Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Int J Mol Sci ; 24(24)2023 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-38139131

RESUMEN

The study of an organism's response to cerebral ischemia at different levels is essential to understanding the mechanism of the injury and protection. A great interest is devoted to finding the links between quantitative metabolic changes and post-ischemic damage. This work aims to summarize the outcomes of the most studied metabolites in brain tissue-lactate, glutamine, GABA (4-aminobutyric acid), glutamate, and NAA (N-acetyl aspartate)-regarding their biological function in physiological conditions and their role after cerebral ischemia/reperfusion. We focused on ischemic damage and post-ischemic recovery in both experimental-including our results-as well as clinical studies. We discuss the role of blood glucose in view of the diverse impact of hyperglycemia, whether experimentally induced, caused by insulin resistance, or developed as a stress response to the cerebral ischemic event. Additionally, based on our and other studies, we analyze and critically discuss post-ischemic alterations in energy metabolites and the elevation of blood ketone bodies observed in the studies on rodents. To complete the schema, we discuss alterations in blood plasma circulating amino acids after cerebral ischemia. So far, no fundamental brain or blood metabolite(s) has been recognized as a relevant biological marker with the feasibility to determine the post-ischemic outcome or extent of ischemic damage. However, studies from our group on rats subjected to protective ischemic preconditioning showed that these animals did not develop post-ischemic hyperglycemia and manifested a decreased metabolic infringement and faster metabolomic recovery. The metabolomic approach is an additional tool for understanding damaging and/or restorative processes within the affected brain region reflected in the blood to uncover the response of the whole organism via interorgan metabolic communications to the stressful cerebral ischemic challenge.


Asunto(s)
Isquemia Encefálica , Hiperglucemia , Ratas , Animales , Isquemia Encefálica/metabolismo , Infarto Cerebral , Encéfalo/metabolismo , Ácido Láctico/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Hiperglucemia/metabolismo
2.
Cells ; 12(16)2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37626897

RESUMEN

Multifactorial interactions, including nutritional state, likely participate in neurodegeneration's pathogenesis and evolution. Dysregulation in methionine (Met) metabolism could lead to the development of hyperhomocysteinaemia (hHcy), playing an important role in neuronal dysfunction, which could potentially lead to the development of Alzheimer's disease (AD)-like pathological features. This study combines proton magnetic resonance spectroscopy (1H MRS) with immunohistochemical analysis to examine changes in the metabolic ratio and histomorphological alterations in the dorsal rat hippocampus (dentate gyrus-DG) subjected to a high Met diet. Male Wistar rats (420-480 g) underwent hHcy evoked by a Met-enriched diet (2 g/kg of weight/day) lasting four weeks. Changes in the metabolic ratio profile and significant histomorphological alterations have been found in the DG of hHcy rats. We have detected increased morphologically changed neurons and glial cells with increased neurogenic markers and apolipoprotein E positivity parallel with a diminished immunosignal for the N-Methyl-D-Aspartate receptor 1 in hHcy animals. A Met diet induced hHcy, likely via direct Hcy neurotoxicity, an interference with one carbon unit metabolism, and/or epigenetic regulation. These conditions lead to the progression of neurodegeneration and the promotion of AD-like pathological features in the less vulnerable hippocampal DG, which presents a plausible therapeutic target.


Asunto(s)
Enfermedad de Alzheimer , Hiperhomocisteinemia , Masculino , Ratas , Animales , Metionina , Epigénesis Genética , Ratas Wistar , Racemetionina , Dieta , Hipocampo
3.
Neurol Res ; 45(10): 912-918, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37581270

RESUMEN

Vitamin D deficiency is involved in the pathogenesis of multiple sclerosis (MS), a severe autoimmune demyelinating disease of the central nervous system. The gene polymorphism Cdx-2 (rs11568820, G/A) seriously influences the trancriptional activity of the vitamin D receptor (VDR) that binds the vitamin D responsive elements of target genes including HLA-DRB1*15. The aim of the present study in Slovaks was to analyse the association of Cdx-2 variants with the risk of MS and disability progression, and to assess the DRB1*15:01 allele as a possible confounding factor. In total, 493 MS patients and 417 healthy controls were involved in this study. The genotyping of Cdx-2 was performed using restriction analysis; DRB1*15:01 positivity was determined by a high-resolution melting analysis of its surrogate marker rs3135388 (G/A). Our results did not prove any allelic association between Cdx-2 and a risk of MS (minor allele A - 0.181 in patients vs. 0.161 in controls, OR = 1.15, .95 CI = 0.90-1.47, p = 0.289). The logistic regression analysis, adjusted for sex and age, showed no differences in Cdx-2 genotype counts when using an additive, dominant or recessive genetic model (p = 0.351, 0.150, 0.240 respectively). The Cdx-2 variants were also not associated with disease disability progression, evaluated using the Multiple Sclerosis Severity Score. The HLA-DRB1*15:01 allele was found to strongly increase the risk of MS in our study (0.300 in patients vs. 0.101 in controls, OR = 3.83, .95 CI = 2.94-4.99, p = 1.016 × 10-26, dominant genetic model OR = 4.62, .95 CI = 3.40-6.26, p = 9.1 × 10-23). In summary, we found the Cdx-2 as a single genetic marker not to be associated with MS development or progression in Slovaks, independently of HLA-DRB1*15:01 status.


Asunto(s)
Predisposición Genética a la Enfermedad , Esclerosis Múltiple , Humanos , Predisposición Genética a la Enfermedad/genética , Cadenas HLA-DRB1/genética , Esclerosis Múltiple/genética , Frecuencia de los Genes , Polimorfismo de Nucleótido Simple/genética , Genotipo , Alelos , Receptores de Calcitriol
4.
Biomedicines ; 11(4)2023 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-37189811

RESUMEN

During aging, heart structure and function gradually deteriorate, which subsequently increases susceptibility to ischemia-reperfusion (IR). Maintenance of Ca2+ homeostasis is critical for cardiac contractility. We used Langendorff's model to monitor the susceptibility of aging (6-, 15-, and 24-month-old) hearts to IR, with a specific focus on Ca2+-handling proteins. IR, but not aging itself, triggered left ventricular changes when the maximum rate of pressure development decreased in 24-month-olds, and the maximum rate of relaxation was most affected in 6-month-old hearts. Aging caused a deprivation of Ca2+-ATPase (SERCA2a), Na+/Ca2+ exchanger, mitochondrial Ca2+ uniporter, and ryanodine receptor contents. IR-induced damage to ryanodine receptor stimulates Ca2+ leakage in 6-month-old hearts and elevated phospholamban (PLN)-to-SERCA2a ratio can slow down Ca2+ reuptake seen at 2-5 µM Ca2+. Total and monomeric PLN mirrored the response of overexpressed SERCA2a after IR in 24-month-old hearts, resulting in stable Ca2+-ATPase activity. Upregulated PLN accelerated inhibition of Ca2+-ATPase activity at low free Ca2+ in 15-month-old after IR, and reduced SERCA2a content subsequently impairs the Ca2+-sequestering capacity. In conclusion, our study suggests that aging is associated with a significant decrease in the abundance and function of Ca2+-handling proteins. However, the IR-induced damage was not increased during aging.

5.
Metabolites ; 13(3)2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36984804

RESUMEN

Metabolomics is a relatively new research area that focuses mostly on the profiling of selected molecules and metabolites within the organism. A SARS-CoV-2 infection itself can lead to major disturbances in the metabolite profile of the infected individuals. The aim of this study was to analyze metabolomic changes in the urine of patients during the acute phase of COVID-19 and approximately one month after infection in the recovery period. We discuss the observed changes in relation to the alterations resulting from changes in the blood plasma metabolome, as described in our previous study. The metabolome analysis was performed using NMR spectroscopy from the urine of patients and controls. The urine samples were collected at three timepoints, namely upon hospital admission, during hospitalization, and after discharge from the hospital. The acute COVID-19 phase induced massive alterations in the metabolic composition of urine was linked with various changes taking place in the organism. Discriminatory analyses showed the feasibility of successful discrimination of COVID-19 patients from healthy controls based on urinary metabolite levels, with the highest significance assigned to citrate, Hippurate, and pyruvate. Our results show that the metabolomic changes persist one month after the acute phase and that the organism is not fully recovered.

6.
Neurochem Int ; 160: 105419, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36113578

RESUMEN

The ever-present risk of brain ischemic events in humans and its full prevention make the detailed studies of an organism's response to ischemia at different levels essential to understanding the mechanism of the injury as well as protection. We used the four-vessel occlusion as an animal model of forebrain ischemia to investigate its impact on the metabolic alterations in both the hippocampus and the blood plasma to see changes on the systemic level. By inducing sublethal ischemic stimuli, we focused on the endogenous phenomena known as ischemic tolerance. NMR spectroscopy was used to analyze relative metabolite levels in tissue extracts from rats' hippocampus and blood plasma in three various ischemic/reperfusion times: 3 h, 24 h, and 72 h. Hippocampal tissues were characterized by postischemically decreased glutamate and GABA (4-aminobutyrate) tissue content balanced with increased glutamine level, with most pronounced changes at 3 h reperfusion time. Glutamate (as well as glutamine) levels recovered towards the control levels on the third day, as if the glutamate re-synthesis would be firstly preferred before GABA. These results are indicating the higher feasibility of re-establishing of glutamatergic transmission three days after an ischemic event, in contrast to GABA-ergic. Tissue levels of N-acetylaspartate (NAA), as well as choline, were decreased without the tendency to recover three days after the ischemic event. Metabolomic analysis of blood plasma revealed that ischemically preconditioned rats, contrary to the non-preconditioned animals, did not show hyperglycemic conditions. Ischemically induced semi-ketotic state, manifested in increased plasma ketone bodies 3-hydroxybutyrate and acetoacetate, seems to be programmed to support the brain tissue revitalization after the ischemic event. These and other metabolites changes found in blood plasma as well as in the hippocampus were observed to a lower extent or recovered faster in preconditioned animals. Some metabolomic changes in hippocampal tissue extract were so strong that even single metabolites were able to differentiate between ischemic, ischemically preconditioned, and control brain tissues.


Asunto(s)
Acetoacetatos , Precondicionamiento Isquémico , Ácido 3-Hidroxibutírico , Animales , Colina , Glutamatos , Glutamina , Hipocampo , Humanos , Precondicionamiento Isquémico/métodos , Prosencéfalo , Ratas , Ratas Wistar , Extractos de Tejidos , Ácido gamma-Aminobutírico
7.
Metabolites ; 12(7)2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35888785

RESUMEN

End-stage kidney disease is preferably treated by kidney transplantation. The suboptimal function of the allograft often results in misbalances in kidney-controlled processes and requires long-term monitoring of allograft function and viability. As the kidneys are organs with a very high metabolomic rate, a metabolomics approach is suitable to describe systematic changes in post-transplant patients and has great potential for monitoring allograft function, which has not been described yet. In this study, we used blood plasma samples from 55 patients after primary kidney transplantation identically treated with immunosuppressants with follow-up 50 months in the mean after surgery and evaluated relative levels of basal plasma metabolites detectable by NMR spectroscopy. We were looking for the correlations between circulating metabolites levels and allograft performance and allograft rejection features. Our results imply a quantitative relationship between restricted renal function, insufficient hydroxylation of phenylalanine to tyrosine, lowered renal glutamine utilization, shifted nitrogen balance, and other alterations that are not related exclusively to the metabolism of the kidney. No link between allograft function and energy metabolism can be concluded, as no changes were found for glucose, glycolytic intermediates, and 3-hydroxybutyrate as a ketone body representative. The observed changes are to be seen as a superposition of changes in the comprehensive inter-organ metabolic exchange, when the restricted function of one organ may induce compensatory effects or cause secondary alterations. Particular differences in plasma metabolite levels in patients with acute cellular and antibody-mediated allograft rejection were considered rather to be related to the loss of kidney function than to the molecular mechanism of graft rejection since they largely follow the alterations observed by restricted allograft function. In the end, we showed using a simple mathematical model, multilinear regression, that the basal plasmatic metabolites correlated with allograft function expressed by the level of glomerular filtration rate (with creatinine: p-value = 4.0 × 10-26 and r = 0.94, without creatinine: p-value = 3.2 × 10-22 and r = 0.91) make the noninvasive estimation of the allograft function feasible.

8.
Biomolecules ; 12(4)2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35454143

RESUMEN

1H Nuclear Magnetic Resonance (NMR) metabolomics is one of the fundamental tools in the fast-developing metabolomics field. It identifies and quantifies the most abundant metabolites, alterations of which can describe energy metabolism, activated immune response, protein synthesis and catabolism, neurotransmission, and many other factors. This paper summarizes our results of the 1H NMR metabolomics approach to characterize the distribution of relevant metabolites and their alterations induced by cerebral ischemic injury or its combination with hyperhomocysteinemia in the affected tissue and blood plasma in rodents. A decrease in the neurotransmitter pool in the brain tissue likely follows the disordered feasibility of post-ischemic neurotransmission. This decline is balanced by the increased tissue glutamine level with the detected impact on neuronal health. The ischemic injury was also manifested in the metabolomic alterations in blood plasma with the decreased levels of glycolytic intermediates, as well as a post-ischemically induced ketosis-like state with increased plasma ketone bodies. As the 3-hydroxybutyrate can act as a likely neuroprotectant, its post-ischemic increase can suggest its supporting role in balancing ischemic metabolic dysregulation. Furthermore, the 1H NMR approach revealed post-ischemically increased 3-hydroxybutyrate in the remote organs, such as the liver and heart, as well as decreased myocardial glutamate. Ischemic preconditioning, as a proposed protective strategy, was manifested in a lower extent of metabolomic changes and/or their faster recovery in a longitudinal study. The paper also summarizes the pre- and post-ischemic metabolomic changes in the rat hyperhomocysteinemic models. Animals are challenged with hyperglycemia and ketosis-like state. A decrease in several amino acids in plasma follows the onset and progression of hippocampal neuropathology when combined with ischemic injury. The 1H NMR metabolomics approach also offers a high potential for metabolites in discriminatory analysis in the search for potential biomarkers of ischemic injury. Based on our results and the literature data, this paper presents valuable findings applicable in clinical studies and suggests the precaution of a high protein diet, especially foods which are high in Met content and low in B vitamins, in the possible risk of human cerebrovascular neuropathology.


Asunto(s)
Isquemia Encefálica , Hiperhomocisteinemia , Precondicionamiento Isquémico , Cetosis , Ácido 3-Hidroxibutírico , Animales , Isquemia Encefálica/metabolismo , Precondicionamiento Isquémico/métodos , Estudios Longitudinales , Ratas
9.
Neurochem Int ; 154: 105293, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35101551

RESUMEN

Radiation-induced brain injury (RII) is a harmful side-effect occurring after conventional radiation therapy (usually fractionated whole-brain irradiation/fWBI) of patients with cerebral tumors and metastases. An important role in the quality of patients' lives plays cognitive, executive, and emotional functions, regulation on which are involved in frontal cortices pathways. This study assessed the morphologic and metabolic alterations in the rodent frontal cortex caused by fWBI with the total dose of 32 Gy in 4 fractions performed by linear accelerator Clinac iX. Nine male Wistar rats underwent radiation procedures, whereas the other nine rats were investigated as a sham-irradiated group. All eighteen animals were examined using magnetic resonance (MR) in three intervals - before, on 2nd, and 70th day after sham/irradiation. After ten weeks of surviving, all rats underwent histopathological analysis determined by image analysis of immunofluorescent stained sections in the frontal cortex. MR examination was performed on 7T MR scanner Bruker BioSpec 70/20 and consisted of MR-volumetry, T2 relaxometry, and single-voxel proton-1 MR spectroscopy localized in the frontal cortex. Both tissue volume and T2 relaxation time of the frontal cortex were significantly lower in animals after 2 and 70 days of exposure than in controls; however, there were no differences between irradiated groups. Similarly, in animals' frontal cortex after fWBI, increased levels of myoinositol and glutamate/glutamine ratios were observed. Ratios of N-acetyl-aspartate, choline, and peaks of lactate and lipids did not change between groups. The histopathological analysis of the frontal cortex showed increased signs of neurodegeneration and a slight increase in astrocytes and microglia in exposed animals. Early (2 days, 10 weeks) after clinically relevant fWBI were in the frontal cortices of exposed rodents confirmed morphologic and metabolic changes indicating neurodegenerative changes, initializing cerebral atrophy, and evident signs of endothelial disruption and dysregulated neurotransmission that may cause a wide range of functional as well as cognitive deficits.


Asunto(s)
Encéfalo , Roedores , Animales , Encéfalo/metabolismo , Lóbulo Frontal/diagnóstico por imagen , Humanos , Masculino , Espectroscopía de Protones por Resonancia Magnética , Ratas , Ratas Wistar
10.
Mol Cell Biochem ; 477(5): 1621-1628, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35220548

RESUMEN

Increased concentration of plasma homocysteine (Hcy) is an independent risk factor of cardiovascular disease, yet the mechanism by which hyperhomocysteinemia (HHcy) causes cardiac dysfunction is largely unknown. The aim of present study was to investigate the contribution of sarcoplasmic reticulum to impaired cardiac contractile function in HHCy. HHcy-induced by subcutaneous injection of Hcy (0.45 µmol/g of body weight) twice a day for a period of 2 weeks resulted in significant decrease in developed left ventricular pressure and maximum rate of ventricular relaxation. Our results show that abundances of SR Ca2+-handling proteins, Ca2+-ATPase (SERCA2), calsequestrin and histidine-rich calcium-binding protein are significantly reduced while the content of phospholamban is unchanged. Moreover, we found that increased PLN:SERCA2 ratio results in the inhibition of SERCA2 activity at low free Ca2+ concentrations. We further discovered that HHcy is not associated with increased oxidative stress in SR. Taken together, these findings suggest that disturbances in SR Ca2+ handling, caused by altered protein contents but not oxidative damage, may contribute to impaired cardiac contractility in HHcy.


Asunto(s)
Hiperhomocisteinemia , Retículo Sarcoplasmático , Animales , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Calsecuestrina/metabolismo , Corazón/fisiología , Hiperhomocisteinemia/inducido químicamente , Contracción Miocárdica , Miocardio/metabolismo , Ratas , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico
11.
Metabolites ; 11(8)2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34436457

RESUMEN

The study of an organism's response to ischemia at different levels is essential to understand the mechanism of the injury as well as protection. We used the occlusion of four vessels as an animal model of global cerebral ischemia to investigate metabolic alterations in cerebral cortex, hippocampus, blood plasma, as well as in a remote organ, the heart, in rats undergoing 24 h postischemic reperfusion. By inducing sublethal ischemic stimuli, we focused on endogenous phenomena known as ischemic tolerance that is currently the best known and most effective way of protecting against ischemic injury. NMR spectroscopy was used to analyze relative metabolite levels in homogenates from rats' cerebral cortex, hippocampus, and heart together with deproteinized blood plasma. In individual animals subjected to global cerebral ischemia, relative concentrations of the essential amino acids isoleucine, valine, phenylalanine, and tyrosine in cerebral cortex correlated with those in blood plasma (p < 0.05, or boundary significant p < 0.09). This did not apply for the hippocampus, suggesting a closer relation between ischemic cortex and metabolomic blood components. Hippocampal non-participation on correlation with blood components may emphasize the observed partial or full normalization the post-ischemically altered levels of a number of metabolites in the preconditioned animals. Remarkably, that was observed for cortex to a lesser extent. As a response to the global cerebral ischemia in heart tissue, we observed decreased glutamate and increased 3-hydroxybutyrate. Ischemically induced semi-ketotic state and other changes found in blood plasma partially normalized when ischemic preconditioning was introduced. Some metabolomic changes were so strong that even individual metabolites were able to differentiate between ischemic, ischemically preconditioned, and control brain tissues.

12.
Int J Mol Sci ; 22(9)2021 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-34066973

RESUMEN

L-methionine, an essential amino acid, plays a critical role in cell physiology. High intake and/or dysregulation in methionine (Met) metabolism results in accumulation of its intermediate(s) or breakdown products in plasma, including homocysteine (Hcy). High level of Hcy in plasma, hyperhomocysteinemia (hHcy), is considered to be an independent risk factor for cerebrovascular diseases, stroke and dementias. To evoke a mild hHcy in adult male Wistar rats we used an enriched Met diet at a dose of 2 g/kg of animal weight/day in duration of 4 weeks. The study contributes to the exploration of the impact of Met enriched diet inducing mild hHcy on nervous tissue by detecting the histo-morphological, metabolomic and behavioural alterations. We found an altered plasma metabolomic profile, modified spatial and learning memory acquisition as well as remarkable histo-morphological changes such as a decrease in neurons' vitality, alterations in the morphology of neurons in the selective vulnerable hippocampal CA 1 area of animals treated with Met enriched diet. Results of these approaches suggest that the mild hHcy alters plasma metabolome and behavioural and histo-morphological patterns in rats, likely due to the potential Met induced changes in "methylation index" of hippocampal brain area, which eventually aggravates the noxious effect of high methionine intake.


Asunto(s)
Conducta Animal , Hipocampo/patología , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/metabolismo , Metabolómica , Animales , Homocisteína/sangre , Hiperhomocisteinemia/patología , Etiquetado Corte-Fin in Situ , Espectroscopía de Resonancia Magnética , Masculino , Metionina , Ratas Wistar , Coloración y Etiquetado
13.
Neoplasma ; 68(4): 852-860, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33904314

RESUMEN

Targeting metabolomic pathways is a promising strategy for cancer treatment. Alterations in the metabolomic state have also an epigenetic impact, making the metabolomic studies even more interesting. We explored metabolomic changes in the blood plasma of patients with primary and secondary lung cancer and tried to explore their origin. We also applied a discrimination algorithm to the data. In the study, blood samples from 132 patients with primary lung cancer, 47 with secondary lung cancer, and 77 subjectively healthy subjects without any cancer history were used. The samples were measured by NMR spectroscopy. PCA and PLS-DA analyses did not distinguish between patients with primary and secondary lung tumors. Accordingly, no significantly changed levels of plasmatic metabolites were found between these groups. When comparing with healthy controls, significantly increased glucose, citrate, acetate, 3-hydroxybutyrate, and creatinine balanced with decreased pyruvate, lactate, alanine, tyrosine, and tryptophan were found as a common feature of both groups. Metabolomic analysis of blood plasma showed considerable proximity of patients with primary and secondary lung cancer. The changes observed can be partially explained as cancer-derived and also as changes showing ischemic nature. Random Forrest discrimination based on the relative concentration of metabolites in blood plasma performed very promising with AUC of 0.95 against controls; however noticeable parts of differencing metabolites are overlapping with those observed after ischemic injury in other studies.


Asunto(s)
Neoplasias Pulmonares , Metabolómica , Humanos , Pulmón , Espectroscopía de Resonancia Magnética , Plasma
14.
Int J Mol Sci ; 21(20)2020 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-33080955

RESUMEN

Elevated concentration of homocysteine (Hcy) in the blood plasma, hyperhomocysteinemia (HHcy), has been implicated in various disorders, including cardiovascular and neurodegenerative diseases. Accumulating evidence indicates that pathophysiology of these diseases is linked with mitochondrial dysfunction. In this review, we discuss the current knowledge concerning the effects of HHcy on mitochondrial homeostasis, including energy metabolism, mitochondrial apoptotic pathway, and mitochondrial dynamics. The recent studies suggest that the interaction between Hcy and mitochondria is complex, and reactive oxygen species (ROS) are possible mediators of Hcy effects. We focus on mechanisms contributing to HHcy-associated oxidative stress, such as sources of ROS generation and alterations in antioxidant defense resulting from altered gene expression and post-translational modifications of proteins. Moreover, we discuss some recent findings suggesting that HHcy may have beneficial effects on mitochondrial ROS homeostasis and antioxidant defense. A better understanding of complex mechanisms through which Hcy affects mitochondrial functions could contribute to the development of more specific therapeutic strategies targeted at HHcy-associated disorders.


Asunto(s)
Encéfalo/irrigación sanguínea , Sistema Cardiovascular/metabolismo , Homocisteína/metabolismo , Mitocondrias/metabolismo , Animales , Metabolismo Energético , Homocisteína/química , Humanos , Estrés Oxidativo
15.
Biomolecules ; 10(8)2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32751764

RESUMEN

Hyperhomocysteinemia (hHcy) represents a strong risk factor for atherosclerosis-associated diseases, like stroke, dementia or Alzheimer's disease. A methionine (Met)-rich diet leads to an elevated level of homocysteine in plasma and might cause pathological alterations across the brain. The hippocampus is being constantly studied for its selective vulnerability linked with neurodegeneration. This study explores metabolic and histo-morphological changes in the rat hippocampus after global ischemia in the hHcy conditions using a combination of proton magnetic resonance spectroscopy and magnetic resonance-volumetry as well as immunohistochemical analysis. After 4 weeks of a Met-enriched diet at a dose of 2 g/kg of animal weight/day, adult male Wistar rats underwent 4-vessel occlusion lasting for 15 min, followed by a reperfusion period varying from 3 to 7 days. Histo-morphological analyses showed that the subsequent ischemia-reperfusion insult (IRI) aggravates the extent of the sole hHcy-induced degeneration of the hippocampal neurons. Decreased volume in the grey matter, extensive changes in the metabolic ratio, deeper alterations in the number and morphology of neurons, astrocytes and their processes were demonstrated in the hippocampus 7 days post-ischemia in the hHcy animals. Our results suggest that the combination of the two risk factors (hHcy and IRI) endorses and exacerbates the rat hippocampal neurodegenerative processes.


Asunto(s)
Isquemia Encefálica/metabolismo , Dieta , Hipocampo/metabolismo , Hiperhomocisteinemia/complicaciones , Metionina/efectos adversos , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Hipocampo/diagnóstico por imagen , Hipocampo/efectos de los fármacos , Hipocampo/patología , Homocisteína/metabolismo , Hiperhomocisteinemia/etiología , Espectroscopía de Resonancia Magnética , Masculino , Metionina/administración & dosificación , Ratas , Ratas Wistar
16.
IUBMB Life ; 72(9): 2010-2023, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32663378

RESUMEN

Cardiac arrest is one of the major causes of death and disability. The aim of the study was to identify dynamic time-dependent metabolomic changes reflected in rat plasma induced by cerebral ischemia and reperfusion with the focus on the protective effect of ischemic preconditionig. Global cerebral ischemia in rats was induced by the four-vessel occlusion. Blood plasma was collected in three reperfusion times: an early post-acute 3 hr, then 24 hr, as an incipient time for delayed neuronal death induction and 72 hr as prolonged reperfusion period. The metabolomic measurements were conducted via untargeted nuclear magnetic resonance spectroscopy. Plasma of ischemized rats manifested dynamic metabolomic changes over the reperfusion time, such as increased levels of ketone bodies, decreased levels of pyruvate, alanine, and citrate. All three branched chain amino acids showed common pattern during reperfusion time: a decrease in 3 hr compared to sham, then a highest level in 24 hr and decrease in 72 hr reperfusion time, similar to their corresponding ketoacids. The protective effect of ischemic preconditioning was demonstrated by a faster tendency of plasma metabolites to normalize. Results also proved the remarkable metabolomic differences between the control (naïve) and sham-operated anesthetized animals, what warrants for critical evaluation of surgery/anaesthesy in the algorithm of metabolomic animal studies.


Asunto(s)
Isquemia Encefálica/patología , Precondicionamiento Isquémico/métodos , Metaboloma , Plasma/metabolismo , Daño por Reperfusión/patología , Animales , Isquemia Encefálica/metabolismo , Masculino , Ratas , Ratas Wistar , Daño por Reperfusión/metabolismo , Factores de Tiempo
17.
Int J Mol Sci ; 20(24)2019 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-31835644

RESUMEN

Hyperhomocysteinemia (hHcy) is regarded as an independent and strong risk factor for cerebrovascular diseases, stroke, and dementias. The hippocampus has a crucial role in spatial navigation and memory processes and is being constantly studied for neurodegenerative disorders. We used a moderate methionine (Met) diet at a dose of 2 g/kg of animal weight/day in duration of four weeks to induce mild hHcy in adult male Wistar rats. A novel approach has been used to explore the hippocampal metabolic changes using proton magnetic resonance spectroscopy (1H MRS), involving a 7T MR scanner in combination with histochemical and immunofluorescence analysis. We found alterations in the metabolic profile, as well as remarkable histo-morphological changes such as an increase of hippocampal volume, alterations in number and morphology of astrocytes, neurons, and their processes in the selective vulnerable brain area of animals treated with a Met-enriched diet. Results of both methodologies suggest that the mild hHcy induced by Met-enriched diet alters volume, histo-morphological pattern, and metabolic profile of hippocampal brain area, which might eventually endorse the neurodegenerative processes.


Asunto(s)
Hipocampo/diagnóstico por imagen , Hiperhomocisteinemia/diagnóstico por imagen , Metaboloma/efectos de los fármacos , Metionina/efectos adversos , Animales , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hiperhomocisteinemia/inducido químicamente , Hiperhomocisteinemia/metabolismo , Masculino , Tamaño de los Órganos/efectos de los fármacos , Espectroscopía de Protones por Resonancia Magnética , Ratas , Ratas Wistar
18.
J Neurol Sci ; 407: 116458, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31622679

RESUMEN

Multiple sclerosis (MS) is a disease characterized by overlapping processes of neuroinflammation and neuro-axonal degeneration. Disturbances of the hypothalamo-pituitary axis in MS are supposed to modulate neuroinflammatory circuits, however, there is insufficient knowledge about the hypothalamic metabolism alterations in early MS. This 1H MRS study performed on a 1.5 T MR-scanner was focused on the hypothalamus of 31 pre-treatment patients after their first clinical MS episode/s, compared to 31 healthy controls. The metabolite ratios of N-acetyl-aspartate &N-acetyl-aspartyl-glutamate (tNAA), glutamate & glutamine (Glx), myo-Inositol (mIns), choline- and creatine-containing compounds (tCho, tCr) were further correlated with the Expanded Disability Status Scale (EDSS). In the hypothalamus of early MS patients compared to controls, we found decreased tNAA/tCr and increased tCho/tNAA, mIns/tNAA, Glx/tCr, and Glx/tNAA. In addition, tCho/tNAA, Glx/tNAA, and mIns/tNAA were positively and tNAA/tCr was negatively correlated with EDSS. Results suggest that the decline of the tNAA ratio, indicating neuro-axonal dysfunction in the hypothalamus, may be linked with glutamate excitotoxicity. Excessive glutamate concentrations may cause microglial activation and myelinated tracts degradation with subsequent gliosis, paralleled by increased mIns and tCho ratios. This indicates that glutamate excitotoxicity can play an important role in MS from its earliest stages.


Asunto(s)
Hipotálamo/metabolismo , Espectroscopía de Resonancia Magnética , Esclerosis Múltiple/metabolismo , Adulto , Ácido Aspártico/metabolismo , Colina/metabolismo , Creatina/metabolismo , Femenino , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Hipotálamo/diagnóstico por imagen , Inositol/metabolismo , Masculino , Esclerosis Múltiple/diagnóstico por imagen , Adulto Joven
19.
Gen Physiol Biophys ; 38(4): 305-314, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31219427

RESUMEN

Blood biomarkers are usually present in low concentration and can be masked by the high-abundance proteins, of which albumin is the predominant one. The purpose of this study was to compare four different albumin removal methods compatible with in-gel based proteomics, applicable for plasma, without requiring specific techniques and high financial input. Plasma underwent albumin depletion with ultrafiltration device Amicon Ultra, commercial ProteoPrep Blue Albumin and IgG Depletion Kit, acetonitrile precipitation method and precipitation with acetonitrile-methanol protocol. All samples were evaluated by 1-D and 2-D gel electrophoresis with subsequent mass spectrometry protein identification. Two of the tested methods (ProteoPrep BlueKit and acetonitrile-methanol precipitation) maintained sufficient protein content for further in-gel analyses. Their 2-D protein profiles were distinctively separated and overlapped with protein profile of crude plasma. Protein spot count showed significant increase in protein spots, compared to crude plasma, only with acetonitrile-methanol precipitation method. Precipitation with acetonitrile-methanol method significantly increased number of protein spots on 2-D protein profile and improved score of mass spectrometry identification. However, albumin was still present and found in number of protein spots.


Asunto(s)
Albúminas/aislamiento & purificación , Proteínas Sanguíneas/análisis , Plasma/química , Proteómica/métodos , Electroforesis en Gel Bidimensional , Humanos
20.
J Physiol Biochem ; 75(2): 217-227, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31115776

RESUMEN

Myocardial ischemia reperfusion is associated with mitochondrial dysfunction and increased formation of reactive oxygen/nitrogen species. The main purpose of this study was to assess the role of tyrosine nitration of mitochondrial proteins in postischemic contractile dysfunction known as myocardial stunning. Isolated Langendorff-perfused rat hearts were subjected to 20-min global ischemia followed by 30-min reperfusion. The reperfused hearts showed marked decline in left ventricular developed pressure, maximal rate of contraction (+dP/dt), and maximal rate of relaxation (-dP/dt). Immunofluorescence and ELISA assays demonstrated enhanced protein tyrosine nitration in reperfused hearts. Using two-dimensional gel electrophoresis and MALDI-TOF/TOF mass spectrometry, eight mitochondrial proteins were identified to be nitrated after ischemia reperfusion. These proteins are crucial in mitochondrial electron transport, fatty acid oxidation, tricarboxylic acid cycle, ATP synthesis, and control of high-energy phosphates. The proteome data also indicated reduced abundance in several of nitrated proteins. The results suggest that these changes may contribute to inhibition of aconitase activity but are unlikely to affect electron transport chain activity. Whether tyrosine nitration of mitochondrial proteins can be considered the contributing factor of postischemic contractile dysfunction remains to be explored.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Tirosina/análogos & derivados , Aconitato Hidratasa/metabolismo , Animales , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Masculino , Espectrometría de Masas , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/química , Contracción Miocárdica , Daño por Reperfusión Miocárdica/fisiopatología , Aturdimiento Miocárdico , Oxidación-Reducción , Ratas , Ratas Wistar , Tirosina/química , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...