scholarly journals Inosine 5′-monophosphate dehydrogenase binds nucleic acids in vitro and in vivo

2004 ◽  
Vol 379 (2) ◽  
pp. 243-251 ◽  
Author(s):  
Jeremy E. McLEAN ◽  
Nobuko HAMAGUCHI ◽  
Peter BELENKY ◽  
Sarah E. MORTIMER ◽  
Martin STANTON ◽  
...  

Inosine 5´-monophosphate dehydrogenase (IMPDH) is the rate-limiting enzyme in the de novo biosynthesis of guanine nucleotides. In addition to the catalytic domain, IMPDH contains a subdomain of unknown function composed of two cystathione β-synthase domains. Our results, using three different assays, show that IMPDHs from Tritrichomonas foetus, Escherichia coli, and both human isoforms bind single-stranded nucleic acids with nanomolar affinity via the subdomain. Approx. 100 nucleotides are bound per IMPDH tetramer. Deletion of the subdomain decreases affinity 10-fold and decreases site size to 60 nucleotides, whereas substitution of conserved Arg/Lys residues in the subdomain with Glu decreases affinity by 20-fold. IMPDH is found in the nucleus of human cells, as might be expected for a nucleic-acid-binding protein. Lastly, immunoprecipitation experiments show that IMPDH binds both RNA and DNA in vivo. These experiments indicate that IMPDH has a previously unappreciated role in replication, transcription or translation that is mediated by the subdomain.

2005 ◽  
Vol 79 (4) ◽  
pp. 2171-2190 ◽  
Author(s):  
Almira Punjabi ◽  
Paula Traktman

ABSTRACT The vaccinia virus F10 protein is one of two virally encoded protein kinases. A phenotypic analysis of infections involving a tetracycline-inducible recombinant (vΔiF10) indicated that F10 is involved in the early stages of virion morphogenesis, as previously reported for the mutants ts28 and ts15. The proteins encoded by ts28 and ts15 have primary defects in enzymatic activity and thermostability, respectively. Using a transient complementation assay, we demonstrated that the enzymatic activity of F10 is essential for its biological function and that both its enzymatic and biological functions depend upon N-terminal sequences that precede the catalytic domain. An execution point analysis indicated that in addition to its role at the onset of morphogenesis, F10 is also required at later stages, when membrane crescents surround virosomal contents and develop into immature virions. The F10 protein is phosphorylated in vivo, appears to be tightly associated with intracellular membranes, and can bind to specific phosphoinositides in vitro. When F10 is repressed or impaired, the phosphorylation of several cellular and viral proteins appears to increase in intensity, suggesting that F10 may normally intersect with cellular signaling cascades via the activation of a phosphatase or the inhibition of another kinase. These cascades may drive the F10-induced remodeling of membranes that accompanies virion biogenesis. Upon the release of ts28-infected cultures from a 40°C-induced block, a synchronous resumption of morphogenesis that culminates in the production of infectious virus can be observed. The pharmacological agents H89 and cerulenin, which are inhibitors of endoplasmic reticulum exit site formation and de novo lipid synthesis, respectively, block this recovery.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3812-3812
Author(s):  
David A. Russler-Germain ◽  
David H Spencer ◽  
Margaret A. Young ◽  
Tamara Lamprecht ◽  
Chris Miller ◽  
...  

Abstract Mutations in DNMT3A (encoding one of two mammalian de novo DNA methyltransferases) are found in >30% of normal karyotype AML cases and correlate with poor clinical outcomes. Most DNMT3A mutations occur at position R882 within the catalytic domain (most commonly R882H) and are virtually always heterozygous. This over-representation suggests that mutations at R882 may result in gain-of-function or dominant-negative activity that contributes to leukemogenesis. However, how DNA methylation might be altered in DNMT3A-mutant cases of AML remains unclear, and no published study to date has addressed the effects of mixing wild-type (WT) and R882H DNMT3A. Importantly, mouse HSPCs deficient in Dnmt3a dramatically expand over time and have a concurrent defect in differentiation (Challen, GA et al. Nat Genet, 2011). Mice haploinsufficient for Dnmt3a, on the other hand, do not have a measurable defect in hematopoiesis. Collectively, these data suggest that the heterozygous R882 mutations probably cause more than a simple loss-of-function phenotype. We purified full-length, human WT and R882H DNMT3A using a mammalian tissue culture system to produce recombinant proteins for biochemical modeling of the de novo methylation potential of a DNMT3A-mutant AML cell. rhR882H DNMT3A exhibits roughly 10-20% of the de novo DNA methyltransferase activity of rhWT DNMT3A, similar to observations by other groups. We added increasing amounts of R882H DNMT3A to a fixed amount of WT DNMT3A and observed a linear increase in the net enzymatic activity, reflecting the summed activity of the two forms of DNMT3A in these 4-hour in vitro reactions. In contrast, 12-hour in vitro DNA methylation assays with mixed WT and R882H DNMT3A demonstrated net methylation less than the predicted summed activity of the two enzymes, suggesting that a dominant-negative effect of R882H DNMT3A may occur with a long equilibration time. To better simulate an AML cell with a heterozygous R882H mutation, we co-transfected HEK293T cells with equal amounts of poly-His-tagged WT and R882H DNMT3A expression vectors. Subsequently co-purified (i.e. in vivo-mixed) WT and R882H DNMT3A exhibited a striking reduction in methyltransferase activity, with total activity similar to R882H DNMT3A alone (Figure 1A). TSQ mass spectrometry allowed us to verify the presence and quantify the relative concentration of WT and R882H DNMT3A in our co-purified samples. We exploited a novel tryptic cleavage site in DNMT3A produced by the R882H mutation to generate standard concentration curves using recombinant peptides distinguishing the two protein forms. Our co-purified enzyme preparations had WT:R882H ratios ranging from 0.79 to 1.60; all demonstrated the dominant-negative effect of R882H. DNMT3A is a processive enzyme, catalyzing multiple methyl-group transfers before dissociating from target DNA. This is dependent on the ability of WT DNMT3A to form homo-oligomers (tetramers and larger), which was recently shown to be disrupted by the R882H mutation using the catalytic domain of DNMT3A produced in E.coli (Holz-Schietinger, C et al. JBC, 2012). We therefore postulated that the dominant-negative effect of R882H may be due to the disruption of WT DNMT3A oligomerization. Using a Superose 6 size exclusion column, we confirmed the tetramerization defect of R882H DNMT3A relative to WT DNMT3A. Notably, in vivo-mixed (co-purified) WT and R882H DNMT3A complexes exhibited a pattern of oligomerization identical to R882H DNMT3A alone. However, WT and R882H DNMT3A mixed in vitro exhibited a distribution of oligomers corresponding to the expected average of the WT and R882H curves (Figure 1B). These data demonstrate that production of equal amounts of WT and R882H DNMT3A within the same cell provides an environment where R882H DNMT3A can exert a potent dominant-negative effect on WT DNMT3A. Furthermore, our data suggest that this effect is associated with diminished formation of tetramers when WT and R882H DNMT3A are complexed together. Thus, the R882H mutation has two distinct consequences that affect DNMT3A activity in AML cells: 1) it severely reduces its own de novo methyltransferase activity, and 2) it disrupts the ability of WT DNMT3A to form functional tetramers. These two effects severely reduce total DNMT3A activity in AML cells, and may explain why this mutation is virtually always heterozygous in AML samples, since homozygosity would not further reduce DNMT3A activity. Disclosures: No relevant conflicts of interest to declare.


2005 ◽  
Vol 25 (14) ◽  
pp. 5947-5954 ◽  
Author(s):  
Violette Morales ◽  
Catherine Regnard ◽  
Annalisa Izzo ◽  
Irene Vetter ◽  
Peter B. Becker

ABSTRACT The male-specific-lethal (MSL) proteins in Drosophila melanogaster serve to adjust gene expression levels in male flies containing a single X chromosome to equal those in females with a double dose of X-linked genes. Together with noncoding roX RNA, MSL proteins form the “dosage compensation complex” (DCC), which interacts selectively with the X chromosome to restrict the transcription-activating histone H4 acetyltransferase MOF (males-absent-on-the-first) to that chromosome. We showed previously that MSL3 is essential for the activation of MOF's nucleosomal histone acetyltransferase activity within an MSL1-MOF complex. By characterizing the MSL3 domain structure and its associated functions, we now found that the nucleic acid binding determinants reside in the N terminus of MSL3, well separable from the C-terminal MRG signatures that form an integrated domain required for MSL1 interaction. Interaction with MSL1 mediates the activation of MOF in vitro and the targeting of MSL3 to the X-chromosomal territory in vivo. An N-terminal truncation that lacks the chromo-related domain and all nucleic acid binding activity is able to trigger de novo assembly of the DCC and establishment of an acetylated X-chromosome territory.


2008 ◽  
Vol 191 (3) ◽  
pp. 862-872 ◽  
Author(s):  
Meghna Mittal ◽  
Silvia Picossi ◽  
Abraham L. Sonenshein

ABSTRACT Citrate synthase, the first and rate-limiting enzyme of the tricarboxylic acid branch of the Krebs cycle, was shown to be required for de novo synthesis of glutamate and glutamine in Listeria monocytogenes. The citrate synthase (citZ) gene was found to be part of a complex operon with the upstream genes lmo1569 and lmo1568. The downstream isocitrate dehydrogenase (citC) gene appears to be part of the same operon as well. Two promoters were shown to drive citZ expression, a distal promoter located upstream of lmo1569 and a proximal promoter located upstream of the lmo1568 gene. Transcription of citZ from both promoters was regulated by CcpC by interaction with a single site; assays of transcription in vivo and assays of CcpC binding in vitro revealed that CcpC interacts with and represses the proximal promoter that drives expression of the lmo1568, citZ, and citC genes and, by binding to the same site, prevents read-through transcription from the distal, lmo1569 promoter. Expression of the lmo1568 operon was not affected by the carbon source but was repressed during growth in complex medium by addition of glutamine.


2020 ◽  
Vol 20 (11) ◽  
pp. 821-830
Author(s):  
Prasad Pofali ◽  
Adrita Mondal ◽  
Vaishali Londhe

Background: Current gene therapy vectors such as viral, non-viral, and bacterial vectors, which are used for cancer treatment, but there are certain safety concerns and stability issues of these conventional vectors. Exosomes are the vesicles of size 40-100 nm secreted from multivesicular bodies into the extracellular environment by most of the cell types in-vivo and in-vitro. As a natural nanocarrier, exosomes are immunologically inert, biocompatible, and can cross biological barriers like the blood-brain barrier, intestinal barrier, and placental barrier. Objective: This review focusses on the role of exosome as a carrier to efficiently deliver a gene for cancer treatment and diagnosis. The methods for loading of nucleic acids onto the exosomes, advantages of exosomes as a smart intercellular shuttle for gene delivery and therapeutic applications as a gene delivery vector for siRNA, miRNA and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and also the limitations of exosomes as a gene carrier are all reviewed in this article. Methods: Mostly, electroporation and chemical transfection are used to prepare gene loaded exosomes. Results: Exosome-mediated delivery is highly promising and advantageous in comparison to the current delivery methods for systemic gene therapy. Targeted exosomes, loaded with therapeutic nucleic acids, can efficiently promote the reduction of tumor proliferation without any adverse effects. Conclusion: In the near future, exosomes can become an efficient gene carrier for delivery and a biomarker for the diagnosis and treatment of cancer.


2020 ◽  
Vol 0 (0) ◽  
Author(s):  
Moein Dehbashi ◽  
Zohreh Hojati ◽  
Majid Motovali-bashi ◽  
Mazdak Ganjalikhani-Hakemi ◽  
Akihiro Shimosaka ◽  
...  

AbstractCancer recurrence presents a huge challenge in cancer patient management. Immune escape is a key mechanism of cancer progression and metastatic dissemination. CD25 is expressed in regulatory T (Treg) cells including tumor-infiltrating Treg cells (TI-Tregs). These cells specially activate and reinforce immune escape mechanism of cancers. The suppression of CD25/IL-2 interaction would be useful against Treg cells activation and ultimately immune escape of cancer. Here, software, web servers and databases were used, at which in silico designed small interfering RNAs (siRNAs), de novo designed peptides and virtual screened small molecules against CD25 were introduced for the prospect of eliminating cancer immune escape and obtaining successful treatment. We obtained siRNAs with low off-target effects. Further, small molecules based on the binding homology search in ligand and receptor similarity were introduced. Finally, the critical amino acids on CD25 were targeted by a de novo designed peptide with disulfide bond. Hence we introduced computational-based antagonists to lay a foundation for further in vitro and in vivo studies.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii98-ii98
Author(s):  
Anne Marie Barrette ◽  
Alexandros Bouras ◽  
German Nudelman ◽  
Zarmeen Mussa ◽  
Elena Zaslavsky ◽  
...  

Abstract Glioblastoma (GBM) remains an incurable disease, in large part due to its malignant infiltrative spread, and current clinical therapy fails to target the invasive nature of tumor cells in disease progression and recurrence. Here, we use the YAP-TEAD inhibitor Verteporfin to target a convergence point for regulating tumor invasion/metastasis and establish the robust anti-invasive therapeutic efficacy of this FDA-approved drug and its survival benefit across several preclinical glioma models. Using patient-derived GBM cells and orthotopic xenograft models (PDX), we show that Verteporfin treatment disrupts YAP/TAZ-TEAD activity and processes related to cell adhesion, migration and epithelial-mesenchymal transition. In-vitro, Verteporfin impairs tumor migration, invasion and motility dynamics. In-vivo, intraperitoneal administration of Verteporfin in mice with orthotopic PDX tumors shows consistent drug accumulation within the brain and decreased infiltrative tumor burden, across three independent experiments. Interestingly, PDX tumors with impaired invasion after Verteporfin treatment downregulate CDH2 and ITGB1 adhesion protein levels within the tumor microenvironment. Finally, Verteporfin treatment confers survival benefit in two independent PDX models: as monotherapy in de-novo GBM and in combination with standard-of-care chemoradiation in recurrent GBM. These findings indicate potential therapeutic value of this FDA-approved drug if repurposed for GBM patients.


2020 ◽  
Vol 11 (7) ◽  
Author(s):  
Nan Huang ◽  
Chang Xu ◽  
Liang Deng ◽  
Xue Li ◽  
Zhixuan Bian ◽  
...  

AbstractPhosphoribosylaminoimidazole carboxylase, phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS), an essential enzyme involved in de novo purine biosynthesis, is connected with formation of various tumors. However, the specific biological roles and related mechanisms of PAICS in gastric cancer (GC) remain unclear. In the present study, we identified for the first time that PAICS was significantly upregulated in GC and high expression of PAICS was correlated with poor prognosis of patients with GC. In addition, knockdown of PAICS significantly induced cell apoptosis, and inhibited GC cell growth both in vitro and in vivo. Mechanistic studies first found that PAICS was engaged in DNA damage response, and knockdown of PAICS in GC cell lines induced DNA damage and impaired DNA damage repair efficiency. Further explorations revealed that PAICS interacted with histone deacetylase HDAC1 and HDAC2, and PAICS deficiency decreased the expression of DAD51 and inhibited its recruitment to DNA damage sites by impairing HDAC1/2 deacetylase activity, eventually preventing DNA damage repair. Consistently, PAICS deficiency enhanced the sensitivity of GC cells to DNA damage agent, cisplatin (CDDP), both in vitro and in vivo. Altogether, our findings demonstrate that PAICS plays an oncogenic role in GC, which act as a novel diagnosis and prognostic biomarker for patients with GC.


Microbiology ◽  
2014 ◽  
Vol 160 (10) ◽  
pp. 2157-2169 ◽  
Author(s):  
Sudarson Sundarrajan ◽  
Junjappa Raghupatil ◽  
Aradhana Vipra ◽  
Nagalakshmi Narasimhaswamy ◽  
Sanjeev Saravanan ◽  
...  

P128 is an anti-staphylococcal protein consisting of the Staphylococcus aureus phage-K-derived tail-associated muralytic enzyme (TAME) catalytic domain (Lys16) fused with the cell-wall-binding SH3b domain of lysostaphin. In order to understand the mechanism of action and emergence of resistance to P128, we isolated mutants of Staphylococcus spp., including meticillin-resistant Staphylococcus aureus (MRSA), resistant to P128. In addition to P128, the mutants also showed resistance to Lys16, the catalytic domain of P128. The mutants showed loss of fitness as shown by reduced rate of growth in vitro. One of the mutants tested was found to show reduced virulence in animal models of S. aureus septicaemia suggesting loss of fitness in vivo as well. Analysis of the antibiotic sensitivity pattern showed that the mutants derived from MRSA strains had become sensitive to meticillin and other β-lactams. Interestingly, the mutant cells were resistant to the lytic action of phage K, although the phage was able to adsorb to these cells. Sequencing of the femA gene of three P128-resistant mutants showed either a truncation or deletion in femA, suggesting that improper cross-bridge formation in S. aureus could be causing resistance to P128. Using glutathione S-transferase (GST) fusion peptides as substrates it was found that both P128 and Lys16 were capable of cleaving a pentaglycine sequence, suggesting that P128 might be killing S. aureus by cleaving the pentaglycine cross-bridge of peptidoglycan. Moreover, peptides corresponding to the reported cross-bridge of Staphylococcus haemolyticus (GGSGG, AGSGG), which were not cleaved by lysostaphin, were cleaved efficiently by P128. This was also reflected in high sensitivity of S. haemolyticus to P128. This showed that in spite of sharing a common mechanism of action with lysostaphin, P128 has unique properties, which allow it to act on certain lysostaphin-resistant Staphylococcus strains.


e-Neuroforum ◽  
2007 ◽  
Vol 13 (4) ◽  
Author(s):  
Lars Fester ◽  
Janine Prange-Kiel ◽  
Gabriele M. Rune

ZusammenfassungUnsere Untersuchungen der letzten Jahre haben gezeigt, dass nicht das Ovar die Quelle für Estrogen induzierte synaptische Plastizität im Hippokampus ist, sondern dieses aus dem Hippokampus selber stammt und haben damit einen Paradigmawechsel eingeleitet, der Estrogen als Neuromodulator unabhängig vom Geschlecht identifiziert. Hippokampale Neurone von Ratten beiderlei Geschlechts sind in der Lage, aus Cholesterol Estrogene de novo zu synthetisieren. Diese hippokampale Estrogensynthese ist sowohl für den Erhalt von Spinesynapsen in vivo als auch in vitro essenziell. Die Hemmung der Estrogensynthese zieht einen Synapsenverlust nach sich und Langzeitpotenzierung ist nicht mehr induzierbar. Die Effekte von hippokampalem Estrogen sind auto-/parakriner Natur, die über die bekannten Estrogenrezeptor-Subtypen, ERα und ERβ, vermittelt werden. Die Regulation der hippokampalen Estrogensynthese erfolgt über GnRH und erklärt die Korrelation der Spinesynapsendichte mit dem weiblichen genitalen Zyklus, die für den Hippokampus spezifisch ist.


Sign in / Sign up

Export Citation Format

Share Document