The database was constructed using the amino acid sequences of all curated non-redundant 3248 hydrogenase catalytic subunits represented in the NCBI RefSeq database in August 20142 (link) (Dataset S1 ). In order to test the classification tool, additional sequences from newly-sequenced archaeal and bacterial phyla were retrieved from the Joint Genome Institute’s Integrated Microbial Genomes database43 (link).
>
Genes & Molecular Sequences
>
Amino Acid Sequence
>
Catalytic Domain
Catalytic Domain
The Catalytic Domain is a discrete structural unit within an enzyme that is responsible for its catalytic activity.
This region of the protein typically contains the active site where the substrate binds and the chemical reaction occurs.
Identifying and understanding the catalytic domain is crucial for studying enzyme function, mechanism, and inhibition.
PubCompare.ai's AI-driven platform can help researchers efforlessly locate and compare catalytic domain protocols from literature, preprints, and patents to optimize their research proccess and make data-driven decisions.
This region of the protein typically contains the active site where the substrate binds and the chemical reaction occurs.
Identifying and understanding the catalytic domain is crucial for studying enzyme function, mechanism, and inhibition.
PubCompare.ai's AI-driven platform can help researchers efforlessly locate and compare catalytic domain protocols from literature, preprints, and patents to optimize their research proccess and make data-driven decisions.
Most cited protocols related to «Catalytic Domain»
Amino Acid Sequence
Archaea
Bacteria
Catalytic Domain
Genome
Genome, Microbial
Hydrogenase
Joints
Acetate
belinostat
Catalytic Domain
Chimera
Crystallography
HC toxin
Ligands
Light
Maltose-Binding Proteins
oxamflatin
Panobinostat
Proteins
Radiation
Solvents
Vertebral Column
Vorinostat
X-Ray Diffraction
2',5'-oligoadenylate
Biological Assay
Catalytic Domain
Cell Motility Assays
Chromatography
Codon
Electrons
Escherichia coli
Homo sapiens
Magnesium Chloride
Molar
Mono-S
Mutagenesis
Mutation
Oligonucleotide Primers
Peptide Hydrolases
phosphoramidite
Proteins
Selenomethionine
The constitutive Cas9 expression construct was derived by subcloning the 5’ 3×FLAG tagged human-codon optimized Cas9 cDNA from Streptococcus pyogenes (addgene: # 49535) into the MSCV-PGK-Puro vector (Clontech: # 634401). The U6-sgRNA-EFS-GFP and the U6-sgRNA-EFS-mCherry vectors were derived from the lentiCRISPR plasmid (addgene: # 49535) by removing the hCas9 cDNA and replacing the Puro cassette with GFP or mCherry. The wild-type RPA3 was PCR cloned directly from human cDNA into the MSCV-IRES-GFP (MigR1) vector. All cloning procedures were performed using the In-Fusion cloning system (Clontech: #638909). sgRNAs were cloned by annealing two DNA oligos and ligating into a BsmB1-digested U6-sgRNA-EFS-GFP/mCherry vectors, as described26 (link). To improve U6 promoter efficiency, we added an extra 5’ G nucleotide to all of the sgRNAs that did not start with a 5’ G.
All sgRNAs in this study were designed usinghttp://crispr.mit.edu/ 25 (link). The majority of sgRNAs used in this study had a quality score above 70 to minimize off-target effects. In Figure 1 , sgRNAs were designed targeting 5’ constitutive coding exons of each target gene. For the chromatin regulatory domain-focused CRISPR screen, sgRNAs were designed to target the catalytic domain or bromodomain of each protein based on the NCBI database annotation.
All sgRNA sequences used in this study are provided in aSupplementary Table 1 .
All sgRNAs in this study were designed using
All sgRNA sequences used in this study are provided in a
2',5'-oligoadenylate
Amber Stop Codon
Catalytic Domain
Chromatin
Cloning Vectors
Clustered Regularly Interspaced Short Palindromic Repeats
DNA, Complementary
Exons
Homo sapiens
Internal Ribosome Entry Sites
Nucleotides
Plasmids
Proteins
RPA3 protein, human
Streptococcus pyogenes
Plants were grown under continuous light conditions. The drm1–1, drm2–1, cmt3–7, rdr2–1, drd1–6, rdr6–1, sgs3/sde2–1, sde3–1, dcl2–1, rdr1–1, nrpd2a-1, and nrpd2b-1 mutants have been previously described [18 (link),20 (link),23 ,27 (link),31 (link)]. drm1–1 and drm2–1 are T-DNA alleles isolated in the Wassilewskija (WS) ecotype—both T-DNAs are predicted to disrupt essential catalytic domains. These mutations were backcrossed five times into Ler prior to this analysis. cmt3–7 is a point mutation isolated in the Ler ecotype that creates a stop codon, truncating the CMT3 protein after 27 amino acids. drm1–2, drm2–2, and cmt3–11 are T-DNA insertions in the predicted methyltransferase domains of DRM1, DRM2, and CMT3 that would be expected to create null mutations (T-DNAs SALK_031705, SALK_150863, and SALK_148381 respectively). kyp-6 is T-DNA SALK_041474. The phenotypes of drm1 drm2 cmt3 in the Ler ecotype were scored at two main stages. Twisted rosette leaf shape was scored at approximately 2 wk, prior to bolting. Short stature was scored at approximate 5–6 wk, once the plants had made the majority of their siliques.
Alleles
Amino Acids
Catalytic Domain
Codon, Terminator
Disease, Dejerine-Sottas
DNA Modification Methylases
DRD1 protein, human
Dwarfism
Ecotype
Insertion Mutation
Light
Mutation
Null Mutation
Phenotype
Plant Leaves
Plants
Point Mutation
Proteins
T-DNA
Most recents protocols related to «Catalytic Domain»
All graphs were drawn using Prism GraphPad. Michaelis−Menten kinetics data was also calculated using Prism. Substrate inhibition curves were plotted using Prism or the Dynafit software and the equations in Scheme 1 [53 (link)], where Ks and Kss are dissociation constants and kcat is the turnover of the enzyme in min−1. The Km and Vmax were determined using Prism, using points below the substrate inhibition concentrations.
Scheme 1. Mechanism for substrate inhibition. This mechanism assumes that enzyme I has two substrate (S) binding sites: a catalytic site for normal catalysis and a second non-catalytic or allosteric binding site. When substrate is bound at both sites (SES), the product (P) is produced at a reduced rate.
Binding Sites
Catalysis
Catalytic Domain
Enzymes
Kinetics
prisma
Psychological Inhibition
The molecular docking simulation was used to determine the binding energy of six antiretrovirals to the RdRp, ExoN-NSP10 and 3CLpro proteins of SARS-CoV-2. These proteins are necessary for viral RNA replication and polyprotein processing [12] (link). The crystal structures of RdRp (Identification code, ID: 6M71) [8] (link), ExoN-NSP10 (ID:7MC6) [37] (link) and 3CLpro (ID: 6M2N) [38] (link) were obtained from the Protein Data Bank (PDB) [39] (link). The resolution structures selected were lower than 3 Å [40] (link). The proteins were subjected to preparation by using Discovery Studio [41] and AutoDockTools (ADT). The active forms of the antiretrovirals [42] (link) were drawn and optimized by using Avogadro software [43] (link) and ADT. Remdesivir [44] (link),[45] (link), pibrentasvir [46] (link) and CQ [47] (link),[48] (link) were used as positive controls of the interaction with RdRp, ExoN-NSP10 and 3CLpro, respectively.
PrankWeb [49] (link) was used to determine the number of pockets and the amino acid residues that comprise them. This program also described the size (volume), depth, surface area or general hydrophobicity of each pocket (Table 1 ). In addition, Protein plus [50] (link) was implemented to verify the number of pockets obtained by PrankWeb [49] (link), and to describe their characteristics (size, shapes, amino acids composition and descriptor functional groups). The pockets were selected according to the active site or catalytic domain for each protein, as based on previous reports [16] (link),[51] (link),[52] (link).
Couplings were carried out using AutoDock Vina version 4.2.6 [53] (link), with an exhaustiveness value of 20 and a grid box of 24 Å × 24 Å × 24 Å, centered at (116.7829 Å, 109.9570 Å, 123.9430 Å) (XYZ coordinates) for RdRp (PDB ID: 6M71), (28.6904 Å, −1.9647 Å, 13.6836 Å) for ExoN-NSP10 (PDB: 7MC6) and (−47.585 Å, 1.135 Å, −5.600 Å) for 3CLpro (PDB ID:6M2N) (Table 1 ). The best docking conformation of protein-ligand interactions was predicted based on the binding energy value (kcal/mol). The docked structures were analyzed and visualized by using BIOVIA Discovery Studio Visualizer 16.1.
PrankWeb [49] (link) was used to determine the number of pockets and the amino acid residues that comprise them. This program also described the size (volume), depth, surface area or general hydrophobicity of each pocket (
Couplings were carried out using AutoDock Vina version 4.2.6 [53] (link), with an exhaustiveness value of 20 and a grid box of 24 Å × 24 Å × 24 Å, centered at (116.7829 Å, 109.9570 Å, 123.9430 Å) (XYZ coordinates) for RdRp (PDB ID: 6M71), (28.6904 Å, −1.9647 Å, 13.6836 Å) for ExoN-NSP10 (PDB: 7MC6) and (−47.585 Å, 1.135 Å, −5.600 Å) for 3CLpro (PDB ID:6M2N) (
Amino Acids
Catalysis
Catalytic Domain
DNA Replication
Exons
Ligands
Molecular Docking Simulation
pibrentasvir
Polyproteins
Protein Domain
Proteins
remdesivir
RNA, Viral
RNA Replication
SARS-CoV-2
Virus Replication
Three-dimensional structure of wild and mutant models of catalytic site domain was constructed using MODELLER v9.24 [33 (link)]. Thereafter, all models were refined using galaxy refine server and validated from Ramachandran plot using PROCHECK. Furthermore, energy minimization of both wild and mutant structures was executed using Chimera 1.1.5 software [34 (link)]. PyMOL software was used for calculating RMSD of mutant models w.r.t wild model.
Catalytic Domain
Chimera
Strains carrying the auxin-inducible degron (AID*) for RNase H2 (catalytic subunit Rnh201) were created as described before27 (link). Plasmids and oligonucleotides are listed in Supplementary Data 3 .
Auxins
Catalytic Domain
Endoribonucleases
Oligonucleotides
Plasmids
Strains
Plasmid MLM3636, expressing single-guide RNAs in mammalian cells, was a kind gift from Keith Joung (Addgene plasmid # 43860). Plasmid pMLM2.0 was constructed from MLM3636 by replacing the sgRNA expression cassette with the sgRNA2.0 expression cassette of the lentiviral plasmid sgRNA (MS2)_zeo backbone (Addgene plasmid # 61427) [26 (link)]. Stretches of 20 bp sgRNA to target UCHL1 were designed using the online tool (http://crispr.mit.edu/ ). Double-stranded oligonucleotides containing these targeting sites (listed in Supplemental Table 2) were cloned into BsmBI-digested pMLM2.0.
The plasmid containing the gene of the mammalian codon-optimized dCas9-VP64 activator (a tetramer of the viral VP16 transcriptional activator) was a kind gift from Keith Joung (Addgene; pMLM3705, #47754). An additional multiple-cloning site was inserted by replacing the VP64 coding sequence in dCas9-VP64 with a sequence containing a PacI restriction site, the new plasmid was referred to as No Effector Domain, pdCas9-NED (Addgene #109358) [53 (link)].
Plasmids expressing C-terminally mCherry-tagged dCas9-MSssI (Q147L/E186A) in mammalian cells were constructed as follows [54 (link)]: to create in-frame fusion between dCas9-MSssI and the P2A-mCherry-tag, the double-stranded oligonucleotide AK473-AK474 (5’-CGCGCCCAT ATGTTAATTAACAATTAA/5’-CCGGTTAATTGTTAATTAACATATGGG) was inserted between the SgsI (AscI) and BshTI (AgeI) restriction sites of pSYC-187 (Addgene#74,794). Insertion of AK473-AK474 preserved the flanking restriction sites, introduced a unique PacI site (underlined) and an in-frame stop codon. To abolish the stop codon, a short oligo-duplex (AK481-AK482, 5’-TAAGGTACCGA/5’-CCGGTCGGTACCTTAAT) was cloned between the PacI and the BshTI (AgeI) sites to obtain the plasmid pMCS-P2A-mCherry. (MCS stands for a sequence containing several restriction sites.) The SgsI (AscI) and Eco105I (SnaBI) fragment encoding the ‘MCS-P2A-mChery’ fragment was excised from pMCS-P2A-mCherry and cloned between the SgsI (AscI) and MssI (PmeI) sites of pdCas9-NED. The resulting plasmid pM-dCas9-(NED)-P2A_mCherry encodes a dCas9-P2A-mCherry fusion. The coding sequences of the MSssI (Q147L) and MSssI (E186A) variants were inserted, on SgsI-PacI fragments, between the SgsI and PacI sites of pdCas9-(NED)-P2A_mCherry. The new plasmids named pM-dCas9-MsssI (Q147L)-P2A-mCherry and pM-dCas9-MsssI (E186A)-P2A-mCherry express the respective dCas9-MSssI variant carrying the self-cleavable mCherry-tag. The catalytic domain of H3K27 histone methyltransferase enhancer of zeste homolog 2 (EZH2) was amplified with overhangs containing AscI and PacI restriction sites by PCR from pdCas9-EZH2. The EZH2 catalytic domain was subcloned into the pM-dCas9-NED-P2A-mCherry plasmid to yield pM-dCas9-EZH2-P2A-mCherry. The P2A-mCherry coding sequence was from the plasmid pSYC-187 [55 (link)], which was a kind gift from Seok-Yong Choi (Addgene plasmid # 74794). The catalytic domain of PRDM9 and DOT1L from dCas9-PRDM9 and dCas9-DOT1L, constructed as previously described [47 (link)], were subcloned into the pM-dCas9-NED-P2A-mCherry plasmid.
UCHL1 full-length cDNA (669 bp) was amplified from BEAS-2B cells and inserted into pcDNA4/HisMaxA between the BamHI and XbaI sites to generate the pcDNA4-UCHL1. Structure of the recombinant plasmids was confirmed by sequencing.
The plasmid containing the gene of the mammalian codon-optimized dCas9-VP64 activator (a tetramer of the viral VP16 transcriptional activator) was a kind gift from Keith Joung (Addgene; pMLM3705, #47754). An additional multiple-cloning site was inserted by replacing the VP64 coding sequence in dCas9-VP64 with a sequence containing a PacI restriction site, the new plasmid was referred to as No Effector Domain, pdCas9-NED (Addgene #109358) [53 (link)].
Plasmids expressing C-terminally mCherry-tagged dCas9-MSssI (Q147L/E186A) in mammalian cells were constructed as follows [54 (link)]: to create in-frame fusion between dCas9-MSssI and the P2A-mCherry-tag, the double-stranded oligonucleotide AK473-AK474 (5’-CGCGCCCAT ATGTTAATTAACAATTAA/5’-CCGGTTAATTGTTAATTAACATATGGG) was inserted between the SgsI (AscI) and BshTI (AgeI) restriction sites of pSYC-187 (Addgene#74,794). Insertion of AK473-AK474 preserved the flanking restriction sites, introduced a unique PacI site (underlined) and an in-frame stop codon. To abolish the stop codon, a short oligo-duplex (AK481-AK482, 5’-TAAGGTACCGA/5’-CCGGTCGGTACCTTAAT) was cloned between the PacI and the BshTI (AgeI) sites to obtain the plasmid pMCS-P2A-mCherry. (MCS stands for a sequence containing several restriction sites.) The SgsI (AscI) and Eco105I (SnaBI) fragment encoding the ‘MCS-P2A-mChery’ fragment was excised from pMCS-P2A-mCherry and cloned between the SgsI (AscI) and MssI (PmeI) sites of pdCas9-NED. The resulting plasmid pM-dCas9-(NED)-P2A_mCherry encodes a dCas9-P2A-mCherry fusion. The coding sequences of the MSssI (Q147L) and MSssI (E186A) variants were inserted, on SgsI-PacI fragments, between the SgsI and PacI sites of pdCas9-(NED)-P2A_mCherry. The new plasmids named pM-dCas9-MsssI (Q147L)-P2A-mCherry and pM-dCas9-MsssI (E186A)-P2A-mCherry express the respective dCas9-MSssI variant carrying the self-cleavable mCherry-tag. The catalytic domain of H3K27 histone methyltransferase enhancer of zeste homolog 2 (EZH2) was amplified with overhangs containing AscI and PacI restriction sites by PCR from pdCas9-EZH2. The EZH2 catalytic domain was subcloned into the pM-dCas9-NED-P2A-mCherry plasmid to yield pM-dCas9-EZH2-P2A-mCherry. The P2A-mCherry coding sequence was from the plasmid pSYC-187 [55 (link)], which was a kind gift from Seok-Yong Choi (Addgene plasmid # 74794). The catalytic domain of PRDM9 and DOT1L from dCas9-PRDM9 and dCas9-DOT1L, constructed as previously described [47 (link)], were subcloned into the pM-dCas9-NED-P2A-mCherry plasmid.
UCHL1 full-length cDNA (669 bp) was amplified from BEAS-2B cells and inserted into pcDNA4/HisMaxA between the BamHI and XbaI sites to generate the pcDNA4-UCHL1. Structure of the recombinant plasmids was confirmed by sequencing.
Catalytic Domain
Cells
Clustered Regularly Interspaced Short Palindromic Repeats
Codon
Codon, Terminator
DNA, Complementary
DOT1L protein, human
Exons
EZH2 protein, human
Genes
Mammals
Methyltransferase, Histone
Oligonucleotides
Open Reading Frames
Paramyotonia Congenita
Plasmids
Reading Frames
RNA, Single Guide
Tetrameres
UCHL1 protein, human
Vertebral Column
Viral Transcription
VP-16
Top products related to «Catalytic Domain»
Sourced in United States, China, Germany, United Kingdom, Canada, Japan, France, Italy, Switzerland, Australia, Spain, Belgium, Denmark, Singapore, India, Netherlands, Sweden, New Zealand, Portugal, Poland, Israel, Lithuania, Hong Kong, Argentina, Ireland, Austria, Czechia, Cameroon, Taiwan, Province of China, Morocco
Lipofectamine 2000 is a cationic lipid-based transfection reagent designed for efficient and reliable delivery of nucleic acids, such as plasmid DNA and small interfering RNA (siRNA), into a wide range of eukaryotic cell types. It facilitates the formation of complexes between the nucleic acid and the lipid components, which can then be introduced into cells to enable gene expression or gene silencing studies.
Sourced in United States, China, Japan, Germany, United Kingdom, Canada, France, Italy, Australia, Spain, Switzerland, Netherlands, Belgium, Lithuania, Denmark, Singapore, New Zealand, India, Brazil, Argentina, Sweden, Norway, Austria, Poland, Finland, Israel, Hong Kong, Cameroon, Sao Tome and Principe, Macao, Taiwan, Province of China, Thailand
TRIzol reagent is a monophasic solution of phenol, guanidine isothiocyanate, and other proprietary components designed for the isolation of total RNA, DNA, and proteins from a variety of biological samples. The reagent maintains the integrity of the RNA while disrupting cells and dissolving cell components.
Sourced in Germany, United States, United Kingdom, Netherlands, China, Switzerland, Italy, Canada, Spain, India
Ni-NTA agarose is a solid-phase affinity chromatography resin designed for the purification of recombinant proteins containing a histidine-tag. It consists of nickel-nitrilotriacetic acid (Ni-NTA) coupled to agarose beads, which selectively bind to the histidine-tagged proteins.
Sourced in United States, Hungary
AutoDock Tools is a software suite designed to perform molecular docking simulations. It provides a graphical user interface (GUI) for preparing input files, running docking calculations, and analyzing the results. The core function of AutoDock Tools is to predict the preferred binding orientations and affinities between a small molecule and a target protein.
Sourced in United States, Germany, United Kingdom, China, Sao Tome and Principe, Italy, Japan, Macao, Spain, Canada, France, Switzerland, Ireland, Sweden, Australia
ATP is a laboratory instrument used to measure the presence and concentration of adenosine triphosphate (ATP) in various samples. ATP is a key molecule involved in energy transfer within living cells. The ATP product provides a reliable and accurate method for quantifying ATP levels, which is useful in applications such as microbial detection, cell viability assessment, and ATP-based assays.
Sourced in United States, Germany, United Kingdom, Canada
PyMOL is a molecular visualization software package for rendering and animating 3D molecular structures. It allows users to display, analyze, and manipulate molecular models, providing a powerful tool for research in fields such as biochemistry, structural biology, and drug design.
Sourced in United States
Maestro is a computational modeling software developed by Schrödinger. It is designed to assist researchers in visualizing and analyzing molecular structures and interactions. The core function of Maestro is to provide a comprehensive platform for molecular modeling and simulation.
Sourced in United States, Sweden, United Kingdom, Germany, Japan
The Superdex 200 column is a size-exclusion chromatography media used for the separation and purification of proteins, peptides, and other biomolecules. It is designed to provide efficient separation and high resolution across a wide range of molecular weights. The column is suitable for a variety of applications, including protein analysis, desalting, and buffer exchange.
Sourced in United States, United Kingdom, Germany, China, Canada, Singapore, Japan, Morocco, France
The Q5 Site-Directed Mutagenesis Kit is a laboratory tool designed for introducing precise mutations into DNA sequences. It provides a streamlined workflow for generating site-specific changes in plasmid or linear DNA templates.
The PKA catalytic subunit is a lab equipment product that serves a core function in the regulation of cellular processes. It is a key component in the protein kinase A (PKA) signaling pathway, responsible for the phosphorylation of various target proteins. The PKA catalytic subunit acts as the active, enzymatic portion of the PKA complex, catalyzing the transfer of phosphate groups to specific substrates.
More about "Catalytic Domain"
Catalytic Domain: The Heart of Enzyme Function Enzymes are the workhorses of biological processes, catalyzing a wide range of chemical reactions essential for life.
At the core of an enzyme's function is its catalytic domain - a discrete structural unit responsible for its catalytic activity.
This region of the protein typically contains the active site where the substrate binds and the chemical transformation occurs.
Understanding the catalytic domain is crucial for studying enzyme mechanism, function, and inhibition.
Researchers utilize various techniques to identify and characterize this crucial domain, including the use of tools like Lipofectamine 2000 for transfection, TRIzol reagent for RNA extraction, Ni-NTA agarose for protein purification, and AutoDock Tools for molecular docking simulations.
The catalytic domain often contains key amino acid residues that participate in catalysis, such as those involved in substrate binding, proton transfer, or stabilizing reaction intermediates.
By studying the structure and dynamics of the catalytic domain, scientists can gain valuable insights into the enzyme's mechanism of action and potential target sites for inhibition, as demonstrated by the use of molecules like ATP and the PyMOL Molecular Graphics System.
Moreover, techniques like site-directed mutagenesis, using tools like the Q5 Site-Directed Mutagenesis Kit, allow researchers to systematically investigate the role of specific amino acids within the catalytic domain, revealing their contributions to enzyme function and potential for therapeutic targeting.
Computational approaches, such as those employing the Maestro software and Superdex 200 column for protein purification, also play a crucial role in understanding catalytic domains.
These methods can help predict and analyze the structure, dynamics, and interactions of the catalytic domain, ultimately leading to the development of more effective enzyme-targeted therapies.
By harnessing the power of PubCompare.ai's AI-driven platform, researchers can effortlessly locate and compare catalytic domain protocols from literature, preprints, and patents, optimizing their research process and making data-driven decisions to advance our understanding of this critical enzyme component.
At the core of an enzyme's function is its catalytic domain - a discrete structural unit responsible for its catalytic activity.
This region of the protein typically contains the active site where the substrate binds and the chemical transformation occurs.
Understanding the catalytic domain is crucial for studying enzyme mechanism, function, and inhibition.
Researchers utilize various techniques to identify and characterize this crucial domain, including the use of tools like Lipofectamine 2000 for transfection, TRIzol reagent for RNA extraction, Ni-NTA agarose for protein purification, and AutoDock Tools for molecular docking simulations.
The catalytic domain often contains key amino acid residues that participate in catalysis, such as those involved in substrate binding, proton transfer, or stabilizing reaction intermediates.
By studying the structure and dynamics of the catalytic domain, scientists can gain valuable insights into the enzyme's mechanism of action and potential target sites for inhibition, as demonstrated by the use of molecules like ATP and the PyMOL Molecular Graphics System.
Moreover, techniques like site-directed mutagenesis, using tools like the Q5 Site-Directed Mutagenesis Kit, allow researchers to systematically investigate the role of specific amino acids within the catalytic domain, revealing their contributions to enzyme function and potential for therapeutic targeting.
Computational approaches, such as those employing the Maestro software and Superdex 200 column for protein purification, also play a crucial role in understanding catalytic domains.
These methods can help predict and analyze the structure, dynamics, and interactions of the catalytic domain, ultimately leading to the development of more effective enzyme-targeted therapies.
By harnessing the power of PubCompare.ai's AI-driven platform, researchers can effortlessly locate and compare catalytic domain protocols from literature, preprints, and patents, optimizing their research process and making data-driven decisions to advance our understanding of this critical enzyme component.