Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Bioinformatics ; 36(12): 3922-3924, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32289149

ABSTRACT

MOTIVATION: Signaling pathways capable of switching between two states are ubiquitous within living organisms. They provide the cells with the means to produce reversible or irreversible decisions. Switch-like behavior of biological systems is realized through biochemical reaction networks capable of having two or more distinct steady states, which are dependent on initial conditions. Investigation of whether a certain signaling pathway can confer bistability involves a substantial amount of hypothesis testing. The cost of direct experimental testing can be prohibitive. Therefore, constraining the hypothesis space is highly beneficial. One such methodology is based on chemical reaction network theory (CRNT), which uses computational techniques to rule out pathways that are not capable of bistability regardless of kinetic constant values and molecule concentrations. Although useful, these methods are complicated from both pure and computational mathematics perspectives. Thus, their adoption is very limited amongst biologists. RESULTS: We brought CRNT approaches closer to experimental biologists by automating all the necessary steps in CRNT4SMBL. The input is based on systems biology markup language (SBML) format, which is the community standard for biological pathway communication. The tool parses SBML and derives C-graph representations of the biological pathway with mass action kinetics. Next steps involve an efficient search for potential saddle-node bifurcation points using an optimization technique. This type of bifurcation is important as it has the potential of acting as a switching point between two steady states. Finally, if any bifurcation points are present, continuation analysis with respect to a user-defined parameter extends the steady state branches and generates a bifurcation diagram. Presence of an S-shaped bifurcation diagram indicates that the pathway acts as a bistable switch for the given optimization parameters. AVAILABILITY AND IMPLEMENTATION: CRNT4SBML is available via the Python Package Index. The documentation can be found at https://crnt4sbml.readthedocs.io. CRNT4SBML is licensed under the Apache Software License 2.0.


Subject(s)
Models, Biological , Systems Biology , Kinetics , Signal Transduction , Software
2.
Viruses ; 12(2)2020 02 02.
Article in English | MEDLINE | ID: mdl-32024281

ABSTRACT

Although seasonal influenza vaccines block most predominant influenza types and subtypes, humans still remain vulnerable to waves of seasonal and new potential pandemic influenza viruses for which no immunity may exist because of viral antigenic drift and/or shift. Previously, we described a human monoclonal antibody (hMAb), KPF1, which was produced in human embryonic kidney 293T cells (KPF1-HEK) with broad and potent neutralizing activity against H1N1 influenza A viruses (IAV) in vitro, and prophylactic and therapeutic activities in vivo. In this study, we produced hMAb KPF1 in tobacco plants (KPF1-Antx) and demonstrated how the plant-produced KPF1-Antx hMAb possesses similar biological activity compared with the mammalian-produced KPF1-HEK hMAb. KPF1-Antx hMAb showed broad binding to recombinant HA proteins and H1N1 IAV, including A/California/04/2009 (pH1N1) in vitro, which was comparable to that observed with KPF1-HEK hMAb. Importantly, prophylactic administration of KPF1-Antx hMAb to guinea pigs prevented pH1N1 infection and transmission in both prophylactic and therapeutic experiments, substantiating its clinical potential to prevent and treat H1N1 infections. Collectively, this study demonstrated, for the first time, a plant-produced influenza hMAb with in vitro and in vivo activity against influenza virus. Because of the many advantages of plant-produced hMAbs, such as rapid batch production, low cost, and the absence of mammalian cell products, they represent an alternative strategy for the production of immunotherapeutics for the treatment of influenza viral infections, including emerging seasonal and/or pandemic strains.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/therapeutic use , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/transmission , Plantibodies/therapeutic use , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/biosynthesis , Female , Guinea Pigs , Orthomyxoviridae Infections/therapy , Nicotiana/metabolism
3.
Biosci Rep ; 35(3)2015 May 19.
Article in English | MEDLINE | ID: mdl-26182383

ABSTRACT

The ATP demand required for muscle development is accommodated by elevations in mitochondrial biogenesis, through the co-ordinated activities of the nuclear and mitochondrial genomes. The most important transcriptional activator of the mitochondrial genome is mitochondrial transcription factor A (Tfam); however, the regulation of Tfam expression during muscle differentiation is not known. Thus, we measured Tfam mRNA levels, mRNA stability, protein expression and localization and Tfam transcription during the progression of muscle differentiation. Parallel 2-fold increases in Tfam protein and mRNA were observed, corresponding with 2-3-fold increases in mitochondrial content. Transcriptional activity of a 2051 bp promoter increased during this differentiation period and this was accompanied by a 3-fold greater Tfam mRNA stabilization. Interestingly, truncations of the promoter at 1706 bp, 978 bp and 393 bp promoter all exhibited 2-3-fold higher transcriptional activity than the 2051 bp construct, indicating the presence of negative regulatory elements within the distal 350 bp of the promoter. Activation of AMP kinase augmented Tfam transcription within the proximal promoter, suggesting the presence of binding sites for transcription factors that are responsive to cellular energy state. During differentiation, the accumulating Tfam protein was progressively distributed to the mitochondrial matrix where it augmented the expression of mtDNA and COX (cytochrome c oxidase) subunit I, an mtDNA gene product. Our data suggest that, during muscle differentiation, Tfam protein levels are regulated by the availability of Tfam mRNA, which is controlled by both transcription and mRNA stability. Changes in energy state and Tfam localization also affect Tfam expression and action in differentiating myotubes.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/cytology , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Binding Sites , Cell Differentiation , Cell Line , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Gene Expression Regulation , Mice , Mitochondria, Muscle/drug effects , Promoter Regions, Genetic , RNA Stability , Ribonucleotides/pharmacology
4.
Am J Physiol Regul Integr Comp Physiol ; 303(4): R408-17, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22718808

ABSTRACT

A change in mRNA stability alters the abundance of mRNA available for translation and is emerging as a critical pathway influencing gene expression. Variations in the stability of functional and regulatory mitochondrial proteins may contribute to the divergent mitochondrial densities observed in striated muscle. Thus we hypothesized that the stability of mRNAs encoding for regulatory nuclear and mitochondrial transcription factors would be inversely proportional to muscle oxidative capacity and would be facilitated by the activity of RNA binding proteins (RBPs). The stability of mitochondrial transcription factor A (Tfam), peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), and nuclear respiratory factor 2α (NRF-2α) mRNA was assessed in striated muscles with distinct oxidative capacities using in vitro decay assays. All three mitochondrial regulators were rapidly degraded in cardiac and slow-twitch red (STR) muscle, resulting in a ∼60-65% lower (P < 0.05) mRNA half-life (t(1/2)) compared with fast-twitch white (FTW) fibers. This accelerated rate of Tfam mRNA decay was matched by a 2.5-fold increase in Tfam transcription in slow- compared with fast-twitch muscle (P = 0.05). Protein expression of four unique RBPs [AU-rich binding factor 1 (AUF1), human antigen R (HuR), KH-homology splicing regulatory protein (KSRP), and CUG binding protein 1 (CUGBP1)] believed to modulate mRNA stability was elevated in cardiac and STR muscles (P < 0.05) and was moderately associated with the decay of Tfam, PGC-1α, and NRF-2α mRNA. Variable rates of transcript degradation were apparent when comparing all transcripts within the same muscle type. Thus the distribution of RBPs appears to follow a fiber-type specific pattern and subsequently functions to alter the stability of specific mitochondrial regulators in a transcript- and tissue-specific fashion.


Subject(s)
Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , RNA Stability/physiology , RNA, Messenger/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electron Transport Complex IV/metabolism , GA-Binding Protein Transcription Factor/genetics , GA-Binding Protein Transcription Factor/metabolism , Male , Mitochondria, Muscle/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Virology ; 391(1): 90-8, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19541337

ABSTRACT

The conserved region 3 (CR3) portion of the human adenovirus (HAdV) 5 E1A protein functions as a potent transcriptional activator that induces expression of viral early genes during infection. Expression of HAdV-5 CR3 in the yeast Saccharomyces cerevisiae inhibits growth, as do the corresponding regions of the HAdV-3, 4, 9, 12 and 40 E1A proteins, which represent the remaining five HAdV subgroups. Growth inhibition is alleviated by disruption of the SAGA transcriptional regulatory complex, suggesting that CR3 targets the yeast SAGA complex. In yeast, transcriptional activation by several, but not all, of the CR3 regions requires the Gcn5 acetyltransferase component of SAGA. The CR3 regions of HAdV-3, 5, 9 and 40, but not HAdV-4 and 12 interact with the pCAF acetyltransferase, a mammalian ortholog of yeast Gcn5. Disruption of the previously described N-terminal pCAF binding site abrogates binding by the HAdV-5 243R E1A protein, but not the larger 289R E1A protein, which is otherwise identical except for the presence of CR3. RNA interference directed against pCAF decreased HAdV-5 CR3 dependent transcriptional activation in mammalian cells. Our results identify a second independent binding site for pCAF in E1A and suggest that it contributes to CR3 dependent transcriptional activation.


Subject(s)
Adenovirus E1A Proteins/metabolism , Adenoviruses, Human/genetics , p300-CBP Transcription Factors/metabolism , Adenovirus E1A Proteins/genetics , Adenoviruses, Human/metabolism , Binding Sites , Conserved Sequence , HeLa Cells , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Activation
6.
J Biol Chem ; 283(17): 11355-63, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18285338

ABSTRACT

Histone deacetylases (HDACs) are protein deacetylases that play a role in repression of gene transcription and are emerging targets in cancer therapy. Here, we characterize the structure and enzymatic activity of the catalytic domain of human HDAC7 (cdHDAC7). Although HDAC7 normally exists as part of a multiprotein complex, we show that cdHDAC7 has a low level of deacetylase activity which can be inhibited by known HDAC inhibitors. The crystal structures of human cdHDAC7 and its complexes with two hydroxamate inhibitors are the first structures of the catalytic domain of class IIa HDACs and demonstrate significant differences with previously reported class I and class IIb-like HDAC structures. We show that cdHDAC7 has an additional class IIa HDAC-specific zinc binding motif adjacent to the active site which is likely to participate in substrate recognition and protein-protein interaction and may provide a site for modulation of activity. Furthermore, a different active site topology results in modified catalytic properties and in an enlarged active site pocket. Our studies provide mechanistic insights into class IIa HDACs and facilitate the design of specific modulators.


Subject(s)
Gene Expression Regulation, Enzymologic , Histone Deacetylases/metabolism , Amino Acid Motifs , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Histone Deacetylases/chemistry , Histone Deacetylases/physiology , Humans , Kinetics , Ligands , Models, Biological , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Zinc/chemistry
7.
EMBO J ; 25(12): 2710-22, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16763564

ABSTRACT

We have determined distinct roles for different proteasome complexes in adenovirus (Ad) E1A-dependent transcription. We show that the 19S ATPase, S8, as a component of 19S ATPase proteins independent of 20S (APIS), binds specifically to the E1A transactivation domain, conserved region 3 (CR3). Recruitment of APIS to CR3 enhances the ability of E1A to stimulate transcription from viral early gene promoters during Ad infection of human cells. The ability of CR3 to stimulate transcription in yeast is similarly dependent on the functional integrity of yeast APIS components, Sug1 and Sug2. The 20S proteasome is also recruited to CR3 independently of APIS and the 26S proteasome. Chromatin immunoprecipitation reveals that E1A, S8 and the 20S proteasome are recruited to both Ad early region gene promoters and early region gene sequences during Ad infection, suggesting their requirement in both transcriptional initiation and elongation. We also demonstrate that E1A CR3 transactivation and degradation sequences functionally overlap and that proteasome inhibitors repress E1A transcription. Taken together, these data demonstrate distinct roles for APIS and the 20S proteasome in E1A-dependent transactivation.


Subject(s)
Adenosine Triphosphatases/metabolism , Adenovirus E1A Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription, Genetic , Adenoviridae/physiology , HCT116 Cells , Half-Life , Humans , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Thermodynamics , Transcriptional Activation/genetics
8.
Proc Natl Acad Sci U S A ; 102(18): 6267-72, 2005 May 03.
Article in English | MEDLINE | ID: mdl-15849266

ABSTRACT

Unliganded thyroid hormone (TH) receptors (TRs) and other nuclear receptors (NRs) repress transcription of hormone-activated genes by recruiting corepressors (CoRs), such as NR CoR (N-CoR) and SMRT. Unliganded TRs also activate transcription of TH-repressed genes. Some evidence suggests that these effects also involve TR/CoR contacts; however, the precise reasons that CoRs activate transcription in these contexts are obscure. Unraveling these mechanisms is complicated by the fact that it is difficult to decipher direct vs. indirect effects of TR-coregulator contacts in mammalian cells. In this study, we used yeast, Saccharomyces cerevisiae, which lack endogenous NRs and NR coregulators, to determine how unliganded TRs can activate transcription. We previously showed that adenovirus 5 early-region 1A coactivates unliganded TRs in yeast, and that these effects are blocked by TH. We show here that human adenovirus type 5 early region 1A (E1A) contains a short peptide (LDQLIEEVL amino acids 20-28) that resembles CoR-NR interaction motifs (CoRNR boxes), and that this motif is required for TR binding and coactivation. Although full-length N-CoR does not coactivate TR in yeast, a naturally occurring N-CoR variant (N-CoR(I)) and an artificial N-CoR truncation (N-CoR(C)) that retain CoRNR boxes but lack N-terminal repressor domains behave as potent and direct TH-repressed coactivators for unliganded TRs. We conclude that E1A and N-CoR(I) are naturally occurring TR coactivators that bind in the typical CoR mode and suggest that similar factors could mediate transcriptional activation by unliganded TRs in mammals.


Subject(s)
Adenovirus E1A Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Thyroid Hormone/metabolism , Repressor Proteins/metabolism , Transcriptional Activation , Amino Acid Motifs/genetics , Glutathione Transferase , Nuclear Receptor Co-Repressor 1 , Saccharomyces cerevisiae , Two-Hybrid System Techniques
9.
Virology ; 316(1): 75-83, 2003 Nov 10.
Article in English | MEDLINE | ID: mdl-14599792

ABSTRACT

The N-terminal/conserved region 1 (CR1) portion of the human adenovirus (Ad) 5 E1A protein was previously shown to inhibit growth in the simple eukaryote Saccharomyces cerevisiae. We now demonstrate that the corresponding regions of the E1A proteins of Ad3,-4,-9,-12, and -40, which represent the remaining five Ad subgroups, also inhibit yeast growth. These results suggest that the E1A proteins of all six human Ad subgroups share a common cellular target(s) conserved in yeast. Growth inhibition induced by either full-length or the N-terminal/CR1 portion of Ad5 E1A was relieved by coexpression of the E1A binding portions of the mammalian p300, CBP, and pCAF acetyltransferases. Similarly, growth inhibition by the N-terminal/CR1 portions of the other Ad E1A proteins was suppressed by expression of the same regions of CBP or pCAF known to bind Ad5 E1A. The physical interaction of each of the different Ad E1A proteins with CBP, p300, and pCAF was confirmed in vitro. Furthermore, deletion of the gene encoding yGcn5, the yeast homolog of pCAF and a subunit of the SAGA transcriptional regulatory complex, restored growth in yeast expressing each of the different Ad E1A proteins. This indicates that the SAGA complex is a conserved target of all Ad E1A proteins. Our results demonstrate for the first time that the p300, CBP, and pCAF acetyltransferases are common targets for the E1A proteins of all six human Ad subgroups, highlighting the importance of these interactions for E1A function.


Subject(s)
Acetyltransferases/metabolism , Adenovirus E1A Proteins/metabolism , Adenoviruses, Human , Gene Expression Regulation , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Acetyltransferases/genetics , Adenoviruses, Human/metabolism , Adenoviruses, Human/pathogenicity , CREB-Binding Protein , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Histone Acetyltransferases , Nuclear Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Trans-Activators/genetics
10.
Mol Endocrinol ; 17(6): 1095-105, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12637585

ABSTRACT

In mammalian cells, the human adenovirus type 5 early region 1A (E1A) oncoprotein functions as a thyroid hormone (TH)-dependent activator of the thyroid hormone receptor (TR). Interestingly, in the cellular context of the yeast Saccharomyces cerevisiae, E1A acts as a TR-specific constitutive coactivator that is down-regulated by TH. TH reduces the interaction of E1A with the TR in yeast but not HeLa cells. The N-terminal 82 amino acids of E1A are sufficient for coactivation in yeast and residues 4-29 are essential. In yeast, expression of the nuclear receptor corepressor (N-CoR) could down-regulate constitutive transcriptional activation of the TR by E1A, whereas expression of the glucocorticoid receptor interacting protein 1 (GRIP-1) coactivator reconstituted the E1A-induced pattern of enhanced TH-dependent gene activation by TR observed in mammalian cells. We further show that the mating type switching gene (SWI)/sucrose nonfermenting (SNF) gene chromatin remodeling complex is required for both TH/GRIP-1- and E1A-dependent coactivator function, whereas the general control nonrepressed protein (GCN5)/alteration/deficiency in activation protein (ADA2) components of the SPT, ADA, GCN5, acetylation (SAGA) transcriptional adaptor complex are required for TH/GRIP-1, but not E1A-dependent activation of the TR. Taken together, these studies demonstrate that the novel TR-specific coactivator function of E1A in yeast depends on the SWI/SNF chromatin remodeling complex and can be further influenced by changes in the cellular complement of transcriptional coregulatory proteins.


Subject(s)
Adenovirus E1A Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation/physiology , Nuclear Proteins/metabolism , Receptors, Thyroid Hormone/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Histone Acetyltransferases , Humans , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Coactivator 2 , Protein Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Transcriptional Activation
11.
J Biol Chem ; 277(34): 30844-51, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12070146

ABSTRACT

Expression of the adenovirus E1A protein in the simple eukaryote Saccharomyces cerevisiae inhibits growth. We tested four regions of E1A that alter growth and transcription in mammalian cells for their effects in yeast when expressed as fusions to the Gal4p DNA binding domain. Expression of the N-terminal/conserved region (CR) 1 or CR3, but not of the CR2 or the C-terminal portion of E1A, inhibited yeast growth. Growth inhibition was relieved by deletion of the genes encoding the yGcn5p, Ngg1p, or Spt7p components of the SAGA transcriptional regulatory complex, but not the Ahc1p component of the related ADA complex, indicating that the N-terminal/CR1 and CR3 regions of E1A target the SAGA complex independently. Expression of the pCAF acetyltransferase, a mammalian homologue of yGcn5p, also suppressed growth inhibition by either portion of E1A. Furthermore, the N-terminal 29 residues and the CR3 portion of E1A interacted independently with yGcn5p and pCAF in vitro. Thus, two separate regions of E1A target the yGcn5p component of the SAGA transcriptional activation complex. A subregion of the N-terminal/CR1 fragment spanning residues 30-69 within CR1 also inhibited yeast growth in a SAGA-dependent fashion. However, this region did not interact with yGcn5p or pCAF, suggesting that it makes a third contact with another SAGA component. Our results provide a new model system to elucidate mechanisms by which E1A and the SAGA complex regulate transcription and growth.


Subject(s)
Acetyltransferases/physiology , Adenovirus E1A Proteins/physiology , DNA-Binding Proteins , Fungal Proteins/physiology , Multienzyme Complexes/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/growth & development , Trans-Activators/physiology , Transcription Factors/physiology , Acetyltransferases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Adenovirus E1A Proteins/chemistry , Histone Acetyltransferases , Protein Kinases/physiology
12.
J Gen Virol ; 83(Pt 3): 517-524, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11842246

ABSTRACT

Adenovirus type 5 E1A proteins interact with cellular regulators of transcription to reprogram gene expression in the infected or transformed cell. Although E1A also interacts with DNA directly in vitro, it is not clear how this relates to its function in vivo. The N-terminal conserved regions 1, 2 and 3 and the C-terminal portions of E1A were prepared as purified recombinant proteins and analyses showed that only the C-terminal region bound DNA in vitro. Deletion of E1A amino acids 201-220 inhibited binding and a minimal fragment encompassing amino acids 201-218 of E1A was sufficient for binding single- and double-stranded DNA. This portion of E1A also bound the cation-exchange resins cellulose phosphate and carboxymethyl Sepharose. As this region contains six basic amino acids, in vitro binding of E1A to DNA probably results from an ionic interaction with the phosphodiester backbone of DNA. Studies in Saccharomyces cerevisiae have shown that expression of a strong transcriptional activation domain fused to a DNA-binding domain can inhibit growth. Although fusion of the C-terminal region of E1A to a strong transcriptional activation domain inhibited growth when expressed in yeast, this was not mediated by the DNA-binding domain identified in vitro. These data suggest that E1A does not bind DNA in vivo.


Subject(s)
Adenovirus E1A Proteins/metabolism , Adenoviruses, Human , Cellulose/analogs & derivatives , DNA-Binding Proteins/metabolism , DNA/metabolism , Adenovirus E1A Proteins/chemistry , Adenovirus E1A Proteins/genetics , Binding Sites , Cell Division , Cellulose/metabolism , Chromatography, Affinity , Chromatography, Ion Exchange , DNA/genetics , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Mutation/genetics , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sepharose/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL