Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 636(Pt 1): 89-95, 2022 12 25.
Article in English | MEDLINE | ID: mdl-36332487

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+), a biological molecule integral to redox reactions involved in multiple cellular processes, has the potential to treat nonalcoholic fatty liver diseases (NAFLDs) and nonalcoholic steatohepatitis (NASH). Nicotinamide mononucleotide adenylyltransferase (Nmnat1), one of the NAD+ biosynthesizing enzymes, plays a central role in all NAD+ metabolic pathways and it is vital to embryonic development. However, the function of Nmnat1 in metabolic pathology and, specifically, in the development and progression of NAFLD and NASH remains unexplored. First, we generated hepatic Nmnat1 knockout (H-Nmnat1-/-) mice to investigate the physiological function of Nmnat1 and found that NAD+ levels were significantly lower in H-Nmnat1-/- mice than control mice. However, H-Nmnat1-/- mice appeared normal with comparable metabolic activity. Next, we used three different diet-induced NASH models to assess the pathophysiological role of Nmant1 in metabolic disorders and discovered that hepatic loos of Nmnat1 decreased 35%-40% of total NAD+ in an obese state. Nevertheless, our analysis of phenotypic variations found comparable body composition, gene expression, and liver histology in all NASH models in H-Nmnat1-/- mice. We also found that aged H-Nmnat1-/- mice exhibited comparable liver phenotypes with control mice. These findings suggest that Nmnat1 has a redundancy to the pathophysiology of obesity-induced hepatic disorders.


Subject(s)
Nicotinamide-Nucleotide Adenylyltransferase , Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , NAD/metabolism , Liver/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Obesity/metabolism , Diet , Mice, Inbred C57BL
2.
Biochem Biophys Res Commun ; 637: 58-65, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36375251

ABSTRACT

Malaria is an infectious disease caused by Plasmodium parasites and has high mortality rates, especially among children in African and Southeast Asian countries. Patients with hemolytic anemia are suggested to adapt protective measures against malarial infection. Nicotinamide adenine dinucleotide (NAD+) is a crucial cofactor associated with numerous biological processes that maintain homeostasis in all living organisms. In a previous study, we had demonstrated that the deficiency of nicotinamide mononucleotide adenylyltransferase 3 (Nmnat3), an enzyme catalyzing NAD+ synthesis, causes hemolytic anemia accompanied by a drastic decline in the NAD+ levels in the erythrocytes. It is well known that hemolytic anemia is linked to a reduced risk of malarial infections. In the present study, we investigated whether hemolytic anemia caused by Nmnat3 deficiency is beneficial against malarial infections. We found that Nmnat3 deficiency exacerbated malarial infection and subsequently caused death. Moreover, we demonstrated that the NAD+ levels in malaria-infected Nmnat3 red blood cells significantly increased and the glycolytic flow was largely enhanced to support the rapid growth of malarial parasites. Our results revealed that hemolytic anemia induced by the deletion of Nmnat3 was harmful rather than protective against malaria.


Subject(s)
Anemia, Hemolytic , Malaria , Nicotinamide-Nucleotide Adenylyltransferase , Child , Humans , Anemia, Hemolytic/complications , Anemia, Hemolytic/genetics , Erythrocytes/metabolism , Malaria/complications , NAD/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Animals
3.
Nat Commun ; 12(1): 6767, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799586

ABSTRACT

Nicotinamide riboside (NR) is one of the orally bioavailable NAD+ precursors and has been demonstrated to exhibit beneficial effects against aging and aging-associated diseases. However, the metabolic pathway of NR in vivo is not yet fully understood. Here, we demonstrate that orally administered NR increases NAD+ level via two different pathways. In the early phase, NR was directly absorbed and contributed to NAD+ generation through the NR salvage pathway, while in the late phase, NR was hydrolyzed to nicotinamide (NAM) by bone marrow stromal cell antigen 1 (BST1), and was further metabolized by the gut microbiota to nicotinic acid, contributing to generate NAD+ through the Preiss-Handler pathway. Furthermore, we report BST1 has a base-exchange activity against both NR and nicotinic acid riboside (NAR) to generate NAR and NR, respectively, connecting amidated and deamidated pathways. Thus, we conclude that BST1 plays a dual role as glycohydrolase and base-exchange enzyme during oral NR supplementation.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Antigens, CD/metabolism , Glycoside Hydrolases/metabolism , Niacinamide/analogs & derivatives , Pyridinium Compounds/pharmacokinetics , A549 Cells , ADP-ribosyl Cyclase/genetics , Administration, Oral , Aging/drug effects , Animals , Antigens, CD/genetics , Dietary Supplements , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gastrointestinal Microbiome , Glycoside Hydrolases/genetics , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Small/metabolism , Intestine, Small/microbiology , Mice , Mice, Knockout , Niacin/metabolism , Niacinamide/administration & dosage , Niacinamide/metabolism , Niacinamide/pharmacokinetics , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Pyridinium Compounds/administration & dosage
4.
Nutr Neurosci ; 24(5): 371-383, 2021 May.
Article in English | MEDLINE | ID: mdl-31280708

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) is an essential coenzyme that mediates various redox reactions. Particularly, mitochondrial NAD plays a critical role in energy production pathways, including the tricarboxylic acid (TCA) cycle, fatty acid oxidation, and oxidative phosphorylation. NAD also serves as a substrate for ADP-ribosylation and deacetylation by poly(ADP-ribose) polymerases (PARPs) and sirtuins, respectively. Thus, NAD regulates energy metabolism, DNA damage repair, gene expression, and stress response. Numerous studies have demonstrated the involvement of NAD metabolism in neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and retinal degenerative diseases. Mitochondrial dysfunction is considered crucial pathogenesis for neurodegenerative diseases such as AD and PD. Maintaining appropriate NAD levels is important for mitochondrial function. Indeed, decreased NAD levels are observed in AD and PD, and supplementation of NAD precursors ameliorates disease phenotypes by activating mitochondrial functions. NAD metabolism also plays an important role in axonal degeneration, a characteristic feature of peripheral neuropathy and neurodegenerative diseases. In addition, dysregulated NAD metabolism is implicated in retinal degenerative diseases such as glaucoma and Leber congenital amaurosis, and NAD metabolism is considered a therapeutic target for these diseases. In this review, we summarize the involvement of NAD metabolism in axon degeneration and various neurodegenerative diseases and discuss perspectives of nutritional intervention using NAD precursors.


Subject(s)
NAD/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy , Animals , Humans , Mitochondria/metabolism , NAD/therapeutic use
5.
Aging Cell ; 17(4): e12798, 2018 08.
Article in English | MEDLINE | ID: mdl-29901258

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) is an important cofactor that regulates various biological processes, including metabolism and gene expression. As a coenzyme, NAD controls mitochondrial respiration through enzymes of the tricarboxylic acid (TCA) cycle, ß-oxidation, and oxidative phosphorylation and also serves as a substrate for posttranslational protein modifications, such as deacetylation and ADP-ribosylation by sirtuins and poly(ADP-ribose) polymerase (PARP), respectively. Many studies have demonstrated that NAD levels decrease with aging and that these declines cause various aging-associated diseases. In contrast, activation of NAD metabolism prevents declines in NAD levels during aging. In particular, dietary supplementation with NAD precursors has been associated with protection against age-associated insulin resistance. However, it remains unclear which NAD synthesis pathway is important and/or efficient at increasing NAD levels in vivo. In this study, Nmnat3 overexpression in mice efficiently increased NAD levels in various tissues and prevented aging-related declines in NAD levels. We also demonstrated that Nmnat3-overexpressing (Nmnat3 Tg) mice were protected against diet-induced and aging-associated insulin resistance. Moreover, in skeletal muscles of Nmnat3 Tg mice, TCA cycle activity was significantly enhanced, and the energy source for oxidative phosphorylation was shifted toward fatty acid oxidation. Furthermore, reactive oxygen species (ROS) generation was significantly suppressed in aged Nmnat3 Tg mice. Interestingly, we also found that concentrations of the NAD analog nicotinamide guanine dinucleotide (NGD) were dramatically increased in Nmnat3 Tg mice. These results suggest that Nmnat3 overexpression improves metabolic health and that Nmnat3 is an attractive therapeutic target for metabolic disorders that are caused by aging.


Subject(s)
Cellular Senescence , Guanine Nucleotides/metabolism , Insulin Resistance , NAD/analogs & derivatives , NAD/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/biosynthesis , Animals , Calorimetry , Guanine Nucleotides/analysis , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , NAD/analysis , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Reactive Oxygen Species/metabolism
6.
Front Oncol ; 8: 622, 2018.
Article in English | MEDLINE | ID: mdl-30631755

ABSTRACT

Cancer cells have a unique energy metabolism for sustaining rapid proliferation. The preference for anaerobic glycolysis under normal oxygen conditions is a unique trait of cancer metabolism and is designated as the Warburg effect. Enhanced glycolysis also supports the generation of nucleotides, amino acids, lipids, and folic acid as the building blocks for cancer cell division. Nicotinamide adenine dinucleotide (NAD) is a co-enzyme that mediates redox reactions in a number of metabolic pathways, including glycolysis. Increased NAD levels enhance glycolysis and fuel cancer cells. In fact, nicotinamide phosphoribosyltransferase (Nampt), a rate-limiting enzyme for NAD synthesis in mammalian cells, is frequently amplified in several cancer cells. In addition, Nampt-specific inhibitors significantly deplete NAD levels and subsequently suppress cancer cell proliferation through inhibition of energy production pathways, such as glycolysis, tricarboxylic acid (TCA) cycle, and oxidative phosphorylation. NAD also serves as a substrate for poly(ADP-ribose) polymerase (PARP), sirtuin, and NAD gylycohydrolase (CD38 and CD157); thus, NAD regulates DNA repair, gene expression, and stress response through these enzymes. Thus, NAD metabolism is implicated in cancer pathogenesis beyond energy metabolism and considered a promising therapeutic target for cancer treatment. In this review, we present recent findings with respect to NAD metabolism and cancer pathogenesis. We also discuss the current and future perspectives regarding the therapeutics that target NAD metabolic pathways.

7.
PLoS One ; 11(1): e0147037, 2016.
Article in English | MEDLINE | ID: mdl-26756334

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) is an essential co-enzyme mediating various enzymatic reactions. Mitochondrial NAD particularly occupies a considerable amount of total NAD in cells, and serves as a co-enzyme in tricarboxylic acid cycle (TCA cycle), ß-oxidation, and oxidative phosphorylation. Despite the importance of mitochondrial NAD, its synthesis pathway remains unknown. It has been proposed that NAD synthesis enzyme, Nmnat3, was localized in mitochondria, but its physiological relevance to the metabolism in mitochondria was not fully elucidated. Previously, we have reported that murine Nmnat3 protein was strongly expressed in the cytoplasm of mature erythrocytes, in which mitochondria were absent, and Nmnat3-deficient mice (Nmnat3-KO mice) exhibited splenomegaly and hemolytic anemia due to reduced NAD levels in mature erythrocytes. These results challenged the role of Nmnat3 in mitochondrial NAD synthesis. In this study, we demonstrated that mitochondrial NAD levels in various tissues, except for red blood cells, were unchanged in Nmnat3-KO mice. We also analyzed the metabolites in glycolysis and TCA cycle and found that there were no differences between Nmnat3-KO and WT mice. In addition, the aged Nmnat3-KO mice had comparable NAD levels to that observed in WT mice. Our results indicated that Nmnat3 is dispensable in the maintenance of mitochondrial NAD levels, and that other NAD regulatory pathways may exist in mitochondria.


Subject(s)
Mitochondria/metabolism , NAD/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Animals , Mice , Mice, Knockout , Nicotinamide-Nucleotide Adenylyltransferase/genetics
8.
Biomed Res ; 36(3): 159-67, 2015.
Article in English | MEDLINE | ID: mdl-26106045

ABSTRACT

Patients chronically infected with hepatitis C virus (HCV) are at risk of developing end-stage liver disease and hepatocellular carcinoma. Development of drugs to inhibit hepatocyte damage and a vaccine against HCV is hampered by the lack of a small animal model. We generated mice in which the viral genome RNA was always present in the hepatocytes using a special transgene. Here we show that the HCV genome RNA transcribed by Pol I polymerase can replicate and produce infectious viruses in mice. We obtained a transgenic mouse with 200 copies per haploid which we named the A line mouse. It produced ~ 3 × 10(6) HCV RNA copies/mL serum, which is at the comparable level as patients with chronic HCV infection. This mouse was immunotolerant to HCV and showed hepatic steatosis without any necroinflammation at the age of 6 months or hepatocellular carcinoma at the age of 15 months. Thus, the A line mouse can be used as an animal model for chronic HCV infection. This will enable better study of the abnormalities in metabolism and signal transduction in infected hepatocytes, and development of drugs that cure abnormalities.


Subject(s)
Fatty Liver/etiology , Genome, Viral , Hepacivirus/physiology , Hepatitis C/complications , Hepatitis C/virology , RNA, Viral , Virus Replication , Animals , Disease Models, Animal , Fatty Liver/pathology , Humans , Mice , Mice, Transgenic , RNA Polymerase I/metabolism , Time Factors , Transcription, Genetic , Viral Load
9.
J Biol Chem ; 289(21): 14796-811, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24739386

ABSTRACT

NAD biosynthesis is of substantial interest because of its important roles in regulating various biological processes. Nicotinamide mononucleotide adenylyltransferase 3 (Nmnat3) is considered a mitochondria-localized NAD synthesis enzyme involved in de novo and salvage pathways. Although the biochemical properties of Nmnat3 are well documented, its physiological function in vivo remains unclear. In this study, we demonstrated that Nmnat3 was localized in the cytoplasm of mature erythrocytes and critically regulated their NAD pool. Deficiency of Nmnat3 in mice caused splenomegaly and hemolytic anemia, which was associated with the findings that Nmnat3-deficient erythrocytes had markedly lower ATP levels and shortened lifespans. However, the NAD level in other tissues were not apparently affected by the deficiency of Nmnat3. LC-MS/MS-based metabolomics revealed that the glycolysis pathway in Nmnat3-deficient erythrocytes was blocked at a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) step because of the shortage of the coenzyme NAD. Stable isotope tracer analysis further demonstrated that deficiency of Nmnat3 resulted in glycolysis stall and a shift to the pentose phosphate pathway. Our findings indicate the critical roles of Nmnat3 in maintenance of the NAD pool in mature erythrocytes and the physiological impacts at its absence in mice.


Subject(s)
Anemia, Hemolytic/metabolism , Erythrocytes/metabolism , Glycolysis , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Adenosine Triphosphate/metabolism , Anemia, Hemolytic/genetics , Animals , Blotting, Western , Chromatography, Liquid , Cytoplasm/enzymology , Erythrocytes/ultrastructure , Metabolic Networks and Pathways/genetics , Metabolomics/methods , Mice , Mice, Knockout , Microscopy, Electron, Scanning , NAD/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Splenomegaly/genetics , Splenomegaly/metabolism , Survival Analysis , Tandem Mass Spectrometry
10.
Biochem Biophys Res Commun ; 417(1): 601-6, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22182413

ABSTRACT

The retinoblastoma (Rb) tumor suppressor encodes a nuclear phosphoprotein that regulates cellular proliferation, apoptosis and differentiation. In order to adapt itself to these biological functions, Rb is subjected to modification cycle, phosphorylation and dephosphorylation. To directly determine the effect of phosphorylation-resistant Rb on liver development and function, we generated transgenic mice expressing phosphorylation-resistant human mutant Rb (mt-Rb) under the control of the rat hepatocyte nuclear factor-1 gene promoter/enhancer. Expression of mt-Rb in the liver resulted in macroscopic neoplastic nodules (adenomas) with ∼50% incidence within 15 months old. Interestingly, quantitative reverse transcriptase-PCR analysis showed that c-Myc was up-regulated in the liver of mt-Rb transgenic mice irrespective of having tumor tissues or no tumor. In tumor tissues, several c-Myc target genes, Foxm1, c-Jun, c-Fos, Bmi1 and Skp2, were also up-regulated dramatically. We determined whether mt-Rb activated the Myc promoter in the HTP9 cells and demonstrated that mt-Rb acted as an inhibitor of wild-type Rb-induced repression on the Myc promoter. Our results suggest that continued upregulation of c-Myc target genes promotes the liver tumor formation after about 1 year of age.


Subject(s)
Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Retinoblastoma Protein/metabolism , Animals , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Humans , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/genetics , Rats , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Up-Regulation
11.
Biomed Res ; 32(2): 143-50, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21551950

ABSTRACT

No suitable mouse model is available for studying chronic liver disease caused by hepatitis C virus (HCV). CD81, claudin-1, scavenger receptor class B type I, and occludin were recently reported to be the important factors in HCV entry into hepatocytes. We made transgenic mice (Alb-CCSO) expressing the four human proteins and examined whether HCV from a patient serum or HCV pseudoparticles (HCVpp) were capable of infecting them. HCV was not detected in the mouse serum after injecting the mice with HCV from a patient serum. We also found no indications of HCVpp entry into primary hepatocytes from Alb-CCSO mice. In addition, HCV-infectible Hep3B cells were fused with HCV-resistant primary mouse hepatocytes and the fused cells showed 35-fold lower infectivity compared to wild-type Hep3B cells, indicating that primary mouse hepatocytes have the inhibitory factor(s) in HCVpp entry. Our results suggest that the expression of the human factors does not confer susceptibility to HCV entry into the liver.


Subject(s)
Hepacivirus/physiology , Hepatitis C/genetics , Hepatocytes/virology , Liver/virology , Receptors, Virus/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Fusion , Cells, Cultured , Claudin-1 , Female , Gene Expression , Hepatitis C/metabolism , Hepatitis C/transmission , Hepatitis C/virology , Hepatocytes/metabolism , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/metabolism , Liver/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Occludin , Receptors, Virus/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Tetraspanin 28 , Virion/genetics , Virion/metabolism , Virus Internalization
12.
Genesis ; 49(1): 2-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21254332

ABSTRACT

Irxl1 (Iroquois-related homeobox like-1) is a newly identified three amino-acid loop extension (TALE) homeobox gene, which is expressed in various mesoderm-derived tissues, particularly in the progenitors of the musculoskeletal system. To analyze the roles of Irxl1 during embryonic development, we generated mice carrying a null allele of Irxl1. Mice homozygous for the targeted allele were viable, fertile, and showed reduced tendon differentiation. Skeletal morphology and skeletal muscle weight in Irxl1-knockout mice appeared normal. Expression patterns of several marker genes for cartilage, tendon, and muscle progenitors in homozygous mutant embryos were unchanged. These results suggest that Irxl1 is required for the tendon differentiation but dispensable for the patterning of the musculoskeletal system in development.


Subject(s)
Homeodomain Proteins/genetics , Tendons/embryology , Animals , Body Patterning/genetics , Mice , Mice, Knockout , Musculoskeletal Development/genetics , Musculoskeletal System/anatomy & histology , Musculoskeletal System/embryology , Recombination, Genetic , Repressor Proteins/genetics
13.
Life Sci ; 87(1-2): 17-25, 2010 Jul 03.
Article in English | MEDLINE | ID: mdl-20553735

ABSTRACT

AIMS: Foxp2 is expressed in the lung during mouse development. A monoclonal anti-mouse Foxp2 antibody was created to determine the expression pattern in the developing lung. Next, transcriptional control of two lung genes, CC10 and surfactant protein C (SPC) genes, by Foxp2 was investigated in H441 and A549 cells. Thirdly, expression patterns of Foxp2 and Foxf2 were compared in the developing lung. Finally, Foxp2 expression was determined in the Foxf2-null mice. MAIN METHODS: Immunohistochemical staining and in situ hybridization were applied to the sections of lungs in the developing embryos. KEY FINDINGS: Monoclonal anti-Foxp2 antibody demonstrated that Foxp2 was expressed in the bronchial epithelium at E10.5 and its expression became restricted to the distal portion of the elongating bronchiolar epithelium and finally to type II alveolar epithelial cells around birth and in the adult. Foxp2 activated the SPC gene promoter in the presence of Nkx2.1 in A549 cells while it repressed the CC10 gene promoter in H441 cells. Next, the expression domains of the Foxp2 and Foxf2 were found to be exclusive in the lung. Finally, the expression of Foxp2 did not change in the lung of Foxf2-null mice. SIGNIFICANCE: The Foxp2 protein is expressed in the growing distal edge of airway epithelium. When the bronchiolus elongates, Foxp2 suppresses CC10 expression. When the lung alveolus is formed, Foxp2 modulates the Nkx2.1-mediated SPC expression in type II alveolar cells. Foxp2 and Foxf2 independently play distinct roles in the alveoli and the mesenchyme, respectively.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Lung/metabolism , Repressor Proteins/genetics , Uteroglobin/genetics , Animals , Antibodies, Monoclonal , Bronchi/embryology , Bronchi/metabolism , Epithelium/metabolism , Forkhead Transcription Factors/metabolism , Lung/embryology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Knockout , Mice, Nude , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein C/genetics , Repressor Proteins/metabolism , Uteroglobin/metabolism
14.
Genesis ; 48(5): 295-302, 2010 May.
Article in English | MEDLINE | ID: mdl-20162674

ABSTRACT

To study the expression/function of Tbx10, a T-box gene, Tbx10(LacZ/+) mice were established by replacing the T-box coding region with a LacZ gene. X-gal staining showed that LacZ(+) cells were localized to two-cell populations in rhombomere 4 and rhombomere 6. No significant differences in the locations of LacZ(+) cells were found between Tbx10(LacZ/+) and Tbx10(LacZ/LacZ) mice, and the Tbx10(LacZ/LacZ) mice were viable and fertile. We found that the LacZ(+) cells are present in both embryonic and adult mice. Histological studies suggest that the rhombomere 4-derived LacZ(+) cells are a subpopulation of the ventral interneurons in the pons.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Rhombencephalon/metabolism , T-Box Domain Proteins/genetics , Alleles , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Immunohistochemistry , In Situ Hybridization , Interneurons/cytology , Interneurons/metabolism , Lac Operon/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Mutation , Pons/cytology , Pons/embryology , Pons/metabolism , Rhombencephalon/cytology , Rhombencephalon/embryology , T-Box Domain Proteins/metabolism , beta-Galactosidase/metabolism
15.
Colloids Surf B Biointerfaces ; 66(2): 195-200, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18653320

ABSTRACT

This study was designed to examine if platinum nanoparticles have an activity similar to mitochondrial complex I, NADH:ubiquinone oxidoreductase. Platinum nanoparticles were prepared by a citrate reduction of H(2)PtCl(6) and protected by citrate itself and pectin (CP-Pt). Time- and dose-dependent decreases in NADH and a time-dependent increase in NAD(+) were observed in the presence of 50 microM CP-Pt; these observations were made using a spectrophotometric method in which the maximum absorption spectra at 340 and 260 nm were used for NADH and NAD(+), respectively. The required platinum concentration in CP-Pt to achieve a 50% oxidation of NADH for 3h was approximately 20 microM, and this NADH oxidation did not require oxygen as an electron acceptor. We also verified NAD(+) formation using an NAD(+)/NADH quantification kit. The absorption peak shift from 278 to 284 nm of 2,3-dimethoxy-5-methyl-6-(3-methyl-2-butenyl)-1,4-benzoquinone (CoQ(1)) was observed by incubating CoQ(1) with CP-Pt in an aqueous buffer. A further analysis with HPLC revealed the reduction of CoQ(1) to CoQ(1)H(2) by CP-Pt. As a whole, platinum nanoparticles have an NADH:ubiquinone oxidoreductase-like activity. This suggests that platinum nanoparticles are a potential medicinal substance for oxidative stress diseases with suppressed mitochondrial complex I.


Subject(s)
Electron Transport Complex I/chemistry , Metal Nanoparticles/chemistry , Mitochondria, Heart/enzymology , Platinum/chemistry , Ubiquinone/chemistry , Citric Acid/chemistry , Oxidation-Reduction , Pectins/chemistry , Surface Properties
16.
J Biochem ; 143(3): 407-16, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18079162

ABSTRACT

Forkhead box (Fox) genes are involved in organogenesis and cell differentiation. A mutation of FOXP2 was discovered in patients with severe defects in speech and language. The medaka FoxP2 was cloned in order to clarify the molecular evolution and difference in the protein structure and function by comparing human/mouse and medaka genes. The result showed that medaka FoxP2 had a 73.7% homology to the human and mouse counterparts, and its zinc finger, leucine zipper and forkhead domain structures were conserved. However, medaka FoxP2 lacked a long polyglutamine repeat and had two insertions of unique amino acid sequences. FoxP2 expression was found in the epiphysis and retina, in addition to the midbrain and cerebellum. The transcriptional assay revealed that medaka FoxP2 showed a very weak repressive activity to the CC10 promoter while mouse Foxp2 exhibited a strong repressive activity. Mutational analyses of medaka FoxP2 showed that the three amino acids of forkhead domain were responsible for the weak repressive activity. These results suggest that medaka FoxP2 may play a different function in the development of the medaka fish.


Subject(s)
Evolution, Molecular , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Language , Oryzias/genetics , Sequence Homology, Amino Acid , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Cell Nucleus/metabolism , Cloning, Molecular , Embryo, Nonmammalian/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Humans , Molecular Sequence Data , Oryzias/embryology , Protein Transport , Speech
17.
Free Radic Res ; 41(6): 615-26, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17516233

ABSTRACT

Bimetallic nanoparticles consisting of gold and platinum were prepared by a citrate reduction method and complementarily stabilized with pectin (CP-Au/Pt). The percent mole ratio of platinum was varied from 0 to 100%. The CP-Au/Pt were alloy-structured. They were well dispersed in water. The average diameter of platinum nanoparticles (CP-Pt) was 4.7 +/- 1.5 nm. Hydrogen peroxide (H(2)O(2)) was quenched by CP-Au/Pt consisting of more than 50% platinum whereas superoxide anion radical (O(2)(-)) was quenched by any CP-Au/Pt. The CP-Au/Pt quenched these two reactive oxygen species in dose-dependent manners. The CP-Pt is the strongest quencher. The CP-Pt decomposed H(2)O(2) and consequently generated O(2) like catalase. The CP-Pt actually quenched O(2)(-) which was verified by a superoxide dismutase (SOD) assay kit. This quenching activity against O(2)(-) persisted like SOD. Taken together, CP-Pt may be a SOD/catalase mimetic which is useful for medical treatment of oxidative stress diseases.


Subject(s)
Free Radical Scavengers/pharmacology , Gold/chemistry , Hydrogen Peroxide/chemistry , Nanoparticles , Oxidants/chemistry , Platinum/chemistry , Superoxides/chemistry , Antioxidants/metabolism , Catalase/metabolism , Free Radical Scavengers/chemistry , Hydrogen Peroxide/metabolism , Nanotechnology , Oxidants/metabolism , Oxidative Stress , Oxygen/metabolism , Platinum/pharmacology , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Superoxides/metabolism , Uric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...