21,988 results on '"Catalytic Domain"'
Search Results
2. The topography of lactose synthesis. 1975.
- Author
-
Kuhn NJ and White A
- Subjects
- Animals, Catalytic Domain, Female, Glucose metabolism, Golgi Apparatus metabolism, History, 20th Century, Lactation, Rats, Rats, Wistar, Biology history, Lactose biosynthesis, Mammary Glands, Animal metabolism
- Published
- 2009
- Full Text
- View/download PDF
3. Structural and mechanistic insights into the substrate specificity and hydrolysis of GH31 α-N-acetylgalactosaminidase
- Author
-
Marina Ikegaya, Takatsugu Miyazaki, and Santiago Alonso-Gil
- Subjects
Glycoside Hydrolases ,Stereochemistry ,N-acetylgalactosamine ,Tn antigen ,Peptide ,Quantum mechanics ,Biochemistry ,Substrate Specificity ,alpha-N-Acetylgalactosaminidase ,N-Acetylgalactosamine ,chemistry.chemical_compound ,Catalytic Domain ,Glycoside hydrolase family 31 ,Gut bacteria ,chemistry.chemical_classification ,biology ,Hydrolysis ,Active site ,General Medicine ,Fetuin ,O-glycan ,Enzyme ,chemistry ,Docking (molecular) ,Mucin ,biology.protein - Abstract
Glycoside hydrolase family 31 (GH31) is a diversified family of anomer-retaining α-glycoside hydrolases, such as α-glucosidase and α-xylosidase, among others. Recently, GH31 α-N-acetylgalactosaminidases (Nag31s) have been identified to hydrolyze the core of mucin-type O-glycans and the crystal structure of a gut bacterium Enterococcus faecalis Nag31 has been reported. However, the mechanisms of substrate specificity and hydrolysis of Nag31s are not well investigated. Herein, we show that E. faecalis Nag31 has the ability to release N-acetylgalactosamine (GalNAc) from O-glycoproteins, such as fetuin and mucin, but has low activity against Tn antigen. Mutational analysis and crystal structures of the Michaelis complexes reveal that residues of the active site work in concert with their conformational changes to act on only α-N-acetylgalactosaminides. Docking simulations using GalNAc-attached peptides suggest that the enzyme mainly recognizes GalNAc and side chains of Ser/Thr, but not strictly other peptide residues. Moreover, quantum mechanics calculations indicate that the enzyme preferred p-nitrophenyl α-N-acetylgalactosaminide to Tn antigen and that the hydrolysis progresses through a conformational itinerary, 4C1 → 1S3 → 4C1, in GalNAc of substrates. Our results provide novel insights into the diversification of the sugar recognition and hydrolytic mechanisms of GH31 enzymes.
- Published
- 2022
- Full Text
- View/download PDF
4. Structure basis of the caffeic acid O-methyltransferase from Ligusiticum chuanxiong to understand its selective mechanism
- Author
-
Yan Zicheng, Simin Song, Anqi Chen, Jianquan Zhu, Hai Liao, Jiayu Zhou, Yamei Yu, and Qiuju An
- Subjects
Models, Molecular ,Protein Conformation ,Stereochemistry ,Biochemistry ,Catalysis ,Substrate Specificity ,Structure-Activity Relationship ,chemistry.chemical_compound ,Residue (chemistry) ,Phenols ,Structural Biology ,Catalytic Domain ,Methyl caffeate ,Caffeic acid ,Ligusticum ,Amino Acid Sequence ,Carboxylate ,Molecular Biology ,Ternary complex ,Phylogeny ,biology ,Chemistry ,Hydrogen bond ,Active site ,Methyltransferases ,General Medicine ,Ligand (biochemistry) ,Recombinant Proteins ,Kinetics ,Mutagenesis, Site-Directed ,biology.protein - Abstract
Caffeic acid O-methyltransferase from Ligusticum chuanxiong (LcCOMT) showed strict regiospecificity despite a relative degree of preference. Compared with caffeic acid, methyl caffeate was the preferential substrate by its low Km and high Kcat. In this study, we obtained the SAM binary (1.80 A) and SAH binary (1.95 A) complex LcCOMT crystal structures, and established the ternary complex structure with methyl caffeate by molecular docking. The active site of LcCOMT included phenolic substrate pocket, SAM/SAH ligand pocket and conserved catalytic residues as well. The regiospecificity of LcCOMT that permitted only 3-hydroxyl group to be methylated arise from the interactions between the active site and the phenyl ring. However, the propanoid tail governed the relative preference of LcCOMT. The ester group in methyl caffeate stabilized the anionic intermediate caused by His268-Asp269 pair, whereas caffeic acid was unable to stabilize the anionic intermediate due to the adjacent carboxylate anion in the propanoid tail. Ser183 residue formed an additional hydrogen bond with SAH and its role was identified by S183A mutation. Ile318 residue might be a potential site for determination of substrate preference, and its mutation led to the change of tertiary conformation. The results supported the selective mechanism of LcCOMT.
- Published
- 2022
- Full Text
- View/download PDF
5. Structural basis for the substrate recognition mechanism of ATP-sulfurylase domain of human PAPS synthase 2
- Author
-
Zhaoyuan Hou, Hai Gao, Liang Zhang, Pan Zhang, Houwen Lin, and Lin Zhang
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Genetic Vectors ,Phosphoadenosine Phosphosulfate ,Biophysics ,Gene Expression ,Phenylalanine ,Crystallography, X-Ray ,Biochemistry ,Substrate Specificity ,chemistry.chemical_compound ,Adenosine Triphosphate ,Sulfation ,Multienzyme Complexes ,Catalytic Domain ,Escherichia coli ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Cloning, Molecular ,Molecular Biology ,chemistry.chemical_classification ,Sequence Homology, Amino Acid ,ATP synthase ,biology ,Drug discovery ,Substrate (chemistry) ,Cell Biology ,Recombinant Proteins ,Sulfate Adenylyltransferase ,Neoplasm Proteins ,3'-Phosphoadenosine-5'-phosphosulfate ,Enzyme ,chemistry ,biology.protein ,Thermodynamics ,Protein Conformation, beta-Strand ,Sequence Alignment ,Function (biology) ,Protein Binding - Abstract
Sulfation is an essential modification on biomolecules in living cells, and 3'-Phosphoadenosine-5'-phosphosulfate (PAPS) is its unique and universal sulfate donor. Human PAPS synthases (PAPSS1 and 2) are the only enzymes that catalyze PAPS production from inorganic sulfate. Unexpectedly, PAPSS1 and PAPSS2 do not functional complement with each other, and abnormal function of PAPSS2 but not PAPSS1 leads to numerous human diseases including bone development diseases, hormone disorder and cancers. Here, we reported the crystal structures of ATP-sulfurylase domain of human PAPSS2 (ATPS2) and ATPS2 in complex with is product 5'-phosphosulfate (APS). We demonstrated that ATPS2 recognizes the substrates by using family conserved residues located on the HXXH and PP motifs, and achieves substrate binding and releasing by employing a non-conserved phenylalanine (Phe550) through a never observed flipping mechanism. Our discovery provides additional information to better understand the biological function of PAPSS2 especially in tumorigenesis, and may facilitate the drug discovery against this enzyme.
- Published
- 2022
- Full Text
- View/download PDF
6. Dimeric architecture of maltodextrin glucosidase (MalZ) provides insights into the substrate recognition and hydrolysis mechanism
- Author
-
Eui-Jeon Woo, Yan An, Kyung-Mo Song, Byung-Ha Oh, Su-Jin Lee, Jong-Tae Park, Woo-Chan Ahn, and Kwang-Hyun Park
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Glycoside Hydrolases ,Stereochemistry ,Genetic Vectors ,Biophysics ,Gene Expression ,Crystallography, X-Ray ,Biochemistry ,Substrate Specificity ,chemistry.chemical_compound ,Residue (chemistry) ,Hydrolysis ,Polysaccharides ,Catalytic Domain ,Escherichia coli ,Protein Interaction Domains and Motifs ,Cloning, Molecular ,Molecular Biology ,chemistry.chemical_classification ,biology ,Escherichia coli Proteins ,Active site ,Substrate (chemistry) ,Cell Biology ,Protein engineering ,Maltose ,Maltodextrin ,Recombinant Proteins ,Glucose ,Enzyme ,chemistry ,Biocatalysis ,biology.protein ,Protein Conformation, beta-Strand ,Protein Multimerization ,Protein Binding - Abstract
Maltodextrin glucosidase (MalZ) is a key enzyme in the maltose utilization pathway in Escherichia coli that liberates glucose from the reducing end of the short malto-oligosaccharides. Unlike other enzymes in the GH13_21 subfamily, the hydrolytic activity of MalZ is limited to maltodextrin rather than long starch substrates, forming various transglycosylation products in α-1,3, α-1,4 or α-1,6 linkages. The mechanism for the substrate binding and hydrolysis of this enzyme is not well understood yet. Here, we present the dimeric crystal structure of MalZ, with the N-domain generating a unique substrate binding groove. The N-domain bears CBM34 architecture and forms a part of the active site in the catalytic domain of the adjacent molecule. The groove found between the N-domain and catalytic domain from the adjacent molecule, shapes active sites suitable for short malto-oligosaccharides, but hinders long stretches of oligosaccharides. The conserved residue of E44 protrudes at subsite +2, elucidating the hydrolysis pattern of the substrate by the glucose unit from the reducing end. The structural analysis provides a molecular basis for the substrate specificity and the enzymatic property, and has potential industrial application for protein engineering.
- Published
- 2022
- Full Text
- View/download PDF
7. Consensus mutagenesis and computational simulation provide insight into the desaturation catalytic mechanism for delta 6 fatty acid desaturase
- Author
-
Xin Tang, Jie Cui, Hao Zhang, Haiqin Chen, Wei Chen, and Yong Q. Chen
- Subjects
Fatty Acid Desaturases ,Protein Conformation ,Genetic Vectors ,Saccharomyces cerevisiae ,Biophysics ,Gene Expression ,Biochemistry ,Substrate Specificity ,Linoleic Acid ,Chlorophyta ,Catalytic Domain ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Cloning, Molecular ,Site-directed mutagenesis ,Molecular Biology ,chemistry.chemical_classification ,Sequence Homology, Amino Acid ,biology ,Algal Proteins ,Mutagenesis ,Cell Biology ,biology.organism_classification ,Recombinant Proteins ,Delta-6-desaturase ,Amino acid ,Molecular Docking Simulation ,Fatty acid desaturase ,Enzyme ,chemistry ,Biocatalysis ,Mutagenesis, Site-Directed ,biology.protein ,Sequence Alignment ,Protein Binding ,Polyunsaturated fatty acid - Abstract
Fatty acid desaturase (FADS) generates double bond at a certain position of the corresponding polyunsaturated fatty acids (PUFAs) with high selectivity, the enzyme activity and PUFAs products of which are essential to biological systems and are associated with a variety of physiological diseases. Little is known about the structure of FADSs and their amino acid residues related to catalytic activities. Identifying key residues of Micromonas pusilla delta 6 desaturase (MpFADS6) provides a point of departure for a better understanding of desaturation. In this study, conserved amino acids were anchored through gene consensus analysis, thereby generating corresponding variants by site-directed mutagenesis. To achieve stable and high-efficiency expression of MpFADS6 and its variants in Saccharomyces cerevisiae, the key points of induced expression were optimized. The contribution of conserved residues to the function of enzyme was determined by analyzing enzyme activity of the variants. Molecular modeling indicated that these residues are essential to catalytic activities, or substrate binding. Mutants MpFADS6[Q409R] and MpFADS6[M242P] abolished desaturation, while MpFADS6[F419V] and MpFADS6[A374Q] significantly reduced catalytic activities. Given that certain residues have been identified to have a significant impact on MpFADS6 activities, it is put forward that histidine-conserved region III of FADS6 is related to electronic transfer during desaturation, while histidine-conserved regions I and II are related to desaturation. These findings provide new insights and methods to determine the structure, mechanism and directed transformation of membrane-bound desaturases.
- Published
- 2022
- Full Text
- View/download PDF
8. Implications of divergence of methionine adenosyltransferase in archaea
- Author
-
Saacnicteh Toledo-Patino, Bhanu Pratap Singh Chouhan, Desirae Martinez, Paola Laurino, and Madhuri Gade
- Subjects
S-Adenosylmethionine ,enzyme evolution ,Sequence analysis ,QH301-705.5 ,Protein domain ,methionine adenosyltransferase ,Biology ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,Methionine ,Biosynthesis ,Three-domain system ,Catalytic Domain ,ancestral sequence reconstruction ,catalytic interface ,Biology (General) ,Ancestral Sequence Reconstruction (ASR) ,Research Articles ,Active site ,biology.organism_classification ,Archaea ,chemistry ,Evolutionary biology ,Methionine Adenosyltransferase ,biology.protein ,divergence ,Research Article - Abstract
Methionine adenosyltransferase (MAT) catalyzes the biosynthesis of S‐adenosyl methionine from l‐methionine and ATP. MAT enzymes are ancient, believed to share a common ancestor, and are highly conserved in all three domains of life. However, the sequences of archaeal MATs show considerable divergence compared with their bacterial and eukaryotic counterparts. Furthermore, the structural significance and functional significance of this sequence divergence are not well understood. In the present study, we employed structural analysis and ancestral sequence reconstruction to investigate archaeal MAT divergence. We observed that the dimer interface containing the active site (which is usually well conserved) diverged considerably between the bacterial/eukaryotic MATs and archaeal MAT. A detailed investigation of the available structures supports the sequence analysis outcome: The protein domains and subdomains of bacterial and eukaryotic MAT are more similar than those of archaea. Finally, we resurrected archaeal MAT ancestors. Interestingly, archaeal MAT ancestors show substrate specificity, which is lost during evolution. This observation supports the hypothesis of a common MAT ancestor for the three domains of life. In conclusion, we have demonstrated that archaeal MAT is an ideal system for studying an enzyme family that evolved differently in one domain compared with others while maintaining the same catalytic activity., We investigated archaeal methionine adenosyltransferase (MAT) divergence by structural analysis and ancestral sequence reconstruction. The dimer interface of MAT containing the active site (usually well conserved) diverged considerably between the bacterial/eukaryotic MATs and archaeal MAT. The archaeal MAT ancestor showed substrate specificity, which was lost during evolution, supporting the hypothesis of a common MAT ancestor for the three domains of life.
- Published
- 2022
- Full Text
- View/download PDF
9. Crystal structure of a novel putative sugar isomerase from the psychrophilic bacterium Paenibacillus sp. R4
- Author
-
Jisub Hwang, Jun Hyuck Lee, Min Ju Lee, Hyun Ji Ha, Hyun Ho Park, Sunghark Kwon, Ji Hye Sung, and Yong Jun Kang
- Subjects
Models, Molecular ,Xylose isomerase ,Glucose-6-phosphate isomerase ,Protein Conformation ,Stereochemistry ,Biophysics ,Isomerase ,Crystallography, X-Ray ,Biochemistry ,Paenibacillus ,Bacterial Proteins ,Catalytic Domain ,TIM barrel ,Amino Acids ,Psychrophile ,Molecular Biology ,Binding Sites ,biology ,Chemistry ,Active site ,Cell Biology ,biology.organism_classification ,Hyperthermophile ,Metals ,biology.protein ,Triose-Phosphate Isomerase - Abstract
Sugar isomerases (SIs) catalyze the reversible conversion of aldoses to ketoses. A novel putative SI gene has been identified from the genome sequence information on the psychrophilic bacterium Paenibacillus sp. R4. Here, we report the crystal structure of the putative SI from Paenibacillus sp. R4 (PbSI) at 2.98 A resolution. It was found that the overall structure of PbSI adopts the triose-phosphate isomerase (TIM) barrel fold. PbSI was also identified to have two heterogeneous metal ions as its cofactors at the active site in the TIM barrel, one of which was confirmed as a Zn ion through X-ray anomalous scattering and inductively coupled plasma mass spectrometry analysis. Structural comparison with homologous SI proteins from mesophiles, hyperthermophiles, and a psychrophile revealed that key residues in the active site are well conserved and that dimeric PbSI is devoid of the extended C-terminal region, which tetrameric SIs commonly have. Our results provide novel structural information on the cold-adaptable SI, including information on the metal composition in the active site.
- Published
- 2021
- Full Text
- View/download PDF
10. Clinical and Molecular Characteristics of PRKACA L206R Mutant Cortisol-Producing Adenomas in Korean Patients
- Author
-
Kwangsoo Kim, Jang-Seok Lee, Moon Woo Seong, Jung Hee Kim, Su Jin Kim, Kyu Eun Lee, Ra-Young Song, Min-Kyeong Gwon, Seongmin Choi, and Insoon Jang
- Subjects
Adenoma ,Adult ,Male ,Hydrocortisone ,Endocrinology, Diabetes and Metabolism ,Adrenal Gland ,Mutant ,Steroid biosynthesis ,Biology ,Diseases of the endocrine glands. Clinical endocrinology ,Transcriptome ,Wnt signaling pathway ,Endocrinology ,Catalytic Domain ,medicine ,Cushing syndrome ,Humans ,PRKACA mutation ,Cyclic AMP-Dependent Protein Kinase Catalytic Subunits ,Adrenocortical adenomas ,Middle Aged ,medicine.disease ,RC648-665 ,Molecular biology ,Fold change ,Gene expression profiling ,Adrenal Cortex Neoplasms ,PRKACA ,Original Article ,Female ,Protein Kinases - Abstract
Background: An activating mutation (c.617A>C/p.Lys206Arg, L206R) in protein kinase cAMP-activated catalytic subunit alpha (PRKACA) has been reported in 35% to 65% of cases of cortisol-producing adenomas (CPAs). We aimed to compare the clinical characteristics and transcriptome analysis between PRKACA L206R mutants and wild-type CPAs in Korea.Methods: We included 57 subjects with CPAs who underwent adrenalectomy at Seoul National University Hospital. Sanger sequencing for PRKACA was conducted in 57 CPA tumor tissues. RNA sequencing was performed in 13 fresh-frozen tumor tissues.Results: The prevalence of the PRKACA L206R mutation was 51% (29/57). The mean age of the study subjects was 42±12 years, and 87.7% (50/57) of the patients were female. Subjects with PRKACA L206R mutant CPAs showed smaller adenoma size (3.3±0.7 cm vs. 3.8±1.2 cm, P=0.059) and lower dehydroepiandrosterone sulfate levels (218±180 ng/mL vs. 1,511±3,307 ng/mL, P=0.001) than those with PRKACA wild-type CPAs. Transcriptome profiling identified 244 differentially expressed genes (DEGs) between PRKACA L206R mutant (n=8) and wild-type CPAs (n=5), including five upregulated and 239 downregulated genes in PRKACA L206R mutant CPAs (|fold change| ≥2, PCTNNB1 was the most significant transcription regulator. In several pathway analyses, the Wnt signaling pathway was downregulated and the steroid biosynthesis pathway was upregulated in PRKACA mutants. Protein-protein interaction analysis also showed that PRKACA downregulates Wnt signaling and upregulates steroid biosynthesis.Conclusion: The PRKACA L206R mutation in CPAs causes high hormonal activity with a limited proliferative capacity, as supported by transcriptome profiling.
- Published
- 2021
11. Catalytic flexibility of rice glycosyltransferase OsUGT91C1 for the production of palatable steviol glycosides
- Author
-
Xinyu Xu, Yi Lin, Wei Cheng, Dan Ke, Yujie Chen, Yuquan Wei, Jinzhu Zhang, Jie Zhou, Minghai Tang, Jianxiong He, Haohao Dong, Wenxian Yang, Xiaofeng Zhu, and James H. Naismith
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Glycosylation ,Glycobiology ,General Physics and Astronomy ,Gene Expression ,Steviol ,Protein Engineering ,Substrate Specificity ,chemistry.chemical_compound ,0302 clinical medicine ,Glucosides ,Catalytic Domain ,Stevia ,Plant Proteins ,2. Zero hunger ,chemistry.chemical_classification ,0303 health sciences ,Multidisciplinary ,biology ,Recombinant Proteins ,Biochemistry ,Carbohydrate Sequence ,Taste ,Enzyme mechanisms ,Diterpenes, Kaurane ,Steviol glycoside ,Protein Binding ,Uridine Diphosphate Glucose ,Glycan ,Science ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Glycosyltransferase ,Humans ,Protein Interaction Domains and Motifs ,030304 developmental biology ,X-ray crystallography ,Glycoside ,Glycosyltransferases ,Oryza ,General Chemistry ,Stevia rebaudiana ,Kinetics ,Enzyme ,Aglycone ,Glucose ,chemistry ,Sweetening Agents ,biology.protein ,Biocatalysis ,Protein Conformation, beta-Strand ,030217 neurology & neurosurgery - Abstract
Steviol glycosides are the intensely sweet components of extracts from Stevia rebaudiana. These molecules comprise an invariant steviol aglycone decorated with variable glycans and could widely serve as a low-calorie sweetener. However, the most desirable steviol glycosides Reb D and Reb M, devoid of unpleasant aftertaste, are naturally produced only in trace amounts due to low levels of specific β (1–2) glucosylation in Stevia. Here, we report the biochemical and structural characterization of OsUGT91C1, a glycosyltransferase from Oryza sativa, which is efficient at catalyzing β (1–2) glucosylation. The enzyme’s ability to bind steviol glycoside substrate in three modes underlies its flexibility to catalyze β (1–2) glucosylation in two distinct orientations as well as β (1–6) glucosylation. Guided by the structural insights, we engineer this enzyme to enhance the desirable β (1–2) glucosylation, eliminate β (1–6) glucosylation, and obtain a promising catalyst for the industrial production of naturally rare but palatable steviol glycosides., Steviol glycosides from the plant Stevia rebaudiana are already used as lowcalorie sweeteners, but the most abundant naturally occurring compounds have a bitter aftertaste. Here, the authors characterize and engineer rice glycosyltransferase OsUGT91C1 to facilitate the large-scale production of naturally rare but palatable glycosides Reb D and Reb M
- Published
- 2021
12. Dynamic equilibria in protein kinases
- Author
-
Jake W. Anderson, Laurel M. Pegram, and Natalie G. Ahn
- Subjects
Protein kinase activation ,biology ,Kinase ,Chemistry ,Allosteric regulation ,Active site ,Crystallography, X-Ray ,Article ,law.invention ,Structural Biology ,Regulatory sequence ,law ,Catalytic Domain ,biology.protein ,Biophysics ,Electron paramagnetic resonance ,Protein Kinases ,Molecular Biology ,Conformational ensembles - Abstract
Structural changes involved in protein kinase activation and ligand binding have been determined from a wealth of X-ray crystallographic evidence. Recent solution studies using NMR, EPR, HX-MS, and fluorescence techniques have deepened this understanding by highlighting the underlying energetics and dynamics of multistate conformational ensembles. This new research is showing how activation mechanisms and ligand binding alter the internal motions of kinases and enable allosteric coupling between distal regulatory regions and the active site.
- Published
- 2021
- Full Text
- View/download PDF
13. Molecular cloning and functional characterization of UGTs from Glycyrrhiza uralensis flavonoid pathway
- Author
-
Yan Yin, Ping Li, Chunsheng Liu, Dan Jiang, and Guangxi Ren
- Subjects
Models, Molecular ,Glycosylation ,Molecular Conformation ,Gene Expression ,Molecular cloning ,Biochemistry ,chemistry.chemical_compound ,Structural Biology ,Catalytic Domain ,Complementary DNA ,Glycyrrhiza uralensis ,Amino Acid Sequence ,Cloning, Molecular ,Molecular Biology ,Chromatography, High Pressure Liquid ,Phylogeny ,Flavonoids ,Molecular Structure ,biology ,Chemistry ,Gene Expression Profiling ,Glycosyltransferases ,General Medicine ,biology.organism_classification ,Recombinant Proteins ,Enzyme Activation ,Flavonoid biosynthesis ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Liquiritigenin ,Metabolic Networks and Pathways ,Liquiritin ,Isoliquiritigenin - Abstract
Glycyrrhiza uralensis Fisch., a well-known medicinal plant, contains flavonoids including liquiritigenin and isoliquiritigenin, and their corresponding glycoside liquiritin and isoliquiritin. Although some genes encoding UDP-glycosyltransferases (UGTs) have been functionally characterized in G. uralensis, other UGTs mechanisms of glycosylation remain to be elucidated. Against this background the aim of the present study included cloning and characterization of two full-length cDNA clones of GuUGT isoforms from the UGT multigene family. These included GuUGT2 (NCBI acc. MK341791) and GuUGT3 (NCBI acc. MK341793) with an ORF of 1473 and 1332 bp, respectively. Multiple alignments and phylogenetic analysis revealed GuUGTs protein of Glycine max had a high homology to that of other plants. Meanwhile, quantitative real-time PCR was performed to detect the transcript levels of GuUGTs in different tissues. The results indicated that GuUGTs was more expressed in roots as compared to the leaves, and significantly up-regulated upon NaCl stress. The recombinant protein was heterologous expressed in Escherichia coli and exhibited a high level of UGT activity, catalyzing formation of isoliquiritin and liquiritin from isoliquiritigenin and liquiritigenin. The key residues of GuUGT2 for liquiritigenin glycosylation (Asn223), isoliquiritigenin (Asp272) were predicted by molecular docking and residue scanning based on simulated mutations. These results could serve as an important reference to understand the function of the UGT family. In addition, the identification of GuUGT2 and GuUGT3 provides a foundation for future studies of flavonoid biosynthesis in G. uralensis.
- Published
- 2021
- Full Text
- View/download PDF
14. Second-Shell Amino Acid R266 Helps Determine N-Succinylamino Acid Racemase Reaction Specificity in Promiscuous N-Succinylamino Acid Racemase/o-Succinylbenzoate Synthase Enzymes
- Author
-
Frank M. Raushel, James C. Sacchettini, Dat P. Truong, Mingzhao Zhu, Daniel Romo, Margaret E. Glasner, Simon Rousseau, Benjamin W. Machala, Kenneth G. Hull, and Jamison P. Huddleston
- Subjects
Models, Molecular ,Stereochemistry ,Amycolatopsis ,Crystallography, X-Ray ,Biochemistry ,Article ,Substrate Specificity ,Evolution, Molecular ,03 medical and health sciences ,Residue (chemistry) ,Bacterial Proteins ,Catalytic Domain ,Enzyme Stability ,Amino Acid Sequence ,Carbon-Carbon Lyases ,Conserved Sequence ,Amino Acid Isomerases ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,ATP synthase ,Chemistry ,030302 biochemistry & molecular biology ,Substrate (chemistry) ,Active site ,biology.organism_classification ,Recombinant Proteins ,Amino acid ,Glutamine ,Kinetics ,Enzyme ,Amino Acid Substitution ,Biocatalysis ,Mutagenesis, Site-Directed ,biology.protein - Abstract
Catalytic promiscuity is the coincidental ability to catalyze non-biological reactions in the same active site as the native biological reaction. Several lines of evidence show that catalytic promiscuity plays a role in the evolution of new enzyme functions. Thus, studying catalytic promiscuity can help identify structural features that predispose an enzyme to evolve new functions. This study identifies a potentially pre-adaptive residue in a promiscuous N-succinylamino acid racemase/o-succinylbenzoate synthase (NSAR/OSBS) enzyme from Amycolatopsis sp. T-1–60. This enzyme belongs to a branch of the OSBS family which includes many catalytically promiscuous NSAR/OSBS enzymes. R266 is conserved in all members of the NSAR/OSBS subfamily. However, the homologous position is usually hydrophobic in other OSBS subfamilies, whose enzymes lack NSAR activity. The second-shell amino acid R266 is close to the catalytic acid/base K263, but it does not contact the substrate, suggesting that R266 could affect the catalytic mechanism. Mutating R266 to glutamine in Amycolatopsis NSAR/OSBS profoundly reduces NSAR activity, but moderately reduces OSBS activity. This is due to a 1000-fold decrease in the rate of proton exchange between the substrate and the general acid/base catalyst K263. This mutation is less deleterious for the OSBS reaction because K263 forms a cation-π interaction with the OSBS substrate and/or the intermediate, rather than acting as a general acid/base catalyst. Together, the data explain how R266 contributes to NSAR reaction specificity and was likely an essential preadaptation for the evolution of NSAR activity.
- Published
- 2021
- Full Text
- View/download PDF
15. Diverse, Potent, and Efficacious Inhibitors That Target the EED Subunit of the Polycomb Repressive Complex 2 Methyltransferase
- Author
-
David Longmire, Haley Woods, Phillip B Rawlins, Jessie Hao-Ru Hsu, Daniel H O' Donovan, Alexandra Borodovsky, Peng Wang, Sharan K Bagal, Shaun M Fillery, Peter Barton, Clare Gregson, Beth Williamson, Samuel C Nash, Andrew Pike, Jon A Read, Kurt Gordon Pike, Andrew Bloecher, Erin Code, Minhui Shen, Sameer Kawatkar, Youfeng Nai, Jia Tang, Chengzhi Li, and James C. Robinson
- Subjects
Methyltransferase ,Protein subunit ,Allosteric regulation ,macromolecular substances ,Ligands ,Cell Line ,Structure-Activity Relationship ,Allosteric Regulation ,Heterocyclic Compounds ,Catalytic Domain ,Drug Discovery ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Enzyme Inhibitors ,Enhancer ,Cell Proliferation ,biology ,Chemistry ,EZH2 ,Polycomb Repressive Complex 2 ,Small molecule ,Rats ,Histone methyltransferase ,biology.protein ,Cancer research ,Molecular Medicine ,PRC2 - Abstract
Aberrant activity of the histone methyltransferase polycomb repressive complex 2 (PRC2) has been linked to several cancers, with small-molecule inhibitors of the catalytic subunit of the PRC2 enhancer of zeste homologue 2 (EZH2) being recently approved for the treatment of epithelioid sarcoma (ES) and follicular lymphoma (FL). Compounds binding to the EED subunit of PRC2 have recently emerged as allosteric inhibitors of PRC2 methyltransferase activity. In contrast to orthosteric inhibitors that target EZH2, small molecules that bind to EED retain their efficacy in EZH2 inhibitor-resistant cell lines. In this paper we disclose the discovery of potent and orally bioavailable EED ligands with good solubilities. The solubility of the EED ligands was optimized through a variety of design tactics, with the resulting compounds exhibiting in vivo efficacy in EZH2-driven tumors.
- Published
- 2021
- Full Text
- View/download PDF
16. Molecular insights into the inhibition of glutamate dehydrogenase by the dicarboxylic acid metabolites
- Author
-
Narayan S. Punekar, Prasenjit Bhaumik, Prem Prakash, and Barsa Kanchan Jyotshna Godsora
- Subjects
Lysine ,Coenzymes ,Tartrate ,Biochemistry ,Cofactor ,chemistry.chemical_compound ,Allosteric Regulation ,Glutamate Dehydrogenase ,Structural Biology ,Catalytic Domain ,Dicarboxylic Acids ,Amino Acid Sequence ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,biology ,Chemistry ,Glutamate dehydrogenase ,Active site ,Kinetics ,Aspergillus ,Malonate ,Dicarboxylic acid ,Metabolome ,biology.protein ,Ketoglutaric Acids ,Aspergillus niger ,NAD+ kinase ,Glutamate Dehydrogenase (NADP+) ,NADP ,Protein Binding - Abstract
Glutamate dehydrogenase (GDH) is a salient metabolic enzyme which catalyzes the NAD+ - or NADP+ -dependent reversible conversion of α-ketoglutarate (AKG) to L-glutamate; and thereby connects the carbon and nitrogen metabolism cycles in all living organisms. The function of GDH is extensively regulated by both metabolites (citrate, succinate, etc.) and non-metabolites (ATP, NADH, etc.) but sufficient molecular evidences are lacking to rationalize the inhibitory effects by the metabolites. We have expressed and purified NADP+ -dependent Aspergillus terreus GDH (AtGDH) in recombinant form. Succinate, malonate, maleate, fumarate and tartrate independently inhibit the activity of AtGDH to different extents. The crystal structures of AtGDH complexed with the dicarboxylic acid metabolites and the coenzyme NADPH have been determined. Although AtGDH structures are not complexed with substrate; surprisingly, they acquire super closed conformation like previously reported for substrate and coenzyme bound catalytically competent Aspergillus niger GDH (AnGDH). These dicarboxylic acid metabolites partially occupy the same binding pocket as substrate; but interact with varying polar interactions and the coenzyme NADPH binds to the Domain-II of AtGDH. The low inhibition potential of tartrate as compared to other dicarboxylic acid metabolites is due to its weaker interactions of carboxylate groups with AtGDH. Our results suggest that the length of carbon skeleton and positioning of the carboxylate groups of inhibitors between two conserved lysine residues at the GDH active site might be the determinants of their inhibitory potency. Molecular details on the dicarboxylic acid metabolites bound AtGDH active site architecture presented here would be applicable to GDHs in general. This article is protected by copyright. All rights reserved.
- Published
- 2021
- Full Text
- View/download PDF
17. Modulation of Phosphoprotein Activity by Phosphorylation Targeting Chimeras (PhosTACs)
- Author
-
Angela Gong, Elvira An, Ifunanya Okeke, Saul Jaime-Figueroa, Zhenyi Hu, Xuanmeng Luo, Craig M. Crews, Po-Han Chen, and Sijin Zheng
- Subjects
Phosphatase ,Hyperphosphorylation ,Biochemistry ,Dephosphorylation ,Structure-Activity Relationship ,Catalytic Domain ,Humans ,Protein phosphorylation ,Phosphorylation ,biology ,Chimera ,Chemistry ,Kinase ,Forkhead Box Protein O3 ,Retinoblastoma protein ,RNA-Binding Proteins ,General Medicine ,Protein phosphatase 2 ,Phosphoproteins ,Cell biology ,Enzyme Activation ,biology.protein ,Molecular Medicine ,Protein Tyrosine Phosphatases ,Apoptosis Regulatory Proteins ,Holoenzymes ,HeLa Cells - Abstract
Protein phosphorylation, which regulates many critical aspects of cell biology, is dynamically governed by kinases and phosphatases. Many diseases are associated with dysregulated hyperphosphorylation of critical proteins, such as retinoblastoma protein in cancer. Although kinase inhibitors have been widely applied in the clinic, growing evidence of off-target effects and increasing drug resistance prompts the need to develop a new generation of drugs. Here, we propose a proof-of-concept study of phosphorylation targeting chimeras (PhosTACs). Similar to PROTACs in their ability to induce ternary complexes, PhosTACs focus on recruiting a Ser/Thr phosphatase to a phosphosubstrate to mediate its dephosphorylation. However, distinct from PROTACs, PhosTACs can uniquely provide target gain-of-function opportunities to manipulate protein activity. In this study, we applied a chemical biology approach to evaluate the feasibility of PhosTACs by recruiting the scaffold and catalytic subunits of the PP2A holoenzyme to protein substrates such as PDCD4 and FOXO3a for targeted protein dephosphorylation. For FOXO3a, this dephosphorylation resulted in the transcriptional activation of a FOXO3a-responsive reporter gene.
- Published
- 2021
- Full Text
- View/download PDF
18. A NYN domain protein directly interacts with DECAPPING1 and is required for phyllotactic pattern
- Author
-
Dominique Gagliardi, Philippe Hammann, Jérôme Mutterer, Lauriane Kuhn, Anthony Gobert, Clara Chicois, Johana Chicher, Aude Pouclet, Marlene Schiaffini, Hélène Zuber, Damien Garcia, Elodie Ubrig, Tiphaine Chartier, univOAK, Archive ouverte, Integrative Molecular and Cellular Biology - - IMCBio2017 - ANR-17-EURE-0023 - EURE - VALID, and Initiative d'excellence - Par-delà les frontières, l'Université de Strasbourg - - UNISTRA2010 - ANR-10-IDEX-0002 - IDEX - VALID
- Subjects
biology ,Regular Issue Content ,Physiology ,Chemistry ,RNA Stability ,Protein subunit ,Protein domain ,Endoribonuclease ,Arabidopsis ,Robustness (evolution) ,MRNA Decay ,Plant Science ,biology.organism_classification ,Cell biology ,Decapping complex ,RNA, Plant ,Catalytic Domain ,Endoribonucleases ,Genetics ,Arabidopsis thaliana ,[SDV.BV] Life Sciences [q-bio]/Vegetal Biology ,Enhancer ,Co-Repressor Proteins - Abstract
In eukaryotes, general mRNA decay requires the decapping complex. The activity of this complex depends on its catalytic subunit, DECAPPING2 (DCP2), and its interaction with decapping enhancers, including its main partner DECAPPING1 (DCP1). Here, we report that in Arabidopsis thaliana, DCP1 also interacts with a NYN domain endoribonuclease, hence named DCP1-ASSOCIATED NYN ENDORIBONUCLEASE 1 (DNE1). Interestingly, we found DNE1 predominantly associated with DCP1, but not with DCP2, and reciprocally, suggesting the existence of two distinct protein complexes. We also showed that the catalytic residues of DNE1 are required to repress the expression of mRNAs in planta upon transient expression. The overexpression of DNE1 in transgenic lines led to growth defects and a similar gene deregulation signature than inactivation of the decapping complex. Finally, the combination of dne1 and dcp2 mutations revealed a functional redundancy between DNE1 and DCP2 in controlling phyllotactic pattern formation. Our work identifies DNE1, a hitherto unknown DCP1 protein partner highly conserved in the plant kingdom and identifies its importance for developmental robustness.
- Published
- 2021
- Full Text
- View/download PDF
19. Mycobacterial membrane protein Large 3‐like <scp>‐family</scp> proteins in bacteria, protozoa, fungi, plants, and animals: A bioinformatics and structural investigation
- Author
-
Satish R. Malwal and Eric Oldfield
- Subjects
Models, Molecular ,Staphylococcus aureus ,Protein Conformation ,Trypanosoma cruzi ,Bioinformatics ,Biochemistry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Glycolipid ,Bacterial Proteins ,Protein Domains ,Structural Biology ,Catalytic Domain ,Amino Acid Sequence ,Molecular Biology ,Ergosterol ,biology ,Fungi ,Membrane Transport Proteins ,Biological Transport ,Mycobacterium tuberculosis ,biology.organism_classification ,Sphingolipid ,Transmembrane protein ,Cholesterol ,Membrane protein ,chemistry ,Cord Factors ,Protozoa ,Bacteria ,Protein Binding - Abstract
Lipid transporters play an important role in most if not all organisms, ranging from bacteria to humans. For example, in Mycobacterium tuberculosis, the trehalose monomycolate transporter MmpL3 is involved in cell wall biosynthesis, while in humans, cholesterol transporters are involved in normal cell function as well as in disease. Here, using structural and bioinformatics information, we propose that there are proteins that also contain "MmpL3-like" (MMPL) transmembrane (TM) domains in many protozoa, including Trypanosoma cruzi, as well as in the bacterium Staphylococcus aureus, where the fatty acid transporter FarE has the same set of "active-site" residues as those found in the mycobacterial MmpL3s, and in T. cruzi. We also show that there are strong sequence and predicted structural similarities between the TM proton-translocation domain seen in the X-ray structures of mycobacterial MmpL3s and several human as well as fungal lipid transporters, leading to the proposal that there are similar proteins in apicomplexan parasites, and in plants. The animal, fungal, apicomplexan, and plant proteins have larger extra-membrane domains than are found in the bacterial MmpL3, but they have a similar TM domain architecture, with the introduction of a (catalytically essential) Phe > His residue change, and a Ser/Thr H-bond network, involved in H+ -transport. Overall, the results are of interest since they show that MMPL-family proteins are present in essentially all life forms: archaea, bacteria, protozoa, fungi, plants and animals and, where known, they are involved in "lipid" (glycolipid, phospholipid, sphingolipid, fatty acid, cholesterol, ergosterol) transport, powered by transmembrane molecular pumps having similar structures.
- Published
- 2021
- Full Text
- View/download PDF
20. An Inhibitor-in-Pieces Approach to DAHP Synthase Inhibition: Potent Enzyme and Bacterial Growth Inhibition
- Author
-
Robert Szabla, Paul J. Berti, Christopher M. Brown, Murray S. Junop, Pallavi Mukherjee, Maren Heimhalt, Rebecca Turner, and Ryan A. Grainger
- Subjects
Stereochemistry ,DAHP synthase ,010402 general chemistry ,01 natural sciences ,Phosphates ,03 medical and health sciences ,chemistry.chemical_compound ,Catalytic Domain ,Escherichia coli ,Aromatic amino acids ,Shikimate pathway ,3-Deoxy-7-Phosphoheptulonate Synthase ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,ATP synthase ,Chemistry ,Active site ,Oxime ,0104 chemical sciences ,Kinetics ,Infectious Diseases ,Enzyme ,biology.protein ,Growth inhibition - Abstract
3-Deoxy-d-arabinoheptulosonate-7-phosphate (DAHP) synthase catalyzes the first step in the shikimate biosynthetic pathway and is an antimicrobial target. We used an inhibitor-in-pieces approach, based on the previously reported inhibitor DAHP oxime, to screen inhibitor fragments in the presence and absence of glycerol 3-phosphate to occupy the distal end of the active site. This led to DAHP hydrazone, the most potent inhibitor to date, Ki = 10 ± 1 nM. Three trifluoropyruvate (TFP)-based inhibitor fragments were efficient inhibitors with ligand efficiencies of up to 0.7 kcal mol-1/atom compared with 0.2 kcal mol-1/atom for a typical good inhibitor. The crystal structures showed the TFP-based inhibitors binding upside down in the active site relative to DAHP oxime, providing new avenues for inhibitor development. The ethyl esters of TFP oxime and TFP semicarbazone prevented E. coli growth in culture with IC50 = 0.21 ± 0.01 and 0.77 ± 0.08 mg mL-1, respectively. Overexpressing DAHP synthase relieved growth inhibition, demonstrating that DAHP synthase was the target. Growth inhibition occurred in media containing aromatic amino acids, suggesting that growth inhibition was due to depletion of some other product(s) of the shikimate pathway, possibly folate.
- Published
- 2021
- Full Text
- View/download PDF
21. Catalytic Control of Spiroketal Formation in Rubromycin Polyketide Biosynthesis
- Author
-
Jörn Piel, Robin Teufel, Raspudin Saleem-Batcha, and Marina Toplak
- Subjects
Stereochemistry ,polyketide synthase ,Flavoprotein ,Catalysis ,antibiotics ,Mixed Function Oxygenases ,chemistry.chemical_compound ,Polyketide ,lenticulone ,Protein Domains ,Biosynthesis ,Catalytic Domain ,Polyketide synthase ,Moiety ,Quinone Reductases ,redox tailoring ,ATP synthase ,biology ,Quinones ,General Chemistry ,General Medicine ,Monooxygenase ,Kinetics ,chemistry ,Mutation ,Biocatalysis ,Flavin-Adenine Dinucleotide ,Mutagenesis, Site-Directed ,biology.protein ,collinone ,Pharmacophore ,Oxidation-Reduction ,NADP ,Ethers ,Protein Binding - Abstract
The medically important bacterial aromatic polyketide natural products typically feature a planar, polycyclic core structure. An exception is found for the rubromycins, whose backbones are disrupted by a bisbenzannulated [5,6]-spiroketal pharmacophore that was recently shown to be assembled by flavin-dependent enzymes. In particular, a flavoprotein monooxygenase proved critical for the drastic oxidative rearrangement of a pentangular precursor and the installment of an intermediate [6,6]-spiroketal moiety. Here we provide structural and mechanistic insights into the control of catalysis by this spiroketal synthase, which fulfills several important functions as reductase, monooxygenase, and presumably oxidase. The enzyme hereby tightly controls the redox state of the substrate to counteract shunt product formation, while also steering the cleavage of three carbon-carbon bonds. Our work illustrates an exceptional strategy for the biosynthesis of stable chroman spiroketals., Angewandte Chemie. International Edition, 60 (52), ISSN:1433-7851, ISSN:1521-3773, ISSN:0570-0833
- Published
- 2021
- Full Text
- View/download PDF
22. Structure of Aedes aegypti carboxypeptidase <scp>B1</scp> ‐inhibitor complex uncover the disparity between mosquito and non‐mosquito insect carboxypeptidase inhibition mechanism
- Author
-
Yeu Khai Choong, Edem Gavor, R. Manjunatha Kini, Chacko Jobichen, Yu-Keung Mok, and J. Sivaraman
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Proteases ,Carboxypeptidases A ,Full‐Length Papers ,medicine.medical_treatment ,Genetic Vectors ,Gene Expression ,Aedes aegypti ,Crystallography, X-Ray ,Biochemistry ,Substrate Specificity ,Species Specificity ,Aedes ,Catalytic Domain ,Escherichia coli ,medicine ,Animals ,Protease Inhibitors ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,cardiovascular diseases ,Cloning, Molecular ,Molecular Biology ,chemistry.chemical_classification ,Protease ,Sequence Homology, Amino Acid ,biology ,Chemistry ,fungi ,Midgut ,biology.organism_classification ,Carboxypeptidase ,Carboxypeptidase B ,Recombinant Proteins ,Potato carboxypeptidase inhibitor ,Kinetics ,surgical procedures, operative ,Enzyme ,Conventional PCI ,biology.protein ,Insect Proteins ,Cattle ,Protein Conformation, beta-Strand ,Sequence Alignment ,Protein Binding - Abstract
Metallocarboxypeptidases (MCPs) in the mosquito midgut play crucial roles in infection, as well as in mosquito dietary digestion, reproduction, and development. MCPs are also part of the digestive system of plant-feeding insects, representing key targets for inhibitor development against mosquitoes/mosquito-borne pathogens or as antifeedant molecules against plant-feeding insects. Notably, some non-mosquito insect B-type MCPs are primarily insensitive to plant protease inhibitors such as the potato carboxypeptidase inhibitor (PCI; MW 4-kDa), an inhibitor explored for cancer treatment and insecticide design. Here, we report the crystal structure of Ae. aegypti carboxypeptidase-B1 (CPBAe1)-PCI complex and compared the binding with that of PCI-insensitive CPBs. We show that PCI accommodation is determined by key differences in the active-site regions of MCPs. In particular, the loop regions α6-α7 (Leu242 -Ser250 ) and β8-α8 (Pro269 -Pro280 ) of CPBAe1 are replaced by α-helices in PCI-insensitive insect H.zea CPBHz. These α-helices protrude into the active-site pocket of CPBHz, restricting PCI insertion and rendering the enzyme insensitive. We further compared our structure with the only other PCI complex available, bovine CPA1-PCI. The potency of PCI against CPBAe1 (Ki = 14.7 nM) is marginally less than that of bovine CPA1 (Ki = 5 nM). Structurally, the above loop regions that accommodate PCI binding in CPBAe1 are similar to that of bovine CPA1, although observed changes in proteases residues that interact with PCI could account for the differences in affinity. Our findings suggest that PCI sensitivity is largely dictated by structural interference, which broadens our understanding of carboxypeptidase inhibition as a mosquito population/parasite control strategy. This article is protected by copyright. All rights reserved.
- Published
- 2021
- Full Text
- View/download PDF
23. Active site variants in STT3A cause a dominant type I congenital disorder of glycosylation with neuromusculoskeletal findings
- Author
-
Rita Barone, Filippo Vairo, Bobby G. Ng, Jaak Jaeken, Gert Matthijs, James Pitt, Thierry Dupré, Lyndon Gallacher, Liesbeth Keldermans, Helen Michelakakis, Marina Ventouratou, Susan M. White, Sze Chern Lim, Melissa Baerenfaenger, Mirian C. H. Janssen, Angel Ashikov, Karin Huijben, Sandrine Vuillaumier-Barrot, Diana Ballhausen, Daisy Rymen, Agustí Rodríguez-Palmero, Blai Morales-Romero, Antonia Ribes, Peter Witters, Heidi Peters, Erika Souche, Eva Morava, Agata Fiumara, Pascale de Lonlay, Matthew P. Wilson, Dirk Lefeber, Wasantha Ranatunga, Alejandro Garanto, Hudson H. Freeze, Christian Thiel, BioAnalytical Chemistry, and AIMMS
- Subjects
Male ,Mutant ,congenital disorders of glycosylation ,chemistry.chemical_compound ,0302 clinical medicine ,Catalytic Domain ,Missense mutation ,Musculoskeletal Diseases ,Genetics (clinical) ,Genes, Dominant ,chemistry.chemical_classification ,Genetics ,0303 health sciences ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Middle Aged ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Pedigree ,Oligosaccharyltransferase complex ,Child, Preschool ,glycosylation ,Female ,Adult ,Heterozygote ,Glycosylation ,Adolescent ,Protein subunit ,Biology ,Article ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,oligosaccharyltransferase complex ,medicine ,Humans ,dominant inheritance ,Amino Acid Sequence ,030304 developmental biology ,Sequence Homology, Amino Acid ,Oligosaccharyltransferase ,Membrane Proteins ,medicine.disease ,chemistry ,Hexosyltransferases ,Nervous System Diseases ,Glycoprotein ,Congenital disorder of glycosylation ,030217 neurology & neurosurgery - Abstract
Congenital disorders of glycosylation (CDGs) form a group of rare diseases characterized by hypoglycosylation. We here report the identification of 16 individuals from nine families who have either inherited or de novo heterozygous missense variants in STT3A, leading to an autosomal-dominant CDG. STT3A encodes the catalytic subunit of the STT3A-containing oligosaccharyltransferase (OST) complex, essential for protein N-glycosylation. Affected individuals presented with variable skeletal anomalies, short stature, macrocephaly, and dysmorphic features; half had intellectual disability. Additional features included increased muscle tone and muscle cramps. Modeling of the variants in the 3D structure of the OST complex indicated that all variants are located in the catalytic site of STT3A, suggesting a direct mechanistic link to the transfer of oligosaccharides onto nascent glycoproteins. Indeed, expression of STT3A at mRNA and steady-state protein level in fibroblasts was normal, while glycosylation was abnormal. In S. cerevisiae, expression of STT3 containing variants homologous to those in affected individuals induced defective glycosylation of carboxypeptidase Y in a wild-type yeast strain and expression of the same mutants in the STT3 hypomorphic stt3-7 yeast strain worsened the already observed glycosylation defect. These data support a dominant pathomechanism underlying the glycosylation defect. Recessive mutations in STT3A have previously been described to lead to a CDG. We present here a dominant form of STT3A-CDG that, because of the presence of abnormal transferrin glycoforms, is unusual among dominant type I CDGs. ispartof: AMERICAN JOURNAL OF HUMAN GENETICS vol:108 issue:11 pages:2130-2144 ispartof: location:United States status: published
- Published
- 2021
- Full Text
- View/download PDF
24. Inhibition Mechanism of SARS‐CoV‐2 Main Protease with Ketone‐Based Inhibitors Unveiled by Multiscale Simulations: Insights for Improved Designs**
- Author
-
Iñaki Tuñón, J. Javier Ruiz-Pernía, and Carlos A. Ramos-Guzmán
- Subjects
Ketone ,Molecular model ,Stereochemistry ,Substituent ,Molecular Dynamics Simulation ,SARS‐CoV‐2 Inhibitors | Hot Paper ,Catalysis ,QM/MM ,3CL protease ,chemistry.chemical_compound ,Catalytic Domain ,inhibitors ,Humans ,Hydroxymethyl ,Protease Inhibitors ,Coronavirus 3C Proteases ,Research Articles ,chemistry.chemical_classification ,PF-00835231 ,Binding Sites ,biology ,SARS-CoV-2 ,molecular modeling ,Active site ,COVID-19 ,General Chemistry ,General Medicine ,Ketones ,COVID-19 Drug Treatment ,Kinetics ,chemistry ,Covalent bond ,Drug Design ,biology.protein ,Thermodynamics ,Oxyanion hole ,Research Article - Abstract
We present the results of classical and QM/MM simulations for the inhibition of SARS‐CoV‐2 3CL protease by a hydroxymethylketone inhibitor, PF‐00835231. In the noncovalent complex the carbonyl oxygen atom of the warhead is placed in the oxyanion hole formed by residues 143 to 145, while P1–P3 groups are accommodated in the active site with interactions similar to those observed for the peptide substrate. According to alchemical free energy calculations, the P1′ hydroxymethyl group also contributes to the binding free energy. Covalent inhibition of the enzyme is triggered by the proton transfer from Cys145 to His41. This step is followed by the nucleophilic attack of the Sγ atom on the carbonyl carbon atom of the inhibitor and a proton transfer from His41 to the carbonyl oxygen atom mediated by the P1′ hydroxyl group. Computational simulations show that the addition of a chloromethyl substituent to the P1′ group may lower the activation free energy for covalent inhibition, Multiscale simulations unveil the binding and reaction mechanism of the SARS‐CoV‐2 main protease inhibitor, PF‐00835231 inhibitor. This compound contains a hydroxymethyl group that plays a relevant role in the formation of the noncovalent and covalent complexes. In silico modifications show a possible strategy for the design of new inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
25. Synthesis of indole derivatives as diabetics II inhibitors and enzymatic kinetics study of α-glucosidase and α-amylase along with their in-silico study
- Author
-
Sridevi Chigurupati, Muhammad Nawaz, Fazal Rahim, Vijayan Venugopal, Noor B. Almandil, Faisal Nawaz, Nizam Uddin, Suprava Das, Abdul Wadood, Ahlam Sayer Alrashedy, Naveed Iqbal, Khalid Mohammed Khan, Muhammad Taha, and El Hassane Anouar
- Subjects
Indoles ,Saccharomyces cerevisiae ,Inhibitory postsynaptic potential ,Biochemistry ,Structure-Activity Relationship ,Structural Biology ,Catalytic Domain ,medicine ,Hypoglycemic Agents ,Computer Simulation ,Glycoside Hydrolase Inhibitors ,Amylase ,Molecular Biology ,Acarbose ,Indole test ,chemistry.chemical_classification ,biology ,Chemistry ,Active site ,Hydrogen Bonding ,alpha-Glucosidases ,General Medicine ,Carbohydrate ,Molecular Docking Simulation ,Kinetics ,Enzyme ,Docking (molecular) ,biology.protein ,alpha-Amylases ,medicine.drug - Abstract
In this study, we have investigated a series of indole-based compounds for their inhibitory study against pancreatic α-amylase and intestinal α-glucosidase activity. Inhibitors of carbohydrate degrading enzymes appear to have an essential role as antidiabetic drugs. All analogous exhibited good to moderate α-amylase (IC50 = 3.80 to 47.50 μM), and α-glucosidase inhibitory interactions (IC50 = 3.10–52.20 μM) in comparison with standard acarbose (IC50 = 12.28 μM and 11.29 μM). The analogues 4, 11, 12, 15, 14 and 17 had good activity potential both for enzymes inhibitory interactions. Structure activity relationships were deliberated to propose the influence of substituents on the inhibitory potential of analogues. Docking studies revealed the interaction of more potential analogues and enzyme active site. Further, we studied their kinetic study of most active compounds showed that compounds 15, 14, 12, 17 and 11 are competitive for α-amylase and non- competitive for α-glucosidase.
- Published
- 2021
- Full Text
- View/download PDF
26. Structural investigation of a thermostable 1,2-β-mannobiose phosphorylase from Thermoanaerobacter sp. X-514
- Author
-
Yuanxia Sun, Rey-Ting Guo, Chun-Chi Chen, Zhenying Chang, Yu Yang, Jiangang Yang, Weidong Liu, Longhai Dai, Lilan Zhang, and Jian-Wen Huang
- Subjects
Glycoside Hydrolases ,Phosphorylases ,Protein Conformation ,Stereochemistry ,Static Electricity ,Biophysics ,Mannose ,Thermoanaerobacter ,Ligands ,Biochemistry ,Catalysis ,Mannans ,chemistry.chemical_compound ,Catalytic Domain ,Mannobiose ,Glycoside hydrolase ,Molecular Biology ,Thermostability ,Ions ,chemistry.chemical_classification ,Binding Sites ,Mannosephosphates ,biology ,Temperature ,beta-Mannosidase ,Reproducibility of Results ,Active site ,Substrate (chemistry) ,Cell Biology ,Protein engineering ,Zinc ,Enzyme ,chemistry ,biology.protein ,Plasmids - Abstract
1,2-β-Mannobiose phosphorylases (1,2-β-MBPs) from glycoside hydrolase 130 (GH130) family are important bio-catalysts in glycochemistry applications owing to their ability in synthesizing oligomannans. Here, we report the crystal structure of a thermostable 1,2-β-MBP from Thermoanaerobacter sp. X-514 termed Teth514_1789 to reveal the molecular basis of its higher thermostability and mechanism of action. We also solved the enzyme complexes of mannose, mannose-1-phosphate (M1P) and 1,4-β-mannobiose to manifest the enzyme-substrate interaction networks of three main subsites. Notably, a Zn ion that should be derived from crystallization buffer was found in the active site and coordinates the phosphate moiety of M1P. Nonetheless, this Zn-coordination should reflect an inhibitory status as supplementing Zn severely impairs the enzyme activity. These results indicate that the effects of metal ions should be taken into consideration when applying Teth514_1789 and other related enzymes. Based on the structure, a reliable model of Teth514_1788 that shares 61.7% sequence identity to Teth514_1789 but displays a different substrate preference was built. Analyzing the structural features of these two closely related enzymes, we hypothesized that the length of a loop fragment that covers the entrance of the catalytic center might regulate the substrate selectivity. In conclusion, these information provide in-depth understanding of GH130 1,2-β-MBPs and should serve as an important guidance for enzyme engineering for further applications.
- Published
- 2021
- Full Text
- View/download PDF
27. Kinetic and Structural Characterization of Sialidases (Kdnases) from Ascomycete Fungal Pathogens
- Author
-
Benjamin Noyovitz, Stephen A. McMahon, Nick P G Gauthier, Andrew J. Bennet, Tracey M. Gloster, Kobra Khazaei, Brock W. Byers, Jason R. Nesbitt, Verena Oehler, Nicholas J. Thornton, Jamie Baker, Ali Nejatie, Margo M. Moore, Wesley F. Zandberg, and Elizabeth Steves
- Subjects
Glycan ,Protein Conformation ,Neuraminidase ,Sialidase ,Biochemistry ,Catalysis ,Substrate Specificity ,Aspergillus fumigatus ,03 medical and health sciences ,chemistry.chemical_compound ,Ascomycota ,Catalytic Domain ,Enzyme Stability ,Glycoside hydrolase ,Aspergillus terreus ,skin and connective tissue diseases ,Fluorescent Dyes ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,030306 microbiology ,Temperature ,Glycoside ,General Medicine ,Hydrogen-Ion Concentration ,bacterial infections and mycoses ,biology.organism_classification ,Culture Media ,Sialic acid ,Kinetics ,Enzyme ,chemistry ,biology.protein ,Molecular Medicine - Abstract
Sialidases catalyze the release of sialic acid from the terminus of glycan chains. We previously characterized the sialidase from the opportunistic fungal pathogen, Aspergillus fumigatus, and showed that it is a Kdnase. That is, this enzyme prefers 3-deoxy-d-glycero-d-galacto-non-2-ulosonates (Kdn glycosides) as the substrate compared to N-acetylneuraminides (Neu5Ac). Here, we report characterization and crystal structures of putative sialidases from two other ascomycete fungal pathogens, Aspergillus terreus (AtS) and Trichophyton rubrum (TrS). Unlike A. fumigatus Kdnase (AfS), hydrolysis with the Neu5Ac substrates was negligible for TrS and AtS; thus, TrS and AtS are selective Kdnases. The second-order rate constant for hydrolysis of aryl Kdn glycosides by AtS is similar to that by AfS but 30-fold higher by TrS. The structures of these glycoside hydrolase family 33 (GH33) enzymes in complex with a range of ligands for both AtS and TrS show subtle changes in ring conformation that mimic the Michaelis complex, transition state, and covalent intermediate formed during catalysis. In addition, they can aid identification of important residues for distinguishing between Kdn and Neu5Ac substrates. When A. fumigatus, A. terreus, and T. rubrum were grown in chemically defined media, Kdn was detected in mycelial extracts, but Neu5Ac was only observed in A. terreus or T. rubrum extracts. The C8 monosaccharide 3-deoxy-d-manno-oct-2-ulosonic acid (Kdo) was also identified in A. fumigatus and T. rubrum samples. A fluorescent Kdn probe was synthesized and revealed the localization of AfS in vesicles at the cell surface.
- Published
- 2021
- Full Text
- View/download PDF
28. Mining of a novel esterase (est3S) gene from a cow rumen metagenomic library with organosphosphorus insecticides degrading capability: Catalytic insights by site directed mutations, docking, and molecular dynamic simulations
- Author
-
Hee Yul Lee, Harun M. Patel, Du Yong Cho, Min Ju Kim, Kye Man Cho, Jea Gack Jung, Iqrar Ahmad, Eun Hye Jeong, and Md. Azizul Haque
- Subjects
Rumen ,Esterase Gene ,Molecular Dynamics Simulation ,Ligands ,Biochemistry ,Esterase ,Substrate Specificity ,Organophosphorus Compounds ,Structural Biology ,Catalytic Domain ,Animals ,Amino Acid Sequence ,Cloning, Molecular ,Lipase ,Molecular Biology ,Phylogeny ,Gene Library ,chemistry.chemical_classification ,Gel electrophoresis ,Base Sequence ,biology ,Esterases ,General Medicine ,Physical Chromosome Mapping ,Recombinant Proteins ,Amino acid ,Molecular Docking Simulation ,Molecular Weight ,Kinetics ,Enzyme ,chemistry ,Docking (molecular) ,Biocatalysis ,Mutagenesis, Site-Directed ,biology.protein ,Cattle ,Metagenomics - Abstract
A novel esterase (est3S) gene, 1026 bp in size, was cloned from a metagenomic library made of uncultured microorganisms from the contents of cow rumen. The esterolytic enzyme (Est3S) is composed of 342 amino acids and shows the highest identity with EstGK1 (71.7%) and EstZ3 (63.78%) esterases from the uncultured bacterium. The Est3S did not cluster in any up-to-date classes (I to XVIII) of esterase and lipase. Est3S protein molecular weight was determined to be 38 kDa by gel electrophoresis and showed optimum activity at pH 7.0 and 40 °C and is partially resistant to organic solvents. Est3S activity was enhanced by K+, Na+, Mg2+, and Ca2+ and its highest activity was observed toward the short-chain p-nitrophenyl esters. Additionally, Est3S can degrade chlorpyrifos (CP) and methyl parathion (70% to 80%) in an hour. A mutated Est3S (Ser132-Ala132) did not show any activity toward CP and ester substrates. Notably, the GHS132QG motif is superimposed with the homolog esterase and cutinase-like esterase. Therefore, Ser132 is the critical amino acid like other esterases. The Est3S is relatively stable with ester compounds, and the methyl parathion complex was confirmed by molecular dynamics simulation. Novelty statement A novel esterase gene (est3S) expressing esters and organophosphorus insecticide degradation traits was isolated from the uncultured bacterium in the contents of cow rumen. The Est3S protein did not cluster in any up-to-date classes (I to XVIII) of esterase/lipase proteins. Est3S was stable with the ligands up to 100 ns during the molecular dynamic simulations.
- Published
- 2021
- Full Text
- View/download PDF
29. Synthesis and Structure-Activity Relationship Studies of N-monosubstituted Aroylthioureas as Urease Inhibitors
- Author
-
Zhu-Ping Xiao, Hui Ouyang, Hai-Liang Zhu, Hai-Lian Fang, Li Fang, Ya-Xi Ye, Dawalamu, Fu Zijuan, Wei-Yi Li, Ke Li, Zhu Wenyan, Wei-Wei Ni, Zou Xia, and Li Liu
- Subjects
Urease ,Stereochemistry ,01 natural sciences ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Bacterial Proteins ,Catalytic Domain ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Indophenol ,Enzyme Inhibitors ,IC50 ,030304 developmental biology ,0303 health sciences ,Binding Sites ,Urea binding ,Helicobacter pylori ,biology ,Acetohydroxamic acid ,Thiourea ,Active site ,Hep G2 Cells ,Surface Plasmon Resonance ,Anti-Bacterial Agents ,0104 chemical sciences ,Molecular Docking Simulation ,Kinetics ,010404 medicinal & biomolecular chemistry ,Solubility ,chemistry ,biology.protein ,medicine.drug - Abstract
Background: Thiourea is a classical urease inhibitor which is usually used as a positive control, and many N,N'-disubstituted thioureas have been determined as urease inhibitors. However, due to steric hindrance, N,N'-disubstituted thiourea motif could not bind urease as thiourea. On the contrary, N-monosubstituted thiourea with a tiny thiourea motif could theoretically bind into the active pocket as thiourea. Objective: A series of N-monosubstituted aroylthioureas were designed and synthesized for evaluation as urease inhibitors. Methods: Urease inhibition was determined by the indophenol method and IC50 values were calculated using computerized linear regression analysis of quantal log dose-probit functions. The kinetic parameters were estimated via surface plasmon resonance (SPR) and by nonlinear regression analysis based on the mixed type inhibition model derived from Michaelis-Menten kinetics. Results: Compounds b2, b11, and b19 reversibly inhibited urease with a mixed mechanism, and showed excellent potency against both cell-free urease and urease in the intact cell, with IC50 values being 90- to 450-fold and 5- to 50-fold lower than the positive control acetohydroxamic acid, respectively. The most potent compound b11 showed an IC50 value of 0.060 ± 0.004μM against cell-free urease, which bound to urea binding site with a very low KD value (0.420±0.003nM) and a very long residence time (6.7 min). Compound b11 was also demonstrated to have very low cytotoxicity to mammalian cells. Conclusion: The results revealed that N-monosubstituted aroylthioureas bound to the active site of urease as expected, and represent a new class of urease inhibitors for the development of potential therapeutics against infections caused by urease-containing pathogens.
- Published
- 2021
- Full Text
- View/download PDF
30. The crystal structure of the domain-swapped dimer of onconase highlights some catalytic and antitumor activity features of the enzyme
- Author
-
Antonello Merlino, Giovanni Gotte, Rachele Campagnari, Domenico Loreto, Federica Calzetti, Ilaria Bettin, Marta Menegazzi, Gotte, G., Campagnari, R., Loreto, D., Bettin, I., Calzetti, F., Menegazzi, M., and Merlino, A.
- Subjects
STAT3 Transcription Factor ,Bcl2 ,RNase P ,Dimer ,Antineoplastic Agents ,Apoptosis ,Biochemistry ,Ribonuclease ,Antineoplastic Agent ,chemistry.chemical_compound ,Ribonucleases ,Structural Biology ,Cell Line, Tumor ,Catalytic Domain ,Humans ,Human melanoma ,Melanoma ,Molecular Biology ,Cell Proliferation ,3D domain swapping ,Onconase ,biology ,Cell growth ,Apoptosi ,General Medicine ,Recombinant Protein ,Recombinant Proteins ,chemistry ,biology.protein ,Biophysics ,Phosphorylation ,Protein quaternary structure ,Src kinase ,STAT-3 ,Signal transduction ,Antitumor activity ,Onconase dimer ,Human ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Onconase (ONC) is a monomeric amphibian "pancreatic-type" RNase endowed with remarkable anticancer activity. ONC spontaneously forms traces of a dimer (ONC-D) in solution, while larger amounts can be formed when ONC is lyophilized from mildly acidic solutions. Here, we report the crystal structure of ONC-D and analyze its catalytic and antitumor activities in comparison to ONC. ONC-D forms via the three-dimensional swapping of the N-terminal α-helix between two monomers, but it displays a significantly different quaternary structure from that previously modeled [Fagagnini A et al., 2017, Biochem J 474, 3767-81], and based on the crystal structure of the RNase A N-terminal swapped dimer. ONC-D presents a variable quaternary assembly deriving from a variable open interface, while it retains a catalytic activity that is similar to that of ONC. Notably, ONC-D displays antitumor activity against two human melanoma cell lines, although it exerts a slightly lower cytostatic effect than the monomer. The inhibition of melanoma cell proliferation by ONC or ONC-D is associated with the reduction of the expression of the anti-apoptotic B cell lymphoma 2 (Bcl2), as well as of the total expression and phosphorylation of the Signal Transducer and Activator of Transcription (STAT)-3. Phosphorylation is inhibited in both STAT3 Tyr705 and Ser727 key-residues, as well as in its upstream tyrosine-kinase Src. Consequently, both ONC species should exert their anti-cancer action by inhibiting the pro-tumor pleiotropic STAT3 effects deriving either by its phospho-tyrosine activation or by its non-canonical signaling pathways. Both ONC species, indeed, increase the portion of A375 cells undergoing apoptotic cell death. This study expands the variety of RNase domain-swapped dimeric structures, underlining the unpredictability of the open interface arrangement upon domain swapping. Structural data also offer valuable insights to analyze the differences in the measured ONC or ONC-D biological activities.
- Published
- 2021
- Full Text
- View/download PDF
31. Switching an active site helix in dihydrofolate reductase reveals limits to subdomain modularity
- Author
-
Daniel L. Hartl, Victor Zhao, João V. Rodrigues, Elena R. Lozovsky, and Eugene I. Shakhnovich
- Subjects
chemistry.chemical_classification ,biology ,Protein Conformation ,Chemistry ,Modularity (biology) ,Biophysics ,Active site ,Articles ,medicine.disease_cause ,Kinetics ,Tetrahydrofolate Dehydrogenase ,Molecular dynamics ,Enzyme ,Catalytic Domain ,Dihydrofolate reductase ,Helix ,Escherichia coli ,biology.protein ,medicine ,Binding site - Abstract
To what degree are individual structural elements within proteins modular such that similar structures from unrelated proteins can be interchanged? We study subdomain modularity by creating 20 chimeras of an enzyme, Escherichia coli dihydrofolate reductase (DHFR), in which a catalytically important, 10-residue α-helical sequence is replaced by α-helical sequences from a diverse set of proteins. The chimeras stably fold but have a range of diminished thermal stabilities and catalytic activities. Evolutionary coupling analysis indicates that the residues of this α-helix are under selection pressure to maintain catalytic activity in DHFR. Reversion to phenylalanine at key position 31 was found to partially restore catalytic activity, which could be explained by evolutionary coupling values. We performed molecular dynamics simulations using replica exchange with solute tempering. Chimeras with low catalytic activity exhibit nonhelical conformations that block the binding site and disrupt the positioning of the catalytically essential residue D27. Simulation observables and in vitro measurements of thermal stability and substrate-binding affinity are strongly correlated. Several E. coli strains with chromosomally integrated chimeric DHFRs can grow, with growth rates that follow predictions from a kinetic flux model that depends on the intracellular abundance and catalytic activity of DHFR. Our findings show that although α-helices are not universally substitutable, the molecular and fitness effects of modular segments can be predicted by the biophysical compatibility of the replacement segment.
- Published
- 2021
- Full Text
- View/download PDF
32. Design, Synthesis and Structural Analysis of Glucocerebrosidase Imaging Agents
- Author
-
Yurong Chen, Imogen Breen, Johannes M. F. G. Aerts, Liang Wu, Wendy A. Offen, Qin Su, Marta Artola, Rhianna J. Rowland, Herman S. Overkleeft, Adrianus M. C. H. van den Nieuwendijk, Gideon J. Davies, and Thomas J. M. Beenakker
- Subjects
genetic structures ,Allosteric regulation ,010402 general chemistry ,01 natural sciences ,Catalysis ,03 medical and health sciences ,chemistry.chemical_compound ,Catalytic Domain ,Mode of action ,Fluorescent Dyes ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Organic Chemistry ,Active site ,General Chemistry ,Aziridine ,Imaging agent ,0104 chemical sciences ,3. Good health ,Biochemistry ,Design synthesis ,Structural biology ,biology.protein ,Glucosylceramidase ,Glucocerebrosidase - Abstract
Gaucher disease (GD) is a lysosomal storage disorder caused by inherited deficiencies in beta-glucocerebrosidase (GBA). Current treatments require rapid disease diagnosis and a means of monitoring therapeutic efficacy, both of which may be supported by the use of GBA-targeting activity-based probes (ABPs). Here, we report the synthesis and structural analysis of a range of cyclophellitol epoxide and aziridine inhibitors and ABPs for GBA. We demonstrate their covalent mechanism-based mode of action and uncover binding of the new N-functionalised aziridines to the ligand binding cleft. These inhibitors became scaffolds for the development of ABPs; the O6-fluorescent tags of which bind in an allosteric site at the dimer interface. Considering GBA's preference for O6- and N-functionalised reagents, a bi-functional aziridine ABP was synthesized as a potentially more powerful imaging agent. Whilst this ABP binds to two unique active site clefts of GBA, no further benefit in potency was achieved over our first generation ABPs. Nevertheless, such ABPs should serve useful in the study of GBA in relation to GD and inform the design of future probes.
- Published
- 2021
- Full Text
- View/download PDF
33. Iron-Based Dual Active Site-Mediated Peroxymonosulfate Activation for the Degradation of Emerging Organic Pollutants
- Author
-
Jianlong Wang, Shizong Wang, and Lejin Xu
- Subjects
inorganic chemicals ,biology ,Extended X-ray absorption fine structure ,Chemistry ,Singlet oxygen ,Iron ,Active site ,chemistry.chemical_element ,General Chemistry ,Photochemistry ,Persulfate ,Nitrogen ,Peroxides ,Catalysis ,chemistry.chemical_compound ,Adsorption ,Catalytic Domain ,biology.protein ,Environmental Chemistry ,Degradation (geology) ,Environmental Pollutants - Abstract
It is still a challenge to synthesize highly efficient and stable catalysts for the Fenton-like reaction. In this study, we constructed an integrated catalyst with highly dispersed iron-based dual active sites, in which Fe2N and single-atom Fe (SA-Fe) were embedded into nitrogen- and oxygen-co-doped graphitic carbon (Fe-N-O-GC-350). Extended X-ray absorption fine structure (EXAFS) confirmed the coordination structure of iron, and line combination fitting (LCF) demonstrated the coexistence of Fe2N and SA-Fe with percentages of 75 and 25%, respectively. Iron-based dual active sites endowed Fe-N-O-GC-350 with superior catalytic activity to activate peroxymonosulfate (PMS) as evidenced by the fast degradation rate of sulfamethoxazole (SMX) (0.24 min-1) in the presence of 0.4 mM PMS and 0.1 g/L Fe-N-O-GC-350. Unlike the reported singlet oxygen and high-valent iron oxo-mediated degradation induced by the SA-Fe catalyst, both surface-bound reactive species and singlet oxygen contributed to SMX degradation, while surface-bound reactive species dominated. Density functional theory (DFT) simulation indicated that Fe2N and SA-Fe enhanced the adsorption of PMS, which played a key role in PMS activation. The Fe-N-O-GC-350/PMS system had resistance to the interference of common inorganic anions and high oxidation capacity to recalcitrant organic contaminants. This study elucidated the important role of Fe2N in PMS activation and provide a clue to design rationally catalysts with iron-based dual active sites to activate PMS for the degradation of emerging organic pollutants.
- Published
- 2021
- Full Text
- View/download PDF
34. Free Energy Landscape and Proton Transfer Pathways of the Transimination Reaction at the Active site of the Serine Hydroxymethyltransferase Enzyme in Aqueous Medium
- Author
-
Kumari Soniya and Amalendu Chandra
- Subjects
Aldimine ,Stereochemistry ,Lysine ,chemical and pharmacologic phenomena ,Serine ,chemistry.chemical_compound ,immune system diseases ,Catalytic Domain ,Materials Chemistry ,Physical and Theoretical Chemistry ,Pyridoxal ,Glycine Hydroxymethyltransferase ,chemistry.chemical_classification ,biology ,Water ,Active site ,Substrate (chemistry) ,nervous system diseases ,Surfaces, Coatings and Films ,Enzyme ,chemistry ,Pyridoxal Phosphate ,Serine hydroxymethyltransferase ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Protons - Abstract
Serine hydroxymethyltransferase (SHMT) is a ubiquitous enzyme belonging to the fold type I or aspartate aminotransferase (AspAT) family of the pyridoxal 5'-phosphate (PLP)-dependent enzymes. Like other PLP-dependent enzymes, SHMT also undergoes the so-called transimination reaction before exhibiting its enzymatic activity. The transimination process constitutes an important pre-step for all PLP-dependent enzymes, where an internal aldimine of the PLP-enzyme complex gets converted to an external aldimine of the substrate-PLP complex at the active site of the enzyme. In case of the transimination reaction involving SHMT, the PLP molecule bound to the active site lysine residue of SHMT (internal aldimine) gets detached from the enzyme by a serine substrate to produce an external aldimine complex, where the PLP is now bound to the serine substrate. In the current study, the free energy surfaces and reaction pathways of different steps of the transimination reaction at the active site of SHMT are investigated by employing hybrid quantum mechanical/molecular mechanical (QM/MM) simulations combined with metadynamics methods of rare event sampling. It is found that the process of transimination involving serine and PLP at the active site of the SHMT enzyme takes place through different elementary steps such as the formation of the first geminal diamine intermediate (GDI1), transfer of a proton from the substrate serine to the phenolic oxygen of PLP, followed by another proton transfer from PLP to the amine nitrogen of lysine with the formation of the second geminal diamine intermediate (GDI2), and finally, detachment of the active site lysine residue from PLP to produce the external aldimine.
- Published
- 2021
- Full Text
- View/download PDF
35. Mechanism of the Clinically Relevant E305G Mutation in Human P450 CYP17A1
- Author
-
Ilia G. Denisov, Stephen G. Sligar, Michael C. Gregory, Yilin Liu, Yelena V. Grinkova, and James R. Kincaid
- Subjects
Stereochemistry ,Hydroxylation ,Spectrum Analysis, Raman ,Polymorphism, Single Nucleotide ,Biochemistry ,Translocation, Genetic ,Article ,Substrate Specificity ,chemistry.chemical_compound ,Catalytic Domain ,medicine ,Humans ,Progesterone ,Bond cleavage ,biology ,Androstenedione ,Steroid 17-alpha-Hydroxylase ,Substrate (chemistry) ,Active site ,Cytochrome P450 ,Hydrogen Bonding ,Dehydroepiandrosterone ,Lyase ,chemistry ,CYP17A1 ,Pregnenolone ,Mutation ,Androgens ,biology.protein ,Steroids ,medicine.drug - Abstract
Steroid metabolism in humans originates from cholesterol and involves several enzyme reactions including dehydrogenation, hydroxylation, and carbon–carbon bond cleavage that occur at regio- and stereo-specific points in the four-membered ring structure. Cytochrome P450s occur at critical junctions that control the production of the male sex hormones (androgens), the female hormones (estrogens) as well as the mineralocorticoids and glucocorticoids. An important branch point in human androgen production is catalyzed by cytochrome P450 CYP17A1 and involves an initial Compound I-mediated hydroxylation at the 17-position of either progesterone (PROG) or pregnenolone (PREG) to form 17-hydroxy derivatives, 17OH-PROG and 17OH-PREG, with approximately similar efficiencies. Subsequent processing of the 17-hydroxy substrates involves a C(17)–C(20) bond scission (lyase) activity that is heavily favored for 17OH-PREG in humans. The mechanism for this lyase reaction has been debated for several decades, some workers favoring a Compound I-mediated process, with others arguing that a ferric peroxo- is the active oxidant. Mutations in CYP17A1 can have profound clinical manifestations. For example, the replacement of the glutamic acid side with a glycine chain at position 305 in the CYP17A1 structure causes a clinically relevant steroidopathy; E305G CYP17A1 displays a dramatic decrease in the production of dehydroepiandrosterone from pregnenolone but surprisingly increases the activity of the enzyme toward the formation of androstenedione from progesterone. To better understand the functional consequences of this mutation, we self-assembled wild-type and the E305G mutant of CYP17A1 into nanodiscs and examined the detailed catalytic mechanism. We measured substrate binding, spin state conversion, and solvent isotope effects in the hydroxylation and lyase pathways for these substrates. Given that, following electron transfer, the ferric peroxo- species is the common intermediate for both mechanisms, we used resonance Raman spectroscopy to monitor the positioning of important hydrogen-bonding interactions of the 17-OH group with the heme-bound peroxide. We discovered that the E305G mutation changes the orientation of the lyase substrate in the active site, which alters a critical hydrogen bonding of the 17-alcohol to the iron-bound peroxide. The observed switch in substrate specificity of the enzyme is consistent with this result if the hydrogen bonding to the proximal peroxo oxygen is necessary for a proposed nucleophilic peroxoanion-mediated mechanism for CYP17A1 in carbon–carbon bond scission.
- Published
- 2021
- Full Text
- View/download PDF
36. Epoxyqueuosine Reductase QueH in the Biosynthetic Pathway to tRNA Queuosine Is a Unique Metalloenzyme
- Author
-
Brian S. MacTavish, Rémi Zallot, You Hu, Valérie de Crécy-Lagard, Daniel J. Payan, Steven D. Bruner, Alexander Angerhofer, Alvaro Montoya, John A. Gerlt, and Qiang Li
- Subjects
Iron-Sulfur Proteins ,chemistry.chemical_classification ,Oxidoreductases Acting on CH-CH Group Donors ,TRNA modification ,biology ,Iron ,Queuosine ,Active site ,biology.organism_classification ,Biochemistry ,Article ,Cofactor ,chemistry.chemical_compound ,Enzyme ,Bacterial Proteins ,chemistry ,Catalytic Domain ,Thermotoga maritima ,Transfer RNA ,biology.protein ,Gene - Abstract
Queuosine is a structurally unique and functionally important tRNA modification, widely distributed in eukaryotes and bacteria. The final step of queuosine biosynthesis is the reduction/deoxygenation of epoxyqueuosine to form the cyclopentene motif of the nucleobase. The chemistry is performed by the structurally and functionally characterized cobalamin-dependent QueG. However, the queG gene is absent from several bacteria that otherwise retain queuosine biosynthesis machinery. Members of the IPR003828 family (previously known as DUF208) have been recently identified as nonorthologous replacements of QueG, and this family was renamed QueH. Here, we present the structural characterization of QueH from Thermotoga maritima. The structure reveals an unusual active site architecture with a [4Fe-4S] metallocluster along with an adjacent coordinated iron metal. The juxtaposition of the cofactor and coordinated metal ion predicts a unique mechanism for a two-electron reduction/deoxygenation of epoxyqueuosine. To support the structural characterization, in vitro biochemical and genomic analyses are presented. Overall, this work reveals new diversity in the chemistry of iron/sulfur-dependent enzymes and novel insight into the last step of this widely conserved tRNA modification.
- Published
- 2021
- Full Text
- View/download PDF
37. Investigation of sequon engineering for improved O-glycosylation by the human polypeptide N-acetylgalactosaminyl transferase T2 isozyme and two orthologues
- Author
-
Samantha Rodriguez Perez, Warren W. Wakarchuk, Leif T. N. LeClaire, and Nicole K. Thompson
- Subjects
Threonine ,Glycan ,Glycosylation ,Interferon alpha-2 ,Protein Engineering ,Biochemistry ,Isozyme ,Serine ,chemistry.chemical_compound ,Polysaccharides ,Catalytic Domain ,Escherichia coli ,Humans ,Amino Acid Sequence ,Molecular Biology ,Peptide sequence ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,Chromatography, Reverse-Phase ,biology ,Human Growth Hormone ,Chemistry ,Mucins ,Cell Biology ,Sequon ,Recombinant Proteins ,Amino acid ,Isoenzymes ,carbohydrates (lipids) ,Kinetics ,biology.protein ,N-Acetylgalactosaminyltransferases ,Synthetic Biology ,Sequence Alignment - Abstract
We have been developing bacterial expression systems for human mucin-type O-glycosylation on therapeutic proteins, which is initiated by the addition of α-linked GalNAc to serine or threonine residues by enzymes in the GT-27 family of glycosyltransferases. Substrate preference across different isoforms of this enzyme is influenced by isoform-specific amino acid sequences at the site of glycosylation, which we have exploited to engineer production of Core 1 glycan structures in bacteria on human therapeutic proteins. Using RP-HPLC with a novel phenyl bonded phase to resolve intact protein glycoforms, the effect of sequon mutation on O-glycosylation initiation was examined through in vitro modification of the naturally O-glycosylated human interferon α-2b, and a sequon engineered human growth hormone. As part of the development of our glycan engineering in the bacterial expression system we are surveying various orthologues of critical enzymes to ensure complete glycosylation. Here we present an in vitro enzyme kinetic profile of three related GT-27 orthologues on natural and engineered sequons in recombinant human interferon α2b and human growth hormone where we show a significant change in kinetic properties with the amino acid changes. It was found that optimizing the protein substrate amino acid sequence using Isoform Specific O-Glycosylation Prediction (ISOGlyP, http://isoglyp.utep.edu/index.php) resulted in a measurable increase in kcat/KM, thus improving glycosylation efficiency. We showed that the Drosophila orthologue showed superior activity with our human growth hormone designed sequons compared with the human enzyme.
- Published
- 2021
- Full Text
- View/download PDF
38. Engineering Leifsonia Alcohol Dehydrogenase for Thermostability and Catalytic Efficiency by Enhancing Subunit Interactions
- Author
-
Zixin Deng, Xudong Qu, Chen-Chen Chang, Wei Deng, Hongmin Ma, Lu Zhu, Yang Song, and Lu Yang
- Subjects
Models, Molecular ,Ketone ,Protein Engineering ,Biochemistry ,Catalysis ,Catalytic Domain ,Enzyme Stability ,Molecular Biology ,Alcohol dehydrogenase ,Thermostability ,chemistry.chemical_classification ,Molecular Structure ,biology ,Chemistry ,Organic Chemistry ,Alcohol Dehydrogenase ,Temperature ,Substrate (chemistry) ,Protein engineering ,Combinatorial chemistry ,Saccharum ,Enzyme ,Biocatalysis ,Alcohols ,biology.protein ,Molecular Medicine - Abstract
Leifsonia alcohol dehydrogenase (LnADH) is a promising biocatalyst for the synthesis of chiral alcohols. However, limitations of wild-type LnADH observed for practical application include low activity and poor stability. In this work, protein engineering was employed to improve its thermostability and catalytic efficiency by altering the subunit interfaces. Residues of T100 and S148 were identified to be significant for thermostability and activity, and the melting temperature (ΔTm) and catalytic efficiency of the mutant T100R/S148I toward ketone substrates were improved by 18.7 °C and 1.8-5.5-fold, respectively. Solving the crystal structures of the wild-type enzyme and T100R/S148L reveals the beneficial effects of mutations on stability and catalytic activity. The most robust mutant T100R/S148I is promising for industrial applications and can produce 200 g liter -1 day -1 chiral alcohols at 50 °C by only a 1:500 ratio of enzyme to substrate.
- Published
- 2021
- Full Text
- View/download PDF
39. Structural biology of human telomerase: progress and prospects
- Author
-
Thi Hoang Duong Nguyen
- Subjects
Models, Molecular ,Telomerase ,Computational biology ,Biology ,Biochemistry ,Dyskeratosis Congenita ,Structural Biology ,Catalytic Domain ,human telomerase ,Humans ,Review Articles ,Ribonucleoprotein ,Genome stability ,Human telomerase ,Cryoelectron Microscopy ,Telomere Homeostasis ,Telomere ,telomeres ,Reverse transcriptase ,Structural biology ,Eukaryotic chromosome fine structure ,Mutation ,Biocatalysis ,RNA ,cryo-EM ,Holoenzymes - Abstract
Telomerase ribonucleoprotein was discovered over three decades ago as a specialized reverse transcriptase that adds telomeric repeats to the ends of linear eukaryotic chromosomes. Telomerase plays key roles in maintaining genome stability; and its dysfunction and misregulation have been linked to different types of cancers and a spectrum of human genetic disorders. Over the years, a wealth of genetic and biochemical studies of human telomerase have illuminated its numerous fascinating features. Yet, structural studies of human telomerase have lagged behind due to various challenges. Recent technical developments in cryo-electron microscopy have allowed for the first detailed visualization of the human telomerase holoenzyme, revealing unprecedented insights into its active site and assembly. This review summarizes the cumulative work leading to the recent structural advances, as well as highlights how the future structural work will further advance our understanding of this enzyme.
- Published
- 2021
40. PTP1B inhibition studies of biological active phloroglucinols from the rhizomes of Dryopteris crassirhizoma: Kinetic properties and molecular docking simulation
- Author
-
Jae Sue Choi, Seo Young Yang, Vu Thi Oanh, Byung Sun Min, Nguyen Viet Phong, and Jeong Ah Kim
- Subjects
Dryopteris ,Dryopteris crassirhizoma ,Stereochemistry ,Proton Magnetic Resonance Spectroscopy ,In silico ,Phloroglucinol ,Biochemistry ,Molecular Docking Simulation ,chemistry.chemical_compound ,Ursolic acid ,Structural Biology ,Catalytic Domain ,Carbon-13 Magnetic Resonance Spectroscopy ,Molecular Biology ,Sodium orthovanadate ,Protein Tyrosine Phosphatase, Non-Receptor Type 1 ,chemistry.chemical_classification ,biology ,General Medicine ,biology.organism_classification ,In vitro ,Kinetics ,Enzyme ,chemistry ,Two-dimensional nuclear magnetic resonance spectroscopy ,Rhizome - Abstract
By various chromatographic methods, 30 phloroglucinols (1−30) were isolated from a methanol extract of Dryopteris crassirhizoma, including two new dimeric phloroglucinols (13 and 25). The structures of the isolates were confirmed by HR−MS, 1D, and 2D NMR as well as by comparison with the literature. The protein tyrosine phosphatase 1B (PTP1B) effects of the isolated compounds (1–30) were evaluated using sodium orthovanadate and ursolic acid as a positive control. Among them, trimeric phloroglucinols 26–28 significantly exhibited the PTP1B inhibitory effects with the IC50 values of 1.19 ± 0.13, 1.00 ± 0.04, 1.23 ± 0.05 μM, respectively. In addition, the kinetic analysis revealed compounds 26–28 acted as competitive inhibitors against PTP1B enzyme with Ki values of 0.63, 0.61, 1.57 μM, respectively. Molecular docking simulations were performed to demonstrate that these active compounds can bind with the catalytic sites of PTP1B with negative binding energies and the results are in accordance with that of the kinetic studies. In vitro and in silico results suggest that D. crassirhizoma rhizomes together with compounds 26–28 are potential candidates for treating type 2 diabetes.
- Published
- 2021
- Full Text
- View/download PDF
41. Structure and inhibition of Cryptococcus neoformans sterylglucosidase to develop antifungal agents
- Author
-
Maurizio Del Poeta, Michael V. Airola, Jinwoo Kim, Timothy Clement, Reece M. Hoffmann, Robert C. Rizzo, John E. Burke, Iwao Ojima, Adam Taouil, and Nivea Pereira de Sa
- Subjects
CD4-Positive T-Lymphocytes ,Models, Molecular ,Antifungal Agents ,Secondary infection ,Science ,Mutant ,General Physics and Astronomy ,Crystallography, X-Ray ,General Biochemistry, Genetics and Molecular Biology ,Virulence factor ,Article ,Microbiology ,Fungal Proteins ,chemistry.chemical_compound ,Mice ,Glycolipid ,Catalytic Domain ,Ergosterol ,Animals ,X-ray crystallography ,Cryptococcus neoformans ,Multidisciplinary ,biology ,Drug discovery ,General Chemistry ,Cryptococcosis ,Pathogenic fungus ,biology.organism_classification ,In vitro ,High-Throughput Screening Assays ,Molecular Docking Simulation ,Disease Models, Animal ,Sterols ,chemistry ,Enzyme mechanisms ,Fungal pathogenesis ,Female ,Glucosidases - Abstract
Pathogenic fungi exhibit a heavy burden on medical care and new therapies are needed. Here, we develop the fungal specific enzyme sterylglucosidase 1 (Sgl1) as a therapeutic target. Sgl1 converts the immunomodulatory glycolipid ergosterol 3β-D-glucoside to ergosterol and glucose. Previously, we found that genetic deletion of Sgl1 in the pathogenic fungus Cryptococcus neoformans (Cn) results in ergosterol 3β-D-glucoside accumulation, renders Cn non-pathogenic, and immunizes mice against secondary infections by wild-type Cn, even in condition of CD4+ T cell deficiency. Here, we disclose two distinct chemical classes that inhibit Sgl1 function in vitro and in Cn cells. Pharmacological inhibition of Sgl1 phenocopies a growth defect of the Cn Δsgl1 mutant and prevents dissemination of wild-type Cn to the brain in a mouse model of infection. Crystal structures of Sgl1 alone and with inhibitors explain Sgl1’s substrate specificity and enable the rational design of antifungal agents targeting Sgl1., Sterylglucosidase 1 (Sgl1) is a virulence factor in Cryptococcus neoformans that modulates fungal pathogenesis and host response. Here, the authors characterize Sgl1 structurally, identify Sgl1 inhibitors, and demonstrate Sgl1 inhibition has efficacy in mouse models of infection.
- Published
- 2021
42. Recombinant Production of Bovine Enteropeptidase Light Chain in SHuffle® T7 Express and Optimization of Induction Parameters
- Author
-
Mohammad Shoae, Mohsen Khorashadizadeh, and Hossein Safarpour
- Subjects
chemistry.chemical_classification ,Enteropeptidase ,Serine protease ,biology ,Protein subunit ,Organic Chemistry ,lac operon ,Bioengineering ,Biochemistry ,Analytical Chemistry ,law.invention ,Kinetics ,Enzyme ,chemistry ,law ,Catalytic Domain ,biology.protein ,Recombinant DNA ,Animals ,Cattle ,Inducer ,Thioredoxin ,Plasmids - Abstract
Enteropeptidase is a duodenum serine protease that triggers the activation of pancreatic enzymes by remarkably specific cleavages after lysine residues of peptidyl substrate (Asp)4-Lys. This high specific cleavage makes the enzyme a widely used biotechnological tool in laboratory researches and industrial scale. Previous studies both in small and large scales were showed low expression and miss-folding of the expressed protein. In this study, the DNA sequence encoding the light chain (catalytic subunit) of bovine enteropeptidase (EPL) was subcloned into plasmid pET-32b, downstream to the DNA encoding the fusion partner thioredoxin immediately after the EPL cleavage site. SHuffle® T7 Express was selected as an expression host due to the ability to promote proper folding and correction of the mis-oxidized bonds. Expression and purification of protein was performed, and the result of biological activity confirmed that the active EPL was obtained. Optimization of protein expression conditions was accomplished by response surface methodology for significant factors including induction temperature, duration of induction, inducer concentration and OD600 of induction. The best conditions were achieved in 1.05 mM IPTG at OD600 of 0.6 for seven h incubation at 26.5 °C, and a high level of protein expression was obtained in the optimized condition.
- Published
- 2021
- Full Text
- View/download PDF
43. Cytochrome P450‐catalyzed biosynthesis of furanoditerpenoids in the bioenergy crop switchgrass ( Panicum virgatum L.)
- Author
-
Yuxuan Chen, Kyle A. Pelot, Kira Tiedge, John T. Lovell, Andrew Muchlinski, Danielle Davisson, Lisl Chew, Jason S. Fell, Philipp Zerbe, Meirong Jia, and Justin B. Siegel
- Subjects
cytochrome P450 monooxygenase ,plant specialized metabolism ,Metabolite ,Plant Biology & Botany ,Plant Biology ,Plant Science ,Panicum ,Plant Roots ,chemistry.chemical_compound ,Cytochrome P-450 Enzyme System ,Biosynthesis ,Catalytic Domain ,Genetics ,Plant Proteins ,Biological Products ,Genome ,biology ,diterpenoid biosynthesis ,fungi ,Mutagenesis ,food and beverages ,Active site ,Cytochrome P450 ,Plant ,Cell Biology ,Monooxygenase ,biology.organism_classification ,Biosynthetic Pathways ,plant natural products ,Panicum virgatum ,chemistry ,Biochemistry ,Biocatalysis ,biology.protein ,Biochemistry and Cell Biology ,Diterpenes ,Diterpene ,Genome, Plant - Abstract
Specialized diterpenoid metabolites are important mediators of plant-environment interactions in monocot crops. To understand metabolite functions in plant environmental adaptation that ultimately can enable crop improvement strategies, a deeper knowledge of the underlying species-specific biosynthetic pathways is required. Here, we report the genomics-enabled discovery of five cytochrome P450 monooxygenases (CYP71Z25-CYP71Z29) that form previously unknown furanoditerpenoids in the monocot bioenergy crop Panicum virgatum (switchgrass). Combinatorial pathway reconstruction showed that CYP71Z25-CYP71Z29 catalyze furan ring addition directly to primary diterpene alcohol intermediates derived from distinct class II diterpene synthase products. Transcriptional co-expression patterns and the presence of select diterpenoids in switchgrass roots support the occurrence of P450-derived furanoditerpenoids in planta. Integrating molecular dynamics, structural analysis and targeted mutagenesis identified active site determinants that contribute to the distinct catalytic specificities underlying the broad substrate promiscuity of CYP71Z25-CYP71Z29 for native and non-native diterpenoids.
- Published
- 2021
- Full Text
- View/download PDF
44. Structural and catalytic characterization of Blastochloris viridis and Pseudomonas aeruginosa homospermidine synthases supports the essential role of cation–π interaction
- Author
-
Axel J. Scheidig and Felix Helfrich
- Subjects
Models, Molecular ,transferases ,Rossmann fold ,Protein Conformation ,Stereochemistry ,Nicotinamide adenine dinucleotide ,Crystallography, X-Ray ,medicine.disease_cause ,Substrate Specificity ,03 medical and health sciences ,chemistry.chemical_compound ,Blastochloris viridis ,Structural Biology ,Catalytic Domain ,Cations ,Polyamines ,homospermidine synthases ,medicine ,putrescine ,ddc:530 ,030304 developmental biology ,Indole test ,chemistry.chemical_classification ,Hyphomicrobiaceae ,0303 health sciences ,Alkyl and Aryl Transferases ,integumentary system ,biology ,Chemistry ,030302 biochemistry & molecular biology ,Tryptophan ,Active site ,cation–π interactions ,NAD ,Research Papers ,3. Good health ,Amino acid ,Pseudomonas aeruginosa ,biology.protein ,NAD+ kinase ,polyamine metabolism - Abstract
The homospermidine synthases from P. aeruginosa and B. viridis, as well as their single-residue variants, are compared based on crystal structures and activity assays. A high structural similarity is demonstrated, suggesting the equivalent involvement of relevant residues in the reaction mechanism and catalytic dependence on cation–π interaction., Polyamines influence medically relevant processes in the opportunistic pathogen Pseudomonas aeruginosa, including virulence, biofilm formation and susceptibility to antibiotics. Although homospermidine synthase (HSS) is part of the polyamine metabolism in various strains of P. aeruginosa, neither its role nor its structure has been examined so far. The reaction mechanism of the nicotinamide adenine dinucleotide (NAD+)-dependent bacterial HSS has previously been characterized based on crystal structures of Blastochloris viridis HSS (BvHSS). This study presents the crystal structure of P. aeruginosa HSS (PaHSS) in complex with its substrate putrescine. A high structural similarity between PaHSS and BvHSS with conservation of the catalytically relevant residues is demonstrated, qualifying BvHSS as a model for mechanistic studies of PaHSS. Following this strategy, crystal structures of single-residue variants of BvHSS are presented together with activity assays of PaHSS, BvHSS and BvHSS variants. For efficient homospermidine production, acidic residues are required at the entrance to the binding pocket (‘ionic slide’) and near the active site (‘inner amino site’) to attract and bind the substrate putrescine via salt bridges. The tryptophan residue at the active site stabilizes cationic reaction components by cation–π interaction, as inferred from the interaction geometry between putrescine and the indole ring plane. Exchange of this tryptophan for other amino acids suggests a distinct catalytic requirement for an aromatic interaction partner with a highly negative electrostatic potential. These findings substantiate the structural and mechanistic knowledge on bacterial HSS, a potential target for antibiotic design.
- Published
- 2021
- Full Text
- View/download PDF
45. Plasmodium falciparum metacaspase-2 capture its natural substrate in a non-canonical way
- Author
-
Dinesh Gupta, Ajay K. Saxena, Inderjeet Kalia, Rajan Pandey, E Srinivasan, Agam P. Singh, Kailash C. Pandey, R Rajaekaran, and Vandana
- Subjects
Cell Death ,biology ,Chemistry ,Plasmodium falciparum ,Apoptosis ,General Medicine ,Substrate (biology) ,biology.organism_classification ,Biochemistry ,Metacaspase ,Non canonical ,Caspases ,Catalytic Domain ,Biophysics ,Humans ,Molecular Biology - Abstract
Programmed cell death (PCD) is a multi-step process initiated by a set of proteases, which interacts and cleaves diverse proteins, thus modulating their biochemical and cellular functions. In metazoans, PCD is mediated by proteolytic enzymes called caspases, which triggered cell death by proteolysis of human Tudor staphylococcus nuclease (TSN). Non-metazoans lack a close homologue of caspases but possess an ancestral family of cysteine proteases termed ‘metacaspases’. Studies supported that metacaspases are involved in PCD, but their natural substrates remain unknown. In this study, we performed the Plasmodium falciparum TSN (PfTSN) cleavage assay using wild and selected mutants of P. falciparum metacaspases-2 (PfMCA-2) in vitro and in vivo. Interestingly, PfMCA-2, cleaved a phylogenetically conserved protein, PfTSN at multiple sites. Deletion or substitution mutation in key interacting residues at the active site, Cys157 and His205 of PfMCA-2, impaired its enzymatic activity with the artificial substrate, z-GRR-AMC. However, the mutant Tyr224A did not affect the activity with z-GRR-AMC but abolished the cleavage of PfTSN. These results indicated that the catalytic dyad, Cys157 and His205 of PfMCA-2 was essential for its enzymatic activity with an artificial substrate, whereas Tyr224 and Cys157 residues were responsible for its interaction with the natural substrate and subsequent degradation of PfTSN. Our results suggested that MCA-2 interacts with TSN substrate in a non-canonical way using non-conserved or conformationally available residues for its binding and cleavage. In future, it would be interesting to explore how this interaction leads to the execution of PCD in the Plasmodium.
- Published
- 2021
- Full Text
- View/download PDF
46. Partial Opening of Cytochrome P450cam (CYP101A1) Is Driven by Allostery and Putidaredoxin Binding
- Author
-
Simon P. Skinner, Emanuele Paci, Thomas L. Poulos, Jeanine J. Houwing-Duistermaat, Alec H. Follmer, Marcellus Ubbink, Skinner S.P., Follmer A.H., Ubbink M., Poulos T.L., Houwing-Duistermaat J.J., and Paci E.
- Subjects
Models, Molecular ,endocrine system ,Magnetic Resonance Spectroscopy ,Camphor 5-Monooxygenase ,Cytochrome ,Protein Conformation ,Stereochemistry ,Allosteric regulation ,Cytochrome P450cam, mixture models ,Crystallography, X-Ray ,Biochemistry ,Article ,Electron transfer ,Camphor ,chemistry.chemical_compound ,Allosteric Regulation ,Bacterial Proteins ,Catalytic Domain ,Binding site ,Ferredoxin ,biology ,Pseudomonas putida ,Chemistry ,Active site ,Catalytic cycle ,biology.protein ,Ferredoxins ,Protein Binding - Abstract
Cytochrome P450cam (CYP101A1) catalyzes the region- and stereo-specific 5-exo-hydroxylation of camphor via a multistep catalytic cycle that involves two-electron transfer steps, with an absolute requirement that the second electron be donated by the ferrodoxin, putidaredoxin (Pdx). Whether P450cam, once camphor has bound to the active site and the substrate entry channel has closed, opens up upon Pdx binding, during the second electron transfer step, or it remains closed is still a matter of debate. A potential allosteric site for camphor binding has been identified and postulated to play a role in the binding of Pdx. Here, we have revisited paramagnetic NMR spectroscopy data and determined a heterogeneous ensemble of structures that explains the data, provides a complete representation of the P450cam/Pdx complex in solution, and reconciles alternative hypotheses. The allosteric camphor binding site is always present, and the conformational changes induced by camphor binding to this site facilitates Pdx binding. We also determined that the state to which Pdx binds comprises an ensemble of structures that have features of both the open and closed state. These results demonstrate that there is a finely balanced interaction between allosteric camphor binding and the binding of Pdx at high camphor concentrations.
- Published
- 2021
- Full Text
- View/download PDF
47. Indane-1,3-diones: As Potential and Selective α-glucosidase Inhibitors, their Synthesis, in vitro and in silico Studies
- Author
-
Jamshed Iqbal, Shahnaz Perveen, Khalid Mohammed Khan, Asma Mukhtar, Shehryar Hameed, Shahid Ullah Khan, Sumera Zaib, Shazia Shah, and Kanwal
- Subjects
chemistry.chemical_classification ,biology ,Stereochemistry ,In silico ,Isatin ,Indane ,Active site ,alpha-Glucosidases ,In vitro ,Molecular Docking Simulation ,Kinetics ,Structure-Activity Relationship ,chemistry.chemical_compound ,Enzyme ,chemistry ,Catalytic Domain ,Indans ,Drug Discovery ,biology.protein ,Humans ,Moiety ,Computer Simulation ,Glycoside Hydrolase Inhibitors ,Knoevenagel condensation - Abstract
Background: Diabetes mellitus is one of the most chronic metabolic disorders. Since past few years, our research group had synthesized and evaluated libraries of heterocyclic compounds against α and β-glucosidase enzymes and found encouraging results. The current study comprises of evaluation of indane-1,3-dione as antidiabetic agents based on our previously reported results obtained from closely related moiety isatin and its derivatives. Objective: A library of twenty three indane-1,3-dione derivatives (1-23) was synthesized and evaluated for α and β-glucosidase inhibitions. Moreover, in silico docking studies were carried out to investigate the putative binding mode of selected compounds with the target enzyme. Methods: The indane-1,3-dione derivatives (1-23) were synthesized by Knoevenagel condensation of different substituted benzaldehydes with indane-1,3-dione under basic condition. The structures of synthetic molecules were deduced by using different spectroscopic techniques, including 1H-, 13C-NMR, EI-MS, and CHN analysis. Compounds (1-23) were evaluated for α and β-glucosidase inhibitions by adopting the literature protocols. Result: Off twenty three, eleven compounds displayed good to moderate activity against α- glucosidase enzyme, nonetheless, all compounds exhibited less than 50% inhibition against β- glucosidase enzyme. Compounds 1, 14, and 23 displayed good activity against α-glucosidase enzyme with IC50 values of 2.80 ± 0.11, 0.76 ± 0.01, and 2.17 ± 0.18 μM, respectively. The results have shown that these compounds have selectively inhibited the α-glucosidase enzyme. The in silico docking studies also supported the above results and showed different types of interactions of synthetic molecules with the active site of enzyme. Conclusion: The compounds 1, 14, and 23 have shown good inhibition against α-glucosidase and may potentially serve as lead for the development of new therapeutic representatives.
- Published
- 2021
- Full Text
- View/download PDF
48. Pyrazole Derivatives of Medically Relevant Phenolic Acids: Insight into Antioxidative and Anti-LOX Activity
- Author
-
Jovica Branković, Dušica Simijonović, Zorica D. Petrović, Vesna Milovanović, Vladimir P. Petrović, Goran A. Bogdanović, Milan Mladenović, and Slađana B. Novaković
- Subjects
DPPH ,Pyrazole ,Ligands ,01 natural sciences ,Antioxidants ,Protocatechuic acid ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Catalytic Domain ,Cell Line, Tumor ,Drug Discovery ,Hydroxybenzoates ,Vanillic acid ,Humans ,Moiety ,Organic chemistry ,Lipoxygenase Inhibitors ,030304 developmental biology ,0303 health sciences ,Catechol ,biology ,Active site ,Ligand (biochemistry) ,0104 chemical sciences ,Molecular Docking Simulation ,010404 medicinal & biomolecular chemistry ,chemistry ,Drug Design ,biology.protein ,Pyrazoles - Abstract
Background: From the point of view of medicinal chemistry, compounds containing phenolic and pyrazolic moiety are significant since they are often constituents of bioactive compounds. Objective: The aims of this study were to synthesize pyrazole derivatives of medically relevant phenolic acids, confirm their structure, and evaluate their antioxidative and anti-LOX activities. Methods: Phenolic pyrazole derivatives were obtained, starting from esters of medically relevant phenolic acids. The structures of all obtained compounds were determined by NMR and IR spectroscopy, and UV-Vis spectrophotometry. In addition, the single-crystal X-ray diffraction was used. Pyrazole derivatives were tested for their in vitro antioxidative (DPPH assay), and lipoxygenase (LOX) inhibitory activities. Radical quenching mechanism was estimated using DFT and thermodynamic approach, while molecular docking was used to estimate the binding mode within the enzyme. Results: Pyrazole derivatives were obtained in high yields. The crystal structure of a new compound 3e was determined. Pyrazole derivative with catechol moiety 3d exhibited excellent radical scavenging activity, while compound 3b exhibited the best anti-LOX activity. Molecular docking study revealed that there is no direct interaction of any ligand with the active site of LOX-Ib, but pyrazoles 3a-e behave as inhibitors blocking the approach of linoleic acid to the active site. Conclusion: In this research, protocatechuic and vanillic acid pyrazole derivatives have been obtained for the first time. In vitro antioxidative assay suggests that pyrazole derivate of protocatechuic acid is a powerful radical scavenger, while anti-LOX assay indicates a pyrazole derivative with 4-hydroxyphenyl moiety.
- Published
- 2021
- Full Text
- View/download PDF
49. Chalcones: As Potent α-amylase Enzyme Inhibitors; Synthesis, In Vitro, and In Silico Studies
- Author
-
Khan Momin, Iqbal Maryam, Wadood Abdul, Ashfaq Ur Rehman, Ali Mahboob, Rafique Rafaila, Zaman Khair, Alam Aftab, Shah Sana, Mohammed Khan Khalid, and Yousaf Muhammad
- Subjects
chemistry.chemical_classification ,Chalcone ,biology ,Molecular model ,Stereochemistry ,Aryl ,Condensation reaction ,Molecular Docking Simulation ,Structure-Activity Relationship ,chemistry.chemical_compound ,Chalcones ,Enzyme ,chemistry ,Catalytic Domain ,Drug Discovery ,medicine ,biology.protein ,Computer Simulation ,Amylase ,Enzyme Inhibitors ,alpha-Amylases ,IC50 ,Acarbose ,medicine.drug - Abstract
Background: The inhibition of α-amylase enzyme is one of the best therapeutic approach for the management of type II diabetes mellitus. Chalcone possesses a wide range of biological activities. Objective: In the current study chalcone derivatives (1-16) were synthesized and evaluated their inhibitory potential against α-amylase enzyme. Methods: For that purpose, a library of substituted (E)-1-(naphthalene-2-yl)-3-phenylprop-2-en-1-ones was synthesized by Claisen-Schmidt condensation reaction of 2-acetonaphthanone and substituted aryl benzaldehyde in the presence of base and characterized via different spectroscopic techniques such as EI-MS, HRESI-MS, 1H-, and 13C-NMR. Results: Sixteen synthetic chalcones were evaluated for in vitro porcine pancreatic α-amylase inhibition. All the chalcones demonstrated good inhibitory activities in the range of IC50 = 1.25 ± 1.05 to 2.40 ± 0.09 μM as compared to the standard commercial drug acarbose (IC50 = 1.34 ± 0.3 μM). Conclusion: Chalcone derivatives (1-16) were synthesized, characterized, and evaluated for their α- amylase inhibition. SAR revealed that electron donating groups in the phenyl ring have more influence on enzyme inhibition. However, to insight the participation of different substituents in the chalcones on the binding interactions with the α-amylase enzyme, in silico (computer simulation) molecular modeling analyses were carried out.
- Published
- 2021
- Full Text
- View/download PDF
50. Beyond the Plateau: pL Dependence of Proton Inventories as a Tool for Studying Ribozyme and Ribonuclease Catalysis
- Author
-
Michael E. Harris and Suhyun Yoon
- Subjects
Acid-Base Equilibrium ,biology ,Proton ,Chemistry ,RNase P ,Ribozyme ,Active site ,Hydrogen-Ion Concentration ,Plateau (mathematics) ,Biochemistry ,Catalysis ,Article ,Kinetics ,Ribonucleases ,Chemical physics ,Catalytic Domain ,Ionization ,Kinetic isotope effect ,Solvents ,biology.protein ,Nucleic Acid Conformation ,RNA, Catalytic ,Ribonuclease ,Hepatitis Delta Virus ,Protons - Abstract
Acid/base catalysis is an important catalytic strategy used by ribonucleases and ribozymes; however, understanding the number and identity of functional groups involved in proton transfer remains challenging. The proton inventory (PI) technique analyzes the dependence of the enzyme reaction rate on the ratio of D(2)O to H(2)O and can provide information about the number of exchangeable sites that produce isotope effects and their magnitude. The Gross–Butler (GB) equation is used to evaluate H/D fractionation factors from PI data typically collected under conditions (i.e., a “plateau” in the pH–rate profile) assuming minimal change in active site residue ionization. However, restricting PI analysis to these conditions is problematic for many ribonucleases, ribozymes, and their variants due to ambiguity in the roles of active site residues, the lack of a plateau within the accessible pL range, or cooperative interactions between active site functional groups undergoing ionization. Here, we extend the integration of species distributions for alternative enzyme states in noncooperative models of acid/base catalysis into the GB equation, first used by Bevilacqua and colleagues for the HDV ribozyme, to develop a general population-weighted GB equation that allows simulation and global fitting of the three-dimensional relationship of the D(2)O ratio (n) versus pL versus k(n)/k(0). Simulations using the GPW-GB equation of PI results for RNase A, HDVrz, and VSrz illustrate that data obtained at multiple selected pL values across the pL–rate profile can assist in the planning and interpreting of solvent isotope effect experiments to distinguish alternative mechanistic models.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.