Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(3)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38338992

ABSTRACT

Knee osteoarthritis (OA), an age-related degenerative disease characterized by severe pain and disability, is treated using polynucleotides (PNs) and hyaluronic acid (HA). The intra-articular (IA) injection of HA has been studied extensively in both animal models and in humans; however, the efficacy and mechanisms of action remain unclear. In addition, there has been a paucity of research regarding the use of PN alone or in combination with HA in OA. To investigate the effect of the combined injection of PN and HA in vivo, pathological and behavioral changes were assessed in an OA model. Anterior cruciate ligament transection and medial meniscectomy were performed in Sprague-Dawley rats to create the OA animal model. The locomotor activity improved following PNHA injection, while the OARSI grade improved in the medial tibia and femur. In mild OA, TNFα levels decreased histologically in the PN, HA, and PNHA groups but only the PNHA group showed behavioral improvement in terms of distance. In conclusion, PNHA exhibited anti-inflammatory effects during OA progression and improved locomotor activity regardless of the OARSI grade.


Subject(s)
Hyaluronic Acid , Osteoarthritis, Knee , Rats , Humans , Animals , Hyaluronic Acid/pharmacology , Polynucleotides/pharmacology , Polynucleotides/therapeutic use , Rats, Sprague-Dawley , Osteoarthritis, Knee/drug therapy , Anterior Cruciate Ligament/surgery , Injections, Intra-Articular
3.
Mol Psychiatry ; 28(10): 4474-4484, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37648779

ABSTRACT

Mitochondrial dysfunction has been implicated in Parkinson's Disease (PD) progression; however, the mitochondrial factors underlying the development of PD symptoms remain unclear. One candidate is CR6-interacting factor1 (CRIF1), which controls translation and membrane insertion of 13 mitochondrial proteins involved in oxidative phosphorylation. Here, we found that CRIF1 mRNA and protein expression were significantly reduced in postmortem brains of elderly PD patients compared to normal controls. To evaluate the effect of Crif1 deficiency, we produced mice lacking the Crif1 gene in dopaminergic neurons (DAT-CRIF1-KO mice). From 5 weeks of age, DAT-CRIF1-KO mice began to show decreased dopamine production with progressive neuronal degeneration in the nigral area. At ~10 weeks of age, they developed PD-like behavioral deficits, including gait abnormalities, rigidity, and resting tremor. L-DOPA, a medication used to treat PD, ameliorated these defects at an early stage, although it was ineffective in older mice. Taken together, the observation that CRIF1 expression is reduced in human PD brains and deletion of CRIF1 in dopaminergic neurons leads to early-onset PD with stepwise PD progression support the conclusion that CRIF1-mediated mitochondrial function is important for the survival of dopaminergic neurons.


Subject(s)
Dopaminergic Neurons , Parkinson Disease , Humans , Mice , Animals , Aged , Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , Levodopa/pharmacology , Dopamine/metabolism , Brain/metabolism , Cell Cycle Proteins/genetics
4.
Pharmacol Res ; 184: 106423, 2022 10.
Article in English | MEDLINE | ID: mdl-36064078

ABSTRACT

BMP2 is clinically used as an ectopic bone inducer and plays a significant role in bone development, formation, and diseases. Chitinase 3-like 1 protein (Chi3L1) is found in the skeletal system. However, Chi3L1-mediated bone metabolism and aging-related bone erosion via BMP2 signaling have not yet been demonstrated. Herein, Chi3L1 increased BMP2-induced osteoblast differentiation in mesenchymal precursor cells and human primary osteoblasts. Chi3L1KO(-/-) showed abnormal bone development, and primary osteoblasts isolated from Chi3L1KO(-/-) exhibited impaired osteoblast differentiation and maturation. Chi3L1 also potentiated BMP2 signaling and RUNX2 expression in primary osteoblasts. Chi3L1 interacted with BMPRIa, which increased the surface expression of BMPRIa and promoted BMP2 signaling to induce osteoblast differentiation. Chi3L1KO(-/-) mice showed bone formation reduced with a decrease in RUNX2 expression in calvarial defects. Chi3L1KO(-/-) mice exhibited aging-related osteoporotic bone loss with decreases in the levels of RUNX2 and OPG, while serum PYD level and osteoclast number increased. Chi3L1 increased OPG via non-canonical BMP2 signaling in osteoblasts, which suppressed osteoclastogenesis in BMMs. Furthermore, ROC analysis showed that serum Chi3L1 level clinically decreased in osteoporosis patients. Our findings demonstrate that Chi3L1 promotes bone formation, suppresses osteoclastogenesis, and prevents aging-related osteoporosis.


Subject(s)
Chitinases , Osteoporosis , Animals , Biomarkers/metabolism , Cell Differentiation , Chitinase-3-Like Protein 1/genetics , Chitinase-3-Like Protein 1/metabolism , Chitinases/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Mice , Osteoblasts/metabolism , Osteogenesis , Osteoporosis/drug therapy , Osteoporosis/metabolism
5.
Int J Mol Sci ; 22(7)2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33810547

ABSTRACT

The hypothalamic regulation of appetite governs whole-body energy balance. Satiety is regulated by endocrine factors including leptin, and impaired leptin signaling is associated with obesity. Despite the anorectic effect of leptin through the regulation of the hypothalamic feeding circuit, a distinct downstream mediator of leptin signaling in neuron remains unclear. Angiopoietin-like growth factor (AGF) is a peripheral activator of energy expenditure and antagonizes obesity. However, the regulation of AGF expression in brain and localization to mediate anorectic signaling is unknown. Here, we demonstrated that AGF is expressed in proopiomelanocortin (POMC)-expressing neurons located in the arcuate nucleus (ARC) of the hypothalamus. Unlike other brain regions, hypothalamic AGF expression is stimulated by leptin-induced signal transducers and activators of transcription 3 (STAT3) phosphorylation. In addition, leptin treatment to hypothalamic N1 cells significantly enhanced the promoter activity of AGF. This induction was abolished by the pretreatment of ruxolitinib, a leptin signaling inhibitor. These results indicate that hypothalamic AGF expression is induced by leptin and colocalized to POMC neurons.


Subject(s)
Angiopoietin-like Proteins/genetics , Angiopoietin-like Proteins/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Signal Transduction , Angiopoietin-Like Protein 6 , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Brain/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Phosphorylation , Pro-Opiomelanocortin/metabolism , STAT3 Transcription Factor/metabolism
6.
J Neurochem ; 156(1): 76-87, 2021 01.
Article in English | MEDLINE | ID: mdl-32639632

ABSTRACT

While recent studies strongly suggest that a single, short anesthetic exposure does not affect neurodevelopment, the effects of multiple exposures remain unclear. Unfortunately, studying "multiple exposures" is challenging as it is an extremely heterogeneous descriptor comprising diverse factors. One potentially important, but unrecognized factor is the interval between anesthetic exposures. In order to evaluate the significance of interval, we exposed post-natal day 16, 17 mice to three sevoflurane exposures (2.5%, 1 hr) with short (2 hr) or long (24 hr) intervals. Changes in synaptic transmission, plasticity, protein expression, and behavior were assessed in male and female mice. We discovered that short-interval exposures induced a female-dependent decrease in miniature inhibitory post-synaptic current (mIPSC) frequency 5 days after the last exposure (control: 18.44 ± 2.86 Hz, sevoflurane:14.65 ± 4.54 Hz). Short-interval sevoflurane exposed mice also displayed long-term behavioral deficits at adult age (hypoactivity, anxiety). These behavioral changes were consistent with the sex-dependent changes in inhibitory transmission, as they were more robust in female mice. Although there was no change in learning and memory, short-interval sevoflurane exposures also impaired LTP in a non-sex-dependent manner (control: 171.10 ± 26.90%, sevoflurane: 149.80 ± 26.48 %). Most importantly, we were unable to find long-lasting consequences in mice that received long-interval sevoflurane exposures. Our study provides novel insights regarding the significance of the interval between multiple exposures, and also suggests that the neurotoxic effects of multiple anesthetic exposures may be reduced by simply increasing the interval between each exposure.


Subject(s)
Anesthetics, Inhalation/toxicity , Behavior, Animal/drug effects , Neuronal Plasticity/drug effects , Sevoflurane/toxicity , Synaptic Transmission/drug effects , Anesthetics, Inhalation/administration & dosage , Animals , Animals, Newborn , Brain/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Sevoflurane/administration & dosage , Sex Characteristics
7.
Nutrients ; 12(4)2020 Mar 25.
Article in English | MEDLINE | ID: mdl-32218327

ABSTRACT

Cognitive decline is observed in aging and neurodegenerative diseases, including Alzheimer's disease (AD) and dementia. Intracellular energy produced via mitochondrial respiration is used in the regulation of synaptic plasticity and structure, including dendritic spine length and density, as well as for the release of neurotrophic factors involved in learning and memory. To date, a few synthetic agents for improving mitochondrial function have been developed for overcoming cognitive impairment. However, no natural compounds that modulate synaptic plasticity by directly targeting mitochondria have been developed. Here, we demonstrate that a mixture of Schisandra chinensis extract (SCE) and ascorbic acid (AA) improved cognitive function and induced synaptic plasticity-regulating proteins by enhancing mitochondrial respiration. Treatment of embryonic mouse hippocampal mHippoE-14 cells with a 4:1 mixture of SCE and AA increased basal oxygen consumption rate. We found that mice injected with the SCE-AA mixture showed enhanced learning and memory and recognition ability. We further observed that injection of the SCE-AA mixture in mice significantly increased expression of postsynaptic density protein 95 (PSD95), an increase that was correlated with enhanced brain-derived neurotrophic factor (BDNF) expression. These results demonstrate that a mixture of SCE and AA improves mitochondrial function and memory, suggesting that this natural compound mixture could be used to alleviate AD and aging-associated memory decline.


Subject(s)
Ascorbic Acid/pharmacology , Cell Respiration/drug effects , Cognition/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Plant Extracts/pharmacology , Schisandra/chemistry , Animals , Cell Line , Drug Synergism , Hippocampus/drug effects , Hippocampus/metabolism , Learning/drug effects , Male , Memory/drug effects , Mice , Oxygen Consumption/drug effects , Plant Extracts/chemistry , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism
8.
Front Cell Neurosci ; 14: 4, 2020.
Article in English | MEDLINE | ID: mdl-32047423

ABSTRACT

Preclinical animal studies have continuously reported the possibility of long-lasting neurotoxic effects after general anesthesia in young animals. Such studies also show that the neurological changes induced by anesthesia in young animals differ by their neurodevelopmental stage. Exposure to anesthetic agents increase dendritic spines and induce sex-dependent changes of excitatory/inhibitory synaptic transmission in late postnatal mice, a critical synaptogenic period. However, the mechanisms underlying these changes remain unclear. Abnormal activation of the mammalian target of rapamycin (mTOR) signaling pathway, an important regulator of neurodevelopment, has also been shown to induce similar changes during neurodevelopment. Interestingly, previous studies show that exposure to general anesthetics during neurodevelopment can activate the mTOR signaling pathway. This study, therefore, evaluated the role of mTOR signaling after exposing postnatal day (PND) 16/17 mice to sevoflurane, a widely used inhalation agent in pediatric patients. We first confirmed that a 2-h exposure of 2.5% sevoflurane could induce widespread mTOR phosphorylation in both male and female mice. Pretreatment with the mTOR inhibitor rapamycin not only prevented anesthesia-induced mTOR phosphorylation, but also the increase in mitochondrial respiration and male-dependent enhancement of excitatory synaptic transmission. However, the changes in inhibitory synaptic transmission that appear after anesthesia in female mice were not affected by rapamycin pretreatment. Our results suggest that mTOR inhibitors may act as potential therapeutic agents for anesthesia-induced changes in the developing brain.

9.
Curr Biol ; 30(2): 276-291.e9, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31928877

ABSTRACT

Current pharmacological treatments for Parkinson's disease (PD) are focused on symptomatic relief, but not on disease modification, based on the strong belief that PD is caused by irreversible dopaminergic neuronal death. Thus, the concept of the presence of dormant dopaminergic neurons and its possibility as the disease-modifying therapeutic target against PD have not been explored. Here we show that optogenetic activation of substantia nigra pars compacta (SNpc) neurons alleviates parkinsonism in acute PD animal models by recovering tyrosine hydroxylase (TH) from the TH-negative dormant dopaminergic neurons, some of which still express DOPA decarboxylase (DDC). The TH loss depends on reduced dopaminergic neuronal firing under aberrant tonic inhibition, which is attributed to excessive astrocytic GABA. Blocking the astrocytic GABA synthesis recapitulates the therapeutic effect of optogenetic activation. Consistently, SNpc of postmortem PD patients shows a significant population of TH-negative/DDC-positive dormant neurons surrounded by numerous GABA-positive astrocytes. We propose that disinhibiting dormant dopaminergic neurons by blocking excessive astrocytic GABA could be an effective therapeutic strategy against PD.


Subject(s)
Astrocytes/metabolism , Dopaminergic Neurons/physiology , Nerve Degeneration/physiopathology , Parkinson Disease/physiopathology , Tyrosine 3-Monooxygenase/metabolism , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Down-Regulation , Female , Humans , Immobility Response, Tonic/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Middle Aged , Parkinson Disease/therapy , Rats , Rats, Wistar , Tyrosine 3-Monooxygenase/antagonists & inhibitors , gamma-Aminobutyric Acid/biosynthesis
10.
J Cereb Blood Flow Metab ; 40(7): 1546-1561, 2020 07.
Article in English | MEDLINE | ID: mdl-31987007

ABSTRACT

Cerebral endothelial cells (ECs) require junctional proteins to maintain blood-brain barrier (BBB) integrity, restricting toxic substances and controlling peripheral immune cells with a higher concentration of mitochondria than ECs of peripheral capillaries. The mechanism underlying BBB disruption by defective mitochondrial oxidative phosphorylation (OxPhos) is unclear in a mitochondria-related gene-targeted animal model. To assess the role of EC mitochondrial OxPhos function in the maintenance of the BBB, we developed an EC-specific CR6-interactin factor1 (Crif1) deletion mouse. We clearly observed defects in motor behavior, uncompacted myelin and leukocyte infiltration caused by BBB maturation and disruption in this mice. Furthermore, we investigated the alteration in the actin cytoskeleton, which interacts with junctional proteins to support BBB integrity. Loss of Crif1 led to reorganization of the actin cytoskeleton and a decrease in tight junction-associated protein expression through an ATP production defect in vitro and in vivo. Based on these results, we suggest that mitochondrial OxPhos is important for the maturation and maintenance of BBB integrity by supplying ATP to cerebral ECs.


Subject(s)
Actins/metabolism , Blood-Brain Barrier/metabolism , Cell Cycle Proteins/metabolism , Endothelial Cells/metabolism , Microvessels/metabolism , Mitochondria/metabolism , Animals , Behavior, Animal , Blood-Brain Barrier/pathology , Capillary Permeability , Cell Culture Techniques , Cell Cycle Proteins/genetics , Endothelial Cells/pathology , Gene Knockdown Techniques , Mice , Mice, Knockout , Mice, Transgenic , Microvessels/ultrastructure , Mitochondria/pathology , Oxygen Consumption/physiology , Transfection
11.
Oxid Med Cell Longev ; 2019: 4174803, 2019.
Article in English | MEDLINE | ID: mdl-31534621

ABSTRACT

Paraquat (PQ), an herbicide considered an environmental contributor to the development of Parkinson's disease (PD), induces dopaminergic neuronal loss through reactive oxygen species (ROS) production and oxidative stress by mitochondrial complex I. Most patients with PQ-induced PD are affected by chronic exposure and require a preventive strategy for modulation of disease progression. To identify drugs that are effective in preventing PD, we screened more than 1000 drugs that are currently used in clinics and in studies employing PQ-treated cells. Of these, chloramphenicol (CP) showed the most powerful inhibitory effect. Pretreatment with CP increased the viability of PQ-treated SN4741 dopaminergic neuronal cells and rat primary cultured dopaminergic neurons compared with control cells treated with PQ only. CP pretreatment also reduced PQ-induced ROS production, implying that mitochondrial complex I is a target of CP. This effect of CP reflected downregulation of the mitochondrial complex I subunit ND1 and diminished PQ recycling, a major mechanism of ROS production, and resulted in the prevention of cell loss. Notably, these effects of CP were not observed in rotenone-pretreated SN4741 cells and Rho-negative cells, in which mitochondrial function is defective. Consistent with these results, CP pretreatment of MPTP-treated PD model mice also ameliorated dopaminergic neuronal cell loss. Our findings indicate that the inhibition of mitochondrial complex I with CP protects dopaminergic neurons and may provide a strategy for preventing neurotoxin-induced PD.


Subject(s)
Chloramphenicol/therapeutic use , Herbicides/adverse effects , Mitochondria/drug effects , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Animals , Chloramphenicol/pharmacology , Disease Models, Animal , Humans , Mice , Oxidative Stress , Parkinson Disease/pathology , Rats
12.
Oncol Rep ; 42(5): 2149-2158, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31545464

ABSTRACT

Primary refractory acute myeloid leukemia (AML) and early recurrence of leukemic cells are among the most difficult hurdles to overcome in the treatment of AML. Moreover, uncertainties surrounding the molecular mechanism underlying refractory AML pose a challenge when it comes to developing novel therapeutic drugs. However, accumulating evidence suggests a contribution of phosphatase and tensin homolog (PTEN)/protein kinase B (AKT) signaling to the development of refractory AML. To assess PTEN/AKT signaling in AML, two types of AML cell lines were evaluated, namely control HL60 cells and KG1α cells, a refractory AML cell line that is resistant to idarubicin and cytarabine (AraC) treatment. Changes in the expression level of glycolysis­ and mitochondrial oxidative phosphorylation­related genes and proteins were evaluated by reverse transcription­quantitative polymerase chain reaction and western blot analyses, respectively. The mitochondrial oxygen consumption and extracellular acidification rates were measured using an XF24 analyzer. CCK8 assay and Annexin V/PI staining were used to analyze cell viability and cellular apoptosis, respectively. The PTEN protein was found to be depleted, whereas AKT phosphorylation levels were elevated in KG1α cells compared with HL60 cells. These changes were associated with increased expression of glucose transporter 1 and hexokinase 2, and increased lactate production. AKT inhibition decreased the proliferation of KG1α cells and decreased extracellular acidification without affecting HL60 cells. Notably, AKT inhibition increased the susceptibility of KG1α cells to chemotherapy with idarubicin and AraC. Taken together, the findings of the present study indicate that activation of AKT by PTEN deficiency sustains the refractory AML status through enhancement of glycolysis and mitochondrial respiration, effects that may be rescued by inhibiting AKT activity.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , Cytarabine/pharmacology , Down-Regulation , Gene Expression Regulation, Neoplastic , Glycolysis/drug effects , HL-60 Cells , Humans , Idarubicin/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Oxidative Phosphorylation , Phosphorylation , Signal Transduction
13.
Int J Mol Sci ; 20(14)2019 Jul 11.
Article in English | MEDLINE | ID: mdl-31336718

ABSTRACT

Current therapeutics for Parkinson's disease (PD) are only effective in providing relief of symptoms such as rigidity, tremors and bradykinesia, and do not exert disease-modifying effects by directly modulating mitochondrial function. Here, we investigated auraptene (AUR) as a potent therapeutic reagent that specifically protects neurotoxin-induced reduction of mitochondrial respiration and inhibits reactive oxygen species (ROS) generation. Further, we explored the mechanism and potency of AUR in protecting dopaminergic neurons. Treatment with AUR significantly increased the viability of substantia nigra (SN)-derived SN4741 embryonic dopaminergic neuronal cells and reduced rotenone-induced mitochondrial ROS production. By inducing antioxidant enzymes AUR treatment also increased oxygen consumption rate. These results indicate that AUR exerts a protective effect against rotenone-induced mitochondrial oxidative damage. We further assessed AUR effects in vivo, investigating tyrosine hydroxylase (TH) expression in the striatum and substantia nigra of MPTP-induced PD model mice and behavioral changes after injection of AUR. AUR treatment improved movement, consistent with the observed increase in the number of dopaminergic neurons in the substantia nigra. These results demonstrate that AUR targets dual pathogenic mechanisms, enhancing mitochondrial respiration and attenuating ROS production, suggesting that the preventative potential of this natural compound could lead to improvement in PD-related neurobiological changes.


Subject(s)
Cell Respiration/drug effects , Coumarins/pharmacology , Free Radical Scavengers/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Animals , Behavior, Animal/drug effects , Biomarkers , Coumarins/chemistry , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Free Radical Scavengers/chemistry , Gene Expression , Mice , Models, Biological , Oxidation-Reduction/drug effects , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Reactive Oxygen Species/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
14.
Lab Invest ; 99(9): 1389-1399, 2019 09.
Article in English | MEDLINE | ID: mdl-31043679

ABSTRACT

High-mobility group box 1 (HMGB1) is actively secreted from inflammatory cells and acts via a non-cell-autonomous mechanism to play an important role in mediating cell proliferation and migration. The HMGB1-RAGE (receptor for advanced glycation end products) axis upregulates tyrosine hydroxylase (TH) expression in response to extracellular insults in dopaminergic neurons in vitro, but little is known about HMGB1 in modulation of dopaminergic neurons in vivo. Here, using immunohistochemistry, we show that HMGB1 and RAGE expression are higher in the nigral area of MPTP (methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-treated mice, a toxin-induced Parkinsonian mouse model, compared with saline-treated controls. HMGB1 was predominantly localized to astrocytes and may affect neighboring dopaminergic neurons in the MPTP mouse model, owing to co-localization of RAGE in these TH-positive cells. In addition, MPTP induced a decrease in TH expression, an effect that was potentiated by inhibition of c-Jun N-terminal kinase (JNK) or RAGE. Moreover, stereotaxic injection of recombinant HMGB1 attenuated the MPTP-induced reduction of TH in a Parkinsonian mouse model. Collectively, our results suggest that an increase of HMGB1, released from astrocytes, upregulates TH expression in an acute MPTP-induced Parkinsonian mouse model, thereby maintaining dopaminergic neuronal functions.


Subject(s)
Astrocytes/metabolism , HMGB1 Protein/metabolism , Parkinsonian Disorders/metabolism , Tyrosine 3-Monooxygenase/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/chemically induced , Receptor for Advanced Glycation End Products/metabolism
15.
Neurotoxicology ; 70: 146-153, 2019 01.
Article in English | MEDLINE | ID: mdl-30502404

ABSTRACT

BACKGROUND: Sex plays an important yet often underexplored role in neurodevelopment and neurotoxicity. While several studies report the importance of sex regarding anesthesia-induced neurotoxicity in neonatal mice, only few have focused on the late postnatal period. Here, to further understand the importance of sex regarding the neurobiological changes after early anesthesia during the critical synaptogenic period, we exposed postnatal day 16, 17 (PND 16, 17) mice to sevoflurane in pediatric patients and performed detailed evaluations in the hippocampus. METHODS: PND 16, 17 mice received a single exposure of oxygen with or without sevoflurane (2.5%) for 2 h. Changes of the hippocampus were analyzed in male and female mice 6 h after exposure: excitatory/inhibitory synaptic transmission, protein/mRNA expression levels of excitatory/inhibitory synaptic molecules (GluR1, GluR2, PSD95, gephyrin, GAD65), and number of excitatory synapses. RESULTS: Sevoflurane exposure increased the frequency of miniature excitatory postsynaptic currents specifically in male mice (control: 0.07 ± 0.04 [Hz]; sevoflurane: 14.72 ± 0.08 [Hz]), while miniature inhibitory postsynaptic currents were affected specifically in female mice. The protein/mRNA expression levels of excitatory synaptic molecules were also increased specifically in male mice. Unexpectedly, protein/mRNA expression levels of inhibitory synaptic molecules were increased in both sexes, and there was no male-specific increase of excitatory synapse number. CONCLUSIONS: Exposure of mice to sevoflurane during the critical, late postnatal period induces sex-dependent changes in the hippocampus. Although often disregarded, our results confirm the importance of sex as a biological variable when studying the changes triggered by early anesthesia.


Subject(s)
Anesthetics, Inhalation/toxicity , Excitatory Postsynaptic Potentials/physiology , Hippocampus/growth & development , Inhibitory Postsynaptic Potentials/physiology , Sex Characteristics , Synapses/physiology , Anesthetics, Inhalation/administration & dosage , Animals , Animals, Newborn , Excitatory Postsynaptic Potentials/drug effects , Female , Hippocampus/drug effects , Hippocampus/ultrastructure , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice , Mice, Inbred C57BL , Sevoflurane/administration & dosage , Sevoflurane/toxicity , Synapses/drug effects , Synapses/ultrastructure
16.
Oncol Rep ; 40(6): 3869-3878, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30272370

ABSTRACT

The identification of large numbers of genetic mutations in immature myeloid cells has made it difficult to identify specific targets for acute myeloid leukemia (AML) therapy. Although current pharmacological targets for controlling cancer are focused on identifying genetic mutations, it is hard to develop the specific drugs to achieve complete remission due to complex and variable genetic mutations. To overcome the failure of the genetic mutation theory, the present study targeted mitochondrial metabolism as a strategy for inducing anti­leukemic activity, based on evidence that AML cells have an abnormally high amount of mitochondria and that somatic mutations can alter metabolic flux in cancer. It was found that L­deprenyl, which is clinically available for the treatment of Parkinson's disease, exerts anti­mitochondria activity in KG­1α cells, as assessed by detection of oxygen consumption rate (OCR) and extracellular acidification (ECAR) using XF analyzer, respectively. Using a luciferase assay for detecting adenosine triphosphate (ATP) content, it was found that suppression of mitochondrial activity led to ATP depletion and was associated with potent cytotoxic activity. L­deprenyl is known to target monoamine oxidase­B (MAO­B) on the outer membrane of mitochondria, therefore, the activity of MAO­A and ­B was measured based on the fluorometric detection of H2O2 produced by the enzyme reaction. Notably, MAO­A and -B activity was low in AML cells and the present findings suggested that the anticancer effect of L­deprenyl was independent of MAO­B. Change of mitochondrial respiration­ and glycolysis­related gene expression levels were measured by reverse transcription­quantitative polymerase chain reaction. Consistent with the aforementioned results, treatment with L­deprenyl reduced the mRNA level of mitochondrial respiration­ and glycolysis­related genes. Collectively, the present results identify L­deprenyl as a novel candidate for the treatment of AML through inhibition of mitochondrial respiration.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Mitochondria/metabolism , Monoamine Oxidase/metabolism , Selegiline/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Male , Mice , Mitochondria/drug effects , Mutation , Oxygen Consumption/drug effects , Selegiline/pharmacology , Xenograft Model Antitumor Assays
17.
Oncol Rep ; 39(1): 239-246, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29192322

ABSTRACT

Among brain tumors, glioblastoma (GBM) is the most aggressive type and is associated with the lowest patient survival rate. Numerous lines of evidence have established that omega-3-polyunsaturated fatty acids (ω3-PUFAs) have potential for the prevention and therapy of several types of cancers. Docosahexaenoic acid (DHA), an ω3-PUFA, was reported to inhibit growth and induce apoptotic and autophagic cell death in several cancer cell lines; however, its effects on GBM cells are still unknown. in the present study, we examined the cytotoxic effect of DHA on the GBM cell lines, D54MG, U87MG, U251MG and GL261. Treatment of GBM cells with DHA induced PARP cleavage, increased the population of sub-G1 cells, and increased the number of TUNEL-positive cells, which are all indicative of apoptosis. Furthermore, treatment of GBM cells with DHA resulted in a significant increase in autophagic activity, as revealed by increased LC3-II levels, GFP-LC3 puncta, and autophagic flux activation, accompanied by activation of 5'-AMP-activated protein kinase (AMPK) and decreases in phosphorylated Akt (p-AktSer473) levels and mTOR activity. In vivo, endogenous expression of Caenorhabditis elegans ω3-desaturase, which converts ω6-PUFAs to ω3-PUFAs, in fat-1 transgenic mice yielded a significant decrease in tumor volume following subcutaneous injection of mouse glioma cells (GL261), when compared with wild-type mice. TUNEL-positive cell numbers and LC3-II levels were elevated in tumor tissue from the fat-1 transgenic mice compared with tumor tissue from the wild-type mice. In addition, p-Akt levels were decreased and p-AMPK levels were increased in tumor tissue from the fat-1 transgenic mice. These results indicate that ω3-PUFAs induce cell death through apoptosis and autophagy in GBM cells; thus, it may be possible to use ω3-PUFAs as chemopreventive and therapeutic agents for GBM.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Docosahexaenoic Acids/administration & dosage , Fatty Acid Desaturases/genetics , Glioblastoma/drug therapy , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Autophagy , Brain Neoplasms/metabolism , Caenorhabditis elegans/enzymology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Docosahexaenoic Acids/pharmacology , Fatty Acid Desaturases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Humans , Mice , Mice, Transgenic , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Xenograft Model Antitumor Assays
18.
Theranostics ; 7(18): 4632-4642, 2017.
Article in English | MEDLINE | ID: mdl-29158850

ABSTRACT

Rationale: Signal transducer and activator of transcription-3 (STAT3) plays a pivotal role in cancer biology. Many small-molecule inhibitors that target STAT3 have been developed as potential anticancer drugs. While designing small-molecule inhibitors that target the SH2 domain of STAT3 remains the leading focus for drug discovery, there has been a growing interest in targeting the DNA-binding domain (DBD) of the protein. Methods: We demonstrated the potential antitumor activity of a novel, small-molecule (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) that directly binds to the DBD of STAT3, in patient-derived non-small cell lung cancer (NSCLC) xenograft model as well as in NCI-H460 cell xenograft model in nude mice. Results: MMPP effectively inhibited the phosphorylation of STAT3 and its DNA binding activity in vitro and in vivo. It induced G1-phase cell cycle arrest and apoptosis through the regulation of cell cycle- and apoptosis-regulating genes by directly binding to the hydroxyl residue of threonine 456 in the DBD of STAT3. Furthermore, MMPP showed a similar or better antitumor activity than that of docetaxel or cisplatin. Conclusion: MMPP is suggested to be a potential candidate for further development as an anticancer drug that targets the DBD of STAT3.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , DNA-Binding Proteins/metabolism , DNA/metabolism , Lung Neoplasms/drug therapy , Phthalic Acids/pharmacology , STAT3 Transcription Factor/metabolism , A549 Cells , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Biochem Biophys Res Commun ; 493(1): 358-364, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28887039

ABSTRACT

The derangement of tyrosine hydroxylase (TH) activity reduces dopamine synthesis and is implicated in the pathogenesis of Parkinson's disease. However, the extracellular modulator and intracellular regulatory mechanisms of TH have yet to be identified. Recently, high-mobility group box 1 (HMGB1) was reported to be actively secreted from glial cells and is regarded as a mediator of dopaminergic neuronal loss. However, the mechanism for how HMGB1 affects TH expression, particularly through the receptor for advanced glycation endproducts (RAGE), has not yet been investigated. We found that recombinant HMGB1 (rHMGB1) upregulates TH mRNA expression via simultaneous activation of JNK phosphorylation, and this induction of TH expression is blocked by inhibitors of RAGE and JNK. To investigate how TH expression levels change through the HMGB1-RAGE axis as a result of MPP+ toxicity, we co-treated SN4741 dopaminergic cells with MPP+ and rHMGB1. rHMGB1 blocked the reduction of TH mRNA following MPP+ treatment without altering cell survival rates. Our results suggest that HMGB1 upregulates TH expression to maintain dopaminergic neuronal function via activating RAGE, which is dependent on JNK phosphorylation.


Subject(s)
Dopaminergic Neurons/physiology , HMGB1 Protein/metabolism , MAP Kinase Kinase 4/metabolism , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/physiology , Tyrosine 3-Monooxygenase/metabolism , Animals , Cell Line , Phosphorylation , Rats , Up-Regulation/physiology
20.
Exp Neurobiol ; 26(2): 104-112, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28442947

ABSTRACT

Movement defects in obesity are associated with peripheral muscle defects, arthritis, and dysfunction of motor control by the brain. Although movement functionality is negatively correlated with obesity, the brain regions and downstream signaling pathways associated with movement defects in obesity are unclear. A dopaminergic neuronal pathway from the substantia nigra (SN) to the striatum is responsible for regulating grip strength and motor initiation through tyrosine hydroxylase (TH) activity-dependent dopamine release. We found that mice fed a high-fat diet exhibited decreased movement in open-field tests and an increase in missteps in a vertical grid test compared with normally fed mice. This motor abnormality was associated with a significant reduction of TH in the SN and striatum. We further found that phosphorylation of c-Jun N-terminal kinase (JNK), which modulates TH expression in the SN and striatum, was decreased under excess-energy conditions. Our findings suggest that high calorie intake impairs motor function through JNK-dependent dysregulation of TH in the SN and striatum.

SELECTION OF CITATIONS
SEARCH DETAIL
...