Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Cell Sci ; 137(16)2024 08 15.
Article in English | MEDLINE | ID: mdl-39140134

ABSTRACT

FEM1B is a substrate-recognition component of the CRL2 E3 ubiquitin-protein ligase. This multi-protein complex targets specific proteins for ubiquitylation, which leads to their degradation. Here, we demonstrate the regulation of FEM1B expression by stop codon readthrough (SCR). In this process, translating ribosomes readthrough the stop codon of FEM1B to generate a C-terminally extended isoform that is highly unstable. A total of 81 nucleotides in the proximal 3'UTR of FEM1B constitute the necessary and sufficient cis-signal for SCR. Also, they encode the amino acid sequence responsible for the degradation of the SCR product. CRISPR-edited cells lacking this region, and therefore SCR of FEM1B, showed increased FEM1B expression. This in turn resulted in reduced expression of SLBP (a target of FEM1B-mediated degradation) and replication-dependent histones (target of SLBP for mRNA stability), causing cell cycle delay. Evolutionary analysis revealed that this phenomenon is specific to the genus Pan and Homo (Hominini). Overall, we show a relatively recently evolved SCR process that relieves the cell cycle from the negative regulation by FEM1B.


Subject(s)
Cell Cycle Proteins , Cell Cycle , Codon, Terminator , Humans , Codon, Terminator/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle/genetics , Protein Biosynthesis/genetics , Animals , 3' Untranslated Regions/genetics , HEK293 Cells , Histones/metabolism , Histones/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nuclear Proteins , mRNA Cleavage and Polyadenylation Factors
2.
EMBO Rep ; 25(4): 2118-2143, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499809

ABSTRACT

Stop codon readthrough (SCR) is the process where translation continues beyond a stop codon on an mRNA. Here, we describe a strategy to enhance or induce SCR in a transcript-selective manner using a CRISPR-dCas13 system. Using specific guide RNAs, we target dCas13 to the region downstream of canonical stop codons of mammalian AGO1 and VEGFA mRNAs, known to exhibit natural SCR. Readthrough assays reveal enhanced SCR of these mRNAs (both exogenous and endogenous) caused by the dCas13-gRNA complexes. This effect is associated with ribosomal pausing, which has been reported for several SCR events. Our data show that CRISPR-dCas13 can also induce SCR across premature termination codons (PTCs) in the mRNAs of green fluorescent protein and TP53. We demonstrate the utility of this strategy in the induction of readthrough across the thalassemia-causing PTC in HBB mRNA and hereditary spherocytosis-causing PTC in SPTA1 mRNA. Thus, CRISPR-dCas13 can be programmed to enhance or induce SCR in a transcript-selective and stop codon-specific manner.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , RNA, Guide, CRISPR-Cas Systems , Animals , Codon, Terminator/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Codon, Nonsense/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Protein Biosynthesis , Mammals/genetics , Mammals/metabolism
3.
J Mol Biol ; 435(21): 168274, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37714299

ABSTRACT

During translation, a stop codon on the mRNA signals the ribosomes to terminate the process. In certain mRNAs, the termination fails due to the recoding of the canonical stop codon, and ribosomes continue translation to generate C-terminally extended protein. This process, termed stop codon readthrough (SCR), regulates several cellular functions. SCR is driven by elements/factors that act immediately downstream of the stop codon. Here, we have analysed the process of SCR using a simple mathematical model to investigate how the kinetics of translating ribosomes influences the efficiency of SCR. Surprisingly, the analysis revealed that the rate of translation inversely regulates the efficiency of SCR. We tested this prediction experimentally in mammalian AGO1 and MTCH2 mRNAs. Reduction in translation either globally by harringtonine or locally by rare codons caused an increase in the efficiency of SCR. Thus, our study has revealed a hitherto unknown mode of regulation of SCR.


Subject(s)
Codon, Terminator , Protein Biosynthesis , RNA, Messenger , Ribosomes , Codon, Terminator/genetics , Codon, Terminator/metabolism , Ribosomes/genetics , Ribosomes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Humans , HEK293 Cells , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism
4.
J Biol Chem ; 299(9): 105184, 2023 09.
Article in English | MEDLINE | ID: mdl-37611826

ABSTRACT

Termination codon readthrough (TCR) is a process in which ribosomes continue to translate an mRNA beyond a stop codon generating a C-terminally extended protein isoform. Here, we demonstrate TCR in mammalian NNAT mRNA, which encodes NNAT, a proteolipid important for neuronal differentiation. This is a programmed event driven by cis-acting RNA sequences present immediately upstream and downstream of the canonical stop codon and is negatively regulated by NONO, an RNA-binding protein known to promote neuronal differentiation. Unlike the canonical isoform NNAT, we determined that the TCR product (NNATx) does not show detectable interaction with the sarco/endoplasmic reticulum Ca2+-ATPase isoform 2 Ca2+ pump, cannot increase cytoplasmic Ca2+ levels, and therefore does not enhance neuronal differentiation in Neuro-2a cells. Additionally, an antisense oligonucleotide that targets a region downstream of the canonical stop codon reduced TCR of NNAT and enhanced the differentiation of Neuro-2a cells to cholinergic neurons. Furthermore, NNATx-deficient Neuro-2a cells, generated using CRISPR-Cas9, showed increased cytoplasmic Ca2+ levels and enhanced neuronal differentiation. Overall, these results demonstrate regulation of neuronal differentiation by TCR of NNAT. Importantly, this process can be modulated using a synthetic antisense oligonucleotide.


Subject(s)
Calcium , Neurons , Protein Biosynthesis , Animals , Calcium/metabolism , Cell Differentiation , Codon, Terminator , Mammals/metabolism , Oligonucleotides, Antisense/metabolism , Receptors, Antigen, T-Cell/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Neurons/cytology
5.
Wiley Interdiscip Rev RNA ; 14(2): e1739, 2023 03.
Article in English | MEDLINE | ID: mdl-35570338

ABSTRACT

Recognition of a stop codon by translation machinery as a sense codon results in translational readthrough instead of termination. This recoding process, termed stop codon readthrough (SCR) or translational readthrough, is found in all domains of life including mammals. The context of the stop codon, local mRNA topology, and molecules that interact with the mRNA region downstream of the stop codon determine SCR. The products of SCR can have localization, stability, and function different from those of the canonical isoforms. In this review, we discuss how recent technological and computational advances have increased our understanding of the SCR process in the mammalian system. Based on the known molecular events that occur during SCR of multiple mRNAs, we propose transient molecular roadblocks on an mRNA downstream of the stop codon as a possible mechanism for the induction of SCR. We argue, with examples, that the insights gained from the natural SCR events can guide us to develop novel strategies for the treatment of diseases caused by premature stop codons. This article is categorized under: Translation > Regulation.


Subject(s)
Protein Biosynthesis , Proteome , Animals , Codon, Terminator/genetics , Proteome/genetics , Mammals/genetics , Mammals/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Mol Biol Cell ; 33(12): br21, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35976696

ABSTRACT

It is generally believed that human mature erythrocytes do not possess functional ribosomes and therefore cannot synthesize proteins. However, the absence of translation is not consistent with the long lifespan of mature erythrocytes. They stay viable and functional for about 115 d in the circulatory system. Here, using a highly pure preparation of human mature erythrocytes, we demonstrate the presence of translation by polysome profiling, [35S]methionine labeling, and RiboPuromycylation. [35S]methionine labeling revealed that the translation in mature erythrocytes is about 10% of that observed in reticulocytes. We could observe polysomes by transmission electron microscopy in these cells. RNA-seq and quantitative real-time PCR performed on polysome fractions of these cells revealed that HBA (α-globin) and HBB (ß-globin) transcripts are translated. Using a luciferase-based reporter assay and mutational studies, we show that the sequence of the 5' untranslated region is crucial for the translation of these transcripts. Furthermore, mature erythrocytes showed reduced expression of globin proteins (α- and ß-) when treated with translation inhibitors. Overall, we provide multiple lines of evidence for translation of globin mRNAs in human mature erythrocytes.


Subject(s)
Erythrocytes , beta-Globins , 5' Untranslated Regions , Erythrocytes/metabolism , Humans , Methionine/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , alpha-Globins/metabolism , beta-Globins/genetics , beta-Globins/metabolism
7.
J Biol Chem ; 298(8): 102173, 2022 08.
Article in English | MEDLINE | ID: mdl-35752360

ABSTRACT

Stop codon readthrough (SCR) is the process of continuation of translation beyond the stop codon, generating protein isoforms with C-terminal extensions. SCR has been observed in viruses, fungi, and multicellular organisms, including mammals. However, SCR is largely unexplored in plants. In this study, we have analyzed ribosome profiling datasets to identify mRNAs that exhibit SCR in Arabidopsis thaliana. Analyses of the ribosome density, ribosome coverage, and three-nucleotide periodicity of the ribosome profiling reads in the mRNA region downstream of the stop codon provided strong evidence for SCR in mRNAs of 144 genes. We show that SCR generated putative evolutionarily conserved nuclear localization signals, transmembrane helices, and intrinsically disordered regions in the C-terminal extensions of several of these proteins. Furthermore, gene ontology functional enrichment analysis revealed that these 144 genes belong to three major functional groups-translation, photosynthesis, and abiotic stress tolerance. Using a luminescence-based readthrough assay, we experimentally demonstrated SCR in representative mRNAs belonging to each of these functional classes. Finally, using microscopy, we show that the SCR product of one gene that contains a nuclear localization signal at the C-terminal extension, CURT1B, localizes to the nucleus as predicted. Based on these observations, we propose that SCR plays an important role in plant physiology by regulating protein localization and function.


Subject(s)
Arabidopsis , Animals , Arabidopsis/genetics , Arabidopsis/metabolism , Codon, Terminator/metabolism , Mammals/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism
8.
Elife ; 112022 03 24.
Article in English | MEDLINE | ID: mdl-35323109

ABSTRACT

The SARS-CoV-2 non-structural protein 1 (Nsp1) contains an N-terminal domain and C-terminal helices connected by a short linker region. The C-terminal helices of Nsp1 (Nsp1-C-ter) from SARS-CoV-2 bind in the mRNA entry channel of the 40S ribosomal subunit and blocks mRNA entry, thereby shutting down host protein synthesis. Nsp1 suppresses host immune function and is vital for viral replication. Hence, Nsp1 appears to be an attractive target for therapeutics. In this study, we have in silico screened Food and Drug Administration (FDA)-approved drugs against Nsp1-C-ter. Among the top hits obtained, montelukast sodium hydrate binds to Nsp1 with a binding affinity (KD) of 10.8 ± 0.2 µM in vitro. It forms a stable complex with Nsp1-C-ter in simulation runs with -95.8 ± 13.3 kJ/mol binding energy. Montelukast sodium hydrate also rescues the inhibitory effect of Nsp1 in host protein synthesis, as demonstrated by the expression of firefly luciferase reporter gene in cells. Importantly, it shows antiviral activity against SARS-CoV-2 with reduced viral replication in HEK cells expressing ACE2 and Vero-E6 cells. We, therefore, propose montelukast sodium hydrate can be used as a lead molecule to design potent inhibitors to help combat SARS-CoV-2 infection.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Antiviral Agents/pharmacology , Drug Delivery Systems , Humans , Pharmaceutical Preparations , RNA, Messenger/metabolism , Viral Nonstructural Proteins/metabolism
9.
J Biol Chem ; 295(50): 17009-17026, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33028634

ABSTRACT

Stop codon read-through (SCR) is a process of continuation of translation beyond a stop codon. This phenomenon, which occurs only in certain mRNAs under specific conditions, leads to a longer isoform with properties different from that of the canonical isoform. MTCH2, which encodes a mitochondrial protein that regulates mitochondrial metabolism, was selected as a potential read-through candidate based on evolutionary conservation observed in the proximal region of its 3' UTR. Here, we demonstrate translational read-through across two evolutionarily conserved, in-frame stop codons of MTCH2 using luminescence- and fluorescence-based assays, and by analyzing ribosome-profiling and mass spectrometry (MS) data. This phenomenon generates two isoforms, MTCH2x and MTCH2xx (single- and double-SCR products, respectively), in addition to the canonical isoform MTCH2, from the same mRNA. Our experiments revealed that a cis-acting 12-nucleotide sequence in the proximal 3' UTR of MTCH2 is the necessary signal for SCR. Functional characterization showed that MTCH2 and MTCH2x were localized to mitochondria with a long t1/2 (>36 h). However, MTCH2xx was found predominantly in the cytoplasm. This mislocalization and its unique C terminus led to increased degradation, as shown by greatly reduced t1/2 (<1 h). MTCH2 read-through-deficient cells, generated using CRISPR-Cas9, showed increased MTCH2 expression and, consistent with this, decreased mitochondrial membrane potential. Thus, double-SCR of MTCH2 regulates its own expression levels contributing toward the maintenance of normal mitochondrial membrane potential.


Subject(s)
3' Untranslated Regions/genetics , Codon, Terminator/genetics , Membrane Potential, Mitochondrial , Mitochondrial Membrane Transport Proteins/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Animals , Aorta/cytology , Aorta/metabolism , Base Sequence , CRISPR-Cas Systems/genetics , Cattle , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , HEK293 Cells , Humans , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Protein Isoforms , Ribosomes/metabolism
10.
Biol Lett ; 16(10): 20200391, 2020 10.
Article in English | MEDLINE | ID: mdl-33050831

ABSTRACT

Naturally occurring fluorescence has been observed in multiple species ranging from bacteria to birds. In macroscopic animals such as birds, fluorescence provides a visual communication signal. However, the functional significance of this phenomenon is unknown in most cases. Though photoprotection is attributed to fluorescence under ultraviolet (UV) light in some organisms, it lacks direct experimental evidence. Here, we demonstrate naturally occurring fluorescence under UV light in a eutardigrade belonging to the genus Paramacrobiotus. Using a natural variant that lacks fluorescence, we show that the fluorescence confers tolerance to lethal UV radiation. Remarkably, the fluorescent extract from Paramacrobiotus sp. could protect the UV-sensitive tardigrade Hypsibius exemplaris and nematode Caenorhabditis elegans from germicidal UV radiation. We propose that Paramacrobiotus sp. possess a protective fluorescent shield that absorbs harmful UV radiation and emits harmless blue light.


Subject(s)
Tardigrada , Ultraviolet Rays , Animals , Fluorescence
11.
Biochemistry ; 59(1): 80-84, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31577420

ABSTRACT

Nonsense mutations that result in premature stop codons in the HBB gene cause ß-thalassemia. This disease is characterized by a reduced hemoglobin level due to the lack of ß-globin. Compounds that induce translational readthrough across the thalassemia-causing premature stop codon will have therapeutic benefits. Currently available molecules that induce translational readthrough lack specificity, and some of them show toxicity after prolonged use. In this study, we have developed an oligonucleotide-based approach to induce translational readthrough across the thalassemia-causing premature stop codon. Oligonucleotides that target HBB mRNA downstream of the premature stop codon could induce translational readthrough, generating a full-length ß-globin protein. We show this effect using fluorescence- and luminescence-based readthrough assays and by Western blot. Remarkably, the amount of oligonucleotide-induced translational readthrough product is comparable to that of the protein generated by normal translation when there was no premature stop codon. Thus, these oligonucleotides, with certain modifications, have the potential to be used as drugs for the treatment of ß-thalassemia. Also, this strategy can be extended to treat other genetic diseases caused by premature stop codons.


Subject(s)
Codon, Terminator/metabolism , Oligodeoxyribonucleotides/pharmacology , Oligonucleotides, Antisense/pharmacology , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , beta-Globins/genetics , HEK293 Cells , Humans , Thalassemia/genetics
12.
EMBO J ; 38(16): e100727, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31330067

ABSTRACT

Translational readthrough generates proteins with extended C-termini, which often possess distinct properties. Here, we have used various reporter assays to demonstrate translational readthrough of AGO1 mRNA. Analysis of ribosome profiling data and mass spectrometry data provided additional evidence for translational readthrough of AGO1. The endogenous readthrough product, Ago1x, could be detected by a specific antibody both in vitro and in vivo. This readthrough process is directed by a cis sequence downstream of the canonical AGO1 stop codon, which is sufficient to drive readthrough even in a heterologous context. This cis sequence has a let-7a miRNA-binding site, and readthrough is promoted by let-7a miRNA. Interestingly, Ago1x can load miRNAs on target mRNAs without causing post-transcriptional gene silencing, due to its inability to interact with GW182. Because of these properties, Ago1x can serve as a competitive inhibitor of miRNA pathway. In support of this, we observed increased global translation in cells overexpressing Ago1x. Overall, our results reveal a negative feedback loop in the miRNA pathway mediated by the translational readthrough product of AGO1.


Subject(s)
Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , MicroRNAs/genetics , Protein Biosynthesis , Argonaute Proteins/chemistry , Autoantigens/metabolism , Binding Sites , Codon, Terminator , Eukaryotic Initiation Factors/chemistry , Feedback, Physiological , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , RNA-Binding Proteins/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Signal Transduction
13.
J Biol Chem ; 293(49): 19148-19156, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30309984

ABSTRACT

About 1 billion years ago, in a single-celled holozoan ancestor of all animals, a gene fusion of two tRNA synthetases formed the bifunctional enzyme, glutamyl-prolyl-tRNA synthetase (EPRS). We propose here that a confluence of metabolic, biochemical, and environmental factors contributed to the specific fusion of glutamyl- (ERS) and prolyl- (PRS) tRNA synthetases. To test this idea, we developed a mathematical model that centers on the precursor-product relationship of glutamic acid and proline, as well as metabolic constraints on free glutamic acid availability near the time of the fusion event. Our findings indicate that proline content increased in the proteome during the emergence of animals, thereby increasing demand for free proline. Together, these constraints contributed to a marked cellular depletion of glutamic acid and its products, with potentially catastrophic consequences. In response, an ancient organism invented an elegant solution in which genes encoding ERS and PRS fused to form EPRS, forcing coexpression of the two enzymes and preventing lethal dysregulation. The substantial evolutionary advantage of this coregulatory mechanism is evidenced by the persistence of EPRS in nearly all extant animals.


Subject(s)
Amino Acyl-tRNA Synthetases/chemistry , Bacterial Proteins/chemistry , Evolution, Molecular , Models, Chemical , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Animals , Bacteria/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Citric Acid Cycle , Gene Fusion , Glutamate-tRNA Ligase/chemistry , Glutamate-tRNA Ligase/genetics , Glutamate-tRNA Ligase/metabolism , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Ketoglutaric Acids/chemistry , Ketoglutaric Acids/metabolism , Proline/chemistry , Proline/metabolism , Protein Biosynthesis/genetics
14.
J Mol Med (Berl) ; 96(11): 1177-1187, 2018 11.
Article in English | MEDLINE | ID: mdl-30155768

ABSTRACT

Endothelial cells respond to mechanical stimuli such as stretch. This property can be exploited with caution to induce angiogenesis which will have immense potential to treat pathological conditions associated with insufficient angiogenesis. The primary aim of this study is to test if low-pressure shock waves can be used to induce angiogenesis. Using a simple diaphragm-based shock tube, we demonstrate that a single pulse of low pressure (0.4 bar) shock wave is enough to induce proliferation in bovine aortic endothelial cells and human pulmonary microvascular endothelial cells. We show that this is associated with enhanced Ca++ influx and phosphorylation of phosphatidylinositol-3-kinase (PI3K) which is normally observed when endothelial cells are exposed to stretch. We also demonstrate the pro-angiogenic effect of shock waves of single pulse (per dose) using murine back punch wound model. Shock wave treated mice showed enhanced wound-induced angiogenesis as reflected by increased vascular area and vessel length. They also showed accelerated wound closure compared to control mice. Overall, our study shows that just a single pulse/shot (per dose) of shock waves can be used to induce angiogenesis. Importantly, we demonstrate this effect using a pulse of low-pressure shock waves (0.4 bar, in vitro and 0.15 bar, in vivo). KEY MESSAGES: Low-pressure single-pulse shock waves can induce endothelial cell migration and proliferation. This effect is endothelial cell specific. These shock waves enhance wound-induced angiogenesis in vivo. These shock waves can also accelerate wound healing in vivo.


Subject(s)
Extracorporeal Shockwave Therapy , Neovascularization, Physiologic , Animals , Cattle , Cell Movement , Cell Proliferation , Endothelial Cells/physiology , Female , Humans , Mice, Inbred BALB C
15.
Adv Mater ; 30(22): e1800429, 2018 May.
Article in English | MEDLINE | ID: mdl-29635828

ABSTRACT

Spatiotemporally controlled active manipulation of external micro-/nanoprobes inside living cells can lead to development of innovative biomedical technologies and inspire fundamental studies of various biophysical phenomena. Examples include gene silencing applications, real-time mechanical mapping of the intracellular environment, studying cellular response to local stress, and many more. Here, for the first time, cellular internalization and subsequent intracellular manipulation of a system of helical nanomotors driven by small rotating magnetic fields with no adverse effect on the cellular viability are demonstrated. This remote method of fuelling and guidance limits the effect of mechanical transduction to cells containing external probes, in contrast to ultrasonically or chemically powered techniques that perturb the entire experimental volume. The investigation comprises three cell types, containing both cancerous and noncancerous types, and is aimed toward analyzing and engineering the motion of helical propellers through the crowded intracellular space. The studies provide evidence for the strong anisotropy, heterogeneity, and spatiotemporal variability of the cellular interior, and confirm the suitability of helical magnetic nanoprobes as a promising tool for future cellular investigations and applications.


Subject(s)
Magnetics , Anisotropy , Magnetic Fields , Motion , Nanotechnology
16.
Chemistry ; 24(33): 8393-8403, 2018 Jun 12.
Article in English | MEDLINE | ID: mdl-29603822

ABSTRACT

Nanomaterials having enzyme-like activity (nanozymes) make them suitable candidates for various biomedical applications. In this study, we demonstrate the morphology-dependent enzyme mimetic activity of Mn3 O4 nanoparticles. It is found that Mn3 O4 nanoparticles mimic the functions of all three cellular antioxidant enzymes: superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). Interestingly, the nanozyme activity of Mn3 O4 depends on various factors including size, morphology, surface area, and the redox properties of the metal ions. The Mn3 O4 nanoflowers exhibited remarkably high activity in all three enzyme systems and the order of multienzyme activity of different morphologies was: flowers ≫ flakes > hexagonal plates≈polyhedrons≈cubes. Interestingly, all five nanoforms are taken up by the mammalian cells and were found to be biocompatible, with very low cytotoxicity. The activity of the most active nanoflowers was studied in primary human umbilical vein endothelial cells (HUVEC) and human pulmonary microvascular endothelial cells (hPMEC) and it was found that Mn3 O4 does not reduce the level of nitric oxide (NO). This is in contrast to the effect of some of the Mn-porphyrin-based SOD mimetics, which are known to scavenge NO in endothelial cells.


Subject(s)
Antioxidants/chemistry , Human Umbilical Vein Endothelial Cells/chemistry , Human Umbilical Vein Endothelial Cells/physiology , Manganese/chemistry , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide/chemistry , Superoxide Dismutase/chemistry , Animals , Catalase , Glutathione Peroxidase , Humans , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL