Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Chem Inf Model ; 64(3): 1030-1042, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38224368

ABSTRACT

The sulfonamide function is used extensively as a general building block in various inhibitory scaffolds and, more specifically, as a zinc-binding group (ZBG) of metalloenzyme inhibitors. Here, we provide biochemical, structural, and computational characterization of a metallopeptidase in complex with inhibitors, where the mono- and bisubstituted sulfamide functions are designed to directly engage zinc ions of a bimetallic enzyme site. Structural data showed that while monosubstituted sulfamides coordinate active-site zinc ions via the free negatively charged amino group in a canonical manner, their bisubstituted counterparts adopt an atypical binding pattern divergent from expected positioning of corresponding tetrahedral reaction intermediates. Accompanying quantum mechanics calculations revealed that electroneutrality of the sulfamide function is a major factor contributing to the markedly lower potency of bisubstituted compounds by considerably lowering their interaction energy with the enzyme. Overall, while bisubstituted uncharged sulfamide functions can bolster favorable pharmacological properties of a given inhibitor, their use as ZBGs in metalloenzyme inhibitors might be less advantageous due to their suboptimal metal-ligand properties.


Subject(s)
Metalloproteins , Protease Inhibitors , Protease Inhibitors/pharmacology , Metalloproteins/chemistry , Zinc/metabolism , Ions
2.
Acta Crystallogr D Struct Biol ; 79(Pt 7): 655-665, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37338420

ABSTRACT

Nine new crystal structures of CG-rich DNA 18-mers with the sequence 5'-GGTGGGGGC-XZ-GCCCCACC-3', which are related to the bacterial repetitive extragenic palindromes, are reported. 18-mer oligonucleotides with the central XZ dinucleotide systematically mutated to all 16 sequences show complex behavior in solution, but all ten so far successfully crystallized 18-mers crystallized as A-form duplexes. The refinement protocol benefited from the recurrent use of geometries of the dinucleotide conformer (NtC) classes as refinement restraints in regions of poor electron density. The restraints are automatically generated at the dnatco.datmos.org web service and are available for download. This NtC-driven protocol significantly helped to stabilize the structure refinement. The NtC-driven refinement protocol can be adapted to other low-resolution data such as cryo-EM maps. To test the quality of the final structural models, a novel validation method based on comparison of the electron density and conformational similarity to the NtC classes was employed.


Subject(s)
DNA , Nucleic Acid Conformation , DNA/chemistry , Cryoelectron Microscopy/methods
3.
J Med Chem ; 64(8): 4810-4840, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33830764

ABSTRACT

Histone deacetylase 6 (HDAC6) is a promising therapeutic target for the treatment of neurodegenerative disorders. SW-100 (1a), a phenylhydroxamate-based HDAC6 inhibitor (HDAC6i) bearing a tetrahydroquinoline (THQ) capping group, is a highly potent and selective HDAC6i that was shown to be effective in mouse models of Fragile X syndrome and Charcot-Marie-Tooth disease type 2A (CMT2A). In this study, we report the discovery of a new THQ-capped HDAC6i, termed SW-101 (1s), that possesses excellent HDAC6 potency and selectivity, together with markedly improved metabolic stability and druglike properties compared to SW-100 (1a). X-ray crystallography data reveal the molecular basis of HDAC6 inhibition by SW-101 (1s). Importantly, we demonstrate that SW-101 (1s) treatment elevates the impaired level of acetylated α-tubulin in the distal sciatic nerve, counteracts progressive motor dysfunction, and ameliorates neuropathic symptoms in a CMT2A mouse model bearing mutant MFN2. Taken together, these results bode well for the further development of SW-101 (1s) as a disease-modifying HDAC6i.


Subject(s)
Charcot-Marie-Tooth Disease/drug therapy , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Quinolines/chemistry , Acetylation , Animals , Benzamides/chemistry , Benzamides/metabolism , Binding Sites , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Crystallography, X-Ray , Disease Models, Animal , Half-Life , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Phenotype , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Quinolines/metabolism , Quinolines/therapeutic use , Structure-Activity Relationship , Tubulin/metabolism
4.
Viruses ; 13(2)2021 01 27.
Article in English | MEDLINE | ID: mdl-33514045

ABSTRACT

Engineered small non-antibody protein scaffolds are a promising alternative to antibodies and are especially attractive for use in protein therapeutics and diagnostics. The advantages include smaller size and a more robust, single-domain structural framework with a defined binding surface amenable to mutation. This calls for a more systematic approach in designing new scaffolds suitable for use in one or more methods of directed evolution. We hereby describe a process based on an analysis of protein structures from the Protein Data Bank and their experimental examination. The candidate protein scaffolds were subjected to a thorough screening including computational evaluation of the mutability, and experimental determination of their expression yield in E. coli, solubility, and thermostability. In the next step, we examined several variants of the candidate scaffolds including their wild types and alanine mutants. We proved the applicability of this systematic procedure by selecting a monomeric single-domain human protein with a fold different from previously known scaffolds. The newly developed scaffold, called ProBi (Protein Binder), contains two independently mutable surface patches. We demonstrated its functionality by training it as a binder against human interleukin-10, a medically important cytokine. The procedure yielded scaffold-related variants with nanomolar affinity.


Subject(s)
Directed Molecular Evolution/methods , Proteins/chemistry , Proteins/metabolism , Amino Acid Sequence , Computer Simulation , Databases, Protein , Interleukin-10/metabolism , Protein Binding , Protein Conformation , Protein Engineering , Protein Stability , Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosomes/metabolism
5.
J Med Chem ; 63(18): 10246-10262, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32815366

ABSTRACT

Selective inhibition of histone deacetylase 6 (HDAC6) is being recognized as a therapeutic approach for cancers. In this study, we designed a new HDAC6 inhibitor, named Suprastat, using in silico simulations. X-ray crystallography and molecular dynamics simulations provide strong evidence to support the notion that the aminomethyl and hydroxyl groups in the capping group of Suprastat establish significant hydrogen bond interactions, either direct or water-mediated, with residues D460, N530, and S531, which play a vital role in regulating the deacetylase function of the enzyme and which are absent in other isoforms. In vitro characterization of Suprastat demonstrates subnanomolar HDAC6 inhibitory potency and a hundred- to a thousand-fold HDAC6 selectivity over the other HDAC isoforms. In vivo studies reveal that a combination of Suprastat and anti-PD1 immunotherapy enhances antitumor immune response, mediated by a decrease of protumoral M2 macrophages and increased infiltration of antitumor CD8+ effector and memory T-cells.


Subject(s)
Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Immunologic Factors/therapeutic use , Melanoma/drug therapy , Phenylurea Compounds/therapeutic use , Animals , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Female , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Humans , Hydrogen Bonding , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/metabolism , Immunologic Factors/chemical synthesis , Immunologic Factors/metabolism , Immunotherapy , Melanoma/therapy , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/metabolism , Protein Binding , Rats
6.
J Med Chem ; 62(18): 8557-8577, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31414801

ABSTRACT

Isoxazole is a five-membered heterocycle that is widely used in drug discovery endeavors. Here, we report the design, synthesis, and structural and biological characterization of SS-208, a novel HDAC6-selective inhibitor containing the isoxazole-3-hydroxamate moiety as a zinc-binding group as well as a hydrophobic linker. A crystal structure of the Danio rerio HDAC6/SS-208 complex reveals a bidentate coordination of the active-site zinc ion that differs from the preferred monodentate coordination observed for HDAC6 complexes with phenylhydroxamate-based inhibitors. While SS-208 has minimal effects on the viability of murine SM1 melanoma cells in vitro, it significantly reduced in vivo tumor growth in a murine SM1 syngeneic melanoma mouse model. These findings suggest that the antitumor activity of SS-208 is mainly mediated by immune-related antitumor activity as evidenced by the increased infiltration of CD8+ and NK+ T cells and the enhanced ratio of M1 and M2 macrophages in the tumor microenvironment.


Subject(s)
Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Isoxazoles/pharmacology , Melanoma/drug therapy , Animals , CD8-Positive T-Lymphocytes/cytology , Catalytic Domain , Cell Line, Tumor , Drug Discovery , Histone Deacetylase Inhibitors/chemistry , Humans , Hydroxamic Acids/chemistry , Isoxazoles/chemistry , Macrophages/cytology , Mice , Microsomes/chemistry , Natural Killer T-Cells/cytology , Transplantation, Isogeneic , Zebrafish , Zinc/chemistry
7.
FASEB J ; 33(3): 4035-4045, 2019 03.
Article in English | MEDLINE | ID: mdl-30496698

ABSTRACT

Histone deacetylase 6 (HDAC6) is a multidomain cytosolic hydrolase acting mostly on nonhistone protein substrates. Investigations of the substrate specificity of HDAC6 are confounded by the presence of 2 catalytically active deacetylase domains (DD1 and DD2). In this study, acetylome peptide microarrays and peptide libraries were used to map the substrate specificity of DD1 and DD2 of human HDAC6. The results show that DD1 is solely responsible for the deacetylation of substrates harboring the acetyllysine at their C terminus, whereas DD2 exclusively deacetylates peptides with an internal acetyllysine residue. Also, statistical analysis of the deacetylation data revealed amino acid preferences at individual positions flanking the acetyllysine, where glycine and arginine residues are favored at positions N-terminal to the central acetyllysine; negatively charged glutamate is strongly disfavored throughout the sequence. Finally, the deacylation activity of HDAC6 was profiled by using a panel of acyl derivatives of the optimized peptide substrate and showed that HDAC6 acts as a proficient deformylase. Our data thus offer a detailed insight into the substrate preferences of the individual HDAC6 domains at the peptide level, and these findings can in turn help in elucidating the biologic roles of the enzyme and facilitate the development of new domain-specific inhibitors as research tools or therapeutic agents.-Kutil, Z., Skultetyova, L., Rauh, D., Meleshin, M., Snajdr, I., Novakova, Z., Mikesova, J., Pavlicek, J., Hadzima, M., Baranova, P., Havlinova, B., Majer, P., Schutkowski, M., Barinka, C. The unraveling of substrate specificity of histone deacetylase 6 domains using acetylome peptide microarrays and peptide libraries.


Subject(s)
Catalytic Domain , Histone Deacetylase 6/chemistry , HEK293 Cells , Histone Deacetylase 6/metabolism , Humans , Lysine/chemistry , Lysine/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Static Electricity , Substrate Specificity
8.
Sci Rep ; 7(1): 11547, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912522

ABSTRACT

Human histone deacetylase 6 (HDAC6) is the major deacetylase responsible for removing the acetyl group from Lys40 of α-tubulin (αK40), which is located lumenally in polymerized microtubules. Here, we provide a detailed kinetic analysis of tubulin deacetylation and HDAC6/microtubule interactions using individual purified components. Our data unequivocally show that free tubulin dimers represent the preferred HDAC6 substrate, with a K M value of 0.23 µM and a deacetylation rate over 1,500-fold higher than that of assembled microtubules. We attribute the lower deacetylation rate of microtubules to both longitudinal and lateral lattice interactions within tubulin polymers. Using TIRF microscopy, we directly visualized stochastic binding of HDAC6 to assembled microtubules without any detectable preferential binding to microtubule tips. Likewise, indirect immunofluorescence microscopy revealed that microtubule deacetylation by HDAC6 is carried out stochastically along the whole microtubule length, rather than from the open extremities. Our data thus complement prior studies on tubulin acetylation and further strengthen the rationale for the correlation between tubulin acetylation and microtubule age.


Subject(s)
Histone Deacetylase 6/metabolism , Microtubules/metabolism , Tubulin/metabolism , Histone Deacetylase 6/chemistry , Humans , Kinetics , Microscopy, Fluorescence , Substrate Specificity
9.
J Med Chem ; 59(10): 4539-50, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27074627

ABSTRACT

Inhibition of glutamate carboxypeptidase II (GCPII) is effective in preclinical models of neurological disorders associated with excessive activation of glutamatergic systems. Here we report synthesis, structural characterization, and biological activity of new hydroxamic acid-based inhibitors with nanomolar affinity for human GCPII. Crystal structures of GCPII/hydroxamate complexes revealed an unprecedented binding mode in which the putative P1' glutarate occupies the spacious entrance funnel rather than the conserved glutamate-binding S1' pocket. This unique binding mode provides a mechanistic explanation for the structure-activity relationship data, most notably the lack of enantiospecificity and the tolerance for bulky/hydrophobic functions as substituents of a canonical glutarate moiety. The in vivo pharmacokinetics profile of one of the inhibitors will be presented along with analgesic efficacy data from the rat chronic constrictive injury model of neuropathic pain.


Subject(s)
Enzyme Inhibitors/pharmacology , Glutamate Carboxypeptidase II/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Antigens, Surface/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glutamate Carboxypeptidase II/metabolism , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 24(10): 2340-5, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24731280

ABSTRACT

Urea-based inhibitors of human glutamate carboxypeptidase II (GCPII) have advanced into clinical trials for imaging metastatic prostate cancer. In parallel efforts, agents with increased lipophilicity have been designed and evaluated for targeting GCPII residing within the neuraxis. Here we report the structural and computational characterization of six complexes between GCPII and P1'-diversified urea-based inhibitors that have the C-terminal glutamate replaced by more hydrophobic moieties. The X-ray structures are complemented by quantum mechanics calculations that provide a quantitative insight into the GCPII/inhibitor interactions. These data can be used for the rational design of novel glutamate-free GCPII inhibitors with tailored physicochemical properties.


Subject(s)
Enzyme Inhibitors/chemistry , Glutamate Carboxypeptidase II/antagonists & inhibitors , Urea/analogs & derivatives , Antigens, Surface/chemistry , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/pharmacology , Glutamate Carboxypeptidase II/chemistry , Humans , Kinetics , Models, Molecular , Molecular Conformation , Protein Conformation , Structure-Activity Relationship , Urea/chemistry , Urea/pharmacology
11.
J Biomol Screen ; 17(8): 1030-40, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22751730

ABSTRACT

Glutamate carboxypeptidase II (GCPII) is an important target for therapeutic and diagnostic interventions aimed at prostate cancer and neurologic disorders. Here we describe the development and optimization of a high-throughput screening (HTS) assay based on fluorescence polarization (FP) that facilitates the identification of novel scaffolds inhibiting GCPII. First, we designed and synthesized a fluorescence probe based on a urea-based inhibitory scaffold covalently linked to a Bodipy TMR fluorophore (TMRGlu). Next, we established and optimized conditions suitable for HTS and evaluated the assay robustness by testing the influence of a variety of physicochemical parameters (e.g., pH, temperature, time) and additives. Using known GCPII inhibitors, the FP assay was shown to be comparable to benchmark assays established in the field. Finally, we evaluated the FP assay by HTS of a 20 000-compound library. The novel assay presented here is robust, highly reproducible (Z' = 0.82), inexpensive, and suitable for automation, thus providing an excellent platform for HTS of small-molecule libraries targeting GCPII.


Subject(s)
Fluorescence Polarization/methods , Glutamate Carboxypeptidase II/antagonists & inhibitors , High-Throughput Screening Assays/methods , Antigens, Surface/genetics , Antigens, Surface/metabolism , Fluorescent Dyes/chemical synthesis , Glutamate Carboxypeptidase II/genetics , Glutamate Carboxypeptidase II/metabolism , Humans , Ligands , Protein Binding , Small Molecule Libraries/pharmacology
12.
Blood Cells Mol Dis ; 45(3): 219-22, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20692194

ABSTRACT

Hemoglobin Haná [ß63(E7) His-Asn] is an unstable hemoglobin variant that was described in a Czech proband and her sister with Heinz body hemolytic anemia. The mother bearing the same mutation was asymptomatic; nevertheless, all three carriers had the same proportion of the mutant globin chains. Assessment of several erythrocyte antioxidant parameters revealed that both symptomatic children, unlike their asymptomatic mother, had significantly decreased glutathione reductase (GR) activity. Their GR activities were restorable in vitro by flavin adenine dinucleotide. The riboflavin supplementation improved their glutathione metabolism and ameliorated their hemolysis. Pre- and post-treatment assessment of the B(2) vitamers indicated suboptimal pre-treatment vitamin B(2) status in both children. This study provides evidence that partial GR deficiency may alter the clinical manifestation of an unstable hemoglobinopathy.


Subject(s)
Anemia, Hemolytic , Family , Glutathione Reductase/metabolism , Heinz Bodies , Hemoglobins, Abnormal/genetics , Mutation, Missense , Riboflavin/administration & dosage , Vitamin B Complex/administration & dosage , Adolescent , Adult , Amino Acid Substitution , Anemia, Hemolytic/blood , Anemia, Hemolytic/drug therapy , Anemia, Hemolytic/genetics , Female , Flavin-Adenine Dinucleotide/pharmacology , Glutathione/metabolism , Glutathione Reductase/genetics , Hemoglobinopathies/blood , Hemoglobinopathies/drug therapy , Hemoglobinopathies/genetics , Humans , Male
13.
BMC Evol Biol ; 10: 154, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20500864

ABSTRACT

BACKGROUND: The arylalkylamine N-acetyltransferase (AANAT) family is divided into structurally distinct vertebrate and non-vertebrate groups. Expression of vertebrate AANATs is limited primarily to the pineal gland and retina, where it plays a role in controlling the circadian rhythm in melatonin synthesis. Based on the role melatonin plays in biological timing, AANAT has been given the moniker "the Timezyme". Non-vertebrate AANATs, which occur in fungi and protists, are thought to play a role in detoxification and are not known to be associated with a specific tissue. RESULTS: We have found that the amphioxus genome contains seven AANATs, all having non-vertebrate type features. This and the absence of AANATs from the genomes of Hemichordates and Urochordates support the view that a major transition in the evolution of the AANATs may have occurred at the onset of vertebrate evolution. Analysis of the expression pattern of the two most structurally divergent AANATs in Branchiostoma lanceolatum (bl) revealed that they are expressed early in development and also in the adult at low levels throughout the body, possibly associated with the neural tube. Expression is clearly not exclusively associated with the proposed analogs of the pineal gland and retina. blAANAT activity is influenced by environmental lighting, but light/dark differences do not persist under constant light or constant dark conditions, indicating they are not circadian in nature. bfAANAT alpha and bfAANAT delta' have unusually alkaline (> 9.0) optimal pH, more than two pH units higher than that of vertebrate AANATs. CONCLUSIONS: The substrate selectivity profiles of bfAANAT alpha and delta' are relatively broad, including alkylamines, arylalkylamines and diamines, in contrast to vertebrate forms, which selectively acetylate serotonin and other arylalkylamines. Based on these features, it appears that amphioxus AANATs could play several roles, including detoxification and biogenic amine inactivation. The presence of seven AANATs in amphioxus genome supports the view that arylalkylamine and polyamine acetylation is important to the biology of this organism and that these genes evolved in response to specific pressures related to requirements for amine acetylation.


Subject(s)
Arylalkylamine N-Acetyltransferase/genetics , Chordata, Nonvertebrate/genetics , Evolution, Molecular , Multigene Family , Amino Acid Sequence , Animals , Chordata, Nonvertebrate/enzymology , DNA, Complementary/genetics , Gene Expression , Likelihood Functions , Molecular Sequence Data , Phylogeny , Sequence Alignment , Sequence Analysis, DNA , Substrate Specificity
14.
J Biol Chem ; 283(21): 14552-8, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18362150

ABSTRACT

Arylalkylamine N-acetyltransferase (AANAT) catalyzes the N-acetylation of serotonin, the penultimate step in the synthesis of melatonin. Pineal AANAT activity increases at night in all vertebrates, resulting in increased melatonin production. This increases circulating levels of melatonin, thereby providing a hormonal signal of darkness. Kinetic and structural analysis of AANAT has determined that one element is floppy. This element, termed Loop 1, is one of three loops that comprise the arylalkylamine binding pocket. During the course of chordate evolution, Loop 1 acquired the tripeptide CPL, and the enzyme became highly active. Here we focused on the functional importance of the CPL tripeptide and found that activity was markedly reduced when it was absent. Moreover, increasing the local flexibility of this tripeptide region by P64G and P64A mutations had the counterintuitive effect of reducing activity and reducing the overall movement of Loop 1, as estimated from Langevin dynamics simulations. Binding studies indicate that these mutations increased the off-rate constant of a model substrate without altering the dissociation constant. The structural kink and local rigidity imposed by Pro-64 may enhance activity by favoring configurations of Loop 1 that facilitate catalysis and do not become immobilized by intramolecular interactions.


Subject(s)
Arylalkylamine N-Acetyltransferase/chemistry , Arylalkylamine N-Acetyltransferase/metabolism , Animals , Arylalkylamine N-Acetyltransferase/genetics , Gene Expression , Guanidine , Models, Molecular , Mutation/genetics , Proline/genetics , Proline/metabolism , Protein Denaturation , Protein Structure, Tertiary , Sheep , Substrate Specificity
15.
Biochemistry ; 42(31): 9295-306, 2003 Aug 12.
Article in English | MEDLINE | ID: mdl-12899616

ABSTRACT

CD69 is the earliest leukocyte activation antigen playing a pivotal role in cellular signaling. Here, we show that a globular C-terminal domain of CD69 belonging to C-type lectins binds calcium through Asp 171, Glu 185, and Glu 187 with K(d) approximately 54 microM. Closure of the calcium-binding site results in a conformational shift of Thr 107 and Lys 172. Interestingly, structural changes in all of these amino acids lead to the formation of high-affinity binding sites for N-acetyl-D-glucosamine. Similarly, a structural change in Glu 185 and Glu 187 contributes to a high-affinity site for N-acetyl-D-galactosamine. Site-directed mutagenesis and molecular modeling allowed us to describe the structural details of binding sites for both carbohydrates. These studies explain the importance of calcium for recognition of carbohydrates by CD69 and provide an important paradigm for the role of weak interactions in the immune system.


Subject(s)
Acetylglucosamine/metabolism , Antigens, CD/chemistry , Antigens, Differentiation, T-Lymphocyte/chemistry , Calcium/metabolism , Lymphocytes/metabolism , Amino Acid Sequence , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , DNA Primers/chemistry , Escherichia coli , Humans , Kinetics , Lectins, C-Type , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Polymerase Chain Reaction , Protein Folding , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...