49 results on '"Samir M. Hamdan"'
Search Results
2. Quick and Easy Assembly of a One-Step qRT-PCR Kit for COVID-19 Diagnostics Using In-House Enzymes
- Author
-
Masateru Takahashi, Muhammad Tehseen, Rahul Salunke, Etsuko Takahashi, Sara Mfarrej, Mohamed A. Sobhy, Fatimah S. Alhamlan, Sharif Hala, Gerardo Ramos-Mandujano, Ahmed A. Al-Qahtani, Fadwa S. Alofi, Afrah Alsomali, Anwar M. Hashem, Asim Khogeer, Naif A. M. Almontashiri, Jae Man Lee, Hiroaki Mon, Kosuke Sakashita, Mo Li, Takahiro Kusakabe, Arnab Pain, and Samir M. Hamdan
- Subjects
Chemistry ,QD1-999 - Published
- 2021
- Full Text
- View/download PDF
3. Cryo-EM structure of human Pol κ bound to DNA and mono-ubiquitylated PCNA
- Author
-
Vlad-Stefan Raducanu, Alfredo De Biasio, Souvika Bakshi, Ramon Crehuet, Masateru Takahashi, Matthew Percival, Samir M. Hamdan, Timothy J. Ragan, Frederick W. Muskett, Mohamed Abdelmaboud Sobhy, Muhammad Tehseen, Claudia Lancey, Kerry Blair, and Ministerio de Economía y Competitividad (España)
- Subjects
Ubiquitin binding ,DNA polymerase ,DNA damage ,viruses ,Science ,General Physics and Astronomy ,DNA-Directed DNA Polymerase ,Protomer ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Ubiquitin ,Cryoelectron microscopy ,Proliferating Cell Nuclear Antigen ,Humans ,Polymerase ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,Ubiquitination ,General Chemistry ,DNA ,Proliferating cell nuclear antigen ,chemistry ,biology.protein ,Biophysics ,030217 neurology & neurosurgery ,DNA Damage ,Protein Binding - Abstract
Y-family DNA polymerase κ (Pol κ) can replicate damaged DNA templates to rescue stalled replication forks. Access of Pol κ to DNA damage sites is facilitated by its interaction with the processivity clamp PCNA and is regulated by PCNA mono-ubiquitylation. Here, we present cryo-EM reconstructions of human Pol κ bound to DNA, an incoming nucleotide, and wild type or mono-ubiquitylated PCNA (Ub-PCNA). In both reconstructions, the internal PIP-box adjacent to the Pol κ Polymerase-Associated Domain (PAD) docks the catalytic core to one PCNA protomer in an angled orientation, bending the DNA exiting the Pol κ active site through PCNA, while Pol κ C-terminal domain containing two Ubiquitin Binding Zinc Fingers (UBZs) is invisible, in agreement with disorder predictions. The ubiquitin moieties are partly flexible and extend radially away from PCNA, with the ubiquitin at the Pol κ-bound protomer appearing more rigid. Activity assays suggest that, when the internal PIP-box interaction is lost, Pol κ is retained on DNA by a secondary interaction between the UBZs and the ubiquitins flexibly conjugated to PCNA. Our data provide a structural basis for the recruitment of a Y-family TLS polymerase to sites of DNA damage., This research was supported by King Abdullah University of Science and Technology through core funding (to S.M.H.) and the Competitive Research Award Grant CRG8 URF/1/4036‐01‐01 (to S.M.H. and A.D.B.), and by the Wellcome Trust (to A.D.B.). R.C. acknowledges funding from the MINECO (CTQ2016-78636-P) and to AGAUR, (2017 SGR 324). The MD project has been carried out using CSUC resources. We acknowledge The Midlands Regional Cryo-EM Facility at the Leicester Institute of Structural and Chemical Biology (LISCB), major funding from MRC (MC_PC_17136). We thank Christos Savva (LISCB, University of Leicester) for his help in cryo-EM data collection and advice on data processing.
- Published
- 2021
4. Vigilant: An Engineered VirD2-Cas9 Complex for Lateral Flow Assay-Based Detection of SARS-CoV2
- Author
-
Ahmed Mahas, Muhammad Tehseen, Magdy M. Mahfouz, Tin Marsic, Samir M. Hamdan, and Zahir Ali
- Subjects
Letter ,Bioengineering ,02 engineering and technology ,Sensitivity and Specificity ,dCas9 ,molecular diagnostics ,chemistry.chemical_compound ,Biotin ,medicine ,Humans ,General Materials Science ,Polymerase ,VirD2 ,medicine.diagnostic_test ,biology ,SARS-CoV-2 ,Chemistry ,Oligonucleotide ,Cas9 ,nucleic acid detection ,Mechanical Engineering ,COVID-19 ,Nucleic acid test ,lateral flow assay ,Reverse Transcription ,General Chemistry ,biosensors ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,Molecular diagnostics ,Molecular biology ,relaxases ,Biotinylation ,Nucleic acid ,biology.protein ,RNA, Viral ,CRISPR-Cas Systems ,RT-RPA ,CRISPR-Cas9 ,0210 nano-technology - Abstract
Rapid, sensitive, and specific point-of-care testing for pathogens is crucial for disease control. Lateral flow assays (LFAs) have been employed for nucleic acid detection, but they have limited sensitivity and specificity. Here, we used a fusion of catalytically inactive SpCas9 endonuclease and VirD2 relaxase for sensitive, specific nucleic acid detection by LFA. In this assay, the target nucleic acid is amplified with biotinylated oligos. VirD2-dCas9 specifically binds the target sequence via dCas9 and covalently binds to a FAM-tagged oligonucleotide via VirD2. The biotin label and FAM tag are detected by a commercially available LFA. We coupled this system, named Vigilant (VirD2-dCas9 guided and LFA-coupled nucleic acid test), to reverse transcription-recombinase polymerase amplification to detect SARS-CoV2 in clinical samples. Vigilant exhibited a limit of detection of 2.5 copies/μL, comparable to CRISPR-based systems, and showed no cross-reactivity with SARS-CoV1 or MERS. Vigilant offers an easy-to-use, rapid, cost-effective, and robust detection platform for SARS-CoV2.
- Published
- 2021
5. Implementing fluorescence enhancement, quenching, and FRET for investigating flap endonuclease 1 enzymatic reaction at the single-molecule level
- Author
-
Mohamed Abdelmaboud Sobhy, Amer Bralic, Masateru Takahashi, Muhammad Tehseen, Samir M. Hamdan, and Alfredo De Biasio
- Subjects
DNA Replication ,DNA Polymerase δ ,Biophysics ,Flap structure-specific endonuclease 1 ,Review ,Cleavage (embryo) ,Biochemistry ,Structural Biology ,Genetics ,Fluorescence enhancement ,Flap endonuclease ,ComputingMethodologies_COMPUTERGRAPHICS ,Nuclease ,Quenching (fluorescence) ,biology ,Okazaki fragments ,Chemistry ,DNA replication ,Single-molecule fluorescence ,Computer Science Applications ,Fluorescence quenching ,Förster resonance energy transfer ,PIFE ,FRET ,biology.protein ,TP248.13-248.65 ,Biotechnology - Abstract
Graphical abstract, Flap endonuclease 1 (FEN1) is an important component of the intricate molecular machinery for DNA replication and repair. FEN1 is a structure-specific 5′ nuclease that cleaves nascent single-stranded 5′ flaps during the maturation of Okazaki fragments. Here, we review our research primarily applying single-molecule fluorescence to resolve important mechanistic aspects of human FEN1 enzymatic reaction. The methodology presented in this review is aimed as a guide for tackling other biomolecular enzymatic reactions by fluorescence enhancement, quenching, and FRET and their combinations. Using these methods, we followed in real-time the structures of the substrate and product and 5′ flap cleavage during catalysis. We illustrate that FEN1 actively bends the substrate to verify its features and continues to mold it to induce a protein disorder-to-order transitioning that controls active site assembly. This mechanism suppresses off-target cleavage of non-cognate substrates and promotes their dissociation with an accuracy that was underestimated from bulk assays. We determined that product release in FEN1 after the 5′ flap release occurs in two steps; a brief binding to the bent nicked-product followed by longer binding to the unbent nicked-product before dissociation. Based on our cryo-electron microscopy structure of the human lagging strand replicase bound to FEN1, we propose how this two-step product release mechanism may regulate the final steps during the maturation of Okazaki fragments.
- Published
- 2021
6. <scp>TSGIT</scp> : An N‐ and C‐terminal tandem tag system for purification of native and intein‐mediated ligation‐ready proteins
- Author
-
Vlad-Stefan Raducanu, Masateru Takahashi, Yujing Ouyang, Samir M. Hamdan, Daniela-Violeta Raducanu, and Muhammad Tehseen
- Subjects
Methods and Applications ,truncated protein ,Recombinant Fusion Proteins ,Intein ,Computational biology ,Protein degradation ,Cleavage (embryo) ,Biochemistry ,Inteins ,Bacteriophage ,03 medical and health sciences ,chemistry.chemical_compound ,protein ligation ,Biotin ,biotin ,protein cleavage ,Protein purification ,Cloning, Molecular ,protein expression ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Expression vector ,biology ,fusion tag ,030302 biochemistry & molecular biology ,biology.organism_classification ,purification tag ,Amino acid ,chemistry ,SUMO ,protein degradation ,IPL - Abstract
A large variety of fusion tags have been developed to improve protein expression, solubilization, and purification. Nevertheless, these tags have been combined in a rather limited number of composite tags and usually these composite tags have been dictated by traditional commercially‐available expression vectors. Moreover, most commercially‐available expression vectors include either N‐ or C‐terminal fusion tags but not both. Here, we introduce TSGIT, a fusion‐tag system composed of both N‐ and a C‐terminal composite fusion tags. The system includes two affinity tags, two solubilization tags and two cleavable tags distributed at both termini of the protein of interest. Therefore, the N‐ and the C‐terminal composite fusion tags in TSGIT are fully orthogonal in terms of both affinity selection and cleavage. For using TSGIT, we streamlined the cloning, expression, and purification procedures. Each component tag is selected to maximize its benefits toward the final construct. By expressing and partially purifying the protein of interest between the components of the TSGIT fusion, the full‐length protein is selected over truncated forms, which has been a long‐standing problem in protein purification. Moreover, due to the nature of the cleavable tags in TSGIT, the protein of interest is obtained in its native form without any additional undesired N‐ or C‐terminal amino acids. Finally, the resulting purified protein is ready for efficient ligation with other proteins or peptides for downstream applications. We demonstrate the use of this system by purifying a large amount of native fluorescent mRuby3 protein and bacteriophage T7 gp2.5 ssDNA‐binding protein.
- Published
- 2020
7. Simplified detection of polyhistidine-tagged proteins in gels and membranes using a UV-excitable dye and a multiple chelator head pair
- Author
-
Jasmeen S. Merzaban, Vlad-Stefan Raducanu, Daniela-Violeta Raducanu, Ioannis Isaioglou, and Samir M. Hamdan
- Subjects
recombinant protein expression ,0301 basic medicine ,Ultraviolet Rays ,Recombinant Fusion Proteins ,Western blot ,Protein tag ,Biochemistry ,UVHis-PAGE ,03 medical and health sciences ,chemistry.chemical_compound ,physical method ,Protein purification ,Humans ,metal-ion-chelating nitrilotriacetate (NTA) ,Polyhistidine-tag ,blot membrane ,Molecular Biology ,Fluorescent Dyes ,Detection limit ,030102 biochemistry & molecular biology ,His-tag detection ,Denaturing Gradient Gel Electrophoresis ,Chemistry ,Methods and Resources ,imaging ,UV transilluminator ,Cell Biology ,UV detection of His-tag protein ,histidine ,Fluorescence ,PAGE ,Blot ,030104 developmental biology ,Membrane ,Small Ubiquitin-Related Modifier Proteins ,Biophysics ,fluorescence ,Naked eye - Abstract
The polyhistidine tag (His-tag) is one of the most popular protein tags used in the life sciences. Traditionally, the detection of His-tagged proteins relies on immunoblotting with anti-His antibodies. This approach is laborious for certain applications, such as protein purification, where time and simplicity are critical. The His-tag can also be directly detected by metal ion-loaded nickel-nitrilotriacetic acid-based chelator heads conjugated to fluorophores, which is a convenient alternative method to immunoblotting. Typically, such chelator heads are conjugated to either green or red fluorophores, the detection of which requires specialized excitation sources and detection systems. Here, we demonstrate that post-run staining is ideal for His-tag detection by metal ion-loaded and fluorescently labeled chelator heads in PAGE and blot membranes. Additionally, by comparing the performances of different chelator heads, we show how differences in microscopic affinity constants translate to macroscopic differences in the detection limits in environments with limited diffusion, such as PAGE. On the basis of these results, we devise a simple approach, called UVHis-PAGE, that uses metal ion-loaded and fluorescently labeled chelator heads to detect His-tagged proteins in PAGE and blot membranes. Our method uses a UV transilluminator as an excitation source, and the results can be visually inspected by the naked eye.
- Published
- 2020
8. Deracemization and Stereoinversion of Alcohols Using Two Mutants of Secondary Alcohol Dehydrogenase from Thermoanaerobacter pseudoethanolicus
- Author
-
Samir M. Hamdan, Etsuko Takahashi, Musa M. Musa, Masateru Takahashi, and Sodiq Adeyeye Nafiu
- Subjects
biology ,Secondary alcohol dehydrogenase ,Stereochemistry ,Chemistry ,Organic Chemistry ,Mutant ,Stereoselectivity ,Physical and Theoretical Chemistry ,Thermoanaerobacter ,biology.organism_classification ,Redox - Published
- 2020
9. Functional binding of E-selectin to its ligands is enhanced by structural features beyond its lectin domain
- Author
-
X. Takahiro Kusakabe, Bader Al Alwan, Fajr A. Aleisa, Samir M. Hamdan, Jae Man Lee, X. Satoshi Habuchi, Jasmeen S. Merzaban, Dina B. AbuSamra, Shuho Nozue, Muhammad Tehseen, and Kosuke Sakashita
- Subjects
glycoprotein ,0301 basic medicine ,cell migration ,carbohydrate-binding protein ,Glycobiology and Extracellular Matrices ,Ligands ,Biochemistry ,conformational extension ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,Protein Domains ,glycobiology ,Polysaccharides ,Epidermal growth factor ,Cell Line, Tumor ,E-selectin ,binding kinetics ,Animals ,Humans ,Cell adhesion ,short consensus repeats ,Molecular Biology ,dimerization ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Lectin ,cell adhesion ,glycoprotein structure ,Cell Biology ,Bombyx ,Receptor–ligand kinetics ,Cell biology ,silkworm expression ,Transplantation ,Kinetics ,adhesion ,Transmembrane domain ,Immobilized Proteins ,030104 developmental biology ,biology.protein ,complement regulatory repeats ,Protein Multimerization ,homing ,Selectin - Abstract
Selectins are key to mediating interactions involved in cellular adhesion and migration, underlying processes such as immune responses, metastasis, and transplantation. Selectins are composed of a lectin domain, an epidermal growth factor (EGF)-like domain, multiple short consensus repeats (SCRs), a transmembrane domain, and a cytoplasmic tail. It is well-established that the lectin and EGF domains are required to mediate interactions with ligands; however, the contributions of the other domains in mediating these interactions remain obscure. Using various E-selectin constructs produced in a newly developed silkworm-based expression system and several assays performed under both static and physiological flow conditions, including flow cytometry, glycan array analysis, surface plasmon resonance, and cell-rolling assays, we show here that a reduction in the number of SCR domains is correlated with a decline in functional E-selectin binding to hematopoietic cell E- and/or L-selectin ligand (HCELL) and P-selectin glycoprotein ligand-1 (PSGL-1). Moreover, the binding was significantly improved through E-selectin dimerization and by a substitution (A28H) that mimics an extended conformation of the lectin and EGF domains. Analyses of the association and dissociation rates indicated that the SCR domains, conformational extension, and dimerization collectively contribute to the association rate of E-selectin–ligand binding, whereas just the lectin and EGF domains contribute to the dissociation rate. These findings provide the first evidence of the critical role of the association rate in functional E-selectin–ligand interactions, and they highlight that the SCR domains have an important role that goes beyond the structural extension of the lectin and EGF domains.
- Published
- 2020
10. Structure of the processive human Pol δ holoenzyme
- Author
-
Vlad-Stefan Raducanu, Muhammad Tehseen, Nekane Merino, Afnan Shirbini, Francisco J. Blanco, Alfredo De Biasio, Fahad Rashid, Manal S. Zaher, Claudia Lancey, Samir M. Hamdan, Timothy J. Ragan, and Christos G. Savva
- Subjects
DNA Replication ,Models, Molecular ,0301 basic medicine ,Flap Endonucleases ,DNA polymerase ,Science ,Protein subunit ,viruses ,Amino Acid Motifs ,Flap structure-specific endonuclease 1 ,General Physics and Astronomy ,Plasma protein binding ,Article ,General Biochemistry, Genetics and Molecular Biology ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Catalytic Domain ,Proliferating Cell Nuclear Antigen ,Humans ,lcsh:Science ,030304 developmental biology ,DNA Polymerase III ,0303 health sciences ,Multidisciplinary ,030102 biochemistry & molecular biology ,biology ,Okazaki fragments ,Cryoelectron Microscopy ,DNA replication ,DNA ,General Chemistry ,Proliferating cell nuclear antigen ,Cell biology ,Protein Subunits ,030104 developmental biology ,chemistry ,Biophysics ,biology.protein ,Human genome ,lcsh:Q ,Holoenzymes ,030217 neurology & neurosurgery ,Protein Binding - Abstract
In eukaryotes, DNA polymerase δ (Pol δ) bound to the proliferating cell nuclear antigen (PCNA) replicates the lagging strand and cooperates with flap endonuclease 1 (FEN1) to process the Okazaki fragments for their ligation. We present the high-resolution cryo-EM structure of the human processive Pol δ–DNA–PCNA complex in the absence and presence of FEN1. Pol δ is anchored to one of the three PCNA monomers through the C-terminal domain of the catalytic subunit. The catalytic core sits on top of PCNA in an open configuration while the regulatory subunits project laterally. This arrangement allows PCNA to thread and stabilize the DNA exiting the catalytic cleft and recruit FEN1 to one unoccupied monomer in a toolbelt fashion. Alternative holoenzyme conformations reveal important functional interactions that maintain PCNA orientation during synthesis. This work sheds light on the structural basis of Pol δ’s activity in replicating the human genome., Pol δ bound to the proliferating cell nuclear antigen (PCNA) replicates the lagging strand in eukaryotes and cooperates with flap endonuclease 1 (FEN1) to process the Okazaki fragments for their ligation. Here, the authors present a Cryo-EM structure of the human 4-subunit Pol δ bound to DNA and PCNA in a replicating state with an incoming nucleotide in the active site.
- Published
- 2020
11. Simultaneous cyclic deracemisation and stereoinversion of alcohols using orthogonal biocatalytic oxidation and reduction reactions
- Author
-
Etsuko Takahashi, Musa M. Musa, Samir M. Hamdan, Masateru Takahashi, and Sodiq Adeyeye Nafiu
- Subjects
chemistry.chemical_classification ,Ketone ,Secondary alcohol dehydrogenase ,biology ,biology.organism_classification ,Redox ,Combinatorial chemistry ,Catalysis ,Cofactor ,chemistry ,biology.protein ,Stereoselectivity ,Thermoanaerobacter - Abstract
We developed a concurrent cyclic deracemisation approach for secondary alcohols that combines a non-stereospecific oxidation step and a stereoselective reduction step using two mutants of Thermoanaerobacter pseudoethanolicus secondary alcohol dehydrogenase (TeSADH) that exhibit various extents of stereoselectivities. In this approach, W110G TeSADH, a sparingly stereoselective mutant, performs the non-stereospecific oxidation step and W110V/G198D TeSADH performs the stereoselective reduction step. The use of orthogonal cofactor regeneration systems allowed for the spontaneous operation of these mutants. (S)-Configured alcohols were obtained in moderate ee's from their racemates using this strategy. To our knowledge, this report provides the first example of a fully enzymatic cyclic deracemisation with a stereoselective reduction step (CD-RS) for alcohols. This approach was further improved into a deracemisation strategy via stereoinversion using concurrent (R)-selective I86A TeSADH-catalysed oxidation that leaves (S)-alcohols untouched and W110V/G198D TeSADH-catalysed stereoselective reduction of the resultant ketone intermediates into the corresponding (S)-configured alcohols. The latter strategy enabled quantitative production of (S)-1-phenylethanol in >99% ee from its racemate. Overall, we show the superiority of the stereoinversion deracemisation approach for alcohols when compared with cyclic deracemisation, which is mainly due to the elimination of futile cycles in the former.
- Published
- 2020
12. Proliferating cell nuclear antigen-agarose column: A tag-free and tag-dependent tool for protein purification affinity chromatography
- Author
-
Masateru Takahashi, Muhammad Tehseen, Afnan Shirbini, Fahad Rashid, Samir M. Hamdan, and Vlad-Stefan Raducanu
- Subjects
DNA Replication ,DNA Repair ,DNA polymerase ,Buffers ,010402 general chemistry ,01 natural sciences ,Biochemistry ,DNA-binding protein ,Chromatography, Affinity ,Analytical Chemistry ,Affinity chromatography ,Proliferating Cell Nuclear Antigen ,Protein purification ,Humans ,DNA Polymerase III ,chemistry.chemical_classification ,DNA ligase ,Chromatography ,biology ,Okazaki fragments ,Chemistry ,Sepharose ,010401 analytical chemistry ,Organic Chemistry ,DNA replication ,General Medicine ,Recombinant Proteins ,0104 chemical sciences ,Proliferating cell nuclear antigen ,Resins, Synthetic ,biology.protein ,Protein Binding - Abstract
Protein purification by affinity chromatography relies primarily on the interaction of a fused-tag to the protein of interest. Here, we describe a tag-free affinity method that employs functional selection interactions to a broad range of proteins. To achieve this, we coupled human DNA-clamp proliferating cell nuclear antigen (PCNA) that interacts with over one hundred proteins to an agarose resin. We demonstrate the versatility of our PCNA-Agarose column at various chromatographic steps by purifying PCNA-binding proteins that are involved in DNA Replication (DNA polymerase δ, flap endonuclease 1 and DNA ligase 1), translesion DNA synthesis (DNA polymerases eta, kappa and iota) and genome stability (p15). We also show the competence of the PCNA-Agarose column to purify non-PCNA binding proteins by fusing the PCNA-binding motif of human p21 as an affinity tag. Finally, we establish that our PCNA-Agarose column is a suitable analytical method for characterizing the binding strength of PCNA-binding proteins. The conservation and homology of PCNA-like clamps will allow for the immediate extension of our method to other species.
- Published
- 2019
13. Cryo-EM structure of Pol κ−DNA−PCNA holoenzyme and implications for polymerase switching in DNA lesion bypass
- Author
-
Ramon Crehuet, Claudia Lancey, Muhammad Tehseen, Samir M. Hamdan, Timothy J. Ragan, Masateru Takahashi, Mohamed Abdelmaboud Sobhy, and Alfredo De Biasio
- Subjects
chemistry.chemical_compound ,chemistry ,biology ,viruses ,Political science ,biology.protein ,Library science ,DNA ,Polymerase ,Proliferating cell nuclear antigen - Abstract
Replacement of the stalled replicative polymerase (Pol δ) at a DNA lesion by the error-prone DNA polymerase κ (Pol κ) restarts synthesis past the lesion to prevent genome instability. The switching from Pol δ to Pol κ is mediated by the processivity clamp PCNA but the structural basis of this mechanism is unknown. We determined the Cryo-EM structures of human Pol κ–DNA–PCNA complex and of a stalled Pol δ–DNA–PCNA complex at 3.9 and 4.7 Å resolution, respectively. In Pol κ complex, the C-terminus of the PAD domain docks the catalytic core to one PCNA protomer in an angled orientation, bending the DNA exiting Pol κ active site through PCNA. In Pol δ complex, the DNA is disengaged from the active site but is retained by the thumb domain. We present a model for polymerase switching facilitated by Pol κ recruitment to PCNA and Pol κ conformational sampling to seize the DNA from stalled Pol δ assisted by PCNA tilting.
- Published
- 2020
14. Expanding the Substrate Specificity of Thermoanaerobacter pseudoethanolicus Secondary Alcohol Dehydrogenase by a Dual Site Mutation
- Author
-
Masateru Takahashi, Samir M. Hamdan, Odey Bsharat, Musa M. Musa, Claire Vieille, and Ibrahim Karume
- Subjects
biology ,Secondary alcohol dehydrogenase ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Library science ,010402 general chemistry ,biology.organism_classification ,01 natural sciences ,Dual site ,0104 chemical sciences ,Substrate specificity ,Physical and Theoretical Chemistry ,Thermoanaerobacter - Abstract
The authors acknowledge the support provided by the Deanship of Scientific Research (DSR) at King Fahd University of Petroleum and Minerals (KFUPM) for funding this work under project number IN151032. They also acknowledge the supported by baseline research fund to S.M.H. by King Abdullah University of Science and Technology.
- Published
- 2018
15. Dynamic structure mediates halophilic adaptation of a DNA polymerase from the deep-sea brines of the Red Sea
- Author
-
Ulrich Stingl, Intikhab Alam, Jasmeen S. Merzaban, Etsuko Takahashi, Anastassja Akal, Luay I. Joudeh, Gobind Das, Samir M. Hamdan, Enzo Di Fabrizio, Muhammad Tehseen, Mohamed Abdelmaboud Sobhy, Masateru Takahashi, Mohamed M. Elshenawy, Monica Marini, Kosuke Sakashita, Joudeh, Luay [0000-0001-9338-205X], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,DNA polymerase engineering ,Circular dichroism ,Halophilic enzymes ,Structural adaptation ,Structure dynamism ,Thermophilic enzymes ,Archaeal Proteins ,DNA-Directed DNA Polymerase ,Indian Ocean ,Thermococcus ,Molecular Dynamics Simulation ,DNA polymerase ,Biochemistry ,03 medical and health sciences ,halophilic enzymes ,Genetics ,structural adaptation ,A-DNA ,Molecular Biology ,Polymerase ,biology ,Chemistry ,Thermophile ,Research ,structure dynamism ,Salt bridge (protein and supramolecular) ,Anoxic waters ,Halophile ,030104 developmental biology ,biology.protein ,Biophysics ,Biotechnology ,thermophilic enzymes - Abstract
The deep-sea brines of the Red Sea are remote and unexplored environments characterized by high temperatures, anoxic water, and elevated concentrations of salt and heavy metals. This environment provides a rare system to study the interplay between halophilic and thermophilic adaptation in biologic macromolecules. The present article reports the first DNA polymerase with halophilic and thermophilic features. Biochemical and structural analysis by Raman and circular dichroism spectroscopy showed that the charge distribution on the protein's surface mediates the structural balance between stability for thermal adaptation and flexibility for counteracting the salt-induced rigid and nonfunctional hydrophobic packing. Salt bridge interactions via increased negative and positive charges contribute to structural stability. Salt tolerance, conversely, is mediated by a dynamic structure that becomes more fixed and functional with increasing salt concentration. We propose that repulsive forces among excess negative charges, in addition to a high percentage of negatively charged random coils, mediate this structural dynamism. This knowledge enabled us to engineer a halophilic version of Thermococcus kodakarensis DNA polymerase.-Takahashi, M., Takahashi, E., Joudeh, L. I., Marini, M., Das, G., Elshenawy, M. M., Akal, A., Sakashita, K., Alam, I., Tehseen, M., Sobhy, M. A., Stingl, U., Merzaban, J. S., Di Fabrizio, E., Hamdan, S. M. Dynamic structure mediates halophilic adaptation of a DNA polymerase from the deep-sea brines of the Red Sea.
- Published
- 2018
16. Asymmetric Reduction of Substituted 2-Tetralones by Thermoanaerobacter pseudoethanolicus Secondary Alcohol Dehydrogenase
- Author
-
Claire Vieille, Musa M. Musa, Sulayman A. Oladepo, Masateru Takahashi, Samir M. Hamdan, and Odey Bsharat
- Subjects
biology ,010405 organic chemistry ,Chemistry ,Stereochemistry ,Organic Chemistry ,Substituent ,010402 general chemistry ,biology.organism_classification ,01 natural sciences ,Catalysis ,0104 chemical sciences ,Enzyme catalysis ,Inorganic Chemistry ,chemistry.chemical_compound ,Biocatalysis ,biology.protein ,Tetralone ,Stereoselectivity ,Physical and Theoretical Chemistry ,Thermoanaerobacter ,Alcohol dehydrogenase ,Tetralones - Abstract
Here, ketones bearing two bulky substituents, named bulky-bulky ketones, were successfully reduced to their corresponding optically-enriched alcohols using various mutants of Thermoanaerobacter pseudoethanolicus secondary alcohol dehydrogenase (TeSADH). Substituted 2-tetralones in particular were reduced to 2-tetralols with high conversion and high enantioselectivity. The pharmacological importance of substituted 2-tetralols as key drug-building blocks makes our biocatalytic reduction method a highly essential tool. We showed that changing the position of the substituent on the aromatic ring of 2-tetralones impacted their binding affinity and the reaction maximum catalytic rate. Docking studies with several TeSADH mutants explained how the position of the substituent on the tetralone influences the binding orientation of substituted 2-tetralones and their reaction stereoselectivity.
- Published
- 2017
17. Two chromatographic schemes for protein purification involving the biotin/avidin interaction under native conditions
- Author
-
Muhammad Tehseen, Vlad-Stefan Raducanu, Samir M. Hamdan, Afnan Shirbini, and Daniela-Violeta Raducanu
- Subjects
Flap Endonucleases ,SUMO-1 Protein ,Flap structure-specific endonuclease 1 ,Biotin ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Protein biotinylation ,Chromatography, Affinity ,Analytical Chemistry ,chemistry.chemical_compound ,DNA Ligase ATP ,Proliferating Cell Nuclear Antigen ,Protein purification ,Humans ,Biotinylation ,chemistry.chemical_classification ,DNA ligase ,Chromatography ,biology ,Chemistry ,Escherichia coli Proteins ,010401 analytical chemistry ,Organic Chemistry ,Proteins ,General Medicine ,Avidin ,0104 chemical sciences ,biology.protein ,Target protein ,Plasmids - Abstract
The strength of the biotin/avidin interaction makes it an ideal tool for the purification of biotin-labeled proteins via avidin-coupled resin with high specificity and selectivity. Nevertheless, this tight binding comes at an extra cost of performing the elution step under denaturing conditions. Weakening the biotin/avidin interaction improves the elution conditions, but only to mild or harsh denaturing buffers with the drawback of reducing the specificity and selectivity of this interaction. Here, we present two chromatographic protein purification schemes that are well-suited for application under native conditions thus preserving the strength of the biotin/avidin interaction. In the first scheme, we introduce a biotin-labeled SUMO-tag to each of human flap endonuclease 1 and Escherichia coli replication termination protein Tus, and elute both proteins by performing on-resin cleavage using SUMO protease. In the second scheme, we immobilize biotin-labeled human proliferating cell nuclear antigen (PCNA) on the avidin-coupled resin and use the resulting resin as a tag-free affinity method to purify the PCNA-binding protein human DNA Ligase 1. Furthermore, we streamlined the protein biotinylation protocol by constructing a single plasmid expression system that ensures high level of expression and solubility for each of the target protein bearing the biotin-tag and the enzyme responsible for the in vivo biotinylation reaction. Both chromatographic schemes resulted in a high yield of pure proteins in their native form.
- Published
- 2019
18. Resolution of the Holliday junction recombination intermediate by human GEN1 at the single-molecule level
- Author
-
Masateru Takahashi, Samir M. Hamdan, Muhammad Tehseen, Amer Bralic, Fahad Rashid, Vlad-Stefan Raducanu, Manal S. Zaher, and Mohamed Abdelmaboud Sobhy
- Subjects
Nuclear Envelope ,Dimer ,Biology ,Cleavage (embryo) ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genetics ,Holliday junction ,Molecule ,Humans ,Homologous Recombination ,030304 developmental biology ,Recombination, Genetic ,0303 health sciences ,DNA, Cruciform ,Endodeoxyribonucleases ,Nucleic Acid Enzymes ,Holliday Junction Resolvases ,Monomer ,Förster resonance energy transfer ,chemistry ,Biophysics ,Homologous recombination ,Dimerization ,030217 neurology & neurosurgery ,Recombination - Abstract
Human GEN1 is a cytosolic homologous recombination protein that resolves persisting four-way Holliday junctions (HJ) after the dissolution of the nuclear membrane. GEN1 dimerization has been suggested to play key role in the resolution of the HJ, but the kinetic details of its reaction remained elusive. Here, single-molecule FRET shows how human GEN1 binds the HJ and always ensures its resolution within the lifetime of the GEN1-HJ complex. GEN1 monomer generally follows the isomer bias of the HJ in its initial binding and subsequently distorts it for catalysis. GEN1 monomer remains tightly bound with no apparent dissociation until GEN1 dimer is formed and the HJ is fully resolved. Fast on- and slow off-rates of GEN1 dimer and its increased affinity to the singly-cleaved HJ enforce the forward reaction. Furthermore, GEN1 monomer binds singly-cleaved HJ tighter than intact HJ providing a fail-safe mechanism if GEN1 dimer or one of its monomers dissociates after the first cleavage. The tight binding of GEN1 monomer to intact- and singly-cleaved HJ empowers it as the last resort to process HJs that escape the primary mechanisms.
- Published
- 2018
19. Deracemization of Secondary Alcohols by using a Single Alcohol Dehydrogenase
- Author
-
Ibrahim Karume, Samir M. Hamdan, Musa M. Musa, and Masateru Takahashi
- Subjects
chemistry.chemical_classification ,Ketone ,biology ,010405 organic chemistry ,Organic Chemistry ,010402 general chemistry ,biology.organism_classification ,01 natural sciences ,Redox ,Catalysis ,0104 chemical sciences ,Enzyme catalysis ,Inorganic Chemistry ,Thermoanaerobacter ethanolicus ,chemistry.chemical_compound ,chemistry ,Alcohol oxidation ,Acetone ,biology.protein ,Organic chemistry ,Stereoselectivity ,Physical and Theoretical Chemistry ,Alcohol dehydrogenase - Abstract
We developed a single-enzyme-mediated two-step approach for deracemization of secondary alcohols. A single mutant of Thermoanaerobacter ethanolicus secondary alcohol dehydrogenase enables the nonstereoselective oxidation of racemic alcohols to ketones, followed by a stereoselective reduction process. Varying the amounts of acetone and 2-propanol cosubstrates controls the stereoselectivities of the consecutive oxidation and reduction reactions, respectively. We used one enzyme to accomplish the deracemization of secondary alcohols with up to >99 % ee and >99.5 % recovery in one pot and without the need to isolate the prochiral ketone intermediate.
- Published
- 2016
20. Dual enzymatic dynamic kinetic resolution by Thermoanaerobacter ethanolicus secondary alcohol dehydrogenase and Candida antarctica lipase B
- Author
-
Samir M. Hamdan, Odey Bsharat, Bassam El Ali, Ibrahim Karume, Masateru Takahashi, and Musa M. Musa
- Subjects
chemistry.chemical_classification ,biology ,010405 organic chemistry ,General Chemical Engineering ,Lipase b ,General Chemistry ,010402 general chemistry ,biology.organism_classification ,01 natural sciences ,0104 chemical sciences ,Kinetic resolution ,Hexane ,Thermoanaerobacter ethanolicus ,chemistry.chemical_compound ,Enzyme ,Enantiopure drug ,chemistry ,Organic chemistry ,Candida antarctica ,Racemization - Abstract
The immobilization of Thermoanaerobacter ethanolicus secondary alcohol dehydrogenase (TeSADH) using sol–gel method enables its use to racemize enantiopure alcohols in organic media. Here, we report the racemization of enantiopure phenyl-ring-containing secondary alcohols using xerogel-immobilized W110A TeSADH in hexane rather than the aqueous medium required by the enzyme. We further showed that this racemization approach in organic solvent was compatible with Candida antarctica lipase B (CALB)-catalyzed kinetic resolution. This compatibility, therefore, allowed a dual enzymatic dynamic kinetic resolution of racemic alcohols using CALB-catalyzed kinetic resolution and W110A TeSADH-catalyzed racemization of phenyl-ring-containing alcohols.
- Published
- 2016
21. Single-Molecule Imaging of PAF15-PCNA using DNA Skybridge
- Author
-
Jong-Bong Lee Lee, Samir M. Hamdan, Alfredo De Biasio, Fahad Rashid, Gayun Bu, Daehyung Kim, Amaia Gonzalez-Magaña, and Francisco J. Blanco
- Subjects
chemistry.chemical_compound ,biology ,Chemistry ,Biophysics ,biology.protein ,Single Molecule Imaging ,DNA ,Proliferating cell nuclear antigen - Published
- 2020
22. A direct fluorescent signal transducer embedded in a DNA aptamer paves the way for versatile metal-ion detection
- Author
-
Vlad-Stefan Raducanu, Jasmeen S. Merzaban, Manal S. Zaher, Yanyan Li, Fahad Rashid, and Samir M. Hamdan
- Subjects
Aptamer ,Metal ions in aqueous solution ,Nanotechnology ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Signal ,Metal ,chemistry.chemical_compound ,Materials Chemistry ,A-DNA ,Electrical and Electronic Engineering ,Instrumentation ,Metals and Alloys ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,Fluorescence ,0104 chemical sciences ,Surfaces, Coatings and Films ,Electronic, Optical and Magnetic Materials ,Transducer ,chemistry ,visual_art ,visual_art.visual_art_medium ,0210 nano-technology ,DNA - Abstract
Using DNA aptamers as sensors for metal ions provide a variety of applications in biology and industry. Many of these sensors are based on guanine-rich DNA sequences that undergo conformational changes upon metal-ion binding. However, these sensors require an exogenous reporter that can recognize such DNA conformational changes and transduce the signal. Here, we bypass the exogenous reporter by embedding a signal transducer in the guanine-rich DNA aptamer that measures directly the DNA conformational changes upon metal-ion binding. Our signal transducer is an environmentally sensitive Cy3 fluorescent dye that is internally coupled to the DNA aptamer. We demonstrate the applicability of our embedded-signal transducer approach using a known potassium-responding aptamer. We next demonstrate the versatility of this approach by designing an aptamer sensor that can detect potassium ions in the low micro-molar range and with high selectivity against a wide range of ions including sodium. The aptamer accurately measured potassium ions concentration in a variety of aqueous and biological test samples. Our embedded-signal transducer approach will pave the way for the development of aptamer sensors for a variety of ligands.
- Published
- 2020
23. Microfluidics-based super-resolution microscopy enables nanoscopic characterization of blood stem cell rolling
- Author
-
Luay I. Joudeh, Bader Al Alwan, Samir M. Hamdan, Jasmeen S. Merzaban, Karmen AbuZineh, Satoshi Habuchi, AbuZineh, Karmen [0000-0003-1459-1422], Joudeh, Luay I [0000-0001-9338-205X], Al Alwan, Bader [0000-0001-6075-3889], Hamdan, Samir M [0000-0001-5192-1852], Merzaban, Jasmeen S [0000-0002-7276-2907], Habuchi, Satoshi [0000-0002-6663-2807], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Fluorescence-lifetime imaging microscopy ,Microfluidics ,Immunology ,03 medical and health sciences ,Membrane Microdomains ,parasitic diseases ,Research Methods ,Humans ,natural sciences ,Progenitor cell ,Cell adhesion ,Lipid raft ,Research Articles ,Microscopy ,Multidisciplinary ,Microscopy, Confocal ,biology ,Chemistry ,CD44 ,technology, industry, and agriculture ,SciAdv r-articles ,Actin cytoskeleton ,Hematopoietic Stem Cells ,Nanostructures ,030104 developmental biology ,Hyaluronan Receptors ,biology.protein ,Biophysics ,E-Selectin ,Selectin ,Homing (hematopoietic) ,Research Article - Abstract
Super-resolution imaging reveals subtle interplay between nanoscopic organization of membrane ligands and cellular interaction., Hematopoietic stem/progenitor cell (HSPC) homing occurs via cell adhesion mediated by spatiotemporally organized ligand-receptor interactions. Although molecules and biological processes involved in this multistep cellular interaction with endothelium have been studied extensively, molecular mechanisms of this process, in particular the nanoscale spatiotemporal behavior of ligand-receptor interactions and their role in the cellular interaction, remain elusive. We introduce a microfluidics-based super-resolution fluorescence imaging platform and apply the method to investigate the initial essential step in the homing, tethering, and rolling of HSPCs under external shear stress that is mediated by selectins, expressed on endothelium, with selectin ligands (that is, CD44) expressed on HSPCs. Our new method reveals transient nanoscale reorganization of CD44 clusters during cell rolling on E-selectin. We demonstrate that this mechanical force-induced reorganization is accompanied by a large structural reorganization of actin cytoskeleton. The CD44 clusters were partly disrupted by disrupting lipid rafts. The spatial reorganization of CD44 and actin cytoskeleton was not observed for the lipid raft–disrupted cells, demonstrating the essential role of the spatial clustering of CD44 on its reorganization during cell rolling. The lipid raft disruption causes faster and unstable cell rolling on E-selectin compared with the intact cells. Together, our results demonstrate that the spatial reorganization of CD44 and actin cytoskeleton is the result of concerted effect of E-selectin–ligand interactions, external shear stress, and spatial clustering of the selectin ligands, and has significant effect on the tethering/rolling step in HSPC homing. Our new experimental platform provides a foundation for characterizing complicated HSPC homing.
- Published
- 2018
24. Positioning the 5′-flap junction in the active site controls the rate of flap endonuclease-1–catalyzed DNA cleavage
- Author
-
Samir M. Hamdan, Bo Song, and Manju M. Hingorani
- Subjects
0301 basic medicine ,DNA repair ,Flap Endonucleases ,Flap structure-specific endonuclease 1 ,DNA and Chromosomes ,Cleavage (embryo) ,Biochemistry ,Catalysis ,03 medical and health sciences ,chemistry.chemical_compound ,Catalytic Domain ,Humans ,Magnesium ,Flap endonuclease ,Molecular Biology ,biology ,Chemistry ,DNA replication ,Active site ,Cell Biology ,DNA ,030104 developmental biology ,Phosphodiester bond ,biology.protein ,Biophysics ,Calcium - Abstract
Flap endonucleases catalyze cleavage of single-stranded DNA flaps formed during replication, repair, and recombination and are therefore essential for genome processing and stability. Recent crystal structures of DNA-bound human flap endonuclease (hFEN1) offer new insights into how conformational changes in the DNA and hFEN1 may facilitate the reaction mechanism. For example, previous biochemical studies of DNA conformation performed under non-catalytic conditions with Ca(2+) have suggested that base unpairing at the 5′-flap:template junction is an important step in the reaction, but the new structural data suggest otherwise. To clarify the role of DNA changes in the kinetic mechanism, we measured a series of transient steps, from substrate binding to product release, during the hFEN1-catalyzed reaction in the presence of Mg(2+). We found that whereas hFEN1 binds and bends DNA at a fast, diffusion-limited rate, much slower Mg(2+)-dependent conformational changes in DNA around the active site are subsequently necessary and rate-limiting for 5′-flap cleavage. These changes are reported overall by fluorescence of 2-aminopurine at the 5′-flap:template junction, indicating that local DNA distortion (e.g. disruption of base stacking observed in structures), associated with positioning the 5′-flap scissile phosphodiester bond in the hFEN1 active site, controls catalysis. hFEN1 residues with distinct roles in the catalytic mechanism, including those binding metal ions (Asp-34 and Asp-181), steering the 5′-flap through the active site and binding the scissile phosphate (Lys-93 and Arg-100), and stacking against the base 5′ to the scissile phosphate (Tyr-40), all contribute to these rate-limiting conformational changes, ensuring efficient and specific cleavage of 5′-flaps.
- Published
- 2018
25. PCNA Structure and Interactions with Partner Proteins
- Author
-
Muse Oke, Samir M. Hamdan, and Manal S. Zaher
- Subjects
chemistry.chemical_compound ,biology ,chemistry ,DNA polymerase ,Docking (molecular) ,biology.protein ,Processivity ,DNA ,Cell biology ,Proliferating cell nuclear antigen - Abstract
Proliferating cell nuclear antigen (PCNA) consists of three identical monomers that topologically encircle double-stranded DNA. PCNA stimulates the processivity of DNA polymerase d and, to a less extent, the intrinsically highly processive DNA polymerase e. It also functions as a platform that recruits and coordinates the activities of a large number of DNA processing proteins. Emerging structural and biochemical studies suggest that the nature of PCNA-partner proteins interactions is complex. A hydrophobic groove at the front side of PCNA serves as a primary docking site for the consensus PIP box motifs present in many PCNA-binding partners. Sequences that immediately flank the PIP box motif or regions that are distant from it could also interact with the hydrophobic groove and other regions of PCNA. Posttranslational modifications on the backside of PCNA could add another dimension to its interaction with partner proteins. An encounter of PCNAwith different DNA structures might also be involved in coordinating its interactions. Finally, the ability of PCNA to bind up to three proteins while topologically linked to DNA suggests that it would be a versatile toolbox in many different DNA processing reactions.
- Published
- 2018
26. Eukaryotic DNA Replicases
- Author
-
Manal S. Zaher, Muse Oke, and Samir M. Hamdan
- Subjects
Biochemistry ,Chemistry ,Eukaryotic DNA replication - Published
- 2018
27. Phosphate steering by Flap Endonuclease 1 promotes 5′-flap specificity and incision to prevent genome instability
- Author
-
Steven J. Shaw, Alexander J. Neil, Fahad Rashid, Altaf H. Sarker, Mark J. Thompson, Jane A. Grasby, Mai Zong Her, Victoria J. B. Gotham, Susan E. Tsutakawa, Emma C. Jardine, Andrew S. Arvai, Samir M. Hamdan, John A. Tainer, Sergei M. Mirkin, L. David Finger, Sana I. Algasaier, and Jane C. Kim
- Subjects
0301 basic medicine ,Genome instability ,DNA Replication ,DNA Repair ,DNA repair ,Flap Endonucleases ,Science ,Flap structure-specific endonuclease 1 ,General Physics and Astronomy ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Genomic Instability ,Phosphates ,Substrate Specificity ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Catalytic Domain ,Genetics ,Humans ,Nucleotide ,Amino Acid Sequence ,Flap endonuclease ,Binding site ,Cancer ,chemistry.chemical_classification ,Multidisciplinary ,Binding Sites ,Human Genome ,DNA replication ,General Chemistry ,DNA ,Molecular biology ,030104 developmental biology ,chemistry ,Mutation ,Sequence Alignment ,030217 neurology & neurosurgery - Abstract
DNA replication and repair enzyme Flap Endonuclease 1 (FEN1) is vital for genome integrity, and FEN1 mutations arise in multiple cancers. FEN1 precisely cleaves single-stranded (ss) 5′-flaps one nucleotide into duplex (ds) DNA. Yet, how FEN1 selects for but does not incise the ss 5′-flap was enigmatic. Here we combine crystallographic, biochemical and genetic analyses to show that two dsDNA binding sites set the 5′polarity and to reveal unexpected control of the DNA phosphodiester backbone by electrostatic interactions. Via ‘phosphate steering’, basic residues energetically steer an inverted ss 5′-flap through a gateway over FEN1’s active site and shift dsDNA for catalysis. Mutations of these residues cause an 18,000-fold reduction in catalytic rate in vitro and large-scale trinucleotide (GAA)n repeat expansions in vivo, implying failed phosphate-steering promotes an unanticipated lagging-strand template-switch mechanism during replication. Thus, phosphate steering is an unappreciated FEN1 function that enforces 5′-flap specificity and catalysis, preventing genomic instability., Flap Endonuclease 1 is a DNA replication and repair enzyme indispensable for maintaining genomic stability. Here the authors provide mechanistic details on how FEN1 selects for 5′-flaps and promotes catalysis to avoid large-scale repeat expansion by a process termed ‘phosphate steering’.
- Published
- 2017
28. Single-molecule FRET unveils induced-fit mechanism for substrate selectivity in flap endonuclease 1
- Author
-
Ivaylo Ivanov, Samir M. Hamdan, Hubert Marek Piwonski, Chunli Yan, John A. Tainer, Satoshi Habuchi, Mohamed Abdelmaboud Sobhy, Paul D. Harris, Luay I. Joudeh, Fahad Rashid, Susan E. Tsutakawa, and Manal S. Zaher
- Subjects
Models, Molecular ,0301 basic medicine ,030103 biophysics ,Human purified proteins ,Flap Endonucleases ,Protein Conformation ,QH301-705.5 ,Science ,Flap structure-specific endonuclease 1 ,Biology ,Models, Biological ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,Substrate Specificity ,03 medical and health sciences ,chemistry.chemical_compound ,None ,Humans ,Protein–DNA interaction ,Biology (General) ,Replication protein A ,Nuclease ,General Immunology and Microbiology ,General Neuroscience ,DNA ,General Medicine ,Single-molecule FRET ,Biophysics and Structural Biology ,Molecular biology ,Single Molecule Imaging ,DNA binding site ,030104 developmental biology ,chemistry ,expressed proteins in E. coli ,biology.protein ,Biophysics ,Nucleic Acid Conformation ,Medicine ,recombinant protein ,In vitro recombination ,Protein Binding ,Research Article - Abstract
Human flap endonuclease 1 (FEN1) and related structure-specific 5’nucleases precisely identify and incise aberrant DNA structures during replication, repair and recombination to avoid genomic instability. Yet, it is unclear how the 5’nuclease mechanisms of DNA distortion and protein ordering robustly mediate efficient and accurate substrate recognition and catalytic selectivity. Here, single-molecule sub-millisecond and millisecond analyses of FEN1 reveal a protein-DNA induced-fit mechanism that efficiently verifies substrate and suppresses off-target cleavage. FEN1 sculpts DNA with diffusion-limited kinetics to test DNA substrate. This DNA distortion mutually ‘locks’ protein and DNA conformation and enables substrate verification with extreme precision. Strikingly, FEN1 never misses cleavage of its cognate substrate while blocking probable formation of catalytically competent interactions with noncognate substrates and fostering their pre-incision dissociation. These findings establish FEN1 has practically perfect precision and that separate control of induced-fit substrate recognition sets up the catalytic selectivity of the nuclease active site for genome stability. DOI: http://dx.doi.org/10.7554/eLife.21884.001, eLife digest When a cell divides it must copy its genetic information, which is found in the form of strands of DNA. Damage to the DNA may lead to cancer or a number of genetic diseases. However, every time a cell divides more than 10 million toxic “flaps” of excess DNA are generated. A protein called flap endonuclease 1 (FEN1) keeps the DNA in good repair by cutting off the flaps in a highly specific and selective manner. Many proteins that interact with DNA are attracted to specific genetic sequences within the DNA strands. However, this is not the case for FEN1 and several other “structure-specific” proteins that help to repair and replicate DNA strands. So how do these proteins select the correct regions of DNA to interact with? Rashid et al. used single-molecule fluorescence measurements to examine how purified FEN1 proteins interact with DNA flaps. The results show that FEN1 can perfectly recognize and correctly remove flaps through a process called “mutual-induced fit”, where the DNA and FEN1 are shaped by each other to produce a highly specific structure. Further work is now needed to examine whether other proteins that are related to FEN1 use a similar process to ensure that they always cut DNA in the same way. More detailed and direct examination of the structure of FEN1 through other experimental methods may also help to reveal how the mutual-induced fit process enables FEN1 to achieve such high levels of precision. This could increase our understanding of how problems with FEN1 and similar proteins lead to different genetic diseases. DOI: http://dx.doi.org/10.7554/eLife.21884.002
- Published
- 2017
29. Author response: Single-molecule FRET unveils induced-fit mechanism for substrate selectivity in flap endonuclease 1
- Author
-
Ivaylo Ivanov, Hubert Marek Piwonski, Luay I. Joudeh, John A. Tainer, Samir M. Hamdan, Chunli Yan, Manal S. Zaher, Mohamed Abdelmaboud Sobhy, Paul D. Harris, Susan E. Tsutakawa, Fahad Rashid, and Satoshi Habuchi
- Subjects
Mechanism (engineering) ,Chemistry ,Biophysics ,Flap structure-specific endonuclease 1 ,Substrate (chemistry) ,Single-molecule FRET ,Selectivity - Published
- 2017
30. DNA skybridge: 3D structure producing a light sheet for high-throughput single-molecule imaging
- Author
-
Manal S. Zaher, I I Hwan Cho, Jong-Bong Lee, Samir M. Hamdan, Yeonmo Cho, Fahad Rashid, Cherlhyun Jeong, and Daehyung Kim
- Subjects
Immobilized Nucleic Acids ,02 engineering and technology ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Optical imaging ,Interference (communication) ,Genetics ,Molecule ,Nanotechnology ,Throughput (business) ,030304 developmental biology ,0303 health sciences ,Immobilized DNA ,business.industry ,Optical Imaging ,DNA ,021001 nanoscience & nanotechnology ,Single Molecule Imaging ,High-Throughput Screening Assays ,chemistry ,Nucleic acid ,Optoelectronics ,Methods Online ,0210 nano-technology ,business - Abstract
Real-time visualization of single-proteins or -complexes on nucleic acid substrates is an essential tool for characterizing nucleic acid binding proteins. Here, we present a novel surface-condition independent and high-throughput single-molecule optical imaging platform called ‘DNA skybridge’. The DNA skybridge is constructed in a 3D structure with 4 μm-high thin quartz barriers in a quartz slide. Each DNA end is attached to the top of the adjacent barrier, resulting in the extension and immobilization of DNA. In this 3D structure, the bottom surface is out-of-focus when the target molecules on the DNA are imaged. Moreover, the DNA skybridge itself creates a thin Gaussian light sheet beam parallel to the immobilized DNA. This dual property allows for imaging a single probe-tagged molecule moving on DNA while effectively suppressing interference with the surface and background signals from the surface.
- Published
- 2019
31. Sequential and Multistep Substrate Interrogation Provides the Scaffold for Specificity in Human Flap Endonuclease 1
- Author
-
Masateru Takahashi, Samir M. Hamdan, Luay I. Joudeh, Xiaojuan Huang, and Mohamed Abdelmaboud Sobhy
- Subjects
DNA Replication ,Models, Molecular ,Flap Endonucleases ,Flap structure-specific endonuclease 1 ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Substrate Specificity ,chemistry.chemical_compound ,Cleave ,Fluorescence Resonance Energy Transfer ,Humans ,Flap endonuclease ,lcsh:QH301-705.5 ,Nuclease ,DNA replication ,DNA ,Enzyme binding ,Kinetics ,Förster resonance energy transfer ,lcsh:Biology (General) ,Biochemistry ,chemistry ,Biophysics ,biology.protein ,Nucleic Acid Conformation - Abstract
Summary Human flap endonuclease 1 (FEN1), one of the structure-specific 5′ nucleases, is integral in replication, repair, and recombination of cellular DNA. The 5′ nucleases share significant unifying features yet cleave diverse substrates at similar positions relative to 5′ end junctions. Using single-molecule Forster resonance energy transfer, we find a multistep mechanism that verifies all substrate features before inducing the intermediary-DNA bending step that is believed to unify 5′ nuclease mechanisms. This is achieved by coordinating threading of the 5′ flap of a nick junction into the conserved capped-helical gateway, overseeing the active site, and bending by binding at the base of the junction. We propose that this sequential and multistep substrate recognition process allows different 5′ nucleases to recognize different substrates and restrict the induction of DNA bending to the last common step. Such mechanisms would also ensure the protection of DNA junctions from nonspecific bending and cleavage.
- Published
- 2013
32. Two Modes of Interaction of the Single-stranded DNA-binding Protein of Bacteriophage T7 with the DNA Polymerase-Thioredoxin Complex
- Author
-
Samir M. Hamdan, Charles C. Richardson, and Sharmistha Ghosh
- Subjects
Models, Molecular ,DNA polymerase ,Static Electricity ,Molecular Conformation ,DNA-Directed DNA Polymerase ,Biochemistry ,chemistry.chemical_compound ,Thioredoxins ,Bacteriophage T7 ,Escherichia coli ,Protein–DNA interaction ,Molecular Biology ,Polymerase ,biology ,Lysine ,DNA replication ,Helicase ,Cell Biology ,Processivity ,Surface Plasmon Resonance ,Recombinant Proteins ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Kinetics ,chemistry ,Enzymology ,biology.protein ,Biophysics ,Thioredoxin ,DNA ,Protein Binding - Abstract
The DNA polymerase encoded by bacteriophage T7 has low processivity. Escherichia coli thioredoxin binds to a segment of 76 residues in the thumb subdomain of the polymerase and increases the processivity. The binding of thioredoxin leads to the formation of two basic loops, loops A and B, located within the thioredoxin-binding domain (TBD). Both loops interact with the acidic C terminus of the T7 helicase. A relatively weak electrostatic mode involves the C-terminal tail of the helicase and the TBD, whereas a high affinity interaction that does not involve the C-terminal tail occurs when the polymerase is in a polymerization mode. T7 gene 2.5 single-stranded DNA-binding protein (gp2.5) also has an acidic C-terminal tail. gp2.5 also has two modes of interaction with the polymerase, but both involve the C-terminal tail of gp2.5. An electrostatic interaction requires the basic residues in loops A and B, and gp2.5 binds to both loops with similar affinity as measured by surface plasmon resonance. When the polymerase is in a polymerization mode, the C terminus of gene 2.5 protein interacts with the polymerase in regions outside the TBD. gp2.5 increases the processivity of the polymerase-helicase complex during leading strand synthesis. When loop B of the TBD is altered, abortive DNA products are observed during leading strand synthesis. Loop B appears to play an important role in communication with the helicase and gp2.5, whereas loop A plays a stabilizing role in these interactions.
- Published
- 2010
33. Peptide ligands specific to the oxidized form of Escherichia coli thioredoxin
- Author
-
Samir M. Hamdan, Charles C. Richardson, Brian K. Kay, Bridget S. Banach, and Michael D. Scholle
- Subjects
animal structures ,Phage display ,Molecular Sequence Data ,Biophysics ,Peptide binding ,Peptide ,DNA-Directed DNA Polymerase ,Biology ,Ligands ,Binding, Competitive ,Biochemistry ,Article ,Substrate Specificity ,Analytical Chemistry ,Inhibitory Concentration 50 ,Thioredoxins ,Peptide Library ,Catalytic Domain ,Escherichia coli ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Peptide library ,Molecular Biology ,Peptide sequence ,Nucleic Acid Synthesis Inhibitors ,chemistry.chemical_classification ,T7 DNA polymerase ,Peptide Fragments ,chemistry ,Thioredoxin ,Oxidation-Reduction ,Protein Binding ,Binding domain - Abstract
Thioredoxin (Trx) is a highly conserved redox protein involved in several essential cellular processes. In this study, our goal was to isolate peptide ligands to Escherichia coli Trx that mimic protein-protein interactions, specifically the T7 polymerase-Trx interaction. To do this, we subjected Trx to affinity selection against a panel of linear and cysteine-constrained peptides using M13 phage display. A novel cyclized conserved peptide sequence, with a motif of C(D/N/S/T/G)D(S/T)-hydrophobic-C-X-hydrophobic-P, was isolated to Trx. These peptides bound specifically to the E. coli Trx when compared to the human and spirulina homologs. An alanine substitution of the active site cysteines (CGPC) resulted in a significant loss of peptide binding affinity to the Cys-32 mutant. The peptides were also characterized in the context of Trx's role as a processivity factor of the T7 DNA polymerase (gp5). As the interaction between gp5 and Trx normally takes place under reducing conditions, which might interfere with the conformation of the disulfide-bridged peptides, we made use of a 22 residue deletion mutant of gp5 in the thioredoxin binding domain (gp5Delta22) that bypassed the requirements of reducing conditions to interact with Trx. A competition study revealed that the peptide selectively inhibits the interaction of gp5Delta22 with Trx, under oxidizing conditions, with an IC50 of approximately 10 microM.
- Published
- 2008
34. Inadequate inhibition of host RNA polymerase restricts T7 bacteriophage growth on hosts overexpressing udk
- Author
-
Samir M. Hamdan, Arkadiusz W. Kulczyk, Udi Qimron, Stanley Tabor, and Charles C. Richardson
- Subjects
RNA-dependent RNA polymerase ,Biology ,Microbiology ,Molecular biology ,chemistry.chemical_compound ,chemistry ,Sigma factor ,Transcription (biology) ,RNA editing ,RNA polymerase ,RNA polymerase I ,medicine ,biology.protein ,T7 RNA polymerase ,Molecular Biology ,Polymerase ,medicine.drug - Abstract
Overexpression of udk, an Escherichia coli gene encoding a uridine/cytidine kinase, interferes with T7 bacteriophage growth. We show here that inhibition of T7 phage growth by udk overexpression can be overcome by inhibition of host RNA polymerase. Overexpression of gene 2, whose product inhibits host RNA polymerase, restores T7 phage growth on hosts overexpressing udk. In addition, rifampicin, an inhibitor of host RNA polymerase, restores the burst size of T7 phage on udk-overexpressing hosts to normal. In agreement with these findings, suppressor mutants that overcome the inhibition arising from udk overexpression gain the ability to grow on hosts that are resistant to inhibition of RNA polymerase by gene 2 protein, and suppressor mutants that overcome a lack of gene 2 protein gain the ability to grow on hosts that overexpress udk. Mutations that eliminate or weaken strong promoters for host RNA polymerase in T7 DNA, and mutations in T7 gene 3.5 that affect its interaction with T7 RNA polymerase, also reduce the interference with T7 growth by host RNA polymerase. We propose a general model for the requirement of host RNA polymerase inhibition.
- Published
- 2007
35. Quantitative Characterization of E-selectin Interaction with Native CD44 and P-selectin Glycoprotein Ligand-1 (PSGL-1) Using a Real Time Immunoprecipitation-based Binding Assay
- Author
-
Samir M. Hamdan, Dina B. AbuSamra, Jasmeen S. Merzaban, Alia Al-Kilani, Samah Zeineb Gadhoum, and Kosuke Sakashita
- Subjects
Immunoprecipitation ,Glycobiology and Extracellular Matrices ,Plasma protein binding ,Biochemistry ,chemistry.chemical_compound ,Cell Movement ,Cell Line, Tumor ,E-selectin ,Protein Interaction Mapping ,Humans ,Protein Interaction Maps ,Cell adhesion ,Molecular Biology ,Membrane Glycoproteins ,biology ,Chemistry ,Ligand binding assay ,Cell Biology ,Sialyl-Lewis X ,Hyaluronan Receptors ,biology.protein ,Biophysics ,P-selectin glycoprotein ligand-1 ,E-Selectin ,Selectin ,Protein Binding - Abstract
Selectins (E-, P-, and L-selectins) interact with glycoprotein ligands to mediate the essential tethering/rolling step in cell transport and delivery that captures migrating cells from the circulating flow. In this work, we developed a real time immunoprecipitation assay on a surface plasmon resonance chip that captures native glycoforms of two well known E-selectin ligands (CD44/hematopoietic cell E-/L-selectin ligand and P-selectin glycoprotein ligand-1) from hematopoietic cell extracts. Here we present a comprehensive characterization of their binding to E-selectin. We show that both ligands bind recombinant monomeric E-selectin transiently with fast on- and fast off-rates, whereas they bind dimeric E-selectin with remarkably slow on- and off-rates. This binding requires the sialyl Lewis x sugar moiety to be placed on both O- and N-glycans, and its association, but not dissociation, is sensitive to the salt concentration. Our results suggest a mechanism through which monomeric selectins mediate initial fast on and fast off kinetics to help capture cells out of the circulating shear flow; subsequently, tight binding by dimeric/oligomeric selectins is enabled to significantly slow rolling.
- Published
- 2015
36. Essential Residues in the C Terminus of the Bacteriophage T7 Gene 2.5 Single-stranded DNA-binding Protein
- Author
-
Samir M. Hamdan, Seung-Joo Lee, Charles C. Richardson, and Boriana Marintcheva
- Subjects
Phenylalanine ,Molecular Sequence Data ,DNA, Single-Stranded ,Biology ,Biochemistry ,Viral Proteins ,chemistry.chemical_compound ,Residue (chemistry) ,Bacteriophage T7 ,Escherichia coli ,Amino Acid Sequence ,Molecular Biology ,Gene ,chemistry.chemical_classification ,C-terminus ,T7 DNA polymerase ,Cell Biology ,Protein Structure, Tertiary ,Amino acid ,DNA-Binding Proteins ,Kinetics ,chemistry ,Mutagenesis ,Replisome ,Gene Deletion ,DNA ,Plasmids ,Protein Binding - Abstract
Gene 2.5 of bacteriophage T7 encodes a single-stranded DNA (ssDNA)-binding protein (gp2.5) that is an essential component of the phage replisome. Similar to other prokaryotic ssDNA-binding proteins, gp2.5 has an acidic C terminus that is involved in protein-protein interactions at the replication fork and in modulation of the ssDNA binding properties of the molecule. We have used genetic and biochemical approaches to identify residues critical for the function of the C terminus of gp2.5. The presence of an aromatic residue in the C-terminal position is essential for gp2.5 function. Deletion of the C-terminal residue, phenylalanine, is detrimental to its function, as is the substitution of this residue with non-aromatic amino acids. Placing the C-terminal phenylalanine in the penultimate position also results in loss of function. Moderate shortening of the length of the acidic portion of the C terminus is tolerated when the aromatic nature of the C-terminal residue is preserved. Gradual removal of the acidic C terminus of gp2.5 results in a higher affinity for ssDNA and a decreased ability to interact with T7 DNA polymerase/thioredoxin. The replacement of the charged residues in the C terminus with neutral amino acids abolishes gp2.5 function. Our data show that both the C-terminal aromatic residue and the overall acidic charge of the C terminus of gp2.5 are critical for its function.
- Published
- 2006
37. Nickel subsulfide is similar to potassium dichromate in protecting normal human fibroblasts from the mutagenic effects of benzo[a]pyrene diolepoxide
- Author
-
David S. Reinhold, Samir M. Hamdan, and Brent Morse
- Subjects
biology ,Epidemiology ,Health, Toxicology and Mutagenesis ,biology.organism_classification ,medicine.disease_cause ,Molecular biology ,Chinese hamster ,Nickel Subsulfide ,chemistry.chemical_compound ,chemistry ,Benzo(a)pyrene ,medicine ,Potassium dichromate ,Antimutagen ,Genetics (clinical) ,Oxidative stress ,Hypoxanthine ,Carcinogen - Abstract
The cellular response to multiple carcinogen treatment has not been extensively studied, even though the effect of individual carcinogens is, in many cases, well known. We have previously shown that potassium dichromate can protect normal human fibroblasts from the mutagenic effects of benzo-[a]pyrene diolepoxide (BPDE), and that this effect may be via an oxidative stress mechanism [Tesfai et al. (1998) Mutat Res 416:159-168]. Here, we extend our previous work by showing that nickel subsulfide can produce the same effect. Normal human fibroblasts, preincubated with nickel subsulfide for 46 hr followed by a coincubation of nickel subsulfide and BPDE for 2 hr, showed a dramatic reduction in the mutant frequency of the hypoxanthine (guanine)phosphoribosyl-transterase (HPRT) gene when compared to cells treated only with BPDE. The preincubation period with nickel subsulfide was necessary to see the antagonistic effect, since it was not observed if the cells were simply incubated with both carcinogens for 2 hr. The extent of the antagonistic effect was nickel subsulfide dose-dependent and also appeared to be species? specific, since the effect was not observed when Chinese hamster fibroblasts were tested. Finally, the antagonistic effect of the nickel subsulfide was eliminated by vitamin E, suggesting that production of reactive oxygen species by the nickel may be required. This data, along with our previous work, suggest that the antagonistic effect we observe is not chromium-specific, and that it could be speciesspecific.
- Published
- 1999
38. Single Molecule Studies of Nucleic Acid Enzymes
- Author
-
Samir M. Hamdan and Antoine M. van Oijen
- Subjects
chemistry.chemical_compound ,Magnetic tweezers ,biology ,Biochemistry ,chemistry ,Base pair ,DNA polymerase ,biology.protein ,Nucleic acid ,Helicase ,RNA ,Polymerase ,DNA - Abstract
This chapter reviews the various single molecule methods used and the type of information obtained by such studies of nucleic acid enzymes. Numerous different enzymes have evolved to catalyze the synthesis, digestion, unwinding, and unlinking of nucleic acids that are central to genomic maintenance. The development of single molecule techniques has allowed researchers to study the activity of nucleic acid enzymes, such as RNA polymerases, DNA polymerases, topoisomerases, exonucleases, andDNA helicases, at an unprecedented level of detail. The ability to observe the activity of RNA and DNA-binding proteins on the single molecule level provides tremendous opportunities in the field of nucleic acid enzymology. The chapter provides an overview of the various techniques and a number of nucleic acid enzyme systems where single molecule approaches have proven to be particularly powerful in unraveling the molecular details of enzymatic activity. The recent development of several methods, such as flow stretching, magnetic tweezers, and optical trapping to mechanically manipulate objects of microscopic scale has allowed researchers to stretch and twist individual DNA molecules. Advances in fluorescence spectroscopy and microscopy have made it possible to detect the fluorescence from a single fluorophore under biological tweezers, transcriptional elongation traces of individual E. coli RNA polymerases could be obtained with high spatial resolution, allowing for a detailed study of the statistics of transcriptional pausing. The technical improvements of optical trapping techniques allowed observation of movements of individual RNA polymerases along their template DNA with a resolution better than a single base pair.
- Published
- 2009
39. Hydrolysis of the 5'-p-nitrophenyl ester of TMP by oligoribonucleases (ORN) from Escherichia coli, Mycobacterium smegmatis, and human
- Author
-
Samir M. Hamdan, N. Liyou, Phil A. Jennings, Ah Young Park, Tamarind E. Hamwood, Christopher M. Elvin, Robert J.K. Wood, and Nicholas E. Dixon
- Subjects
inorganic chemicals ,Exonuclease ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Mycobacterium smegmatis ,medicine.disease_cause ,Cofactor ,Nitrophenols ,Hydrolysis ,Escherichia ,medicine ,Escherichia coli ,Thymidine Monophosphate ,Humans ,Histidine ,Amino Acid Sequence ,chemistry.chemical_classification ,biology ,biology.organism_classification ,Kinetics ,Protein Subunits ,Enzyme ,Biochemistry ,chemistry ,Solubility ,Exoribonucleases ,biology.protein ,Chromatography, Gel ,Oligopeptides ,Sequence Alignment ,Biotechnology - Abstract
Escherichia coli oligoribonuclease (EcoORN), encoded by the orn gene, is a 3'-5' exonuclease that degrades short single-stranded oligoribonucleotides to rNMPs in the final step of RNA degradation. The orn gene is essential in E. coli, but not in higher organisms, and close homologues are present in other genomes from the beta and gamma subdivisions of the Protobacteriaceae, including many pathogenic species. We report here the expression in E. coli of orn and homologues from Mycobacterium smegmatis and human, and large-scale purification of the three enzymes. All three were found to promote the hydrolysis of the 5'-p-nitrophenyl ester of TMP (pNP-TMP) with similar values of Michaelis-Menten parameters (k(cat)=100-650 min(-1), K(M)=0.4-2.0 mM, at pH 8.00 and 25 degrees C, with 1 mM Mn(2+)). Hydrolysis by pNP-TMP by all three enzymes depended on a divalent metal ion, with Mn(2+) being preferred over Mg(2+) as cofactor, and was inhibited by Ni(2+). The concentration dependency of Mn(2+) was examined, giving K(Mn) values of 0.2-0.6 mM. The availability of large amounts of the purified enzymes and a simple spectrophotometric assay for ORN activity should facilitate large-scale screening for new inhibitors of bacterial oligoribonucleases.
- Published
- 2007
40. Mechanism of DNA binding and sequence recognition by T7 DNA primase
- Author
-
Charles C. Richardson, Seung-Joo Lee, and Samir M. Hamdan
- Subjects
chemistry.chemical_compound ,Chemistry ,Mechanism (biology) ,Genetics ,Computational biology ,Primase ,Molecular Biology ,Biochemistry ,DNA ,Biotechnology ,Sequence (medicine) - Published
- 2006
41. Role of the linker between the zinc binding domain and the polymerization domain of the bacteriophage T7 DNA primase
- Author
-
Samir M. Hamdan, Seung-Joo Lee, Charles C. Richardson, and Udi Qimron
- Subjects
Zinc binding ,biology ,HMG-box ,Chemistry ,biology.organism_classification ,Biochemistry ,Molecular biology ,Domain (software engineering) ,Bacteriophage ,Polymerization ,Genetics ,Biophysics ,Primase ,Molecular Biology ,Linker ,Biotechnology ,Binding domain - Published
- 2006
42. Application of electrospray ionization mass spectrometry to study the hydrophobic interaction between the ɛ and θ subunits of DNA polymerase III
- Author
-
Margaret M. Sheil, Samir M. Hamdan, Nicholas E. Dixon, Rajesh Gupta, and Jennifer L. Beck
- Subjects
chemistry.chemical_classification ,Circular dichroism ,Spectrometry, Mass, Electrospray Ionization ,biology ,Hydrogen bond ,Electrospray ionization ,Escherichia coli Proteins ,Static Electricity ,Analytical chemistry ,Active site ,Nuclear magnetic resonance spectroscopy ,Biochemistry ,Article ,Hydrophobic effect ,Dissociation constant ,Crystallography ,Protein Subunits ,chemistry ,Enzyme Stability ,biology.protein ,Non-covalent interactions ,Protein Structure, Quaternary ,Molecular Biology ,Hydrophobic and Hydrophilic Interactions ,DNA Polymerase III - Abstract
Electrospray ionization mass spectrometry (ESI-MS) has widespread routine use as a tool in proteomics for confirmation of primary structure determination and for characterization of purified proteins (Griffiths et al. 2001). Application of ESI-MS to detection and characterization of non-covalent complexes of biomolecules is not as well established, but there are now many examples of noncovalent interactions that have been studied in the gas phase, including those between protein subunits, proteins and nucleic acids, and enzymes and substrates (Veenstra 1999; Burkitt et al. 2003; Sanglier et al. 2003). The most obvious use of ESI-MS for study of these complexes is in determination of the stoichiometry of binding partners, and this has recently been extended to monitor subunit exchange between small heat shock proteins in real time (Sobott et al. 2002). The establishment of stoichiometry is a prelude to more detailed structural determination of biomolecules in complexes. The stability of the complex (dissociation constants), the types of noncovalent interactions (e.g., polar vs. nonpolar), and conformational changes in the binding partners upon complex formation are important when considering the mechanism of biological action of biomolecular complexes (e.g., protein–protein, protein–DNA). There is a suite of biophysical techniques that can be applied to study these properties. These range from monitoring of changes in fluorescence or surface plasmon resonance (SPR) for determination of dissociation constants and use of circular dichroism and NMR spectroscopy for following conformational changes, ultimately to determination of complete structures of complexes by NMR, X-ray crystallography, or cryo-electron microscopy. ESI-MS offers speed and sensitivity in monitoring components of equilibrium mixtures. Consequently, there are increasing numbers of reports of its use for determination of dissociation constants or relative binding affinities of non-covalent complexes (Jorgensen et al. 1998; Kapur et al. 2002; Bligh et al. 2003). Furthermore, there are ESI-MS studies where the stabilities of noncovalent complexes have been assessed by their resistance to dissociation in the mass spectrometer using CID (collision-induced dissociation) or thermal denaturation experiments (Gupta et al. 2001; de Brouwer et al. 2002; Benesch et al. 2003). Nevertheless, data obtained from ESI-MS studies need to be interpreted with caution. First, the ionization process itself might perturb equilibria (Wang and Agnes 1999). Second, there is a paucity of information about changes in the strength or specificity of noncovalent interactions that occur on transfer from the condensed to the gas phase during the ionization process. The stabilities of complexes between biological macromolecules involve contributions from ionic, hydrogen bonding, hydrophobic, and/or van der Waals interactions. Several ESI-MS studies support the proposal that electrostatic interactions are strengthened in vacuo, while hydrophobic interactions are unaffected or weakened through loss of water during desolvation and/or ionization (Loo 1997). For these reasons, it is important to study the behavior of noncovalent complexes that have been well characterized in solution to enable evaluation of data from ESI-MS experiments. Recently, as part of a study aimed at investigating the behavior of noncovalent complexes on transferal from solution to the gas phase, we used ESI-MS to study the well-characterized Tus-Ter (protein–DNA) complex that terminates DNA replication in Escherichia coli (Kapur et al. 2002). We showed that relative binding affinities of mutant Tus proteins for double-stranded TerB DNA were the same in the gas phase as in solution, and conversely, that the relative affinities of wild-type Tus for various double-stranded DNA sequences were unchanged on transferal to the gas phase. Both the X-ray structure (Kamada et al. 1996) and SPR studies of the ionic strength dependence of dissociation of the Tus-TerB complex (Neylon et al. 2000) show there are substantial polar and electrostatic contacts between the binding partners. Consistent with this, ESI mass spectra showed that dissociation of the complex required high concentrations of ammonium acetate, in the range of 1–2 M (Kapur et al. 2002). For the present work, we used ESI-MS to investigate the predominantly hydrophobic interactions between two protein subunits of E. coli DNA polymerase III: the θ subunit, and the N-terminal domain (residues 2–186) of the ɛ subunit (ɛ186). DNA polymerase III is a multisubunit enzyme that is the major replicative polymerase of E. coli (Kelman and O’Donnell 1995; McHenry 2003). Three of the 10 subunits, α, ɛ, and θ, comprise the catalytic core: the large α-subunit contains the polymerase active site, and ɛ contributes the proofreading 3′→5′ exonuclease activity, while the precise function of θ is not known (Studwell-Vaughan and O’Donnell 1993; Kunkel and Bebenek 2000). The ɛ subunit consists of two domains (Perrino et al. 1999; Taft-Benz and Schaaper 1999; Hamdan et al. 2000). The N-terminal domain (ɛ186) contains the exonuclease active site and forms a stable 1:1 complex with θ (Perrino et al. 1999; Hamdan et al. 2002a). The complex forms readily and essentially quantitatively on mixing of the two subunits, and is sufficiently stable that it can be isolated by ion-exchange chromatography (Hamdan et al. 2002a). It is stable for extended periods at 25°C in aqueous solution under conditions required for NMR studies (Pintacuda et al. 2004). Although there is no high-resolution structure yet available for the ɛ186–θ complex, we have reported the crystal structure of ɛ186 (Hamdan et al. 2002b) and the solution structure of θ (Keniry et al. 2000). NMR chemical shift mapping experiments in the latter study suggested that a series of small hydrophobic residues on the external face of the first helix of θ (residues 21–27, AAAGVAF) are involved in its association with ɛ186, and this has been confirmed in the more recent NMR structure of θ in the ɛ186–θ complex (M. Keniry, pers. comm.). Moreover, recent comparisons of NMR spectra of free ɛ186 and ɛ186–θ have identified hydrophobic residues in ɛ at the θ-binding interface, including Ile31, Val50, Val58, Ile68, Leu74, Ile154, Leu161, Leu165, and Leu166 (DeRose et al. 2003). It appears, therefore, that interaction between the two proteins is mediated largely via aliphatic side chains, and the forces that hold them together are largely hydrophobic in nature. In the present work, we aimed to use ESI-MS to supply “snapshots” of components of mixtures of ɛ186 and θ. The stability of the ɛ186–θ complex was studied under various solution and instrumental conditions. Its stability at high ionic strength is consistent with a dominant contribution of nonpolar interactions, in contrast with that of the Tus-Ter complexes, where interactions are primarily polar and electrostatic in nature and are disrupted at relatively low ionic strength (Kapur et al. 2002). In addition, ESI-MS experiments suggested that the ɛ subunit protects ɛ186 from aggregation in organic solvent/water mixtures. This is consistent with earlier experiments in which θ was shown to stabilize ɛ186 against thermal inactivation (Hamdan et al. 2002a).
- Published
- 2004
43. Hydrolysis of the 5'-p-nitrophenyl ester of TMP by the proofreading exonuclease (epsilon) subunit of Escherichia coli DNA polymerase III
- Author
-
Samir M. Hamdan, Ji Yeon Yang, Paul D. Carr, P. E. Lilley, Phillip R. Thompson, Nicholas E. Dixon, Esther M. M. Bulloch, Jeffrey A. Crowther, David L. Ollis, Susan E. Brown, and Jennifer L. Beck
- Subjects
Exonuclease ,Exodeoxyribonuclease V ,DNA polymerase ,Stereochemistry ,Cations, Divalent ,Biochemistry ,Catalysis ,Divalent ,Hydrolysis ,Escherichia coli ,Thymidine Monophosphate ,Magnesium ,Exodeoxyribonuclease ,Klenow fragment ,DNA Polymerase III ,chemistry.chemical_classification ,Manganese ,biology ,Chemistry ,Active site ,Hydrogen-Ion Concentration ,Molecular biology ,Kinetics ,Exodeoxyribonucleases ,biology.protein ,Titration - Abstract
The core of DNA polymerase III, the replicative polymerase in Escherichia coli, consists of three subunits (alpha, epsilon, and theta). The epsilon subunit is the 3'-5' proofreading exonuclease that associates with the polymerase (alpha) through its C-terminal region and theta through a 185-residue N-terminal domain (epsilon 186). A spectrophotometric assay for measurement of epsilon activity is described. Proteins epsilon and epsilon 186 and the epsilon 186.theta complex catalyzed the hydrolysis of the 5'-p-nitrophenyl ester of TMP (pNP-TMP) with similar values of k(cat) and K(M), confirming that the N-terminal domain of epsilon bears the exonuclease active site, and showing that association with theta has little direct effect on the chemistry occurring at the active site of epsilon. On the other hand, formation of the complex with theta stabilized epsilon 186 by approximately 14 degrees C against thermal inactivation. For epsilon 186, k(cat) = 293 min(-)(1) and K(M) = 1.08 mM at pH 8.00 and 25 degrees C, with a Mn(2+) concentration of 1 mM. Hydrolysis of pNP-TMP by epsilon 186 depended absolutely on divalent metal ions, and was inhibited by the product TMP. Dependencies on Mn(2+) and Mg(2+) concentrations were examined, giving a K(Mn) of 0.31 mM and a k(cat) of 334 min(-1) for Mn(2+) and a K(Mg) of 6.9 mM and a k(cat) of 19.9 min(-1) for Mg(2+). Inhibition by TMP was formally competitive [K(i) = 4.3 microM (with a Mn(2+) concentration of 1 mM)]. The pH dependence of pNP-TMP hydrolysis by epsilon 186, in the pH range of 6.5-9.0, was found to be simple. K(M) was essentially invariant between pH 6.5 and 8.5, while k(cat) depended on titration of a single group with a pK(a) of 7.7, approaching limiting values of 50 min(-1) at pH6.5 and 400 min(-1) at pH9.0. These data are used in conjunction with crystal structures of the complex of epsilon 186 with TMP and two Mn(II) ions bound at the active site to develop insights into the mechanisms of pNP-TMP hydrolysis by epsilon at high and low pH values.
- Published
- 2002
44. Structural basis for proofreading during replication of the Escherichia coli chromosome
- Author
-
Paul D. Carr, Nicholas E. Dixon, Samir M. Hamdan, Susan E. Brown, and David L. Ollis
- Subjects
Exonuclease ,DNA Replication ,Models, Molecular ,Exodeoxyribonuclease V ,DNA polymerase ,Stereochemistry ,Molecular Sequence Data ,Crystallography, X-Ray ,Protein Structure, Secondary ,dnaQ ,chemistry.chemical_compound ,Structural Biology ,Escherichia coli ,Amino Acid Sequence ,Molecular Biology ,X-ray crystallography ,DNA Polymerase III ,RecBCD ,biology ,Molecular Structure ,Circular bacterial chromosome ,Escherichia coli Proteins ,DNA replication ,Chromosomes, Bacterial ,Protein Structure, Tertiary ,DnaQ ,Exodeoxyribonucleases ,chemistry ,Biochemistry ,biology.protein ,Proofreading ,binuclear metallohydrolase ,exonuclease (3′–5′) ,Sequence Alignment ,DNA ,manganese metalloenzyme - Abstract
The epsilon subunit of the Escherichia coli replicative DNA polymerase III is the proofreading 3'-5' exonuclease. Structures of its catalytic N-terminal domain (epsilon186) were determined at two pH values (5.8 and 8.5) at resolutions of 1.7-1.8 A, in complex with two Mn(II) ions and a nucleotide product of its reaction, thymidine 5'-monophosphate. The protein structure is built around a core five-stranded beta sheet that is a common feature of members of the DnaQ superfamily. The structures were identical, except for differences in the way TMP and water molecules are coordinated to the binuclear metal center in the active site. These data are used to develop a mechanism for epsilon and to produce a plausible model of the complex of epsilon186 with DNA.
- Published
- 2002
45. Single-molecule Observations of Replisome Structure and Function
- Author
-
M. van Antoine Oijen, Samir M. Hamdan, Joseph J. Loparo, Charles C. Richardson, and Zernike Institute for Advanced Materials
- Subjects
Quantitative Biology::Biomolecules ,Total internal reflection fluorescence microscope ,biology ,Biophysics ,DNA replication ,Helicase ,T7 DNA polymerase ,Molecular biology ,Primer extension ,chemistry.chemical_compound ,chemistry ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,biology.protein ,Replisome ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,Polymerase ,DNA - Abstract
DNA replication requires the coordinated activity of a large number of enzymes at the replication fork. Understanding the mechanisms controlling this organization requires a direct probing of the dynamics of fully functional replisomes during replication. Observations at the single-molecule level provide the most direct way to visualize the complex biochemistry of the replisome and to quantify the many transient intermediates essential to replication. We present a novel assay that combines the observation of individual fluorescently labeled proteins with the mechanical manipulation of DNA. Surface-tethered DNAs labeled with quantum dots are hydrodynamically stretched and imaged with a TIRF microscope. Activity of the replisome is observed as a change in the DNA length due to the differing force-dependent extension of single- and double-stranded DNA at low pico-Newton forces. We employ a two-color imaging scheme to monitor DNA length in real-time and to stroboscopically image fluorescently labeled single proteins interacting with DNA. Observation of labeled proteins in an ongoing replication reaction allows us to pose structural questions about the stoichiometry and exchange of proteins at the prokaryotic replication fork. We will discuss preliminary results on primer extension by the T7 DNA polymerase and strand-displacement synthesis by the coupled activity of the T7 helicase and polymerase.
- Published
- 2009
46. Real-time single-molecule observation of rolling-circle DNA replication
- Author
-
Samir M. Hamdan, Nathan A. Tanner, Nicholas E. Dixon, Joseph J. Loparo, Slobodan Jergic, Antoine M. van Oijen, and Zernike Institute for Advanced Materials
- Subjects
DNA Replication ,DNA, Bacterial ,DNA-Directed DNA Polymerase ,Biology ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Multienzyme Complexes ,Bacteriophage T7 ,Replication (statistics) ,Genetics ,medicine ,Rolling circle DNA replication ,Escherichia coli ,Organic Chemicals ,030304 developmental biology ,Fluorescent Dyes ,0303 health sciences ,Total internal reflection fluorescence microscope ,DNA synthesis ,DNA replication ,Deoxyguanine Nucleotides ,Molecular biology ,3. Good health ,Kinetics ,chemistry ,Microscopy, Fluorescence ,DNA, Viral ,Biophysics ,Replisome ,Methods Online ,030217 neurology & neurosurgery ,DNA ,Dideoxynucleotides - Abstract
We present a simple technique for visualizing replication of individual DNA molecules in real time. By attaching a rolling-circle substrate to a TIRF microscope-mounted flow chamber, we are able to monitor the progression of single-DNA synthesis events and accurately measure rates and processivities of single T7 and Escherichia coli replisomes as they replicate DNA. This method allows for rapid and precise characterization of the kinetics of DNA synthesis and the effects of replication inhibitors.
- Published
- 2009
47. Racemization of enantiopure secondary alcohols by Thermoanaerobacter ethanolicus secondary alcohol dehydrogenase
- Author
-
Samir M. Hamdan, Maris Laivenieks, Claire Vieille, Masateru Takahashi, Musa M. Musa, and Robert S. Phillips
- Subjects
Molecular Structure ,biology ,Chemistry ,Stereochemistry ,Organic Chemistry ,Stereoisomerism ,Thermoanaerobacter ,biology.organism_classification ,Biochemistry ,Cofactor ,Alcohol Oxidoreductases ,Thermoanaerobacter ethanolicus ,Enantiopure drug ,Biocatalysis ,Alcohols ,biology.protein ,Organic chemistry ,Molecule ,Physical and Theoretical Chemistry ,Enantiomer ,Racemization - Abstract
Controlled racemization of enantiopure phenyl-ring-containing secondary alcohols is achieved in this study using W110A secondary alcohol dehydrogenase from Thermoanaerobacter ethanolicus (W110A TeSADH) and in the presence of the reduced and oxidized forms of its cofactor nicotinamide-adenine dinucleotide. Racemization of both enantiomers of alcohols accepted by W110A TeSADH, not only with low, but also with reasonably high, enantiomeric discrimination is achieved by this method. Furthermore, the high tolerance of TeSADH to organic solvents allows TeSADH-catalyzed racemization to be conducted in media containing up to 50% (v/v) of organic solvents.
- Published
- 2013
48. Dynamics Of DNA Replication Loops Reveal Temporal Control Of Lagging-strand Synthesis
- Author
-
Charles C. Richardson, Samir M. Hamdan, Antoine M. van Oijen, Joseph J. Loparo, Masateru Takahashi, and Zernike Institute for Advanced Materials
- Subjects
DNA Replication ,Time Factors ,DNA polymerase ,DNA polymerase II ,Biophysics ,Eukaryotic DNA replication ,DNA-Directed DNA Polymerase ,Antiparallel (biochemistry) ,Article ,DnaG ,Multienzyme Complexes ,Bacteriophage T7 ,Directionality ,Polymerase ,Genetics ,Multidisciplinary ,DNA clamp ,biology ,Okazaki fragments ,Chemistry ,DNA replication ,Bacteriophage lambda ,Cell biology ,Microscopy, Fluorescence ,Prokaryotic DNA replication ,DNA, Viral ,biology.protein ,Replisome ,Primer (molecular biology) - Abstract
In all organisms, the protein machinery responsible for the replication of DNA, the replisome, is faced with a directionality problem. The antiparallel nature of duplex DNA permits the leading-strand polymerase to advance in a continuous fashion, but forces the lagging-strand polymerase to synthesize in the opposite direction. By extending RNA primers, the lagging-strand polymerase restarts at short intervals and produces Okazaki fragments1,2. At least in prokaryotic systems, this directionality problem is solved by the formation of a loop in the lagging strand of the replication fork to reorient the lagging-strand DNA polymerase so that it advances in parallel with the leading-strand polymerase. The replication loop grows and shrinks during each cycle of Okazaki-fragment synthesis3. Here, we employ single-molecule techniques to visualize, in real time, the formation and release of replication loops by individual replisomes of bacteriophage T7 supporting coordinated DNA replication. Analysis of the distributions of loop sizes and lag times between loops reveals that initiation of primer synthesis and the completion of an Okazaki fragment each serve as a trigger for loop release. The presence of two triggers may represent a fail-safe mechanism ensuring the timely reset of the replisome after the synthesis of every Okazaki fragment.
- Published
- 2009
49. Studies of the Structures of Core Subunits of Escherichia coli DNA Polymerase III Holoenzyme
- Author
-
Samir M. Hamdan, Gottfried Otting, David L. Ollis, Paul D. Carr, Nicholas E. Dixon, Susan E. Brown, and Max A. Keniry
- Subjects
Core (optical fiber) ,biology ,Biochemistry ,Chemistry ,biology.protein ,Escherichia coli DNA ,Polymerase - Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.