Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Aging Neurosci ; 13: 674688, 2021.
Article in English | MEDLINE | ID: mdl-34276338

ABSTRACT

In mouse models of Alzheimer's disease (AD), normobaric intermittent hypoxia training (IHT) can preserve neurobehavioral function when applied before deficits develop, but IHT's effectiveness after onset of amyloid-ß (Aß) accumulation is unclear. This study tested the hypothesis that IHT improves learning-memory behavior, diminishes Aß accumulation in cerebral cortex and hippocampus, and enhances cerebrocortical contents of the neuroprotective trophic factors erythropoietin and brain-derived neurotrophic factor (BDNF) in mice manifesting AD traits. Twelve-month-old female 3xTg-AD mice were assigned to untreated 3xTg-AD (n = 6), AD+IHT (n = 6), and AD+sham-IHT (n = 6) groups; 8 untreated wild-type (WT) mice also were studied. AD+IHT mice alternately breathed 10% O2 for 6 min and room air for 4 min, 10 cycles/day for 21 days; AD+sham-IHT mice breathed room air. Spatial learning-memory was assessed by Morris water maze. Cerebrocortical and hippocampal Aß40 and Aß42 contents were determined by ELISA, and cerebrocortical erythropoietin and BDNF were analyzed by immunoblotting and ELISA. The significance of time (12 vs. 12 months + 21 days) and treatment (IHT vs. sham-IHT) was evaluated by two-factor ANOVA. The change in swimming distance to find the water maze platform after 21 d IHT (-1.6 ± 1.8 m) differed from that after sham-IHT (+5.8 ± 2.6 m). Cerebrocortical and hippocampal Aß42 contents were greater in 3xTg-AD than WT mice, but neither time nor treatment significantly affected Aß40 or Aß42 contents in the 3xTg-AD mice. Cerebrocortical erythropoietin and BDNF contents increased appreciably after IHT as compared to untreated 3xTg-AD and AD+sham-IHT mice. In conclusion, moderate, normobaric IHT prevented spatial learning-memory decline and restored cerebrocortical erythropoietin and BDNF contents despite ongoing Aß accumulation in 3xTg-AD mice.

2.
Exp Biol Med (Maywood) ; 245(8): 740-747, 2020 04.
Article in English | MEDLINE | ID: mdl-32299228

ABSTRACT

IMPACT STATEMENT: The effects of intermittent hypoxic training or conditioning on many pathological conditions have been widely investigated. One of the pathological conditions dealt with intermittent hypoxic training is ischemic stroke. Well-known mechanisms of intermittent hypoxia-induced protection are related to increased energy metabolism and the enhanced antioxidant effects. In the last decades, the role of microglia in the progress of ischemic stroke-related brain damage has been focused. The dual-edge function of microglia indicates that the microglia-mediated inflammatory response is definitely beneficial in the early stage of ischemic stroke, but long-term activation of microglia is rather detrimental during the recovery process. The effect of IHT on microglia polarization is not investigated. This study focused on whether IHT regulates the polarization of microglia without dampening its classic phagocytic function. This study will provide pivotal information regarding the effects of IHT on the long-term effects on the recovery process from ischemic stroke.


Subject(s)
Cell Hypoxia , Microglia/metabolism , Phagocytosis , Animals , Cells, Cultured , Cytokines/metabolism , Glucose/deficiency , Mice , Oxygen/metabolism , Phenotype , Reactive Oxygen Species/metabolism
3.
Am J Physiol Renal Physiol ; 316(5): F889-F897, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30810354

ABSTRACT

Sex is an important biological variable that impacts diverse physiological and pathological processes, including the progression of diabetic nephropathy. Diabetic nephropathy is one of the most common complications of diabetes mellitus and is the leading cause of end-stage renal disease. The endothelial nitric oxide synthase-deficient (eNOS-/-) db/db mouse is an appropriate and valuable model to study mechanisms in the development of diabetic nephropathy because of the similarities of the features of diabetic kidney disease in this model to those in humans. The aim of the present study was to determine whether there was a sex difference in renal injury in eNOS-/-db/db mice. Both male and female eNOS-/-db/db mice showed hyperglycemia, obesity, and renal hypertrophy. However, there was no significant difference in those variables between male and female mice. Furthermore, both male and female diabetic mice showed progressive albuminuria and significantly greater levels of serum creatinine and blood urea nitrogen compared with the same sex of wild-type mice (nondiabetic controls). Although all three variables in female eNOS-/-db/db mice had a tendency to be greater than those in male eNOS-/-db/db mice, those sex differences were not statistically significant. Moreover, both male and female eNOS-/-db/db mice showed significant mesangial expansion, higher glomerular injury scores, profound renal fibrosis, and substantial accumulation of fibronectin and collagen type IV proteins. However, sex differences in those structural changes were not observed. Similarly, survival rates of male and female eNOS-/-db/db mice were comparable. Taken together, the results from the present study suggest no sex difference in renal structural and functional damage in eNOS-/-db/db mice.


Subject(s)
Diabetic Nephropathies/enzymology , Kidney/enzymology , Nitric Oxide Synthase Type III/deficiency , Animals , Blood Glucose/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Disease Progression , Extracellular Matrix Proteins/metabolism , Female , Fibrosis , Genetic Predisposition to Disease , Hyperglycemia/blood , Hyperglycemia/enzymology , Hyperglycemia/genetics , Kidney/pathology , Kidney/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Obesity/enzymology , Obesity/genetics , Obesity/physiopathology , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Sex Factors , Urination , Weight Gain
5.
Methods Mol Biol ; 1717: 229-235, 2018.
Article in English | MEDLINE | ID: mdl-29468596

ABSTRACT

Ischemia-reperfusion syndromes of the heart and brain are the leading cause of death and long-term disability worldwide. Development of effective treatments for myocardial infarction, stroke, cardiac arrest and their sequelae requires preclinical models that replicate specific features of ischemia-reperfusion. The complexities of intact animals, including the integrated function of organ systems, autonomic innervation and endocrine factors, often preclude detailed study of specific components of ischemia-reperfusion injury cascades. Ischemia represents the interruption of metabolic fuel and oxygen delivery to support cellular oxidative metabolism; reintroduction of oxygen upon reperfusion of ischemic tissue triggers oxidative stress which initiates the reperfusion injury cascade culminating in injury and death of cells and tissues. Thus, cultured cells subjected to hypoxia, fuel deprivation and reoxygenation replicate the cardinal features of ischemia-reperfusion, while accommodating interventions such as siRNA suppression of specific genes and pharmacological activation or inhibition of signaling cascades that are not feasible in more complex preparations, especially intact animals. This chapter describes an in vitro OGD-reoxygenation cell culture model, an excellent preparation to examine the cellular mechanisms mediating ischemia-reperfusion injury and/or cytoprotection.


Subject(s)
Cytoprotection , Glucose , Neurons , Oxygen Consumption , Oxygen/metabolism , Reperfusion Injury , Animals , Cell Hypoxia , Glucose/deficiency , Glucose/metabolism , Humans , Mice , Neurons/metabolism , Neurons/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/therapy
6.
Vitam Horm ; 105: 197-232, 2017.
Article in English | MEDLINE | ID: mdl-28629519

ABSTRACT

The human brain requires uninterrupted delivery of blood-borne oxygen and nutrients to sustain its function. Focal ischemia, particularly, ischemic stroke, and global ischemia imposed by cardiac arrest disrupt the brain's fuel supply. The resultant ATP depletion initiates a complex injury cascade encompassing intracellular Ca2+ overload, glutamate excitotoxicity, oxido-nitrosative stress, extracellular matrix degradation, and inflammation, culminating in neuronal and astroglial necrosis and apoptosis, neurocognitive deficits, and even death. Unfortunately, brain ischemia has proven refractory to pharmacological intervention. Many promising treatments afforded brain protection in animal models of focal and global ischemia, but failed to improve survival and neurocognitive recovery of stroke and cardiac arrest patients in randomized clinical trials. The culprits are the blood-brain barrier (BBB) that limits transferral of medications to the brain parenchyma, and the sheer complexity of the injury cascade, which presents a daunting array of targets unlikely to respond to monotherapies. Erythropoietin is a powerful neuroprotectant capable of interrupting multiple aspects of the brain injury cascade. Preclinical research demonstrates erythropoietin's ability to suppress glutamate excitotoxicity and intracellular Ca2+ overload, dampen oxidative stress and inflammation, interrupt the apoptotic cascade, and preserve BBB integrity. However, the erythropoietin dosages required to traverse the BBB and achieve therapeutically effective concentrations in the brain parenchyma impose untoward side effects. Recent discoveries that hypoxia induces erythropoietin production within the brain and that neurons, astroglia, and cerebrovascular endothelium harbor membrane erythropoietin receptors, raise the exciting prospect of harnessing endogenous erythropoietin to protect the brain from the ravages of ischemia-reperfusion.


Subject(s)
Brain Ischemia/metabolism , Erythropoietin/physiology , Animals , Brain , Humans , Neuroprotective Agents , Oxidative Stress , Reperfusion Injury/metabolism
7.
Am J Physiol Regul Integr Comp Physiol ; 313(1): R10-R18, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28490448

ABSTRACT

Abrupt cessation of chronic alcohol consumption triggers signaling cascades that harm vulnerable brain regions and produce neurobehavioral deficits. We have demonstrated that a program of intermittent, normobaric hypoxia training (IHT) in rats prevents brain damage and neurobehavioral impairment resulting from abrupt ethanol withdrawal (EW). Moreover, EW induced expression of stress-activated protein kinase p38 and presenilin 1 (PS1), the catalytic subunit of γ-secretase that produces the neurotoxic amyloid-ß (Aß) peptides Aß40 and Aß42. We tested the hypotheses that 1) IHT limits EW-induced activation of the p38-PS1 axis, thereby attenuating γ-secretase activation and Aß accumulation, and 2) EW disables heat shock protein 25 (HSP25), a p38 substrate, molecular chaperone, and antioxidant, and provokes protein carbonylation in a manner suppressed by IHT. Adult male rats completed two cycles of a 4-wk ethanol diet (6.5% wt/vol) and a 3-wk EW or an isocaloric, dextrin-based control diet. A 20-day IHT program (5-8 daily cycles of 5-10 min of 9.5-10% fractional inspired O2 + 4 min of 21% fractional inspired O2) was administered during the first EW phase. After the second EW phase, the brain was excised and the prefrontal cortex extracted. PS1, phosphorylated p38 (p-p38), and HSP25 were analyzed by immunoblot, PS1 messenger RNA by quantitative polymerase chain reaction, protein carbonyl content by spectrometry, and Aß40 and Aß42 contents by enzyme-linked immunosorbent assay. IHT attenuated the EW-associated increases in PS1, p-p38, Aß40, Aß42, and protein carbonyl contents, but not that of PS1 messenger RNA, while preserving functionally competent HSP25 dimers in EW rats. Collectively, these findings suggest that IHT may attenuate EW-induced γ-secretase overactivation by suppressing activation of the p38-PS1 axis and by preventing oxidative protein damage.


Subject(s)
Amyloid beta-Peptides/metabolism , Cerebral Cortex/metabolism , Ethanol/toxicity , Hypoxia/metabolism , Presenilin-1/metabolism , Animals , Cerebral Cortex/drug effects , Gene Expression Regulation , HSP27 Heat-Shock Proteins/metabolism , Ischemic Preconditioning , Male , Oxygen , Presenilin-1/genetics , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/prevention & control , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Exp Biol Med (Maywood) ; 242(10): 1095-1103, 2017 05.
Article in English | MEDLINE | ID: mdl-28361585

ABSTRACT

Cardiac arrest (CA) and cardiocerebral resuscitation (CCR)-induced ischemia-reperfusion imposes oxidative and carbonyl stress that injures the brain. The ischemic shift to anaerobic glycolysis, combined with oxyradical inactivation of glyceraldehyde 3-phosphate dehydrogenase (GAPDH), provokes excessive formation of the powerful glycating agent, methylglyoxal. The glyoxalase (GLO) system, comprising the enzymes glyoxalase 1 (GLO1) and GLO2, utilizes reduced glutathione (GSH) supplied by glutathione reductase (GR) to detoxify methylglyoxal resulting in reduced protein glycation. Pyruvate, a natural antioxidant that augments GSH redox status, could sustain the GLO system in the face of ischemia-reperfusion. This study assessed the impact of CA-CCR on the cerebral GLO system and pyruvate's ability to preserve this neuroprotective system following CA. Domestic swine were subjected to 10 min CA, 4 min closed-chest CCR, defibrillation and 4 h recovery, or to a non-CA sham protocol. Sodium pyruvate or NaCl control was infused (0.1 mmol/kg/min, intravenous) throughout CCR and the first 60 min recovery. Protein glycation, GLO1 content, and activities of GLO1, GR, and GAPDH were analyzed in frontal cortex biopsied at 4 h recovery. CA-CCR produced marked protein glycation which was attenuated by pyruvate treatment. GLO1, GR, and GAPDH activities fell by 86, 55, and 30%, respectively, after CA-CCR with NaCl infusion. Pyruvate prevented inactivation of all three enzymes. CA-CCR sharply lowered GLO1 monomer content with commensurate formation of higher molecular weight immunoreactivity; pyruvate preserved GLO1 monomers. Thus, ischemia-reperfusion imposed by CA-CCR disabled the brain's antiglycation defenses. Pyruvate preserved these enzyme systems that protect the brain from glycation stress. Impact statement Recent studies have demonstrated a pivotal role of protein glycation in brain injury. Methylglyoxal, a by-product of glycolysis and a powerful glycating agent in brain, is detoxified by the glutathione-catalyzed glyoxalase (GLO) system, but the impact of cardiac arrest (CA) and cardiocerebral resuscitation (CCR) on the brain's antiglycation defenses is unknown. This study in a swine model of CA and CCR demonstrated for the first time that the intense cerebral ischemia-reperfusion imposed by CA-resuscitation disabled glyoxalase-1 and glutathione reductase (GR), the source of glutathione for methylglyoxal detoxification. Moreover, intravenous administration of pyruvate, a redox-active intermediary metabolite and antioxidant in brain, prevented inactivation of glyoxalase-1 and GR and blunted protein glycation in cerebral cortex. These findings in a large mammal are first evidence of GLO inactivation and the resultant cerebral protein glycation after CA-resuscitation, and identify novel actions of pyruvate to minimize protein glycation in postischemic brain.


Subject(s)
Brain/pathology , Heart Arrest/therapy , Neuroprotective Agents/administration & dosage , Pyruvaldehyde/toxicity , Pyruvic Acid/administration & dosage , Reperfusion Injury/prevention & control , Resuscitation/adverse effects , Animals , Cerebral Cortex/pathology , Disease Models, Animal , Glutathione Reductase/analysis , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/analysis , Glycosylation , Lactoylglutathione Lyase/analysis , Oxidative Stress , Swine , Treatment Outcome
9.
Mol Neurobiol ; 53(2): 1254-1265, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25616953

ABSTRACT

Neuroglobin (Ngb) is a recently discovered globin with preferential localization to neurons. Growing evidence indicates that Ngb has distinct physiological functions separate from the oxygen storage and transport roles of other globins, such as hemoglobin and myoglobin. We found increased ATP production and decreased glycolysis in Ngb-overexpressing immortalized murine hippocampal cell line (HT-22), in parallel with inhibition of AMP-activated protein kinase (AMPK) signaling and activation of acetyl-CoA carboxylase (ACC). In addition, lipid and glycogen content was increased in Ngb-overexpressing HT-22 cells. AMPK signaling was also inhibited in the brain and heart from Ngb-overexpressing transgenic mice. Although Ngb overexpression did not change glycogen content in whole brain, glycogen synthase was activated in cortical neurons of Ngb-overexpressing mouse brain and Ngb overexpression primary neurons. Moreover, lipid and glycogen content was increased in hearts derived from Ngb-overexpressing mice. These findings suggest that Ngb functions as a metabolic regulator and enhances cellular anabolism through the inhibition of AMPK signaling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Globins/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Adenosine Triphosphate/biosynthesis , Animals , Cell Line , Cerebral Cortex/cytology , Enzyme Activation , Glucose/deficiency , Glutamic Acid/toxicity , Glycogen/metabolism , Glycogen Synthase/metabolism , Glycolysis/drug effects , Humans , Lipids/chemistry , Mice, Transgenic , Models, Biological , Myocardium/enzymology , Neuroglobin , Neurons/metabolism , Oxygen , Phosphorylation/drug effects , Signal Transduction/drug effects
10.
PLoS One ; 10(4): e0123096, 2015.
Article in English | MEDLINE | ID: mdl-25848957

ABSTRACT

Astrocytes outnumber neurons and serve many metabolic and trophic functions in the mammalian brain. Preserving astrocytes is critical for normal brain function as well as for protecting the brain against various insults. Our previous studies have indicated that methylene blue (MB) functions as an alternative electron carrier and enhances brain metabolism. In addition, MB has been shown to be protective against neurodegeneration and brain injury. In the current study, we investigated the protective role of MB in astrocytes. Cell viability assays showed that MB treatment significantly protected primary astrocytes from oxygen-glucose deprivation (OGD) & reoxygenation induced cell death. We also studied the effect of MB on cellular oxygen and glucose metabolism in primary astrocytes following OGD-reoxygenation injury. MB treatment significantly increased cellular oxygen consumption, glucose uptake and ATP production in primary astrocytes. In conclusion our study demonstrated that MB protects astrocytes against OGD-reoxygenation injury by improving astrocyte cellular respiration.


Subject(s)
Astrocytes/physiology , Cell Respiration/drug effects , Glucose/metabolism , Methylene Blue/pharmacology , Neuroprotective Agents/pharmacology , Adenosine Triphosphate/biosynthesis , Animals , Astrocytes/drug effects , Cell Hypoxia , Cell Survival , Cells, Cultured , Cytoprotection , Drug Evaluation, Preclinical , Glycogen/biosynthesis , Hexokinase/metabolism , Mice, Inbred C57BL , Oxygen/metabolism , Oxygen Consumption
11.
PLoS One ; 9(6): e99545, 2014.
Article in English | MEDLINE | ID: mdl-24911518

ABSTRACT

HIV-1 infection enhances HCV replication and as a consequence accelerates HCV-mediated hepatocellular carcinoma (HCC). However, the precise molecular mechanism by which this takes place is currently unknown. Our data showed that infectious HIV-1 failed to replicate in human hepatocytic cell lines. No discernible virus replication was observed, even when the cell lines transfected with HIV-1 proviral DNA were co-cultured with Jurkat T cells, indicating that the problem of liver deterioration in the co-infected patient is not due to the replication of HIV-1 in the hepatocytes of the HCV infected host. Instead, HIV-1 Nef protein was transferred from nef-expressing T cells to hepatocytic cells through conduits, wherein up to 16% (average 10%) of the cells harbored the transferred Nef, when the hepatocytic cells were co-cultured with nef-expressing Jurkat cells for 24 h. Further, Nef altered the size and numbers of lipid droplets (LD), and consistently up-regulated HCV replication by 1.5∼2.5 fold in the target subgenomic replicon cells, which is remarkable in relation to the initially indolent viral replication. Nef also dramatically augmented reactive oxygen species (ROS) production and enhanced ethanol-mediated up-regulation of HCV replication so as to accelerate HCC. Taken together, these data indicate that HIV-1 Nef is a critical element in accelerating progression of liver pathogenesis via enhancing HCV replication and coordinating modulation of key intra- and extra-cellular molecules for liver decay.


Subject(s)
Hepacivirus/physiology , Hepatocytes/metabolism , Hepatocytes/virology , T-Lymphocytes/metabolism , Virus Replication , nef Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , Coculture Techniques , Coinfection , Genes, Reporter , HIV Infections , HIV-1 , Humans , Protein Transport , Reactive Oxygen Species/metabolism , T-Lymphocytes/virology , nef Gene Products, Human Immunodeficiency Virus/genetics
12.
Exp Biol Med (Maywood) ; 239(11): 1461-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24595981

ABSTRACT

Ischemic brain injury inflicted by stroke and cardiac arrest ranks among the leading causes of death and long-term disability in the United States. The brain consumes large amounts of metabolic substrates and oxygen to sustain its energy requirements. Consequently, the brain is exquisitely sensitive to interruptions in its blood supply, and suffers irreversible damage after 10-15 min of severe ischemia. Effective treatments to protect the brain from stroke and cardiac arrest have proven elusive, due to the complexities of the injury cascades ignited by ischemia and reperfusion. Although recombinant tissue plasminogen activator and therapeutic hypothermia have proven efficacious for stroke and cardiac arrest, respectively, these treatments are constrained by narrow therapeutic windows, potentially detrimental side-effects and the limited availability of hypothermia equipment. Mounting evidence demonstrates the cytokine hormone erythropoietin (EPO) to be a powerful neuroprotective agent and a potential adjuvant to established therapies. Classically, EPO originating primarily in the kidneys promotes erythrocyte production by suppressing apoptosis of proerythroid progenitors in bone marrow. However, the brain is capable of producing EPO, and EPO's membrane receptors and signaling components also are expressed in neurons and astrocytes. EPO activates signaling cascades that increase the brain's resistance to ischemia-reperfusion stress by stabilizing mitochondrial membranes, limiting formation of reactive oxygen and nitrogen intermediates, and suppressing pro-inflammatory cytokine production and neutrophil infiltration. Collectively, these mechanisms preserve functional brain tissue and, thus, improve neurocognitive recovery from brain ischemia. This article reviews the mechanisms mediating EPO-induced brain protection, critiques the clinical utility of exogenous EPO to preserve brain threatened by ischemic stroke and cardiac arrest, and discusses the prospects for induction of EPO production within the brain by the intermediary metabolite, pyruvate.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Erythropoietin/therapeutic use , Neuroprotective Agents/therapeutic use , Humans , United States
13.
Brain Res ; 1551: 45-58, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24440774

ABSTRACT

Reactive astrogliosis is an essential feature of astrocytic response to all forms of central nervous system (CNS) injury and disease, which may benefit or harm surrounding neural and non-neural cells. Despite extensive study, its molecular triggers remain largely unknown in term of ischemic stroke. In the current study we investigated the role p38 mitogen-activated protein kinase (MAPK) in astrogliosis both in vitro and in vivo. In a mouse model of middle cerebral artery occlusion (MCAO), p38 MAPK activation was observed in the glia scar area, along with increased glial fibrillary acidic protein (GFAP) expression. In primary astrocyte cultures, hypoxia and scratch injury-induced astrogliosis was attenuated by both p38 inhibition and knockout of p38 MAPK. In addition, both knockout and inhibition of p38 MAPK also reduced astrocyte migration, but did not affect astrocyte proliferation. In a mouse model of permanent MCAO, no significant difference in motor function recovery and lesion volume was observed between conditional GFAP/p38 MAPK knockout mice and littermates. While a significant reduction of astrogliosis was observed in the GFAP/p38 knockout mice compared with the littermates. Our findings suggest that p38 MAPK signaling pathway plays an important role in the ischemic stroke-induced astrogliosis and thus may serve as a novel target to control glial scar formation.


Subject(s)
Astrocytes/enzymology , Brain Ischemia/enzymology , Gliosis/enzymology , Stroke/enzymology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Astrocytes/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Gliosis/etiology , Gliosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stroke/complications , Stroke/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Appl Toxicol ; 34(8): 903-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24037965

ABSTRACT

Cadmium is a toxic metal with no biological function in higher-order mammals. Humans are exposed to cadmium environmental contamination and the mechanism underlying the cadmium's cytotoxicity is unclear. To better understand this mechanism, we employed murine hippocampal HT-22 cells to test the in vitro effects of cadmium toxicity. Our study indicated that cadmium inhibits both mitochondria oxidative phosphorylation and glycolysis. In turn, this causes depolarization of mitochondrial membrane potential, increase of superoxide production and decrease of ATP generation. Furthermore, we demonstrated that the detrimental action of cadmium in bioenergetics could be mitigated by pyruvate, an intermediate metabolic product. Pyruvate decreased superoxide production, maintained mitochondrial membrane potential, restored glycolysis, mitigated the decrease in cellular ATP and attenuated cadmium cytotoxicity. Our study provides the first evidence that pyruvate might offer promising therapy for cadmium poisoning.


Subject(s)
Cadmium/toxicity , Hippocampus/cytology , Hippocampus/drug effects , Pyruvic Acid/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Glycolysis/drug effects , Hippocampus/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Oxidative Stress/drug effects , Phosphorylation/drug effects , Superoxides/antagonists & inhibitors , Superoxides/metabolism
15.
Brain Res ; 1530: 66-75, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23891792

ABSTRACT

Clinical application of recombinant tissue plasminogen activator (rtPA) for stroke is limited by hemorrhagic transformation, which narrows rtPA's therapeutic window. In addition, mounting evidence indicates that rtPA is potentially neurotoxic if it traverses a compromised blood brain barrier. Here, we demonstrated that pyruvate protects cultured HT22 neuronal and primary microvascular endothelial cells co-cultured with primary astrocytes from oxygen glucose deprivation (OGD)/reoxygenation stress and rtPA cytotoxicity. After 3 or 6h OGD, cells were reoxygenated with 11mmol/L glucose±pyruvate (8mmol/L) and/or rtPA (10µg/ml). Measured variables included cellular viability (calcein AM and annexin-V/propidium iodide), reactive oxygen species (ROS; mitosox red and 2',7'-dichlorofluorescein diacetate), NADPH, NADP(+) and ATP contents (spectrophotometry), matrix metalloproteinase-2 (MMP2) activities (gelatin zymography), and cellular contents of MMP2, tissue inhibitor of metalloproteinase-2 (TIMP2), and phosphor-activation of anti-apoptotic p70s6 kinase, Akt and Erk (immunoblot). Pyruvate prevented the loss of HT22 cells after 3h OGD±rtPA. After 6h OGD, rtPA sharply lowered cell viability; pyruvate dampened this effect. Three hours OGD and 4h reoxygenation with rtPA increased ROS formation by about 50%. Pyruvate prevented this ROS formation and doubled cellular NADPH/NADP(+) ratio and ATP content. In endothelial cell monolayers, 3h OGD and 24h reoxygenation increased FITC-dextran leakage, indicating disruption of intercellular junctions. Although rtPA exacerbated this effect, pyruvate prevented it while sharply lowering MMP2/TIMP2 ratio and increasing phosphorylation of p70s6 kinase, Akt and Erk. Pyruvate protects neuronal cells and microvascular endothelium from hypoxia-reoxygenation and cytotoxic action of rtPA while reducing ROS and activating anti-apoptotic signaling. These results support the proposed use of pyruvate as an adjuvant to dampen the side effects of rtPA treatment, thereby extending rtPA's therapeutic window.


Subject(s)
Endothelial Cells/drug effects , Glucose/metabolism , Oxygen/metabolism , Pyruvic Acid/pharmacology , Tissue Plasminogen Activator/pharmacology , Animals , Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Hypoxia/metabolism , Mice , Neuroprotective Agents/metabolism , Reactive Oxygen Species/metabolism , Tissue Plasminogen Activator/metabolism
16.
Brain Res ; 1503: 97-107, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23399685

ABSTRACT

Emerging evidence suggests a decline of ERß expression in various peripheral cancers. ERß has been proposed as a cancer brake that inhibits tumor proliferation. In the current study, we have identified ERß5 as the predominant isoform of ERß in human glioma and its expression was significantly increased in human glioma as compared with non-neoplastic brain tissue. Hypoxia and activation of hypoxia inducible factor (HIF) increased ERß transcription in U87 cells, suggesting elevated ERß expression in glioma might be induced by the hypoxic stress in the tumor. Over-expression of either ERß1 or ERß5 increased PTEN expression and inhibited activation of the PI3K/AKT/mTOR pathway. In addition, ERß5 inhibited the MAPK/ERK pathway. In U87 cells, ERß1 and ERß5 inhibit cell proliferation and reduced cells in the S+G2/M phase. Our findings suggest hypoxia induced ERß5 expression in glioma as a self-protective mechanism against tumor proliferation and that ERß5 might serve as a therapeutic target for the treatment of glioma.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glioma/metabolism , Glioma/pathology , Analysis of Variance , Cell Cycle , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation , Disease Progression , Estrogen Receptor beta/classification , Estrogen Receptor beta/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Oxygen Consumption/physiology , RNA, Messenger , Signal Transduction/physiology , Transfection
17.
J Biol Chem ; 288(13): 9153-64, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23408428

ABSTRACT

Glioblastoma multiforme (GBM), like most cancers, possesses a unique bioenergetic state of aerobic glycolysis known as the Warburg effect. Here, we documented that methylene blue (MB) reverses the Warburg effect evidenced by the increasing of oxygen consumption and reduction of lactate production in GBM cell lines. MB decreases GBM cell proliferation and halts the cell cycle in S phase. Through activation of AMP-activated protein kinase, MB inactivates downstream acetyl-CoA carboxylase and decreases cyclin expression. Structure-activity relationship analysis demonstrated that toluidine blue O, an MB derivative with similar bioenergetic actions, exerts similar action in GBM cell proliferation. In contrast, two other MB derivatives, 2-chlorophenothiazine and promethazine, exert no effect on cellular bioenergetics and do not inhibit GBM cell proliferation. MB inhibits cell proliferation in both temozolomide-sensitive and -insensitive GBM cell lines. In a human GBM xenograft model, a single daily dosage of MB does not activate AMP-activated protein kinase signaling, and no tumor regression was observed. In summary, the current study provides the first in vitro proof of concept that reversal of Warburg effect might be a novel therapy for GBM.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Adenosine Triphosphate/metabolism , Animals , Annexin A5/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Energy Metabolism , Female , Humans , Lactates/metabolism , Methylene Blue/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Oxygen Consumption , Structure-Activity Relationship , Temozolomide , Tolonium Chloride/pharmacology
18.
Stroke ; 43(4): 1101-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22282883

ABSTRACT

BACKGROUND AND PURPOSE: Pyruvate is known to be cytoprotective through antioxidant and anti-inflammatory mechanisms. We tested the hypothesis that pyruvate protects the brain against ischemia-reperfusion injury by inducing endogenous erythropoietin (EPO) expression. METHODS: Pyruvate's protective effect was evaluated in C6 glioma cells and HT22 neuronal cells subjected to transient oxygen glucose deprivation. Cell viability (calcein AM assay) and expression of hypoxia-inducible factor-1α, EPO, Akt and Erk (immunoblot), and EPO receptor (reverse transcription-polymerase chain reaction) were analyzed. Transient focal cerebral ischemia in rats was induced by 2 hours middle cerebral artery occlusion followed by 24 hours reperfusion. Pyruvate or saline was infused from 60 minutes occlusion until 30 minutes reperfusion. Lesion volume and DNA fragmentation were assessed by 2,3,5-triphenyltetrazolium staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay, respectively. Immunoblots were conducted to determine cerebral EPO contents. RESULTS: Pyruvate increased cell viability, hypoxia-inducible factor-1α, EPO, and Akt phosphorylation. Small interfering RNA suppression of hypoxia-inducible factor-1α and EPO abolished pyruvate-induced cytoprotection. In the rat stroke model, pyruvate reduced lesion volume by 84% and DNA fragmentation by 77% versus controls; increased EPO content paralleled these cerebroprotective actions of pyruvate. CONCLUSIONS: Pyruvate activation of the hypoxia-inducible factor-1α-EPO signaling cascade in neurons and glia could protect the brain from ischemia-reperfusion injury.


Subject(s)
Brain/metabolism , Erythropoietin/metabolism , Gene Expression Regulation/drug effects , Pyruvic Acid/pharmacology , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Stroke/therapy , Animals , Brain/pathology , Cell Line, Tumor , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Erythropoietin/metabolism , Reperfusion Injury/pathology , Stroke/metabolism
19.
Ann Thorac Surg ; 90(5): 1529-35, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20971256

ABSTRACT

BACKGROUND: Cardiopulmonary bypass-induced oxidative stress initiates inflammation that can damage the myocardium. This study tested whether cardioplegia enriched with the intermediary metabolite and antioxidant pyruvate dampens postbypass myocardial inflammation. METHODS: Pigs were maintained on cardiopulmonary bypass while their hearts were arrested for 60 minutes with 4:1 blood:crystalloid cardioplegia, in which the crystalloid contained 188 mM glucose ± 24 mM pyruvate. Pigs were weaned from bypass after 30 minutes of whole blood reperfusion and recovered for 4 hours. Glutathione (GSH) and glutathione disulfide (GSSG) were measured in coronary sinus plasma to indirectly monitor myocardial GSH redox state (GSH/GSSG). Left ventricular myocardium was sampled 4 hours after cardiopulmonary bypass for analyses of C-reactive protein, matrix metalloproteinases 2 and 9 and tissue inhibitor of metalloproteinase-2 (TIMP-2), and to assess neutrophil infiltration by histology and myeloperoxidase assay. RESULTS: Coronary sinus GSH/GSSG fell 70% after cardiopulmonary bypass with control cardioplegia, but pyruvate cardioplegia produced a robust increase in coronary sinus GSH/GSSG that persisted for 4 hours after bypass. Myocardial C-reactive protein content increased 5.6-fold after control bypass, and neutrophil infiltration and myeloperoxidase activity also increased, but pyruvate-fortified cardioplegia prevented these inflammatory effects. Control cardioplegia lowered myocardial TIMP-2 content by 59% and increased matrix metalloproteinase-9 activity by 35% versus nonbypass sham values, but pyruvate cardioplegia increased TIMP-2 content ninefold versus control cardioplegia and prevented the increase in matrix metalloproteinase-9. Matrix metalloproteinase-2 was not affected by bypass ± pyruvate. CONCLUSIONS: Pyruvate-enriched cardioplegia dampens cardiopulmonary bypass-induced myocardial inflammation. Increased GSH/GSSG and TIMP-2 may mediate pyruvate's effects.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Heart Arrest, Induced , Myocarditis/prevention & control , Pyruvic Acid/pharmacology , Animals , Antioxidants/pharmacology , C-Reactive Protein/analysis , Female , Glutathione/blood , Glutathione Disulfide/blood , Male , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 9/analysis , Myocarditis/etiology , Myocardium/pathology , Neutrophil Infiltration , Swine , Tissue Inhibitor of Metalloproteinase-2/analysis
20.
Am J Physiol Heart Circ Physiol ; 297(5): H1914-22, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19767525

ABSTRACT

Pyruvate-fortified cardioplegia protects myocardium and hastens postsurgical recovery of patients undergoing cardiopulmonary bypass (CPB). Pyruvate reportedly suppresses degradation of the alpha-subunit of hypoxia-inducible factor-1 (HIF-1), an activator of the gene encoding the cardioprotective cytokine erythropoietin (EPO). This study tested the hypothesis that pyruvate-enriched cardioplegia evoked EPO expression and mobilized EPO signaling mechanisms in myocardium. Hearts of pigs maintained on CPB were arrested for 60 min with 4:1 blood-crystalloid cardioplegia. The crystalloid component contained 188 mM glucose + or - 24 mM pyruvate. After 30-min cardiac reperfusion with cardioplegia-free blood, the pigs were weaned from CPB. Left ventricular myocardium was sampled 4 h after CPB for immunoblot assessment of HIF-1alpha, EPO and its receptor, the signaling kinases Akt and ERK, and endothelial nitric oxide synthase (eNOS), an effector of EPO signaling. Pyruvate-fortified cardioplegia stabilized arterial pressure post-CPB, induced myocardial EPO mRNA expression, and increased HIF-1alpha, EPO, and EPO-R protein contents by 60, 58, and 123%, respectively, vs. control cardioplegia (P < 0.05). Pyruvate cardioplegia also increased ERK phosphorylation by 61 and 118%, respectively, vs. control cardioplegia-treated and non-CPB sham myocardium (P < 0.01), but did not alter Akt phosphorylation. Nitric oxide synthase (NOS) activity and eNOS content fell 32% following control CPB vs. sham, but pyruvate cardioplegia prevented these declines, yielding 49 and 80% greater NOS activity and eNOS content vs. respective control values (P < 0.01). Pyruvate-fortified cardioplegia induced myocardial EPO expression and mobilized the EPO-ERK-eNOS mechanism. By stabilizing HIF-1alpha, pyruvate-fortified cardioplegia may evoke sustained activation of EPO's cardioprotective signaling cascade in myocardium.


Subject(s)
Cardioplegic Solutions/pharmacology , Cardiopulmonary Bypass , Erythropoietin/metabolism , Heart Arrest, Induced/methods , Heart Diseases/prevention & control , Myocardium/metabolism , Pyruvic Acid/pharmacology , Signal Transduction/drug effects , Animals , Blood Pressure/drug effects , Cardioplegic Solutions/metabolism , Cardiopulmonary Bypass/adverse effects , Edema, Cardiac/etiology , Edema, Cardiac/metabolism , Edema, Cardiac/prevention & control , Energy Metabolism , Erythropoietin/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glutathione/metabolism , Heart Arrest, Induced/adverse effects , Heart Diseases/etiology , Heart Diseases/metabolism , Heart Diseases/physiopathology , Heart Rate/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Models, Animal , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pyruvic Acid/metabolism , RNA, Messenger/metabolism , Receptors, Erythropoietin/metabolism , Swine , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...