Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Can J Physiol Pharmacol ; 97(10): 999-1005, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31295412

ABSTRACT

Molecular hydrogen (H2) was believed to be an inert and nonfunctional molecule in mammalian cells; however, we overturned the concept by reporting the therapeutic effects of H2 against oxidative stress. Subsequently, extensive studies revealed multiple functions of H2 by exhibiting the efficacies of H2 in various animal models and clinical studies. Here, we investigated the effect of H2 on free-radical-induced cytotoxicity using tert-butyl hydroperoxide in a human acute monocytic leukemia cell line, THP-1. Cell membrane permeability was determined using lactate dehydrogenase release assay and Hoechst 33342 and propidium iodide staining. Fatty acid peroxidation and mitochondrial viability were measured using 2 kinds of fluorescent dyes, Liperfluo and C11-BODIPY, and using the alamarBlue assay based on the reduction of resazurin to resorufin by mainly mitochondrial succinate dehydrogenase, respectively. Mitochondrial membrane potential was evaluated using tetramethylrhodamine methyl ester. As a result, H2 protected the cultured cells against the cytotoxic effects induced by tert-butyl hydroperoxide; H2 suppressed cellular fatty acid peroxidation and cell membrane permeability, mitigated the decline in mitochondrial oxidoreductase activity and mitochondrial membrane potential, and protected cells against cell death evaluated using propidium iodide staining. These results suggested that H2 suppresses free-radical-induced cell death through protection against fatty acid peroxidation and mitochondrial dysfunction.


Subject(s)
Hydrogen/pharmacology , Mitochondria/drug effects , tert-Butylhydroperoxide/toxicity , Apoptosis/drug effects , Atherosclerosis/drug therapy , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Fatty Acids/metabolism , Humans , Hydrogen/therapeutic use , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects
2.
Curr Alzheimer Res ; 15(5): 482-492, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29110615

ABSTRACT

BACKGROUND: Oxidative stress is one of the causative factors in the pathogenesis of neurodegenerative diseases including mild cognitive impairment (MCI) and dementia. We previously reported that molecular hydrogen (H2) acts as a therapeutic and preventive antioxidant. OBJECTIVE: We assess the effects of drinking H2-water (water infused with H2) on oxidative stress model mice and subjects with MCI. METHODS: Transgenic mice expressing a dominant-negative form of aldehyde dehydrogenase 2 were used as a dementia model. The mice with enhanced oxidative stress were allowed to drink H2-water. For a randomized double-blind placebo-controlled clinical study, 73 subjects with MCI drank ~300 mL of H2-water (H2-group) or placebo water (control group) per day, and the Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-cog) scores were determined after 1 year. RESULTS: In mice, drinking H2-water decreased oxidative stress markers and suppressed the decline of memory impairment and neurodegeneration. Moreover, the mean lifespan in the H2-water group was longer than that of the control group. In MCI subjects, although there was no significant difference between the H2- and control groups in ADAS-cog score after 1 year, carriers of the apolipoprotein E4 (APOE4) genotype in the H2-group were improved significantly on total ADAS-cog score and word recall task score (one of the sub-scores in the ADAS-cog score). CONCLUSION: H2-water may have a potential for suppressing dementia in an oxidative stress model and in the APOE4 carriers with MCI.


Subject(s)
Antioxidants/administration & dosage , Cognitive Dysfunction/drug therapy , Hydrogen/administration & dosage , Administration, Oral , Aged , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Animals , Apolipoproteins E/genetics , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Disease Models, Animal , Double-Blind Method , Drinking Water , Female , Humans , Male , Mental Processes/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/psychology , Neuroprotective Agents/administration & dosage , Oxidative Stress , Treatment Outcome
3.
Plast Reconstr Surg ; 140(6): 1195-1206, 2017 12.
Article in English | MEDLINE | ID: mdl-30998612

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury is one of the leading causes of tissue damage and dysfunction, in particular, free tissue transfer, traumatically amputated extremity, and prolonged tourniquet application during extremity surgery. In this study, the authors investigated the therapeutic effects of hydrogen gas on skeletal muscle ischemia-reperfusion injury. METHODS: The authors compared the concentration of hydrogen in a muscle on intraperitoneal administration of hydrogen-rich saline and on inhalation of hydrogen gas. Animals were subjected to ischemia-reperfusion. Mice were treated with inhalation of hydrogen gas, and the hind gastrocnemius muscle was collected. Muscle morphology and inflammatory change were evaluated after ischemia-reperfusion. Moreover, a footprint test was performed to assess the functional effect of hydrogen. RESULTS: Hydrogen concentration of tissue was significantly higher, and the elevated level was maintained longer by hydrogen gas inhalation than by intraperitoneal administration of hydrogen-rich saline. Infarct zone and area with loss of tissue structure and marked cellular infiltration were significantly decreased in groups treated by hydrogen gas inhalation during ischemia-reperfusion; however, these effects were not observed by posttreatment of hydrogen. One week after ischemia-reperfusion, mice that had been pretreated with hydrogen gas recovered faster and achieved smoother walking in appearance compared with mice in the other groups as assessed by the footprint test. CONCLUSIONS: Inhalation of hydrogen gas attenuates muscle damage, inhibits inflammatory response, and enhances functional recovery. These findings suggest that the optimal route for hydrogen delivery is continuous inhalation of hydrogen gas, which could be a novel clinical mode of treatment in ischemia-reperfusion injury.


Subject(s)
Hydrogen/administration & dosage , Muscle, Skeletal/pathology , Reperfusion Injury/therapy , Administration, Inhalation , Animals , Disease Models, Animal , Hindlimb/blood supply , Hindlimb/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/blood supply , Muscle, Skeletal/physiopathology , Recovery of Function , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Treatment Outcome
4.
Sci Rep ; 6: 18971, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26739257

ABSTRACT

We previously showed that H2 acts as a novel antioxidant to protect cells against oxidative stress. Subsequently, numerous studies have indicated the potential applications of H2 in therapeutic and preventive medicine. Moreover, H2 regulates various signal transduction pathways and the expression of many genes. However, the primary targets of H2 in the signal transduction pathways are unknown. Here, we attempted to determine how H2 regulates gene expression. In a pure chemical system, H2 gas (approximately 1%, v/v) suppressed the autoxidation of linoleic acid that proceeds by a free radical chain reaction, and pure 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphocholine (PAPC), one of the major phospholipids, was autoxidized in the presence or absence of H2. H2 modified the chemical production of the autoxidized phospholipid species in the cell-free system. Exposure of cultured cells to the H2-dependently autoxidized phospholipid species reduced Ca(2+) signal transduction and mediated the expression of various genes as revealed by comprehensive microarray analysis. In the cultured cells, H2 suppressed free radical chain reaction-dependent peroxidation and recovered the increased cellular Ca(2+), resulting in the regulation of Ca(2+)-dependent gene expression. Thus, H2 might regulate gene expression via the Ca(2+) signal transduction pathway by modifying the free radical-dependent generation of oxidized phospholipid mediators.


Subject(s)
Free Radicals/pharmacology , Gene Expression Regulation/drug effects , Hydrogen/pharmacology , Phosphatidylcholines/metabolism , Calcium Signaling , Cell Line , Humans , Linoleic Acid/metabolism , NFATC Transcription Factors/metabolism , Oxidation-Reduction , Phosphatidylcholines/chemistry , Transcriptome/drug effects
5.
Brain Res ; 1605: 49-58, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25698614

ABSTRACT

Stem cell transplantation therapy is currently in clinical trials for the treatment of ischemic stroke, and several beneficial aspects have been reported. Similarly, in Alzheimer's disease (AD), stem cell therapy is expected to provide an efficient therapeutic approach. Indeed, the intracerebral transplantation of stem cells reduced amyloid-ß (Aß) deposition and rescued memory deficits in AD model mice. Here, we show that intravenous transplantation of bone marrow-derived mononuclear cells (BMMCs) improves cognitive function in two different AD mouse models, DAL and APP mice, and prevents neurodegeneration. GFP-positive BMMCs were isolated from tibiae and femurs of 4-week-old mice and then transplanted intravenously into DAL and APP mice. Transplantation of BMMCs suppressed neuronal loss and restored memory impairment of DAL mice to almost the same level as in wild-type mice. Transplantation of BMMCs to APP mice reduced Aß deposition in the brain. APP mice treated with BMMCs performed significantly better on behavioral tests than vehicle-injected mice. Moreover, the effects were observed even with transplantation after the onset of cognitive impairment in DAL mice. Together, our results indicate that intravenous transplantation of BMMCs has preventive effects against the cognitive decline in AD model mice and suggest a potential therapeutic effect of BMMC transplantation therapy.


Subject(s)
Alzheimer Disease/therapy , Bone Marrow Transplantation/methods , Cognition Disorders/prevention & control , Leukocytes, Mononuclear/transplantation , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Bone Marrow Cells , Cognition Disorders/etiology , Disease Models, Animal , Female , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Plaque, Amyloid/metabolism
6.
Neurosci Lett ; 587: 126-31, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25529196

ABSTRACT

Oxidative stress is known to play a prominent role in the onset and early stage progression of Alzheimer's disease (AD). For example, protein oxidation and lipid peroxidation levels are increased in patients with mild cognitive impairment. Here, we created a double-transgenic mouse model of AD to explore the pathological and behavioral effects of oxidative stress. Double transgenic (APP/DAL) mice were constructed by crossing Tg2576 (APP) mice, which express a mutant form of human amyloid precursor protein (APP), with DAL mice expressing a dominant-negative mutant of mitochondrial aldehyde dehydrogenase 2 (ALDH2), in which oxidative stress is enhanced. Y-maze and object recognition tests were performed at 3 and 6 months of age to evaluate learning and memory. The accumulation of amyloid plaques, deposition of phosphorylated-tau protein, and number of astrocytes in the brain were assessed histopathologically at 3, 6, 9, and 12-15 months of age. The life span of APP/DAL mice was significantly shorter than that of APP or DAL mice. In addition, they showed accelerated amyloid deposition, tau phosphorylation, and gliosis. Furthermore, these mice showed impaired performance on Y-maze and object recognition tests at 3 months of age. These data suggest that oxidative stress accelerates cognitive dysfunction and pathological insults in the brain. APP/DAL mice could be a useful model for exploring new approaches to AD treatment.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid/metabolism , Memory Disorders/psychology , Oxidative Stress , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Gliosis , Learning , Memory Disorders/genetics , Mice, Transgenic , Mitochondrial Proteins/genetics , Phosphorylation , tau Proteins/metabolism
7.
BMC Complement Altern Med ; 14: 390, 2014 Oct 11.
Article in English | MEDLINE | ID: mdl-25305781

ABSTRACT

BACKGROUND: Oxidative stress is involved in age-related muscle atrophy, such as sarcopenia. Since Chlorella, a unicellular green alga, contains various antioxidant substances, we used a mouse model of enhanced oxidative stress to investigate whether Chlorella could prevent muscle atrophy. METHODS: Aldehyde dehydrogenase 2 (ALDH2) is an anti-oxidative enzyme that detoxifies reactive aldehydes derived from lipid peroxides such as 4-hydroxy-2-nonenal (4-HNE). We therefore used transgenic mice expressing a dominant-negative form of ALDH2 (ALDH2*2 Tg mice) to selectively decrease ALDH2 activity in the muscles. To evaluate the effect of Chlorella, the mice were fed a Chlorella-supplemented diet (CSD) for 6 months. RESULTS: ALDH2*2 Tg mice exhibited small body size, muscle atrophy, decreased fat content, osteopenia, and kyphosis, accompanied by increased muscular 4-HNE levels. The CSD helped in recovery of body weight, enhanced oxidative stress, and increased levels of a muscle impairment marker, creatine phosphokinase (CPK) induced by ALDH2*2. Furthermore, histological and histochemical analyses revealed that the consumption of the CSD improved skeletal muscle atrophy and the activity of the mitochondrial cytochrome c oxidase. CONCLUSIONS: This study suggests that long-term consumption of Chlorella has the potential to prevent age-related muscle atrophy.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Chlorella/chemistry , Chlorella/metabolism , Mitochondria/enzymology , Muscle, Skeletal/enzymology , Muscular Atrophy/prevention & control , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Atrophy/diet therapy , Muscular Atrophy/enzymology , Muscular Atrophy/metabolism , Oxidative Stress
8.
J Invest Dermatol ; 134(6): 1701-1709, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24129062

ABSTRACT

Oxidative stress is involved in many age-associated diseases, as well as in the aging process itself. The development of interventions to reduce oxidative stress is hampered by the absence of sensitive detection methods that can be used in live animals. We generated transgenic mice expressing ratiometric redox-sensitive green fluorescent protein (roGFP) in the cytosol or mitochondria of several tissues, including skin epidermal keratinocytes. Crossbreeding into hairless albino mice allowed noninvasive optical measurement of skin oxidative state. Topical application of hydrogen peroxide emulsion shifted the keratinocyte redox state toward oxidation within minutes and could be observed in real time by fluorescence ratio imaging. Exposing skin to 365 nm UVA radiation oxidized roGFP localized in keratinocyte mitochondria, but not when roGFP was localized in the cytosol. This suggests that significant amounts of the endogenous photosensitizers that mediate UVA-induced oxidative stress are located in the mitochondria. UVR is the major environmental cause of skin aging and UVA-mediated oxidative stress has been associated with the development of wrinkles in humans. Direct measurements of redox state in defined cell compartments of live animals should be a powerful and convenient tool for evaluating treatments that aim to modulate oxidative stress.


Subject(s)
Oxidative Stress , Skin/metabolism , Skin/physiopathology , Animals , Cytosol/metabolism , Emulsions , Epidermis/metabolism , Green Fluorescent Proteins/metabolism , Humans , Hydrogen Peroxide/pharmacology , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Optics and Photonics , Oxidation-Reduction , Oxygen/metabolism , Transgenes , Ultraviolet Rays
9.
Intern Med ; 51(24): 3351-7, 2012.
Article in English | MEDLINE | ID: mdl-23257519

ABSTRACT

OBJECTIVE: Post-transcriptional taurine modification at the first anticodon ("wobble") nucleotide is deficient in A3243G-mutant mitochondrial (mt) tRNA(Leu(UUR)) of patients with myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS). Wobble nucleotide modifications in tRNAs have recently been identified to be important in the accurate and efficient deciphering of codons. We herein examined whether taurine can alleviate mitochondrial dysfunction in patient-derived pathogenic cells and prevent clinical symptoms in MELAS patients. METHODS AND RESULTS: The addition of taurine to the culture media ameliorated the reduced oxygen consumption, decreased the mitochondrial membrane potential, and increased the oxidative stress in MELAS patient-derived cells. Moreover, high dose oral administration of taurine (0.25 g/kg/day) completely prevented stroke-like episodes in two MELAS patients for more than nine years. CONCLUSION: Taurine supplementation may be a novel potential treatment option for preventing the stroke-like episodes associated with MELAS.


Subject(s)
MELAS Syndrome/complications , MELAS Syndrome/physiopathology , Mitochondria/drug effects , Mitochondria/physiology , Stroke/etiology , Stroke/prevention & control , Taurine/therapeutic use , Adult , Cells, Cultured , Female , Humans , Taurine/pharmacology , Young Adult
10.
Obesity (Silver Spring) ; 19(7): 1396-403, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21293445

ABSTRACT

Recent extensive studies have revealed that molecular hydrogen (H(2)) has great potential for improving oxidative stress-related diseases by inhaling H(2) gas, injecting saline with dissolved H(2), or drinking water with dissolved H(2) (H(2)-water); however, little is known about the dynamic movement of H(2) in a body. First, we show that hepatic glycogen accumulates H(2) after oral administration of H(2)-water, explaining why consumption of even a small amount of H(2) over a short span time efficiently improves various disease models. This finding was supported by an in vitro experiment in which glycogen solution maintained H(2). Next, we examined the benefit of ad libitum drinking H(2)-water to type 2 diabetes using db/db obesity model mice lacking the functional leptin receptor. Drinking H(2)-water reduced hepatic oxidative stress, and significantly alleviated fatty liver in db/db mice as well as high fat-diet-induced fatty liver in wild-type mice. Long-term drinking H(2)-water significantly controlled fat and body weights, despite no increase in consumption of diet and water. Moreover, drinking H(2)-water decreased levels of plasma glucose, insulin, and triglyceride, the effect of which on hyperglycemia was similar to diet restriction. To examine how drinking H(2)-water improves obesity and metabolic parameters at the molecular level, we examined gene-expression profiles, and found enhanced expression of a hepatic hormone, fibroblast growth factor 21 (FGF21), which functions to enhance fatty acid and glucose expenditure. Indeed, H(2) stimulated energy metabolism as measured by oxygen consumption. The present results suggest the potential benefit of H(2) in improving obesity, diabetes, and metabolic syndrome.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Energy Metabolism , Fibroblast Growth Factors/metabolism , Hydrogen/therapeutic use , Liver/metabolism , Obesity/therapy , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Fatty Liver/etiology , Fatty Liver/prevention & control , Fibroblast Growth Factors/genetics , Glycogen/chemistry , Glycogen/metabolism , Hydrogen/administration & dosage , Hydrogen/analysis , Hydrogen/metabolism , Hyperglycemia/etiology , Hyperglycemia/prevention & control , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Oxidative Stress , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Water/chemistry
11.
Circ Res ; 105(11): 1118-27, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19815821

ABSTRACT

RATIONALE: Aldehyde accumulation is regarded as a pathognomonic feature of oxidative stress-associated cardiovascular disease. OBJECTIVE: We investigated how the heart compensates for the accelerated accumulation of aldehydes. METHODS AND RESULTS: Aldehyde dehydrogenase 2 (ALDH2) has a major role in aldehyde detoxification in the mitochondria, a major source of aldehydes. Transgenic (Tg) mice carrying an Aldh2 gene with a single nucleotide polymorphism (Aldh2*2) were developed. This polymorphism has a dominant-negative effect and the Tg mice exhibited impaired ALDH activity against a broad range of aldehydes. Despite a shift toward the oxidative state in mitochondrial matrices, Aldh2*2 Tg hearts displayed normal left ventricular function by echocardiography and, because of metabolic remodeling, an unexpected tolerance to oxidative stress induced by ischemia/reperfusion injury. Mitochondrial aldehyde stress stimulated eukaryotic translation initiation factor 2alpha phosphorylation. Subsequent translational and transcriptional activation of activating transcription factor-4 promoted the expression of enzymes involved in amino acid biosynthesis and transport, ultimately providing precursor amino acids for glutathione biosynthesis. Intracellular glutathione levels were increased 1.37-fold in Aldh2*2 Tg hearts compared with wild-type controls. Heterozygous knockout of Atf4 blunted the increase in intracellular glutathione levels in Aldh2*2 Tg hearts, thereby attenuating the oxidative stress-resistant phenotype. Furthermore, glycolysis and NADPH generation via the pentose phosphate pathway were activated in Aldh2*2 Tg hearts. (NADPH is required for the recycling of oxidized glutathione.) CONCLUSIONS: The findings of the present study indicate that mitochondrial aldehyde stress in the heart induces metabolic remodeling, leading to activation of the glutathione-redox cycle, which confers resistance against acute oxidative stress induced by ischemia/reperfusion.


Subject(s)
Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehydes/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Oxidative Stress/physiology , Activating Transcription Factor 4/metabolism , Adaptation, Physiological/physiology , Aldehyde Dehydrogenase, Mitochondrial , Animals , Disease Models, Animal , Echocardiography , Enzyme Activation/physiology , Enzyme Induction/physiology , Glucose/metabolism , Glutathione/metabolism , Metabolome/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/physiology , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Oligonucleotide Array Sequence Analysis , Pentose Phosphate Pathway/physiology , Transcription, Genetic/physiology
12.
Biochem Biophys Res Commun ; 377(4): 1195-8, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18996093

ABSTRACT

Oxidative stress is implicated in atherogenesis; however most clinical trials with dietary antioxidants failed to show marked success in preventing atherosclerotic diseases. We have found that hydrogen (dihydrogen; H(2)) acts as an effective antioxidant to reduce oxidative stress [I. Ohsawa, M. Ishikawa, K. Takahashi, M. Watanabe, K. Nishimaki, K. Yamagata, K. Katsura, Y. Katayama, S, Asoh, S. Ohta, Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals, Nat. Med. 13 (2007) 688-694]. Here, we investigated whether drinking H(2)-dissolved water at a saturated level (H(2)-water) ad libitum prevents arteriosclerosis using an apolipoprotein E knockout mouse (apoE(-/-)), a model of the spontaneous development of atherosclerosis. ApoE(-/-) mice drank H(2)-water ad libitum from 2 to 6 month old throughout the whole period. Atherosclerotic lesions were significantly reduced by ad libitum drinking of H(2)-water (p=0.0069) as judged by Oil-Red-O staining series of sections of aorta. The oxidative stress level of aorta was decreased. Accumulation of macrophages in atherosclerotic lesions was confirmed. Thus, consumption of H(2)-dissolved water has the potential to prevent arteriosclerosis.


Subject(s)
Antioxidants/administration & dosage , Atherosclerosis/prevention & control , Hydrogen/administration & dosage , Water/administration & dosage , Animals , Antioxidants/chemistry , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Drinking , Hydrogen/chemistry , Mice , Mice, Knockout , Oxidative Stress , Water/chemistry
13.
J Neurosci ; 28(24): 6239-49, 2008 Jun 11.
Article in English | MEDLINE | ID: mdl-18550766

ABSTRACT

Oxidative stress may underlie age-dependent memory loss and cognitive decline. Toxic aldehydes, including 4-hydroxy-2-nonenal (HNE), an end product of lipid peroxides, are known to accumulate in the brain in neurodegenerative disease. We have previously shown that mitochondrial aldehyde dehydrogenase 2 (ALDH2) detoxifies HNE by oxidizing its aldehyde group. To investigate the role of such toxic aldehydes, we produced transgenic mice, which expressed a dominant-negative form of ALDH2 in the brain. The mice had decreased ability to detoxify HNE in their cortical neurons and accelerated accumulation of HNE in the brain. Consequently, their lifespan was shortened and age-dependent neurodegeneration and hyperphosphorylation of tau were observed. Object recognition and Morris water maze tests revealed that the onset of cognitive impairment correlated with the degeneration, which was further accelerated by APOE (apolipoprotein E) knock-out; therefore, the accumulation of toxic aldehydes is by itself critical in the progression of neurodegenerative disease, which could be suppressed by ALDH2.


Subject(s)
Aging/physiology , Aldehyde Dehydrogenase/deficiency , Memory Disorders/complications , Memory Disorders/genetics , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Analysis of Variance , Animals , Behavior, Animal/physiology , Cells, Cultured , Cerebral Cortex/pathology , Embryo, Mammalian , Exploratory Behavior/physiology , Gene Expression Regulation, Developmental/physiology , Humans , In Situ Nick-End Labeling/methods , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Pattern Recognition, Visual/physiology , Peptide Elongation Factor 1/genetics , Psychomotor Performance/physiology
14.
Endocr J ; 55(3): 509-14, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18445996

ABSTRACT

Mitochondrial diabetes is characterized by diabetes and hearing loss in maternal transmission with a heteroplasmic A3243G mutation in the mitochondrial gene. In patients with the mutation, it has been reported that hepatic involvement is rarely observed. We demonstrated a case of hypertrophic cardiomyopathy and hepatic failure with mitochondrial diabetes. To clarify the pathogenesis we analyzed the mitochondrial ultrastructure in the myocytes, the reactive oxygen species (ROS) production in the liver and the status of heteroplasmy of the mitochondrial A3243G mutation in the organs involved. In cardiomyocytes and skeletal muscle, electron microscopic analysis demonstrated typical morphological mitochondrial abnormalities. Immunohistochemical analysis demonstrated enhanced ROS production associated with marked steatosis in the liver, which is often associated with mitochondrial dysfunction. Analysis of the A3243G mutation revealed a substantial ratio of heteroplasmy in these organs including the liver. The presence of steatosis and enhanced oxidative stress in the liver suggested that hepatic failure was associated with mitochondrial dysfunction.


Subject(s)
Diabetes Mellitus/genetics , Genes, Mitochondrial , Liver Failure/etiology , Oxidative Stress/physiology , Autopsy , Diabetes Complications/genetics , Fatal Outcome , Genes, Mitochondrial/physiology , Humans , Liver Failure/genetics , Liver Failure/pathology , Male , Middle Aged , Mutation/physiology , Oxidative Stress/genetics , RNA, Transfer, Leu/genetics
15.
Life Sci ; 82(3-4): 218-25, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18164732

ABSTRACT

Many anticancer drugs attack rapidly dividing cells, including not only malignant cells but also hair follicle cells, and induce alopecia. Chemotherapy-induced alopecia (CIA) is an emotionally distressing side effect of cancer chemotherapy. There is currently no useful preventive therapy for CIA. We have previously constructed anti-death rFNK protein from rat Bcl-x(L) by site-directed mutagenesis to strengthen cytoprotective activity. When fused to the protein transduction domain (PTD) of HIV/Tat, the fusion protein PTD (TAT)-rFNK successfully entered cells from the outside in vitro and in vivo to exhibit anti-death activity against apoptosis and necrosis. Here, we show that topical application of FNK protected against CIA in a newborn rat model. The protective activity against hair-loss was observed in 30-1000 nM TAT-rFNK administrative groups in a dose-dependent manner. Furthermore, a human version of FNK (hFNK) fused to other PTD peptides exhibited a protective ability. These results suggest that PTD-FNK possesses protective activity against CIA and is not restricted to a sequence of PTD peptides or species of FNK. Thus, PTD-FNK represents potential to develop a useful method for preventing CIA in cancer patients.


Subject(s)
Alopecia/prevention & control , Antineoplastic Agents, Phytogenic/toxicity , Cell Cycle Proteins/therapeutic use , Etoposide/toxicity , Protein Serine-Threonine Kinases/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Alopecia/chemically induced , Alopecia/pathology , Animals , Animals, Newborn , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Hair Follicle/drug effects , Hair Follicle/metabolism , Hair Follicle/pathology , Humans , Injections, Intraperitoneal , Injections, Subcutaneous , Protective Agents , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
16.
Nat Med ; 13(6): 688-94, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17486089

ABSTRACT

Acute oxidative stress induced by ischemia-reperfusion or inflammation causes serious damage to tissues, and persistent oxidative stress is accepted as one of the causes of many common diseases including cancer. We show here that hydrogen (H(2)) has potential as an antioxidant in preventive and therapeutic applications. We induced acute oxidative stress in cultured cells by three independent methods. H(2) selectively reduced the hydroxyl radical, the most cytotoxic of reactive oxygen species (ROS), and effectively protected cells; however, H(2) did not react with other ROS, which possess physiological roles. We used an acute rat model in which oxidative stress damage was induced in the brain by focal ischemia and reperfusion. The inhalation of H(2) gas markedly suppressed brain injury by buffering the effects of oxidative stress. Thus H(2) can be used as an effective antioxidant therapy; owing to its ability to rapidly diffuse across membranes, it can reach and react with cytotoxic ROS and thus protect against oxidative damage.


Subject(s)
Antioxidants/therapeutic use , Cerebral Infarction/therapy , Hydrogen/therapeutic use , Hydroxyl Radical/antagonists & inhibitors , Hydroxyl Radical/toxicity , Reperfusion Injury/therapy , Administration, Inhalation , Animals , Antioxidants/administration & dosage , Disease Models, Animal , Humans , Hydrogen/administration & dosage , Hydroxyl Radical/metabolism , Male , Membrane Potential, Mitochondrial/physiology , Oxidation-Reduction , PC12 Cells , Rats , Rats, Sprague-Dawley
17.
J Neurooncol ; 72(3): 231-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15937645

ABSTRACT

OBJECTIVE: Survivin has been identified as a protein expressed in cancer cells and a member of the inhibitor-of-apoptosis protein family. Recent studies suggest that the expression of survivin increases during the G2/M phase of the cell cycle, and may be used in clinical prognosis. We examined whether survivin expression in human gliomas would be a correlative of prognosis. METHODS: We prepared polyclonal anti-survivin serum to establish a survivin index for stained sections, using an immunohistochemical procedure, according to the method used for scoring MIB-1 index, and then stained 29 paraffin-embedded sections from surgical specimens of 29 patients who were classified into three grades of World Health Organization with the mean age of low grade astocytoma (grade II) being 34.7; anaplastic astrocytoma (grade III), 48.8; and glioblastoma multiform (grade IV), 58.4. RESULTS: On staining with the anti-survivin antiserum, all specimens contained positive cells, but the survivin index was heterogeneous among grades. The mean percentage of immunoreactive cells in each specimen was 70.0 (SD 18.2) in grade II, 81.3 (16.5) in grade III, and 85.0 (13.6) in grade IV. Then we compared the survivin index to the MIB-1 index and found that in low-grade gliomas (grade II and III), the difference in survival times between the high and low survivin indexes was significant (P=0.007), whereas that between the high and low MIB-1 indexes was not significant (P=0.092). ONCLUSION: Survivin is more sensitive marker than MIB-1 for the evaluation of low-grade gliomas in that it helps to predict patient survival. Much larger glioma patient series are needed to validate the findings of our limited study.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Ki-67 Antigen/analysis , Microtubule-Associated Proteins/metabolism , Adult , Aged , Aged, 80 and over , Antibodies/analysis , Biomarkers, Tumor , Blotting, Western , Brain Neoplasms/pathology , Child , DNA, Complementary/genetics , Disease Progression , Female , Glioma/pathology , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Male , Microtubule-Associated Proteins/analysis , Middle Aged , Neoplasm Proteins , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Survivin , Treatment Outcome , Tumor Cells, Cultured
18.
EMBO J ; 22(12): 2913-23, 2003 Jun 16.
Article in English | MEDLINE | ID: mdl-12805207

ABSTRACT

Dihydrolipoamide succinyltransferase (DLST) is a subunit enzyme of the alpha-ketoglutarate dehydrogenase complex of the Krebs cycle. While studying how the DLST genotype contributes to the pathogenesis of Alzheimer's disease (AD), we found a novel mRNA that is transcribed starting from intron 7 in the DLST gene. The novel mRNA level in the brain of AD patients was significantly lower than that of controls. The truncated gene product (designated MIRTD) localized to the intermembrane space of mitochondria. To investigate the function of MIRTD, we established human neuroblastoma SH-SY5Y cells expressing a maxizyme, a kind of ribozyme, that specifically digests the MIRTD mRNA. The expression of the maxizyme specifically eliminated the MIRTD protein and the resultant MIRTD-deficient cells exhibited a marked decrease in the amounts of subunits of complexes I and IV of the mitochondrial respiratory chain, resulting in a decline of activity. A pulse-label experiment revealed that the loss of the subunits is a post-translational event. Thus, the DLST gene is bifunctional and MIRTD transcribed from the gene contributes to the biogenesis of the mitochondrial respiratory complexes.


Subject(s)
Acyltransferases/genetics , Acyltransferases/metabolism , Alzheimer Disease/genetics , Electron Transport/physiology , RNA, Messenger/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Animals , Base Sequence , Brain/metabolism , Gene Expression Regulation, Enzymologic , Haplotypes , Humans , Middle Aged , Mitochondria, Liver/physiology , Molecular Sequence Data , Nucleic Acid Conformation , Oxidative Stress , PC12 Cells , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Catalytic/metabolism , RNA, Messenger/genetics , Rats , Tumor Cells, Cultured
19.
J Neurochem ; 84(5): 1110-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12603834

ABSTRACT

Mitochondrial aldehyde dehydrogenase 2 (ALDH2) plays a major role in acetaldehyde detoxification. The alcohol sensitivity is associated with a genetic deficiency of ALDH2. We have previously reported that this deficiency influences the risk for late-onset Alzheimer's disease. However, the biological effects of the deficiency on neuronal cells are poorly understood. Thus, we obtained ALDH2-deficient cell lines by introducing mouse mutant Aldh2 cDNA into PC12 cells. The mutant ALDH2 repressed mitochondrial ALDH activity in a dominant negative fashion, but not cytosolic activity. The resultant ALDH2-deficient transfectants were highly vulnerable to exogenous 4-hydroxy-2-nonenal, an aldehyde derivative generated by the reaction of superoxide with unsaturated fatty acid. In addition, the ALDH2-deficient transfectants were sensitive to oxidative insult induced by antimycin A, accompanied by an accumulation of proteins modified with 4-hydroxy-2-nonenal. Thus, these findings suggest that mitochondrial ALDH2 functions as a protector against oxidative stress.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Mitochondria/enzymology , Pheochromocytoma/enzymology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Aldehydes/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antimycin A/pharmacology , Cell Death/drug effects , Genes, Dominant , Mice , Mitochondria/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , PC12 Cells , Pheochromocytoma/drug therapy , Rats , Reactive Oxygen Species/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...