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










Publication year range
1.
ACS Chem Biol ; 15(6): 1604-1612, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32378881

ABSTRACT

Protein-protein interactions (PPIs) play a critical role in fundamental biological processes. Competitive inhibition of these interfaces requires compounds that can access discontinuous binding epitopes along a large, shallow binding surface area. Conformationally defined protein surface mimics present a viable route to target these interactions. However, the development of minimal protein mimics that engage intracellular targets with high affinity remains a major challenge because mimicry of a portion of the binding interface is often associated with the loss of critical binding interactions. Covalent targeting provides an attractive approach to overcome the loss of noncovalent contacts but have the inherent risk of dominating noncovalent contacts and increasing the likelihood of nonselective binding. Here, we report the iterative design of a proteolytically stable α3ß chimeric helix mimic that covalently targets oncogenic Ras G12C as a model system. We explored several electrophiles to optimize preferential alkylation with the desired C12 on Ras. The designed lead peptide modulates nucleotide exchange, inhibits activation of the Ras-mediated signaling cascade, and is selectively toxic toward mutant Ras G12C cancer cells. The relatively high frequency of acquired cysteines as missense mutations in cancer and other diseases suggests that covalent peptides may offer an untapped therapeutic approach for targeting aberrant protein interactions.


Subject(s)
Drug Delivery Systems , Drug Design , Peptidomimetics/pharmacology , ras Proteins/drug effects , Biophysical Phenomena , Cell Line, Tumor , Humans , Ligands , Peptidomimetics/chemistry , Protein Conformation , Protein Interaction Maps , Proteolysis , Signal Transduction
2.
Nature ; 576(7787): 477-481, 2019 12.
Article in English | MEDLINE | ID: mdl-31827278

ABSTRACT

Oncogenic activation of RAS is associated with the acquisition of a unique set of metabolic dependencies that contribute to tumour cell fitness. Cells that express oncogenic RAS are able to internalize and degrade extracellular protein via a fluid-phase uptake mechanism termed macropinocytosis1. There is increasing recognition of the role of this RAS-dependent process in the generation of free amino acids that can be used to support tumour cell growth under nutrient-limiting conditions2. However, little is known about the molecular steps that mediate the induction of macropinocytosis by oncogenic RAS. Here we identify vacuolar ATPase (V-ATPase) as an essential regulator of RAS-induced macropinocytosis. Oncogenic RAS promotes the translocation of V-ATPase from intracellular membranes to the plasma membrane via a pathway that requires the activation of protein kinase A by a bicarbonate-dependent soluble adenylate cyclase. Accumulation of V-ATPase at the plasma membrane is necessary for the cholesterol-dependent plasma-membrane association of RAC1, a prerequisite for the stimulation of membrane ruffling and macropinocytosis. These observations establish a link between V-ATPase trafficking and nutrient supply by macropinocytosis that could be exploited to curtail the metabolic adaptation capacity of RAS-mutant tumour cells.


Subject(s)
Cell Membrane/enzymology , Oncogene Protein p21(ras)/metabolism , Pinocytosis , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Bicarbonates/metabolism , Carcinogenesis , Cell Line, Tumor , Cell Membrane/metabolism , Cholesterol/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Sodium-Bicarbonate Symporters/metabolism
4.
J Biol Chem ; 291(12): 6534-45, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26814130

ABSTRACT

The small GTPase DiRas1 has tumor-suppressive activities, unlike the oncogenic properties more common to small GTPases such as K-Ras and RhoA. Although DiRas1 has been found to be a tumor suppressor in gliomas and esophageal squamous cell carcinomas, the mechanisms by which it inhibits malignant phenotypes have not been fully determined. In this study, we demonstrate that DiRas1 binds to SmgGDS, a protein that promotes the activation of several oncogenic GTPases. In silico docking studies predict that DiRas1 binds to SmgGDS in a manner similar to other small GTPases. SmgGDS is a guanine nucleotide exchange factor for RhoA, but we report here that SmgGDS does not mediate GDP/GTP exchange on DiRas1. Intriguingly, DiRas1 acts similarly to a dominant-negative small GTPase, binding to SmgGDS and inhibiting SmgGDS binding to other small GTPases, including K-Ras4B, RhoA, and Rap1A. DiRas1 is expressed in normal breast tissue, but its expression is decreased in most breast cancers, similar to its family member DiRas3 (ARHI). DiRas1 inhibits RhoA- and SmgGDS-mediated NF-κB transcriptional activity in HEK293T cells. We also report that DiRas1 suppresses basal NF-κB activation in breast cancer and glioblastoma cell lines. Taken together, our data support a model in which DiRas1 expression inhibits malignant features of cancers in part by nonproductively binding to SmgGDS and inhibiting the binding of other small GTPases to SmgGDS.


Subject(s)
GTP Phosphohydrolases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , GTP Phosphohydrolases/chemistry , Guanine Nucleotide Exchange Factors/chemistry , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , HEK293 Cells , Humans , MCF-7 Cells , Molecular Docking Simulation , NF-kappa B/metabolism , Protein Binding , Protein Structure, Secondary , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Proteins/chemistry , rhoA GTP-Binding Protein
5.
Mol Carcinog ; 54(3): 203-15, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24115212

ABSTRACT

Aggressive dissemination and metastasis of pancreatic ductal adenocarcinoma (PDAC) results in poor prognosis and marked lethality. Rho monomeric G protein levels are increased in pancreatic cancer tissue. As the mechanisms underlying PDAC malignancy are little understood, we investigated the role for cAMP in regulating monomeric G protein regulated invasion and migration of pancreatic cancer cells. Treatment of PDAC cells with cAMP elevating agents that activate adenylyl cyclases, forskolin, protein kinase A (PKA), 6-Bnz-cAMP, or the cyclic nucleotide phosphodiesterase inhibitor cilostamide significantly decreased migration and Matrigel invasion of PDAC cell lines. Inhibition was dose-dependent and not significantly different between forskolin or cilostamide treatment. cAMP elevating drugs not only blocked basal migration, but similarly abrogated transforming-growth factor-ß-directed PDAC cell migration and invasion. The inhibitory effects of cAMP were prevented by the pharmacological blockade of PKA. Drugs that increase cellular cAMP levels decreased levels of active RhoA or RhoC, with a concomitant increase in phosphorylated RhoA. Diminished Rho signaling was correlated with the appearance of thickened cortical actin bands along the perimeter of non-motile forskolin or cilostamide-treated cells. Decreased migration did not reflect alterations in cell growth or programmed cell death. Collectively these data support the notion that increased levels of cAMP specifically hinder PDAC cell motility through F-actin remodeling.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cyclic AMP/metabolism , Pancreatic Neoplasms/pathology , 1-Methyl-3-isobutylxanthine/pharmacology , Amides/pharmacology , Apoptosis , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Colforsin/pharmacology , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Neoplasm Invasiveness , Phosphodiesterase Inhibitors/pharmacology , Pyridines/pharmacology , Quinolones/pharmacology , Vasodilator Agents/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein , Gemcitabine
6.
Cell Cycle ; 13(6): 941-52, 2014.
Article in English | MEDLINE | ID: mdl-24552806

ABSTRACT

Oncogenic mutation or misregulation of small GTPases in the Ras and Rho families can promote unregulated cell cycle progression in cancer. Post-translational modification by prenylation of these GTPases allows them to signal at the cell membrane. Splice variants of SmgGDS, named SmgGDS-607 and SmgGDS-558, promote the prenylation and membrane trafficking of multiple Ras and Rho family members, which makes SmgGDS a potentially important regulator of the cell cycle. Surprisingly little is known about how SmgGDS-607 and SmgGDS-558 affect cell cycle-regulatory proteins in cancer, even though SmgGDS is overexpressed in multiple types of cancer. To examine the roles of SmgGDS splice variants in the cell cycle, we compared the effects of the RNAi-mediated depletion of SmgGDS-558 vs. SmgGDS-607 on cell cycle progression and the expression of cyclin D1, p27, and p21 in pancreatic, lung, and breast cancer cell lines. We show for the first time that SmgGDS promotes proliferation of pancreatic cancer cells, and we demonstrate that SmgGDS-558 plays a greater role than SmgGDS-607 in cell cycle progression as well as promoting cyclin D1 and suppressing p27 expression in multiple types of cancer. Silencing both splice variants of SmgGDS in the cancer cell lines produces an alternative signaling profile compared with silencing SmgGDS-558 alone. We also show that loss of both SmgGDS-607 and SmgGDS-558 simultaneously decreases tumorigenesis of NCI-H1703 non-small cell lung carcinoma (NSCLC) xenografts in mice. These findings indicate that SmgGDS promotes cell cycle progression in multiple types of cancer, making SmgGDS a valuable target for cancer therapeutics.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle , Guanine Nucleotide Exchange Factors/metabolism , Lung Neoplasms/metabolism , Pancreatic Neoplasms/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Guanine Nucleotide Exchange Factors/genetics , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, SCID , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
7.
J Biol Chem ; 289(10): 6862-6876, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24415755

ABSTRACT

Ras family small GTPases localize at the plasma membrane, where they can activate oncogenic signaling pathways. Understanding the mechanisms that promote membrane localization of GTPases will aid development of new therapies to inhibit oncogenic signaling. We previously reported that SmgGDS splice variants promote prenylation and trafficking of GTPases containing a C-terminal polybasic region and demonstrated that SmgGDS-607 interacts with nonprenylated GTPases, whereas SmgGDS-558 interacts with prenylated GTPases in cells. The mechanism that SmgGDS-607 and SmgGDS-558 use to differentiate between prenylated and nonprenylated GTPases has not been characterized. Here, we provide evidence that SmgGDS-607 associates with GTPases through recognition of the last amino acid in the CAAX motif. We show that SmgGDS-607 forms more stable complexes in cells with nonprenylated GTPases that will become geranylgeranylated than with nonprenylated GTPases that will become farnesylated. These binding relationships similarly occur with nonprenylated SAAX mutants. Intriguingly, farnesyltransferase inhibitors increase the binding of WT K-Ras to SmgGDS-607, indicating that the pharmacological shunting of K-Ras into the geranylgeranylation pathway promotes K-Ras association with SmgGDS-607. Using recombinant proteins and prenylated peptides corresponding to the C-terminal sequences of K-Ras and Rap1B, we found that both SmgGDS-607 and SmgGDS-558 directly bind the GTPase C-terminal region, but the specificity of the SmgGDS splice variants for prenylated versus nonprenylated GTPases is diminished in vitro. Finally, we present structural homology models and data from functional prediction software to define both similar and unique features of SmgGDS-607 when compared with SmgGDS-558.


Subject(s)
Guanine Nucleotide Exchange Factors/chemistry , Monomeric GTP-Binding Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Models, Chemical , Molecular Sequence Data , Monomeric GTP-Binding Proteins/genetics , Prenylation , Sequence Analysis, Protein/methods , Software
8.
Mol Cancer Res ; 12(1): 130-42, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24197117

ABSTRACT

UNLABELLED: Breast cancer malignancy is promoted by the small GTPases RhoA and RhoC. SmgGDS is a guanine nucleotide exchange factor that activates RhoA and RhoC in vitro. We previously reported that two splice variants of SmgGDS, SmgGDS-607, and SmgGDS-558, have different characteristics in binding and transport of small GTPases. To define the role of SmgGDS in breast cancer, we tested the expression of SmgGDS in breast tumors, and the role of each splice variant in proliferation, tumor growth, Rho activation, and NF-κB transcriptional activity in breast cancer cells. We show upregulated SmgGDS protein expression in breast cancer samples compared with normal breast tissue. In addition, Kaplan-Meier survival curves indicated that patients with high SmgGDS expression in their tumors had worse clinical outcomes. Knockdown of SmgGDS-558, but not SmgGDS-607, in breast cancer cells decreased proliferation, in vivo tumor growth, and RhoA activity. Furthermore, we found that SmgGDS promoted a Rho-dependent activation of the transcription factor NF-κB, which provides a potential mechanism to define how SmgGDS-mediated activation of RhoA promotes breast cancer. This study demonstrates that elevated SmgGDS expression in breast tumors correlates with poor survival, and that SmgGDS-558 plays a functional role in breast cancer malignancy. Taken together, these findings define SmgGDS-558 as a unique promoter of RhoA and NF-κB activity and a novel therapeutic target in breast cancer. IMPLICATIONS: This study defines a new mechanism to regulate the activities of RhoA and NF-κB in breast cancer cells, and identifies SmgGDS-558 as a novel promoter of breast cancer malignancy.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Guanine Nucleotide Exchange Factors/genetics , NF-kappa B/genetics , rhoA GTP-Binding Protein/genetics , Animals , Breast Neoplasms/mortality , Carcinoma, Intraductal, Noninfiltrating/mortality , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Female , Guanine Nucleotide Exchange Factors/biosynthesis , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Mice , Mice, SCID , Neoplasm Transplantation , Prognosis , Protein Binding/genetics , Protein Isoforms/genetics , Protein Transport/genetics , RNA Interference , RNA Splicing , RNA, Messenger/biosynthesis , RNA, Small Interfering , Signal Transduction/genetics , Transcription, Genetic , Transplantation, Heterologous , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein
9.
Sci Signal ; 6(277): ra39, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23716716

ABSTRACT

During metastasis, cancer cells acquire the ability to dissociate from each other and migrate, which is recapitulated in vitro as cell scattering. The small guanosine triphosphatase (GTPase) Rap1 opposes cell scattering by promoting cell-cell adhesion, a function that requires its prenylation, or posttranslational modification with a carboxyl-terminal isoprenoid moiety, to enable its localization at cell membranes. Thus, signaling cascades that regulate the prenylation of Rap1 offer a mechanism to control the membrane localization of Rap1. We identified a signaling cascade initiated by adenosine A2B receptors that suppressed the prenylation of Rap1B through phosphorylation of Rap1B, which decreased its interaction with the chaperone protein SmgGDS (small GTPase guanosine diphosphate dissociation stimulator). These events promoted the cytosolic and nuclear accumulation of nonprenylated Rap1B and diminished cell-cell adhesion, resulting in cell scattering. We found that nonprenylated Rap1 was more abundant in mammary tumors than in normal mammary tissue in rats and that activation of adenosine receptors delayed Rap1B prenylation in breast, lung, and pancreatic cancer cell lines. Our findings support a model in which high concentrations of extracellular adenosine, such as those that arise in the tumor microenvironment, can chronically activate A2B receptors to suppress Rap1B prenylation and signaling at the cell membrane, resulting in reduced cell-cell contact and promoting cell scattering. Inhibiting A2B receptors may be an effective method to prevent metastasis.


Subject(s)
Adenosine/metabolism , Cell Movement/physiology , Models, Biological , Neoplasm Metastasis/physiopathology , Signal Transduction/physiology , Tumor Microenvironment , rap GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cell Adhesion/physiology , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Confocal , Molecular Sequence Data , Prenylation , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2B/metabolism , rap GTP-Binding Proteins/genetics
10.
Cancer Biol Ther ; 13(8): 647-56, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22549160

ABSTRACT

The small GTPase Rac1 regulates many cellular processes, including cytoskeletal reorganization, cell migration, proliferation, and survival. Additionally, Rac1 plays a major role in activating NF-κB-mediated transcription. Both Rac1 and NF-κB regulate many properties of the malignant phenotype, including anchorage-independent proliferation and survival, metastasis, and angiogenesis. Despite these findings, the roles of Rac1and NF-κB in non-small cell lung carcinoma, a leading cause of cancer deaths, have not been thoroughly investigated. Here, we compared the effects of Rac1 siRNA to that of the Rac1 inhibitor NSC23766 on multiple features of the NSCLC malignant phenotype, including NF-κB activity. We show that the siRNA-mediated silencing of Rac1 in lung cancer cells results in decreased cell proliferation and migration. The decrease in proliferation was observed in both anchorage-dependent and anchorage-independent assays. Furthermore, cells with decreased Rac1 expression have a slowed progression through the G 1 phase of the cell cycle. These effects induced by Rac1 siRNA correlated with a decrease in NF-κB transcriptional activity. Additionally, inhibition of NF-κB signaling with BAY 11-7082 inhibited proliferation; indicating that the loss of cell proliferation and migration induced by the silencing of Rac1 expression may be attributed in part to loss of NF-κB activity. Interestingly, treatment with the Rac1 inhibitor NSC23766 strongly inhibits cell proliferation, cell cycle progression, and NF-κB activity in lung cancer cells, to an even greater extent than the inhibition induced by Rac1 siRNA. These findings indicate that Rac1 plays an important role in lung cancer cell proliferation and migration, most likely through its ability to promote NF-κB activity, and highlight Rac1 pathways as therapeutic targets for the treatment of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Lung Neoplasms/metabolism , NF-kappa B/metabolism , rac1 GTP-Binding Protein/metabolism , Aminoquinolines/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Gene Silencing , Humans , Lung Neoplasms/genetics , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Pyrimidines/pharmacology , Sulfones/pharmacology , Transcription, Genetic/drug effects , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics
11.
Cancer Biol Ther ; 12(8): 707-17, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21799303

ABSTRACT

Mito-CP11, a mitochondria-targeted nitroxide formed by conjugating a triphenylphosphonium cation to a five-membered nitroxide, carboxy-proxyl (CP), has been used as a superoxide dismutase (SOD) mimetic. In this study, we investigated the antiproliferative and cytotoxic properties of submicromolar levels of Mito-CP11 alone and in combination with fluvastatin, a well known cholesterol lowering drug, in breast cancer cells. Mito-CP11, but not CP or CP plus the cationic ligand, methyl triphenylphosphonium (Me-TPP+), inhibited MCF-7 breast cancer cell proliferation. Mito-CP11 had only minimal effect on MCF-10A, non-tumorigenic mammary epithelial cells. Mito-CP11, however, significantly enhanced fluvastatin-mediated cytotoxicity in MCF-7 cells. Mito-CP11 alone and in combination with fluvastatin inhibited nuclear factor kappa-B activity mainly in MCF-7 cells. We conclude that mitochondria-targeted nitroxide antioxidant molecules (such as Mito-CP11) that are non-toxic to non-tumorigenic cells could enhance the cytostatic and cytotoxic effects of statins in breast cancer cells. This strategy of combining mitochondria-targeted non-toxic molecules with cytotoxic chemotherapeutic drugs may be successfully used to enhance the efficacy of antitumor therapies in breast cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Fatty Acids, Monounsaturated/pharmacology , Indoles/pharmacology , Mitochondria/drug effects , Nitrogen Oxides/pharmacology , Antioxidants/administration & dosage , Antioxidants/chemistry , Antioxidants/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Delivery Systems , Drug Synergism , Fatty Acids, Monounsaturated/administration & dosage , Female , Fluvastatin , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Indoles/administration & dosage , Mevalonic Acid/administration & dosage , Mevalonic Acid/pharmacology , Mitochondria/metabolism , Mitochondria/pathology , Nitrogen Oxides/administration & dosage , Nitrogen Oxides/chemistry , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Reactive Oxygen Species/metabolism
12.
Biochemistry ; 48(18): 3838-46, 2009 May 12.
Article in English | MEDLINE | ID: mdl-19290655

ABSTRACT

The active site residue Thr-201 in toluene 4-monooxygenase hydroxylase (T4moH) has a structural counterpart in the active sites of all diiron monooxygenases. Thus, our previous finding that mutation of this residue to Ala, Gly, or Ser had no impact on steady-state catalysis or coupling was surprising. In this work, we provide kinetic, biochemical, and structural evidence that one role of Thr-201 may be to stabilize a peroxo-level intermediate during enzyme catalysis. During reactions in the absence of substrate, T201 T4moH slowly consumed O(2) but only a negligible amount of H(2)O(2) was released. In contrast, T201A T4moH gave stoichometric release of H(2)O(2) during reaction in the absence of substrate. Both enzyme isoforms were tightly coupled during steady-state catalysis with saturating toluene and other optimal substrates and exhibited near-identical kinetic parameters. However, rapid mix single-turnover studies showed that T201A T4moH had a faster first-order rate constant for product formation than T201 T4moH did. Comparison of X-ray crystal structures of resting and reduced T201A T4moH in complex with T4moD with comparable structures of T201 T4moHD revealed changes in the positions of several key active site residues relative to the comparable structures of T201 T4moH with T4moD. This combination of catalytic and structural studies offers important new insight into the role of the role of conserved Thr-201, and its contributions to the catalytic reaction cycle.


Subject(s)
Oxygenases/metabolism , Threonine/metabolism , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Hydrolysis , Kinetics , Models, Molecular , Oxygen/metabolism , Oxygenases/chemistry , Solubility , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...