Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neurochem Int ; 150: 105191, 2021 11.
Article in English | MEDLINE | ID: mdl-34547325

ABSTRACT

Spinal cord ischemia-reperfusion injury (SCIRI) can cause dramatic neuron loss and lead to paraplegia in patients. In this research, the role of mGluR5, a member of the metabotropic glutamate receptors (mGluRs) family, was investigated both in vitro and in vivo to explore a possible method to treat this complication. In vitro experiment, after activating mGluR5 via pretreating cells with (RS)-2-Chloro-5-hydroxyphenylglycine (CHPG) and 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB), excitotoxicity induced by glutamate (Glu) was attenuated in primary spinal cord neurons, evidenced by higher neuron viability, decreased lactate dehydrogenase (LDH) release and less detected TUNEL-positive cells. According to Western Blot (WB) results, Glu treatment resulted in a high level of large-conductance Ca2+- and voltage-activated K+ (BK) channels, with activation relying on the mGluR5-IP3R (inositol triphosphate) pathway. In vivo part, a rat model of SCIRI was built to further investigate the role of mGluR5. After pretreating them with CHPG and CDPPB, the rats showed markedly lower spinal water content, attenuated motor neuron injury in the spinal cord of L4 segments, and better neurological function. This effect could be partially reversed by paxilline, a blocker of BK channels. In addition, activating BK channels alone using specific openers: NS1619 or NS11021 can protect spinal cord neurons from injury induced by either SCIRI or Glu. In conclusion, in this research, we proved that mGluR5 exerts a protective role in SCIRI, and this effect partially works via IP3R-mediated activation of BK channels.


Subject(s)
Adenosylhomocysteinase/biosynthesis , Large-Conductance Calcium-Activated Potassium Channels/biosynthesis , Neuroprotection/physiology , Receptor, Metabotropic Glutamate 5/biosynthesis , Reperfusion Injury/metabolism , Spinal Cord Ischemia/metabolism , Animals , Benzamides/pharmacology , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Neuroprotection/drug effects , Paxillin/pharmacology , Pyrazoles/pharmacology , Rats , Receptor, Metabotropic Glutamate 5/agonists , Reperfusion Injury/prevention & control , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord Ischemia/prevention & control
2.
Neuroscience ; 406: 38-49, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30849448

ABSTRACT

Enhancing the migration and phagocytosis of microglial cells is of great significance for the reducing of the risk of the neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD). The effect of mouse selenoprotein K (mSELENOK) on the migration and phagocytosis of BV2 microglial cells and its mechanism were studied. The results showed that the over-expression of mSELENOK can increase the migratory and phagocytic abilities of the microglial cells, while the knockdown of mSELENOK can decrease the migratory and phagocytic abilities of the cells. The cytosolic free Ca2+ level and inositol trisphosphate receptor (IP3R) mRNA transcript and protein expression were also increased significantly as the consequence of the over-expression of mSELENOK in the microglial cells. On the contrary, the level of cytosolic free Ca2+ and the mRNA transcript and protein expression of IP3R in mSELENOK knockdown cells were decreased significantly. 2-aminoethoxydiphenyl borate (2-APB), an antagonist of IP3R, could prevent the increased migration, phagocytosis, and cytosolic free Ca2+ level of mSELENOK over-expressed microglial cells, and knockdown of IP3R3 could reduce the increased cytosolic Ca2+ level in mSELENOK over-expressed microglial cells. Further studies revealed that selenium supplement (Na2SeO3) can increase the expression of mSELENOK in microglial cells significantly. In summary, these data suggest that mSELENOK can increase cytosolic free Ca2+ level of microglial cells by up-regulating the expression of IP3R, thus enhancing the migration and phagocytosis of microglial cells. Our results indicated that mSELENOK is an important selenoprotein, which plays a role in trace element selenium's functions and can enhance the migration and phagocytosis of microglial cells.


Subject(s)
Adenosylhomocysteinase/biosynthesis , Cell Movement/physiology , Cytosol/metabolism , Microglia/metabolism , Phagocytosis/physiology , Selenoproteins/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Survival/physiology , Mice , Up-Regulation/physiology
3.
BMC Nephrol ; 19(1): 140, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29907098

ABSTRACT

BACKGROUND: The mechanism of podocyte apoptosis is not fully understood. In addition, the role of the inositol 1,4,5-triphosphate receptor (IP3R)/glucose-regulated protein 75 (Grp75)/voltage-dependent anion channel 1 (VDAC1)/mitochondrial calcium uniporter (MCU) calcium regulation axis, which is located at sites of endoplasmic reticulum (ER) mitochondria coupling, in the mechanism of podocyte apoptosis is unclear. This study aimed to understand the roles of this axis in podocyte apoptosis and explore potential targets for podocyte protection. METHODS: The expression of IP3R, Grp75, VDAC1, and MCU and mitochondrial Ca2+ were analyzed during Adriamycin- or angiotensin II-induced apoptosis in cultured mouse podocytes. The interaction between IP3R, Grp75, and VDAC1 was investigated using co-immunoprecipitation experiments. The effects of IP3R, Grp75, and MCU agonists and antagonists on mitochondrial Ca2+ and apoptosis were investigated in cultured podocytes. The podocyte-protective effects of an MCU inhibitor were further investigated in rats with Adriamycin-induced nephropathy. RESULTS: Increased expression of IP3R, Grp75, VDAC1 and MCU, enhanced interaction among the IP3R-Grp75-VDAC1 complex, mitochondrial Ca2+ overload, and increased active caspase-3 levels were confirmed during Adriamycin- or angiotensin II-induced mouse podocyte apoptosis. Agonists of this axis facilitated mitochondrial Ca2+ overload and podocyte apoptosis, whereas specific antagonists against IP3R, Grp75, or MCU prevented mitochondrial Ca2+ overload and podocyte apoptosis. A specific MCU inhibitor prevented Adriamycin-induced proteinuria and podocyte foot process effacement in rats. CONCLUSIONS: This study identified a novel pathway in which the IP3R-Grp75-VDAC1-MCU calcium regulation axis mediated podocyte apoptosis by facilitating mitochondrial Ca2+ overload. Antagonists that inhibit Ca2+ transfer from ER to mitochondria protected mouse podocytes from apoptosis. An MCU inhibitor protected podocytes and decreased proteinuria in rats with Adriamycin-induced nephropathy. Therefore, antagonists to this pathway have promise as novel podocyte-protective drugs.


Subject(s)
Calcium/physiology , Doxorubicin/toxicity , Kidney Diseases/metabolism , Macrocyclic Compounds/pharmacology , Oxazoles/pharmacology , Podocytes/metabolism , Proteinuria/metabolism , Adenosylhomocysteinase/antagonists & inhibitors , Adenosylhomocysteinase/biosynthesis , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Apoptosis/physiology , Calcium Channels/biosynthesis , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/biosynthesis , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Macrocyclic Compounds/therapeutic use , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxazoles/therapeutic use , Podocytes/drug effects , Proteinuria/drug therapy , Rats , Rats, Sprague-Dawley , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/biosynthesis
4.
FASEB J ; 32(3): 1591-1601, 2018 03.
Article in English | MEDLINE | ID: mdl-29127188

ABSTRACT

The substantial rise in the prevalence of nonalcoholic steatohepatitis (NASH), an advanced form of nonalcoholic fatty liver disease, and the strong association between NASH and the development of hepatocellular carcinoma indicate the urgent need for a better understanding of the underlying mechanisms. In the present study, by using the Stelic animal model of NASH and NASH-derived liver carcinogenesis, we investigated the role of the folate-dependent 1-carbon metabolism in the pathogenesis of NASH. We demonstrated that advanced NASH and NASH-related liver carcinogenesis are characterized by a significant dysregulation of 1-carbon homeostasis, with diminished expression of key 1-carbon metabolism genes, especially a marked inhibition of the S-adenosylhomocysteine hydrolase ( Ahcy) gene and an increased level of S-adenosyl-l-homocysteine (SAH). The reduction in Ahcy expression was associated with gene-specific cytosine DNA hypermethylation and enrichment of the gene promoter by trimethylated histone H3 lysine 27 and deacetylated histone H4 lysine 16, 2 main transcription-inhibiting markers. These results indicate that epigenetically mediated inhibition of Ahcy expression may be a driving force in causing SAH elevation and subsequent downstream disturbances in transsulfuration and transmethylation pathways during the development and progression of NASH.-Pogribny, I. P., Dreval, K., Kindrat, I., Melnyk, S., Jimenez, L., de Conti, A., Tryndyak, V., Pogribna, M., Ortega, J. F., James, S. J., Rusyn, I., Beland, F. A. Epigenetically mediated inhibition of S-adenosylhomocysteine hydrolase and the associated dysregulation of 1-carbon metabolism in nonalcoholic steatohepatitis and hepatocellular carcinoma.


Subject(s)
Adenosylhomocysteinase/biosynthesis , Carcinoma, Hepatocellular/enzymology , Epigenesis, Genetic , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Liver Neoplasms/enzymology , Neoplasm Proteins/biosynthesis , Non-alcoholic Fatty Liver Disease/enzymology , Adenosylhomocysteinase/genetics , Animals , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Male , Mice , Neoplasm Proteins/genetics , Non-alcoholic Fatty Liver Disease/pathology , S-Adenosylhomocysteine/metabolism
5.
Pestic Biochem Physiol ; 123: 64-73, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26267054

ABSTRACT

RNA interference (RNAi) is a promising approach to control Leptinotarsa decemlineata. In this study, RNAi efficiency by double-stranded RNA (dsRNA) targeting S-adenosyl-L-homocysteine hydrolase (LdSAHase) was compared among L. decemlineata first- to fourth-instar larvae. Ingesting dsLdSAHase successfully decreased the target gene expression, caused lethality, inhibited growth and impaired pupation in an instar- and concentration-dependent manner. To study the role of Dicer2 and Argonaute2 genes in RNAi efficiency, we identified LdDcr2a, LdDcr2b, LdAgo2a and LdAgo2b. Their expression levels were higher in young larvae than those in old ones. Exposure to dsegfp for 6 h significantly elevated LdDcr2a, LdDcr2b, LdAgo2a and LdAgo2b mRNA levels in the first-, second-, third- and fourth-instar larvae. When the exposure periods were extended, however, the expression levels were gradually reduced. Continuous exposure for 72 h significantly repressed the expression of LdAgo2a and LdAgo2b in the first, second and third larval instars, and the four genes in final instars. Moreover, we found that dsLdSAHase-caused LdSAHase suppressions and larval mortalities were influenced by previous dsegfp exposure: 12 h of previous exposure increased LdSAHase silencing and mortality of the final instar larvae, whereas 72 h of exposure reduced LdSAHase silencing and mortality. Thus, it seems the activities of core RNAi-machinery proteins affect RNAi efficiency in L. decemlineata.


Subject(s)
Coleoptera/metabolism , RNA Interference , Adenosylhomocysteinase/biosynthesis , Animals , Argonaute Proteins/biosynthesis , Coleoptera/genetics , Insect Proteins/biosynthesis , Larva , Ribonuclease III/biosynthesis
6.
FASEB J ; 28(8): 3411-22, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24739303

ABSTRACT

Hyperhomocysteinemia (HHcy) is prevalent in patients with hypertension and is an independent risk factor for aortic pathologies. HHcy is known to cause an imbalance between matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs), leading to the accumulation of collagen in the aorta and resulting in stiffness and development of hypertension. Although the exact mechanism of extracellular matrix (ECM) remodeling is unclear, emerging evidence implicates epigenetic regulation involving DNA methylation. Our purpose was to investigate whether 5-aza-2'-deoxycytidine (Aza), a DNA methyltransferase (DNMT1) inhibitor, reduces high blood pressure (BP) by regulating aortic ECM remodeling in HHcy. Wild-type and cystathionine ß-synthase (CBS)(+/-) HHcy mice were treated with Aza (0.5 mg/kg body weight). In HHcy mice, Aza treatment normalized the plasma homocysteine (Hcy) level and BP. Thoracic and abdominal aorta ultrasound revealed a reduction in the resistive index and wall-to-lumen ratio. Vascular response to phenylephrine, acetylcholine, and sodium nitroprusside improved after Aza in HHcy mice. Histology showed a marked reduction in collagen deposition in the aorta. Aza treatment decreased the expression of DNMT1, MMP9, TIMP1, and S-adenosyl homocysteine hydrolase (SAHH) and upregulated methylene tetrahydrofolate reductase (MTHFR). We conclude that reduction of DNA methylation by Aza in HHcy reduces adverse aortic remodeling to mitigate hypertension.


Subject(s)
Aorta/physiopathology , Azacitidine/analogs & derivatives , DNA Methylation , Epigenesis, Genetic/physiology , Hyperhomocysteinemia/genetics , Hypertension/prevention & control , Vascular Resistance/drug effects , Acetylcholine/pharmacology , Adenosylhomocysteinase/biosynthesis , Adenosylhomocysteinase/genetics , Animals , Aorta/chemistry , Aorta/diagnostic imaging , Aorta/drug effects , Azacitidine/pharmacology , Collagen/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/drug effects , Decitabine , Endothelium, Vascular/physiopathology , Epigenesis, Genetic/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Homocystinuria/complications , Homocystinuria/drug therapy , Homocystinuria/genetics , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/physiopathology , Hypertension/etiology , Hypertension/genetics , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/biosynthesis , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics , Ultrasonography , Vascular Resistance/genetics
7.
J Biol Chem ; 287(43): 36455-64, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22932905

ABSTRACT

Cellular methylation processes enable expression of gluconeogenic enzymes and metabolism of the nutrient selenium. Selenium status has been proposed to relate to type II diabetes risk, and plasma levels of selenoprotein P (SEPP1) have been positively correlated with insulin resistance. Increased expression of gluconeogenic enzymes glucose-6-phosphatase (G6PC) and phosphoenolpyruvate carboxykinase 1 (PCK1) has negative consequences for blood glucose management in type II diabetics. Transcriptional regulation of SEPP1 is directed by the same transcription factors that control the expression of G6PC and PCK1, and these factors are activated by methylation of arginine residues. We sought to determine whether expression of SEPP1 and the aforementioned glucoconeogenic enzymes are regulated by protein methylation, the levels of which are reliant upon adequate S-adenosylmethionine (SAM) and inhibited by S-adenosylhomocysteine (SAH). We treated a human hepatocyte cell line, HepG2, with inhibitors of adenosylhomocysteine hydrolase (AHCY) known to increase concentration of SAH before analysis of G6PC, PCK1, and SEPP1 expression. Increasing SAH decreased 1) the SAM/SAH ratio, 2) protein-arginine methylation, and 3) expression of SEPP1, G6PC, and PCK1 transcripts. Furthermore, hormone-dependent induction of gluconeogenic enzymes was reduced by inhibition of protein methylation. When protein-arginine methyltransferase 1 expression was reduced by siRNA treatment, G6PC expression was inhibited. These findings demonstrate that hepatocellular SAM-dependent protein methylation is required for both SEPP1 and gluconeogenic enzyme expression and that inhibition of protein arginine methylation might provide a route to therapeutic interventions in type II diabetes.


Subject(s)
Gene Expression Regulation , Gluconeogenesis , S-Adenosylmethionine/metabolism , Selenoprotein P/biosynthesis , Adenosylhomocysteinase/biosynthesis , Adenosylhomocysteinase/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Glucose-6-Phosphate/genetics , Glucose-6-Phosphate/metabolism , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Methylation , Phosphoenolpyruvate Carboxykinase (GTP)/biosynthesis , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Protein-Arginine N-Methyltransferases/biosynthesis , Protein-Arginine N-Methyltransferases/genetics , S-Adenosylmethionine/genetics , Selenoprotein P/genetics
8.
Hum Mutat ; 30(4): E555-65, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19177456

ABSTRACT

This paper reports studies of two novel, allelic missense mutations found in the S-adenosylhomocysteine hydrolase (AHCY) gene from a new case of AHCY deficiency in an infant girl who died at age four months. The mutations lead to replacement of arginine with cysteine (p.Arg49Cys) and aspartic acid with glycine (p.Asp86Gly). Functional analysis of recombinant proteins containing the mutations detected showed that both dramatically reduce AHCY activity. The p.Arg49Cys mutant protein forms intermolecular disulphide bonds, leading to macromolecular structures that can be prevented by reducing agent DTT. The p.Asp86Gly protein tends to form enzymatically inactive aggregates and the loss of a single negative charge as a result of the mutation is involved in enzyme inactivation. We show that replacing Gly86 with negatively charged Glu86 in mutant protein restores enzymatic activity to 70% of wild-type, whereas changing Gly86 to positively charged Lys86 or uncharged Leu86 does not improve enzyme activity, indicating that the negative charge is important for maintenance of such activity. These studies significantly extend knowledge about the importance of residue 86 for AHCY activity. Residue 86 has not been implicated before in this way and the results suggest that the present model of S- adenosylhomocysteine (AdoHcy) hydrolysis may need refinement. Our functional studies provide novel insight into the molecular defect underlying AHCY deficiency and reveal that both low enzyme activity and protein stability of AHCY contribute to the clinical phenotype.


Subject(s)
Adenosylhomocysteinase/deficiency , Adenosylhomocysteinase/genetics , Mutation , Adenosylhomocysteinase/biosynthesis , DNA Mutational Analysis , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Fatal Outcome , Female , Humans , Infant , Kinetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
9.
J Biol Chem ; 283(35): 23989-99, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18591246

ABSTRACT

In eukaryotes, S-adenosyl-L-homocysteine hydrolase (Sah1) offers a single way for degradation of S-adenosyl-L-homocysteine, a product and potent competitive inhibitor of S-adenosyl-L-methionine (AdoMet)-dependent methyltransferases. De novo phosphatidylcholine (PC) synthesis requires three AdoMet-dependent methylation steps. Here we show that down-regulation of SAH1 expression in yeast leads to accumulation of S-adenosyl-L-homocysteine and decreased de novo PC synthesis in vivo. This decrease is accompanied by an increase in triacylglycerol (TG) levels, demonstrating that Sah1-regulated methylation has a major impact on cellular lipid homeostasis. TG accumulation is also observed in cho2 and opi3 mutants defective in methylation of phosphatidylethanolamine to PC, confirming that PC de novo synthesis and TG synthesis are metabolically coupled through the efficiency of the phospholipid methylation reaction. Indeed, because both types of lipids share phosphatidic acid as a precursor, we find in cells with down-regulated Sah1 activity major alterations in the expression of the INO1 gene as well as in the localization of Opi1, a negative regulatory factor of phospholipid synthesis, which binds and is retained in the endoplasmic reticulum membrane by phosphatidic acid in conjunction with VAMP/synaptobrevin-associated protein, Scs2. The addition of homocysteine, by the reversal of the Sah1-catalyzed reaction, also leads to TG accumulation in yeast, providing an attractive model for the role of homocysteine as a risk factor of atherosclerosis in humans.


Subject(s)
Adenosylhomocysteinase/biosynthesis , Atherosclerosis/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Phosphatidylcholines/biosynthesis , Saccharomyces cerevisiae/enzymology , Triglycerides/biosynthesis , Adenosylhomocysteinase/genetics , Atherosclerosis/genetics , Down-Regulation/genetics , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Fungal/genetics , Homeostasis/genetics , Homocysteine/genetics , Homocysteine/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Methylation , Myo-Inositol-1-Phosphate Synthase/biosynthesis , Myo-Inositol-1-Phosphate Synthase/genetics , Phosphatidylcholines/genetics , Phosphatidylethanolamine N-Methyltransferase/biosynthesis , Phosphatidylethanolamine N-Methyltransferase/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Risk Factors , S-Adenosylhomocysteine/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Triglycerides/genetics
10.
Biosci Rep ; 28(3): 135-44, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18532926

ABSTRACT

A cDNA clone encoding AmphiSAHH [amphioxus SAHH (S-adenosylhomocysteine hydrolase)] protein was isolated from a cDNA library from the gut of Branchiostoma belcheri tsingtaunese. It contained a 1305 bp open reading frame corresponding to a deduced protein of 434 amino acid residues, with a predicted molecular mass of approx. 47.8 kDa. Phylogenetic analysis showed that AmphiSAHH and sea-urchin SAHH joined together and positioned at the base of the vertebrate SAHH clade, suggesting that both AmphiSAHH and sea-urchin SAHH might share some characteristics of the archetype of vertebrate SAHH proteins. The genomic DNA sequence of AmphiSAHH contained eight exons and seven introns, which was similar to B. floridae and sea-urchin SAHH exon/intron organization. Sequence comparison suggested the evolutionary appearance of the ten exon/nine intron organization of SAHH genes after the split of invertebrates and vertebrates, after which it has been highly conserved. AmphiSAHH has been successfully expressed in Escherichia coli and purified. Western blotting confirmed that the enzyme has a native molecular mass of approx. 48 kDa, and the catalytic activities and NAD(+)/NADH binding affinity of recombinant AmphiSAHH were measured. Immunohistochemistry analysis showed that SAHH was strongly expressed in hepatic caecum, gill, spermary and ovary of amphioxus.


Subject(s)
Adenosylhomocysteinase/isolation & purification , Chordata, Nonvertebrate/enzymology , Adenosylhomocysteinase/analysis , Adenosylhomocysteinase/biosynthesis , Adenosylhomocysteinase/genetics , Amino Acid Sequence , Animals , Chordata, Nonvertebrate/genetics , Conserved Sequence , DNA, Complementary/genetics , Evolution, Molecular , Exons/genetics , Humans , Introns/genetics , Molecular Sequence Data , NAD/metabolism , Phylogeny , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Sea Urchins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Vertebrates/genetics
11.
J Biol Chem ; 283(28): 19342-50, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18487201

ABSTRACT

After protracted low level arsenic exposure, the normal human prostate epithelial cell line RWPE-1 acquires a malignant phenotype with DNA hypomethylation, indicative of disrupted methyl metabolism, and shows arsenic adaptation involving glutathione overproduction and enhanced arsenic efflux. Thus, the interplay between methyl and glutathione metabolism during this progressive arsenic adaptation was studied. Arsenic-treated cells showed a time-dependent increase in LC50 and a marked increase in homocysteine (Hcy) levels. A marked suppression of S-adenosylmethionine (SAM) levels occurred with decreased methionine adenosyltransferase 2A (converts methionine to SAM) expression and increased negative regulator methionine adenosyltransferase B, suggesting reduced conversion of Hcy to SAM. Consistent with Hcy overproduction, activity and expression of S-adenosylhomocysteine hydrolase (converts S-adenosylhomocysteine to Hcy) were both increased. Expression of cystathionine beta-synthase, a key gene in the transsulfuration pathway, and various glutathione production genes were increased, resulting in a 5-fold increase in glutathione. Arsenic efflux increased along with expression of ATP-binding cassette protein C1, which effluxes arsenic as a glutathione conjugate. Evidence of genomic DNA hypomethylation was observed during early arsenic exposure, indicating that the disruption in methyl metabolism had a potential impact related to oncogenesis. Thus, cellular arsenic adaptation is a dynamic, progressive process that involves decreased SAM recycling and concurrent accumulation of Hcy, which is channeled via transsulfuration to increase glutathione and enhance arsenic efflux but may also impact the carcinogenic process.


Subject(s)
Arsenites/pharmacology , Cell Transformation, Neoplastic/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Prostate/enzymology , Sodium Compounds/pharmacology , Adenosylhomocysteinase/biosynthesis , Cell Line , Cell Transformation, Neoplastic/pathology , Cystathionine beta-Synthase/biosynthesis , DNA Methylation/drug effects , Epithelial Cells/pathology , Glutathione/metabolism , Homocysteine/metabolism , Humans , Male , Methionine Adenosyltransferase/biosynthesis , Multidrug Resistance-Associated Proteins/biosynthesis , Prostate/pathology , S-Adenosylmethionine/metabolism
12.
Bioorg Med Chem ; 15(23): 7281-7, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17845853

ABSTRACT

Ribavirin (1,2,4-triazole-3-carboxamide riboside) is a well-known antiviral drug. Ribavirin has also been reported to inhibit human S-adenosyl-L-homocysteine hydrolase (Hs-SAHH), which catalyzes the conversion of S-adenosyl-L-homocysteine to adenosine and homocysteine. We now report that ribavirin, which is structurally similar to adenosine, produces time-dependent inactivation of Hs-SAHH and Trypanosoma cruzi SAHH (Tc-SAHH). Ribavirin binds to the adenosine-binding site of the two SAHHs and reduces the NAD(+) cofactor to NADH. The reversible binding step of ribavirin to Hs-SAHH and Tc-SAHH has similar K(I) values (266 and 194 microM), but the slow inactivation step is 5-fold faster with Tc-SAHH. Ribavirin may provide a structural lead for design of more selective inhibitors of Tc-SAHH as potential anti-parasitic drugs.


Subject(s)
Adenosylhomocysteinase/antagonists & inhibitors , Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Ribavirin/pharmacology , Trypanosoma cruzi/enzymology , Adenosylhomocysteinase/biosynthesis , Adenosylhomocysteinase/isolation & purification , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Binding Sites , Chromatography, High Pressure Liquid/methods , Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Kinetics , Molecular Conformation , NAD/chemistry , NAD/drug effects , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Ribavirin/chemical synthesis , Ribavirin/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Structure-Activity Relationship , Time Factors
13.
Reprod Biol Endocrinol ; 1: 23, 2003 Feb 14.
Article in English | MEDLINE | ID: mdl-12646053

ABSTRACT

During late peri-implantation development, porcine conceptuses undergo a rapid (2-3 hrs) morphological transformation from a 10 mm sphere to a thin filamentous form greater than 150 mm in length. Elongation of the conceptus is important for establishing adequate placental surface area needed for embryo and fetal survival throughout gestation. Genes involved with triggering this unique transition in conceptus development are not well defined. Objective of the present study was to utilize suppression subtractive hybridization (SSH) to characterize the change in gene expression during conceptus transformation from spherical (8-9 mm) to tubular (15-40 mm) to early filamentous (>150 mm) morphology. Spherical, tubular, and filamentous conceptuses were collected from pregnant gilts and subjected to SSH. Forward and reverse subtractions were performed to identify candidate genes differentially expressed during spherical to tubular and tubular to filamentous transition. A total of 384 transcripts were differentially screened to ensure unique expression. Of the transcripts screened, sequences were obtained for 142 that were confirmed to be differentially expressed among the various morphologies. Gene expression profiles during rapid trophoblastic elongation were generated for selected mRNAs using quantitative real-time PCR. During the transition from tubular to early filamentous conceptuses, s-adenosylhomocysteine hydrolase and heat shock cognate 70 kDa expression were significantly enhanced. A novel unknown gene was isolated and shown to be significantly up-regulated at the onset of rapid trophoblastic elongation and further enhanced in filamentous conceptuses.


Subject(s)
Fetal Proteins/biosynthesis , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Nucleic Acid Hybridization/methods , Swine/genetics , Trophoblasts/metabolism , Adenosylhomocysteinase/biosynthesis , Adenosylhomocysteinase/genetics , Animals , DNA, Complementary/genetics , Female , Fetal Proteins/genetics , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , Morphogenesis/genetics , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Subtraction Technique , Swine/metabolism , Trophoblasts/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL