their structure and function to enhance the flow of sensory information. These results complement our knowledge of the causes of atypical nystagmus in microgravity.


Adaptation, Physiological/physiology , Motor Neurons/ultrastructure , Space Flight , Trochlear Nerve/ultrastructure , Weightlessness/adverse effects , Animals , Male , Mice , Mice, Inbred C57BL
18.
J Mol Neurosci ; 71(3): 613-617, 2021 Mar.
Article En | MEDLINE | ID: mdl-32803646

Anti-inflammatory cytokine interleukin-10 (IL-10) plays a crucial role in controlling the resolution of inflammation. In this study, we aimed to assess gene expression and the level of IL-10 in the hippocampus and prefrontal cortex of rats, after a single injection of neurotoxicant trimethyltin chloride (TMT). It was shown that 4 weeks after the treatment with TMT, the level of IL-10 in the prefrontal cortex, but not in the hippocampus of TMT-treated rats, was increased. However, expression level of IL-10 mRNA was upregulated both in the hippocampus and in the prefrontal cortex 3 weeks after the injection. Concomitantly, within the same post-treatment period, the expression level of the cyclooxygenase-2 was upregulated in both brain structures, indicating the induction of neuroinflammation. Considering that TMT leads to the death of neurons mainly in the hippocampus, we assume that in contrast to the prefrontal cortex, the level of anti-inflammatory cytokine IL-10 in the hippocampus is not sufficiently increased to prevent the damaging effect of the neurotoxicant. Therefore, an exogenous increase in the level of IL-10 may be useful for the survival of neurons in conditions of neurotoxic damage to the hippocampus.


Hippocampus/metabolism , Interleukin-10/metabolism , Prefrontal Cortex/metabolism , Trimethyltin Compounds/toxicity , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Interleukin-10/genetics , Male , Prefrontal Cortex/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
19.
Biology (Basel) ; 9(10)2020 Sep 24.
Article En | MEDLINE | ID: mdl-32987717

S-15176, a potent derivative of the anti-ischemic agent trimetazidine, was reported to have multiple effects on the metabolism of mitochondria. In the present work, the effect of S-15176 (1.5 mg/kg/day i.p.) on the ultrastructure and functions of liver mitochondria of C57BL/6 mice with type 2 diabetes mellitus (T2DM) induced by a high-fat diet combined with a low-dose streptozotocin injection was examined. An electron microscopy study showed that T2DM induced mitochondrial swelling and a reduction in the number of liver mitochondria. The number of mtDNA copies in the liver in T2DM decreased. The expression of Drp1 slightly increased, and that of Mfn2 and Opa1 somewhat decreased. The treatment of diabetic animals with S-15176 prevented the mitochondrial swelling, normalized the average mitochondrial size, and significantly decreased the content of the key marker of lipid peroxidation malondialdehyde in liver mitochondria. In S-15176-treated T2DM mice, a two-fold increase in the expression of the PGC-1α and a slight decrease in Drp 1 expression in the liver were observed. The respiratory control ratio, the level of mtDNA, and the number of liver mitochondria of S-15176-treated diabetic mice tended to restore. S-15176 did not affect the decrease in expression of Parkin and Opa1 in the liver of diabetic animals, but slightly suppressed the expression of these proteins in the control. The modulatory effect of S-15176 on dysfunction of liver mitochondria in T2DM can be related to the stimulation of mitochondrial biogenesis and the inhibition of lipid peroxidation in the organelles.

20.
Biochim Biophys Acta Biomembr ; 1862(10): 183383, 2020 10 01.
Article En | MEDLINE | ID: mdl-32522531

The paper considers the effects of plant triterpenoid betulin and its derivative betulonic acid on rat liver mitochondria and liposomes. It was found that betulonic acid and, to a lesser extent, betulin, activate mitochondrial respiration in states 2 and 4 and inhibit ADP- and DNP-stimulated (uncoupled) respiration. The effect of betulonic acid resulted in a significant decrease of the respiratory control and ADP/O ratios and decrease in Δψ. The effects of both compounds were most pronounced in the case of succinate-fueled mitochondrial respiration. This may include both the possible protonophore effect of betulonic acid and the inhibition of respiratory chain complexes by both compounds. Both agents enhanced H2O2 production in succinate-fueled mitochondria, while betulonic acid exerted an antioxidant effect with NAD-dependent substrates. Betulin was found to induce mitochondrial aggregation, but had no effect on membrane permeability. A similar pattern was found on liposomes. As revealed by the laurdan generalized polarization (GP) technique, betulin increased laurdan GP in lecithin liposomes, indicating a decrease in membrane fluidity. Measurements of GP as a function of fluorescence excitation wavelength gave an ascending line for high concentrations of betulin, which can be interpreted as phase heterogeneity of the lipid/betulin system. High concentrations of betulin (> 60 mol%) was also demonstrated to cause permeabilization of lecithin liposomes. Betulonic acid was much less effective in inducing the aggregation of mitochondria and liposomes and had no effect on membrane permeability. The possible mechanisms of betulin and betulonic acid effect on rat liver mitochondria and liposomes are discussed.


Liposomes , Mitochondria, Liver/drug effects , Oleanolic Acid/analogs & derivatives , Triterpenes/pharmacology , Animals , Electron Transport , Energy Metabolism/drug effects , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial/drug effects , Oleanolic Acid/pharmacology , Oxidative Phosphorylation , Rats
...