Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502042

ABSTRACT

The membrane domain of eukaryotic HMG-CoA reductase (HMGR) has the conserved capacity to induce endoplasmic reticulum (ER) proliferation and membrane association into Organized Smooth Endoplasmic Reticulum (OSER) structures. These formations develop in response to overexpression of particular proteins, but also occur naturally in cells of the three eukaryotic kingdoms. Here, we characterize OSER structures induced by the membrane domain of Arabidopsis HMGR (1S domain). Immunochemical confocal and electron microscopy studies demonstrate that the 1S:GFP chimera co-localizes with high levels of endogenous HMGR in several ER compartments, such as the ER network, the nuclear envelope, the outer and internal membranes of HMGR vesicles and the OSER structures, which we name ER-HMGR domains. After high-pressure freezing, ER-HMGR domains show typical crystalloid, whorled and lamellar ultrastructural patterns, but with wide heterogeneous luminal spaces, indicating that the native OSER is looser and more flexible than previously reported. The formation of ER-HMGR domains is reversible. OSER structures grow by incorporation of ER membranes on their periphery and progressive compaction to the inside. The ER-HMGR domains are highly dynamic in their formation versus their disassembly, their variable spherical-ovoid shape, their fluctuating borders and their rapid intracellular movement, indicating that they are not mere ER membrane aggregates, but active components of the eukaryotic cell.


Subject(s)
Arabidopsis Proteins/metabolism , Endoplasmic Reticulum/ultrastructure , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Arabidopsis , Arabidopsis Proteins/chemistry , Endoplasmic Reticulum/metabolism , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/chemistry , Plant Leaves/metabolism , Plant Leaves/ultrastructure , Protein Domains
2.
J Exp Bot ; 70(8): 2325-2338, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30753728

ABSTRACT

Sucrose non-fermenting 1 (SNF1)-related protein kinase 1.1 (SnRK1.1; also known as KIN10 or SnRK1α) has been identified as the catalytic subunit of the complex SnRK1, the Arabidopsis thaliana homologue of a central integrator of energy and stress signalling in eukaryotes dubbed AMPK/Snf1/SnRK1. A nuclear localization of SnRK1.1 has been previously described and is in line with its function as an integrator of energy and stress signals. Here, using two biological models (Nicotiana benthamiana and Arabidopsis thaliana), native regulatory sequences, different microscopy techniques, and manipulations of cellular energy status, it was found that SnRK1.1 is localized dynamically between the nucleus and endoplasmic reticulum (ER). This distribution was confirmed at a spatial and temporal level by co-localization studies with two different fluorescent ER markers, one of them being the SnRK1.1 phosphorylation target HMGR. The ER and nuclear localization displayed a dynamic behaviour in response to perturbations of the plastidic electron transport chain. These results suggest that an ER-associated SnRK1.1 fraction might be sensing the cellular energy status, being a point of crosstalk with other ER stress regulatory pathways.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Protein Serine-Threonine Kinases/metabolism , Arabidopsis/cytology , Chloroplasts/metabolism , Electron Transport , Energy Metabolism , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Plants, Genetically Modified/cytology , Plants, Genetically Modified/metabolism , Signal Transduction/physiology , Stress, Physiological , Nicotiana/cytology , Nicotiana/metabolism , Transcription Factors/metabolism
3.
Biochemistry ; 57(5): 654-662, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29224355

ABSTRACT

The key mevalonate pathway enzyme 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (HMGR) uses the cofactor NAD(P)H to reduce HMG-CoA to mevalonate in the production of countless metabolites and natural products. Although inhibition of HMGR by statin drugs is well-understood, several mechanistic details of HMGR catalysis remain unresolved, and the structural basis for the wide range of cofactor specificity for either NADH or NADPH among HMGRs from different organisms is also unknown. Here, we present crystal structures of HMGR from Streptococcus pneumoniae (SpHMGR) alongside kinetic data of the enzyme's cofactor preferences. Our structure of SpHMGR bound with its kinetically preferred NADPH cofactor suggests how NADPH-specific binding and recognition are achieved. In addition, our structure of HMG-CoA-bound SpHMGR reveals large, previously unknown conformational domain movements that may control HMGR substrate binding and enable cofactor exchange without intermediate release during the catalytic cycle. Taken together, this work provides critical new insights into both the HMGR reaction mechanism and the structural basis of cofactor specificity.


Subject(s)
Acyl Coenzyme A/metabolism , Bacterial Proteins/chemistry , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/chemistry , NADP/metabolism , Streptococcus pneumoniae/enzymology , Bacterial Proteins/metabolism , Binding Sites , Coenzymes/metabolism , Crystallography, X-Ray , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Kinetics , Models, Molecular , NAD/metabolism , Oxidation-Reduction , Protein Binding , Protein Conformation , Protein Domains , Recombinant Proteins/chemistry , Structure-Activity Relationship , Substrate Specificity
4.
Molecules ; 23(3)2018 Mar 06.
Article in English | MEDLINE | ID: mdl-29509695

ABSTRACT

As the main bioactive constituents of Panax species, ginsenosides possess a wide range of notable medicinal effects such as anti-cancer, anti-oxidative, antiaging, anti-inflammatory, anti-apoptotic and neuroprotective activities. However, the increasing medical demand for ginsenosides cannot be met due to the limited resource of Panax species and the low contents of ginsenosides. In recent years, biotechnological approaches have been utilized to increase the production of ginsenosides by regulating the key enzymes of ginsenoside biosynthesis, while synthetic biology strategies have been adopted to produce ginsenosides by introducing these genes into yeast. This review summarizes the latest research progress on cloning and functional characterization of key genes dedicated to the production of ginsenosides, which not only lays the foundation for their application in plant engineering, but also provides the building blocks for the production of ginsenosides by synthetic biology.


Subject(s)
Ginsenosides/biosynthesis , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Biosynthetic Pathways/genetics , Cloning, Molecular , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Farnesyl-Diphosphate Farnesyltransferase/genetics , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Ginsenosides/chemistry , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Intramolecular Transferases/genetics , Intramolecular Transferases/metabolism , Squalene Monooxygenase/genetics , Squalene Monooxygenase/metabolism
5.
BMC Plant Biol ; 17(1): 88, 2017 05 22.
Article in English | MEDLINE | ID: mdl-28532507

ABSTRACT

BACKGROUND: Latex from the dandelion species Taraxacum brevicorniculatum contains many high-value isoprenoid end products, e.g. triterpenes and polyisoprenes such as natural rubber. The isopentenyl pyrophosphate units required as precursors for these isoprenoids are provided by the mevalonate (MVA) pathway. The key enzyme in this pathway is 3-hydroxy-methyl-glutaryl-CoA reductase (HMGR) and its activity has been thoroughly characterized in many plant species including dandelion. However, two enzymes acting upstream of HMGR have not been characterized in dandelion latex: ATP citrate lyase (ACL), which provides the acetyl-CoA utilized in the MVA pathway, and acetoacetyl-CoA thiolase (AACT), which catalyzes the first step in the pathway to produce acetoacetyl-CoA. Here we isolated ACL and AACT genes from T. brevicorniculatum latex and characterized their expression profiles. We also overexpressed the well-characterized HMGR, ACL and AACT genes from Arabidopsis thaliana in T. brevicorniculatum to determine their impact on isoprenoid end products in the latex. RESULTS: The spatial and temporal expression profiles of T. brevicorniculatum ACL and AACT revealed their pivotal role in the synthesis of precursors necessary for isoprenoid biosynthesis in latex. The overexpression of A. thaliana ACL and AACT and HMGR in T. brevicorniculatum latex resulted in the accumulation of all three enzymes, increased the corresponding enzymatic activities and ultimately increased sterol levels by ~5-fold and pentacyclic triterpene and cis-1,4-isoprene levels by ~2-fold. Remarkably high levels of the triterpene precursor squalene were also detected in the triple-transgenic lines (up to 32 mg/g root dry weight) leading to the formation of numerous lipid droplets which were observed in root cross-sections. CONCLUSIONS: We could show the effective expression of up to three transgenes in T. brevicorniculatum latex which led to increased enzymatic activity and resulted in high level squalene accumulation in the dandelion roots up to an industrially relevant amount. Our data provide insight into the regulation of the MVA pathway in dandelion latex and can be used as a basis for metabolic engineering to enhance the production of isoprenoid end products in this specialized tissue.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Latex/metabolism , Taraxacum/metabolism , Terpenes/metabolism , ATP Citrate (pro-S)-Lyase/genetics , Acetyl-CoA C-Acetyltransferase/genetics , Gene Expression Regulation, Plant , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Mevalonic Acid/metabolism , Pentacyclic Triterpenes/metabolism , Phytosterols/metabolism , Squalene/metabolism , Taraxacum/genetics
6.
Plant Physiol ; 170(1): 194-210, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26589673

ABSTRACT

Plants respond to stresses by producing a broad spectrum of bioactive specialized metabolites. Hormonal elicitors, such as jasmonates, trigger a complex signaling circuit leading to the concerted activation of specific metabolic pathways. However, for many specialized metabolic pathways, the transcription factors involved remain unknown. Here, we report on two homologous jasmonate-inducible transcription factors of the basic helix-loop-helix family, TRITERPENE SAPONIN BIOSYNTHESIS ACTIVATING REGULATOR1 (TSAR1) and TSAR2, which direct triterpene saponin biosynthesis in Medicago truncatula. TSAR1 and TSAR2 are coregulated with and transactivate the genes encoding 3-HYDROXY-3-METHYLGLUTARYL-COENZYME A REDUCTASE1 (HMGR1) and MAKIBISHI1, the rate-limiting enzyme for triterpene biosynthesis and an E3 ubiquitin ligase that controls HMGR1 levels, respectively. Transactivation is mediated by direct binding of TSARs to the N-box in the promoter of HMGR1. In transient expression assays in tobacco (Nicotiana tabacum) protoplasts, TSAR1 and TSAR2 exhibit different patterns of transactivation of downstream triterpene saponin biosynthetic genes, hinting at distinct functionalities within the regulation of the pathway. Correspondingly, overexpression of TSAR1 or TSAR2 in M. truncatula hairy roots resulted in elevated transcript levels of known triterpene saponin biosynthetic genes and strongly increased the accumulation of triterpene saponins. TSAR2 overexpression specifically boosted hemolytic saponin biosynthesis, whereas TSAR1 overexpression primarily stimulated nonhemolytic soyasaponin biosynthesis. Both TSARs also activated all genes of the precursor mevalonate pathway but did not affect sterol biosynthetic genes, pointing to their specific role as regulators of specialized triterpene metabolism in M. truncatula.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Medicago truncatula/metabolism , Plant Proteins/metabolism , Saponins/biosynthesis , Binding Sites , Cyclopentanes/metabolism , Gene Expression Regulation, Plant , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Medicago truncatula/genetics , Mevalonic Acid/metabolism , Oxylipins/metabolism , Plant Proteins/genetics , Plant Roots/genetics , Plant Roots/metabolism , Plants, Genetically Modified , Promoter Regions, Genetic , Saponins/genetics , Saponins/metabolism , Sequence Analysis, RNA , Nicotiana/genetics , Triterpenes/metabolism
7.
J Cell Biochem ; 117(9): 2036-44, 2016 09.
Article in English | MEDLINE | ID: mdl-27392312

ABSTRACT

During differentiation, neurons acquire their typical shape and functional properties. At present, it is unclear, whether this important developmental step involves metabolic changes. Here, we studied the contribution of the mevalonate (MVA) pathway to neuronal differentiation using the mouse neuroblastoma cell line N1E-115 as experimental model. Our results show that during differentiation, the activity of 3-hydroxy 3-methylglutaryl Coenzyme A reductase (HMGR), a key enzyme of MVA pathway, and the level of Low Density Lipoprotein receptor (LDLr) decrease, whereas the level of LDLr-related protein-1 (LRP1) and the dimerization of Scavanger Receptor B1 (SRB-1) rise. Pharmacologic inhibition of HMGR by simvastatin accelerated neuronal differentiation by modulating geranylated proteins. Collectively, our data suggest that during neuronal differentiation, the activity of the MVA pathway decreases and we postulate that any interference with this process impacts neuronal morphology and function. Therefore, the MVA pathway appears as an attractive pharmacological target to modulate neurological and metabolic symptoms of developmental neuropathologies. J. Cell. Biochem. 117: 2036-2044, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation/physiology , Cholesterol/biosynthesis , Neurons/metabolism , Terpenes/metabolism , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cholesterol/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Receptors, LDL/genetics , Receptors, LDL/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Simvastatin/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Plant Physiol ; 168(3): 899-914, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26015445

ABSTRACT

The enzyme 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR) has a key regulatory role in the mevalonate pathway for isoprenoid biosynthesis and is composed of an endoplasmic reticulum (ER)-anchoring membrane domain with low sequence similarity among eukaryotic kingdoms and a conserved cytosolic catalytic domain. Organized smooth endoplasmic reticulum (OSER) structures are common formations of hypertrophied tightly packed ER membranes devoted to specific biosynthetic and secretory functions, the biogenesis of which remains largely unexplored. We show that the membrane domain of plant HMGR suffices to trigger ER proliferation and OSER biogenesis. The proliferating membranes become highly enriched in HMGR protein, but they do not accumulate sterols, indicating a morphogenetic rather than a metabolic role for HMGR. The N-terminal MDVRRRPP motif present in most plant HMGR isoforms is not required for retention in the ER, which was previously proposed, but functions as an ER morphogenic signal. Plant OSER structures are morphologically similar to those of animal cells, emerge from tripartite ER junctions, and mainly build up beside the nuclear envelope, indicating conserved OSER biogenesis in high eukaryotes. Factors other than the OSER-inducing HMGR construct mediate the tight apposition of the proliferating membranes, implying separate ER proliferation and membrane association steps. Overexpression of the membrane domain of Arabidopsis (Arabidopsis thaliana) HMGR leads to ER hypertrophy in every tested cell type and plant species, whereas the knockout of the HMG1 gene from Arabidopsis, encoding its major HMGR isoform, causes ER aggregation at the nuclear envelope. Our results show that the membrane domain of HMGR contributes to ER morphogenesis in plant cells.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Endoplasmic Reticulum/metabolism , Hydroxymethylglutaryl CoA Reductases/chemistry , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/chemistry , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Morphogenesis , Plant Cells/enzymology , Amino Acid Motifs , Arabidopsis/genetics , Arabidopsis/ultrastructure , Cell Nucleus/metabolism , Endoplasmic Reticulum/ultrastructure , Genes, Plant , Green Fluorescent Proteins/metabolism , Molecular Sequence Data , Plants, Genetically Modified , Protein Structure, Tertiary , Sterols/metabolism , Structure-Activity Relationship , Nicotiana/metabolism
9.
Int J Mol Sci ; 17(11)2016 Nov 09.
Article in English | MEDLINE | ID: mdl-27834848

ABSTRACT

The aim of this study was to investigate the mechanisms underlying the inhibitory effects of berberine (BBR) on olanzapine (OLZ)-induced adipogenesis in a well-replicated 3T3-L1 cell model. Oil-Red-O (ORO) staining showed that BBR significantly decreased OLZ-induced adipogenesis. Co-treatment with OLZ and BBR decreased the accumulation of triglyceride (TG) and total cholesterol (TC) by 55.58% ± 3.65% and 49.84% ± 8.31%, respectively, in 3T3-L1 adipocytes accompanied by reduced expression of Sterol regulatory element binding proteins 1 (SREBP1), fatty acid synthase (FAS), peroxisome proliferator activated receptor-γ (PPARγ), SREBP2, low-density lipoprotein receptor (LDLR), and hydroxymethylglutaryl-coenzyme A reductase (HMGR) genes compared with OLZ alone. Consistently, the co-treatment downregulated protein levels of SREBP1, SREBP2, and LDLR by 57.71% ± 9.42%, 73.05% ± 11.82%, and 59.46% ± 9.91%, respectively. In addition, co-treatment reversed the phosphorylation level of AMP-activated protein kinase-α (AMPKα), which was reduced by OLZ, determined via the ratio of pAMPKα:AMPKα (94.1%) compared with OLZ alone. The results showed that BBR may prevent lipid metabolism disorders caused by OLZ by reversing the degree of SREBP pathway upregulated and the phosphorylation of AMPKα downregulated. Collectively, these results indicated that BBR could be used as a potential adjuvant to prevent dyslipidemia and obesity caused by the use of second-generation antipsychotic medication.


Subject(s)
AMP-Activated Protein Kinases/genetics , Adipocytes/drug effects , Antipsychotic Agents/antagonists & inhibitors , Benzodiazepines/antagonists & inhibitors , Berberine/pharmacology , Hypolipidemic Agents/pharmacology , Sterol Regulatory Element Binding Protein 1/genetics , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Cell Differentiation , Cholesterol/biosynthesis , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Gene Expression Regulation , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Mice , Olanzapine , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation/drug effects , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Triglycerides/antagonists & inhibitors , Triglycerides/biosynthesis
10.
Chem Soc Rev ; 43(19): 6672-82, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-24710074

ABSTRACT

Cholesterol and metal ions have been suggested to be associated with the onset and progression of Alzheimer's disease (AD). Moreover, recent findings have demonstrated a potential interconnection between these two factors. For example, (a) cholesterol has been shown to be misregulated in AD-afflicted brains, and the aberrant activity of proteins (particularly, apolipoprotein E (ApoE) and 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase (HMGR)) has been linked to cholesterol-related AD exacerbation; (b) dyshomeostasis of metal ions associated with misfolded proteins (i.e., amyloid-ß (Aß) aggregates) found in the brains of AD patients is shown to promote oxidative stress leading to the malfunction of multiple proteins, including cytochrome c oxidase (CcO), and Cu/Zn superoxide dismutase (SOD1); (c) metal ion misregulation has also been observed to disrupt the activity of proteins (e.g., HMGR, low-density lipoproteins (LDL)), required for cholesterol production and regulation. Herein, we briefly discuss the potential involvement of cholesterol and metal ions in AD neuropathogenesis in both individual and interrelated manners.


Subject(s)
Alzheimer Disease/pathology , Cholesterol/metabolism , Metals/metabolism , Alzheimer Disease/metabolism , Apolipoproteins E/metabolism , Brain/metabolism , Cholesterol/chemistry , Humans , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Metals/chemistry , Reactive Oxygen Species/metabolism
11.
Curr Opin Lipidol ; 24(3): 221-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23571733

ABSTRACT

PURPOSE OF REVIEW: The reduction in cardiovascular disease risk by statins is well established. This risk reduction has mostly been attributed to decreases in plasma LDL cholesterol and other pleiotropic effects of statins. Emerging evidence indicates that statins exert multiple effects on lipoprotein metabolism, including chylomicrons and HDLs. RECENT FINDINGS: Kinetic and in-vitro studies have documented that the effects of statins on the metabolism of different lipoproteins are for the most part the direct consequence of cholesterol biosynthesis inhibition and the subsequent change in transcription factors and cell signaling, regulating different aspects of lipoprotein metabolism. Differences in pharmacokinetics and pharmacodynamics among statins lead to diverse biological outcomes. SUMMARY: The current review summarizes recent experimental evidence highlighting the different effects of statins on cellular pathways regulating gene expression. Understanding the basic mechanisms by which different statins regulate lipoprotein metabolism will lead to improved strategies for the prevention and treatment of specific lipoprotein disorders.


Subject(s)
Cardiovascular Diseases/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Lipid Metabolism/drug effects , Transcription Factors/genetics , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Cardiovascular Diseases/blood , Cardiovascular Diseases/physiopathology , Cholesterol, HDL/agonists , Cholesterol, HDL/blood , Cholesterol, LDL/antagonists & inhibitors , Cholesterol, LDL/blood , Chylomicrons/blood , Gene Expression Regulation , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Proprotein Convertase 9 , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors
12.
Antimicrob Agents Chemother ; 57(1): 375-81, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23114753

ABSTRACT

Acanthamoeba is an opportunistic pathogen in humans, whose infections most commonly manifest as Acanthamoeba keratitis or, more rarely, granulomatous amoebic encephalitis. Although there are many therapeutic options for the treatment of Acanthamoeba, they are generally lengthy and/or have limited efficacy. Therefore, there is a requirement for the identification, validation, and development of novel therapeutic targets against these pathogens. Recently, RNA interference (RNAi) has been widely used for these validation purposes and has proven to be a powerful tool for Acanthamoeba therapeutics. Ergosterol is one of the major sterols in the membrane of Acanthamoeba. 3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase is an enzyme that catalyzes the conversion of HMG-CoA to mevalonate, one of the precursors for the production of cholesterol in humans and ergosterol in plants, fungi, and protozoa. Statins are compounds which inhibit this enzyme and so are promising as chemotherapeutics. In order to validate whether this enzyme could be an interesting therapeutic target in Acanthamoeba, small interfering RNAs (siRNAs) against HMG-CoA were developed and used to evaluate the effects induced by the inhibition of Acanthamoeba HMG-CoA. It was found that HMG-CoA is a potential drug target in these pathogenic free-living amoebae, and various statins were evaluated in vitro against three clinical strains of Acanthamoeba by using a colorimetric assay, showing important activities against the tested strains. We conclude that the targeting of HMG-CoA and Acanthamoeba treatment using statins is a novel powerful treatment option against Acanthamoeba species in human disease.


Subject(s)
Acanthamoeba castellanii/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Protozoan Proteins/metabolism , Acanthamoeba castellanii/enzymology , Acanthamoeba castellanii/genetics , Acanthamoeba castellanii/growth & development , Acyl Coenzyme A/antagonists & inhibitors , Acyl Coenzyme A/metabolism , Amino Acid Sequence , Catalytic Domain , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Assays , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Inhibitory Concentration 50 , Mevalonic Acid/metabolism , Molecular Sequence Data , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , RNA, Small Interfering/genetics
13.
Ann Neurol ; 72(4): 510-6, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23109145

ABSTRACT

OBJECTIVE: Primary lateral sclerosis (PLS) is a motor neuron disorder that exclusively affects upper motor neurons leading to their degeneration. Mutations in the ALS2 gene encoding the protein Alsin have been described previously in the juvenile form of the disease. In this study, we identify mutation of the ERLIN2 gene in juvenile PLS patients and describe an in vitro model for loss of ERLIN2 function. METHODS: Single nucleotide polymorphism arrays were used for homozygosity mapping. DNA sequencing of candidate genes was used to detect the underlying mutation. Level of ERLIN2 mRNA was measured by quantitative real time polymerase chain reaction. Knocking down ERLIN2 in NSC34 cells was accomplished by short-hairpin RNA interference. RESULTS: We identified a splice junction mutation in the ERLIN2 gene-a component of the endoplasmic reticulum (ER) lipid rafts-that resulted in abnormal splicing of ERLIN2 transcript and nonsense-mediated decay of ERLIN2 mRNA. Knocking down ERLIN2 in NSC34 cells suppressed their growth in culture. INTERPRETATION: Recently, we found that mutation of SIGMAR1, a component of ER lipid rafts, leads to juvenile amyotrophic lateral sclerosis. The identification of mutation in another component of the ER lipid rafts in juvenile PLS patients emphasizes their role in motor neuron function. Furthermore, the discovered effect of ERLIN2 loss on cell growth may advance understanding of the mechanism behind motor neuron degeneration in PLS.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/physiology , Motor Neuron Disease/genetics , Adolescent , Cell Count , Cells, Cultured , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 8/genetics , DNA/genetics , DNA/isolation & purification , Endoplasmic Reticulum-Associated Degradation/genetics , Endoplasmic Reticulum-Associated Degradation/physiology , Female , Humans , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Infant , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , RNA Interference/physiology , RNA Splice Sites/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
14.
Appl Environ Microbiol ; 78(2): 568-74, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22081574

ABSTRACT

We report here a novel selectable marker for the hyperthermophilic crenarchaeon Sulfolobus islandicus. The marker cassette is composed of the sac7d promoter and the hmg gene coding for the 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (P(sac7d)-hmg), which confers simvastatin resistance to this crenarchaeon. The basic plasmid vector pSSR was constructed by substituting the pyrEF gene of the expression vector pSeSD for P(sac7d)-hmg with which the Sulfolobus expression plasmids pSSRlacS, pSSRAherA, and pSSRNherA were constructed. Characterization of Sulfolobus transformants carrying pSSRlacS indicated that the plasmid was properly maintained under selection. High-level expression of the His(6)-tagged HerA helicase was obtained with the cells harboring pSSRAherA. The establishment of two efficient selectable markers (pyrEF and hmg) was subsequently exploited for genetic analysis. A herA merodiploid strain of S. islandicus was constructed using pyrEF marker and used as the host to obtain pSSRNherA transformant with simvastatin selection. While the gene knockout (ΔherA) cells generated from the herA merodiploid cells failed to form colonies in the presence of 5-fluoroorotic acid (5-FOA), the mutant cells could be rescued by expression of the gene from a plasmid (pSSRNherA), because their transformants formed colonies on a solid medium containing 5-FOA and simvastatin. This demonstrates that HerA is essential for cell viability of S. islandicus. To our knowledge, this is the first application of an antibiotic selectable marker in genetic study for a hyperthermophilic acidophile and in the crenarchaeal lineage.


Subject(s)
Drug Resistance , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Selection, Genetic , Simvastatin/metabolism , Simvastatin/toxicity , Sulfolobus/growth & development , Sulfolobus/metabolism , Molecular Biology/methods
15.
Appl Environ Microbiol ; 78(22): 7968-76, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22941092

ABSTRACT

Ganoderic acids produced by Ganoderma lucidum, a well-known traditional Chinese medicinal mushroom, exhibit antitumor and antimetastasis activities. Genetic modification of G. lucidum is difficult but critical for the enhancement of cellular accumulation of ganoderic acids. In this study, a homologous genetic transformation system for G. lucidum was developed for the first time using mutated sdhB, encoding the iron-sulfur protein subunit of succinate dehydrogenase, as a selection marker. The truncated G. lucidum gene encoding the catalytic domain of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR) was overexpressed by using the Agrobacterium tumefaciens-mediated transformation system. The results showed that the mutated sdhB successfully conferred carboxin resistance upon transformation. Most of the integrated transfer DNA (T-DNA) appeared as a single copy in the genome. Moreover, deregulated constitutive overexpression of the HMGR gene led to a 2-fold increase in ganoderic acid content. It also increased the accumulation of intermediates (squalene and lanosterol) and the upregulation of downstream genes such as those of farnesyl pyrophosphate synthase, squalene synthase, and lanosterol synthase. This study demonstrates that transgenic basidiomycete G. lucidum is a promising system to achieve metabolic engineering of the ganoderic acid pathway.


Subject(s)
Genetics, Microbial/methods , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Metabolic Engineering , Reishi/enzymology , Reishi/metabolism , Triterpenes/metabolism , Agrobacterium tumefaciens/genetics , DNA, Fungal/chemistry , DNA, Fungal/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Molecular Sequence Data , Reishi/genetics , Sequence Analysis, DNA , Transformation, Genetic
16.
Poult Sci ; 91(4): 829-36, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22399721

ABSTRACT

In this longitudinal study with Single Comb White Leghorn chickens, we investigated the effects of stress conditions in birds that were subjected to a high stocking density with feed restrictions on the quantity of telomeric DNA, the rate of DNA damage, and the expression levels of heat shock proteins (HSP) and hydroxyl-3-methyl-glutaryl coenzyme A reductase (HMGCR) genes. The telomere length and telomere-shortening rates were analyzed by quantitative fluorescence in situ hybridization on the nuclei of lymphocytes. The DNA damage rate of lymphocytes was quantified by the comet assay. The expression levels of HSP70, HSP90, and HMGCR genes were measured by quantitative real-time PCR in lymphocytes. The telomere-shortening rate of the lymphocytes was significantly higher in the stress group than in the control. The DNA damage also increased in birds raised under stress conditions, as compared with the control group. The stress conditions had a significant effect on the expressions of HMGCR and HSP90α in lymphocytes but had no significance on HSP70 and HSP90ß in blood. We conclude that the telomere length, especially the telomere-shortening rates, the quantification of total DNA damage, and the expression levels of the HMGCR and HSP90α genes can be used as sensitive physiological stress markers in chickens.


Subject(s)
Chickens/physiology , Gene Expression Regulation , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Animals , Chickens/genetics , Comet Assay/veterinary , DNA/analysis , DNA Damage , Food Deprivation , In Situ Hybridization, Fluorescence/veterinary , Longitudinal Studies , Lymphocytes/metabolism , Population Density , Random Allocation , Real-Time Polymerase Chain Reaction/veterinary , Telomere/metabolism
17.
J Plant Physiol ; 267: 153543, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34678642

ABSTRACT

Autoregulation of nodulation (AON) plays a central role in nodulation by inhibiting the formation of excess number of legume root nodules. In this study, the effect of hydroxymethylglutaryl-coenzyme A reductase 1 (GmHMGR1) gene expression on nodulation and the AON system in Glycine max (L.) Merr was investigated. Wild-type soybean (cultivar Bragg) and its near-isogenic supernodulating mutant (nitrate tolerant symbiotic) nts1007 were selected to identify the expression pattern of this gene in rootlets after inoculation by its microsymbiont Bradyrhizobium. For further analysis, the full length of GmHMGR1 and its promoter were cloned after amplification by inverse-PCR and BAC library screening. Also, we constructed an intron hairpin RNA interference (ihpRNAi) and a GmHMGR1 promoter: ß-glucuronidase fusion constructs, consequently for suppression of GmHMGR1 and histochemical analysis in transgenic soybean hairy roots induced by Agrobacterium rhizogenes strain K599. The GmHMGR1 gene was functional during the early stages of nodulation with the AON system having a negative effect on GmHMGR1 expression and nodule formation in wild-type rootlets. GmHMGR1 was particularly expressed in the developing phloem within the root, nodules and nodule lenticels. Expression of GmHMGR1 in transgenic hairy roots was suppressed by RNAi silencing approximately 85% as compared to empty vector controls. This suggests that the GmHMGR1 gene has an important role in triggering nodule formation as its suppression caused a reduction of nodule formation in nts mutant lines with a deficient AON system.


Subject(s)
Glycine max , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent , Plant Proteins , Plant Root Nodulation , Gene Expression Regulation, Plant , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Root Nodulation/genetics , Plants, Genetically Modified/metabolism , Root Nodules, Plant/genetics , Root Nodules, Plant/metabolism , Glycine max/genetics , Glycine max/metabolism
18.
J Nat Prod ; 73(2): 208-12, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20085309

ABSTRACT

In the course of our screening program for isoprenoids of marine actinobacterial origin, 523 actinobacterial strains were isolated from marine samples. Actinobacteria usually use the 2-C-methyl-d-erythritol 4-phosphate pathway for the production of primary metabolites, but some have been reported to use the mevalonate (MVA) pathway for the production of isoprenoids as secondary metabolites. 3-Hydroxy-3-methyl glutaryl coenzyme A reductase (HMGR) is a key enzyme and plays an important role in the MVA pathway. Therefore, we screened strains possessing the HMGR gene from the 523 strains mentioned above and also investigated isoprenoid compounds from cultures of strains possessing HMGR genes. As a result, Streptomyces sp. SpC080624SC-11 isolated from a marine sponge, Cinachyra sp., was shown to possess the HMGR gene and produce novel isoprenoids, JBIR-46 (1), -47 (2), and -48 (3). On the basis of extensive NMR and MS analyses, the structures of 1-3 were determined to be phenazine derivatives harboring dimethylallyl moieties. Furthermore, the isoprene units of 2 and 3 were confirmed to be synthesized via the MVA pathway in a feeding experiment using [1-(13)C]acetate.


Subject(s)
Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Mevalonic Acid/metabolism , Porifera/microbiology , Streptomyces/chemistry , Terpenes/isolation & purification , Animals , Erythritol/analogs & derivatives , Erythritol/metabolism , Marine Biology , Molecular Structure , Sequence Homology, Nucleic Acid , Streptomyces/enzymology , Streptomyces/genetics , Sugar Phosphates/metabolism , Terpenes/chemistry , Terpenes/metabolism
19.
Appl Environ Microbiol ; 75(17): 5536-43, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19592534

ABSTRACT

(E, E, E)-Geranylgeraniol (GGOH) is a valuable starting material for perfumes and pharmaceutical products. In the yeast Saccharomyces cerevisiae, GGOH is synthesized from the end products of the mevalonate pathway through the sequential reactions of farnesyl diphosphate synthetase (encoded by the ERG20 gene), geranylgeranyl diphosphate synthase (the BTS1 gene), and some endogenous phosphatases. We demonstrated that overexpression of the diacylglycerol diphosphate phosphatase (DPP1) gene could promote GGOH production. We also found that overexpression of a BTS1-DPP1 fusion gene was more efficient for producing GGOH than coexpression of these genes separately. Overexpression of the hydroxymethylglutaryl-coenzyme A reductase (HMG1) gene, which encodes the major rate-limiting enzyme of the mevalonate pathway, resulted in overproduction of squalene (191.9 mg liter(-1)) rather than GGOH (0.2 mg liter(-1)) in test tube cultures. Coexpression of the BTS1-DPP1 fusion gene along with the HMG1 gene partially redirected the metabolic flux from squalene to GGOH. Additional expression of a BTS1-ERG20 fusion gene resulted in an almost complete shift of the flux to GGOH production (228.8 mg liter(-1) GGOH and 6.5 mg liter(-1) squalene). Finally, we constructed a diploid prototrophic strain coexpressing the HMG1, BTS1-DPP1, and BTS1-ERG20 genes from multicopy integration vectors. This strain attained 3.31 g liter(-1) GGOH production in a 10-liter jar fermentor with gradual feeding of a mixed glucose and ethanol solution. The use of bifunctional fusion genes such as the BTS1-DPP1 and ERG20-BTS1 genes that code sequential enzymes in the metabolic pathway was an effective method for metabolic engineering.


Subject(s)
Biosynthetic Pathways/genetics , Diterpenes/metabolism , Genetic Engineering/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Geranyltranstransferase/genetics , Geranyltranstransferase/metabolism , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Squalene/metabolism
20.
Antonie Van Leeuwenhoek ; 95(1): 55-64, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18853273

ABSTRACT

In this study, the gene hmgR encoding the 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoA reductase) was cloned and characterized in the zygomycete fungus Rhizomucor miehei. The hmgR gene comprises a total of 3,585 bp including the coding sequence of a 1,058 amino acids length putative protein and five introns (137, 83, 59, 60 and 69 bp in length) dispersed in the whole coding region. Southern hybridization analysis revealed that the gene is present only in one copy in the R. miehei genome. The isolated Rhizomucor gene was expressed in the related fungus, Mucor circinelloides. Transformants harbouring the Rhizomucor hmgR gene in an autoreplicative plasmid proved to be more tolerant to statins (e.g. lovastatin, simvastatin, and fluvastatin), the competitive inhibitors of the HMG-CoA reductase, than the original M. circinelloides strain. At the same time, heterologous expression of the Rhizomucor hmgR did not affect the carotenoid production of M. circinelloides.


Subject(s)
Cloning, Molecular , Fungal Proteins/genetics , Gene Expression , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Mucor/genetics , Rhizomucor/enzymology , Amino Acid Sequence , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fungi/classification , Fungi/enzymology , Fungi/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/chemistry , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Molecular Sequence Data , Mucor/metabolism , Phylogeny , Protein Structure, Tertiary , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL