Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Article in English | MEDLINE | ID: mdl-39093823

ABSTRACT

Many phytopathogenic bacteria require a type three secretion system (TTSS) to activate effector triggered immunity (ETI). We identified a calcium binding protein, EfhXXfa, in the citrus pathogen, X. citri subsp. aurantifolii, that does not require a TTSS to activate reactive oxygen species (ROS) and elicit a hypersensitive reaction (HR) in tomato leaves following infection. Purified, recombinant EfhXXfa was shown to bind two moles of calcium per mole of protein, whereas mutation of the first of two EF-hands did not bind calcium . EfhXXfa expression was determined to be inducible in hrp-inducing medium. Additionally, growth of X. perforans transconjugants with and without the efhXXfa gene in hrp-inducing medium differed in intracellular calcium concentration; the transconjugant without efhXXfa yielded higher cell pellet masses and higher increased intracellular calcium concentrations relative to cells expressing EfhXXfa. An EfhXXfa homolog, EfhXXe, present in the pepper pathogen, X. euvesicatoria, when expressed in the tomato pathogen, X. perforans, triggered ROS production and an HR in tomato leaves and is a host-limiting factor. Interestingly, all tested X. perforans and X. euvesicatoria strains pathogenic on tomato contain a stop codon immediately upstream of the first EF-hand domain in the efhXXe gene, whereas most X. euvesicatoria strains pathogenic on pepper do not.

2.
Acta Crystallogr D Struct Biol ; 80(Pt 3): 194-202, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38411550

ABSTRACT

The combination of X-ray free-electron lasers (XFELs) with serial femtosecond crystallography represents cutting-edge technology in structural biology, allowing the study of enzyme reactions and dynamics in real time through the generation of `molecular movies'. This technology combines short and precise high-energy X-ray exposure to a stream of protein microcrystals. Here, the XFEL structure of carbonic anhydrase II, a ubiquitous enzyme responsible for the interconversion of CO2 and bicarbonate, is reported, and is compared with previously reported NMR and synchrotron X-ray and neutron single-crystal structures.


Subject(s)
Carbonic Anhydrase II , Carbonic Anhydrase II/chemistry , Crystallography, X-Ray , Proteins/chemistry , Synchrotrons , X-Rays , Humans
3.
JCI Insight ; 8(10)2023 04 25.
Article in English | MEDLINE | ID: mdl-37097751

ABSTRACT

Although thymidylate synthase (TYMS) inhibitors have served as components of chemotherapy regimens, the currently available inhibitors induce TYMS overexpression or alter folate transport/metabolism feedback pathways that tumor cells exploit for drug resistance, limiting overall benefit. Here we report a small molecule TYMS inhibitor that i) exhibited enhanced antitumor activity as compared with current fluoropyrimidines and antifolates without inducing TYMS overexpression, ii) is structurally distinct from classical antifolates, iii) extended survival in both pancreatic xenograft tumor models and an hTS/Ink4a/Arf null genetically engineered mouse tumor model, and iv) is well tolerated with equal efficacy using either intraperitoneal or oral administration. Mechanistically, we verify the compound is a multifunctional nonclassical antifolate, and using a series of analogs, we identify structural features allowing direct TYMS inhibition while maintaining the ability to inhibit dihydrofolate reductase. Collectively, this work identifies nonclassical antifolate inhibitors that optimize inhibition of thymidylate biosynthesis with a favorable safety profile, highlighting the potential for enhanced cancer therapy.


Subject(s)
Folic Acid Antagonists , Mice , Animals , Humans , Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/therapeutic use , Folic Acid Antagonists/chemistry , Enzyme Inhibitors/pharmacology , Drug Resistance , Thymidylate Synthase
4.
Acta Crystallogr F Struct Biol Commun ; 78(Pt 11): 395-402, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36322425

ABSTRACT

Carbonic anhydrases (CAs) are drug targets for a variety of diseases. While many clinically relevant CA inhibitors are sulfonamide-based, novel CA inhibitors are being developed that incorporate alternative zinc-binding groups, such as carboxylic acid moieties, to develop CA isoform-specific inhibitors. Here, the X-ray crystal structure of human CA II (hCA II) in complex with the carboxylic acid ibuprofen [2-(4-isobutylphenyl)propanoic acid, a common over-the-counter nonsteroidal anti-inflammatory drug] is reported to 1.54 Šresolution. The binding of ibuprofen is overlaid with the structures of other carboxylic acids in complex with hCA II to compare their inhibition mechanisms by direct or indirect (via a water) binding to the active-site zinc. Additionally, enzyme-inhibition assays using ibuprofen, nicotinic acid and ferulic acid were performed with hCA II to determine their IC50 values and were compared with those of other carboxylic acid binders. This study discusses the potential development of CA inhibitors utilizing the carboxylic acid moiety.


Subject(s)
Carbonic Anhydrase II , Carbonic Anhydrase Inhibitors , Humans , Carbonic Anhydrase II/chemistry , Carbonic Anhydrase Inhibitors/chemistry , Ibuprofen/pharmacology , Crystallography, X-Ray , Carboxylic Acids/chemistry , Zinc , Structure-Activity Relationship , Carbonic Anhydrase IX/metabolism
5.
J Enzyme Inhib Med Chem ; 37(1): 930-939, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35306936

ABSTRACT

Human (h) carbonic anhydrase (CAs, EC 4.2.1.1) isoforms IX and XII were recently confirmed as anticancer targets against solid hypoxic tumours. The "three-tails approach" has been proposed as an extension of the forerunner "tail" and "dual-tail approach" to fully exploit the amino acid differences at the medium/outer active site rims among different hCAs and to obtain more isoform-selective inhibitors. Many three-tailed inhibitors (TTIs) showed higher selectivity against the tumour-associated isoforms hCA IX and XII with respect to the off-targets hCA I and II. X-ray crystallography studies were performed to investigate the binding mode of four TTIs in complex with a hCA IX mimic. The ability of the most potent and selective TTIs to reduce in vitro the viability of colon cancer (HT29), prostate adenocarcinoma (PC3), and breast cancer (ZR75-1) cell lines was evaluated in normoxic (21% O2) and hypoxic (3% O2) conditions demonstrating relevant anti-proliferative effects.


Subject(s)
Antineoplastic Agents/pharmacology , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Sulfonamides/pharmacology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
6.
J Med Chem ; 65(1): 824-837, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34958217

ABSTRACT

We report a one-pot procedure for the synthesis of asymmetrical ureido-containing benzenesulfonamides based on in situ generation of the corresponding isocyanatobenezenesulfonamide species, which were trapped with the appropriate amines. A library of new compounds was generated and evaluated in vitro for their inhibition properties against a representative panel of the human (h) metalloenzymes carbonic anhydrases (EC 4.2.1.1), and the best performing compounds on the isozyme II (i.e., 7c, 9c, 11g, and 12c) were screened for their ability to reduce the intraocular pressure in glaucomatous rabbits. In addition, the binding modes of 7c, 11f, and 11g were assessed by means of X-ray crystallography.


Subject(s)
Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/pharmacology , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Animals , Carbonic Anhydrase II/antagonists & inhibitors , Crystallography, X-Ray , Drug Design , Glaucoma/drug therapy , Intraocular Pressure/drug effects , Male , Models, Molecular , Protein Binding , Rabbits , Structure-Activity Relationship , Benzenesulfonamides
7.
IUCrJ ; 8(Pt 2): 329, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33708409

ABSTRACT

[This corrects the article DOI: 10.1107/S2052252520000986.].

8.
J Med Chem ; 64(3): 1713-1724, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33523653

ABSTRACT

Carbonic anhydrase IX (CAIX) is considered a target for therapeutic intervention in solid tumors. In this study, the efficacy of the inhibitor, 4-(3-(2,4-difluorophenyl)-oxoimidazolidin-1-yl)benzenesulfonamide (SLC-149), is evaluated on CAIX and a CAIX-mimic. We show that SLC-149 is a better inhibitor than acetazolamide against CAIX. Binding of SLC-149 thermally stabilizes CAIX-mimic at lower concentrations compared to that of CAII. Structural examinations of SLC-149 bound to CAIX-mimic and CAII explain binding preferences. In cell culture, SLC-149 is a more effective inhibitor of CAIX activity in a triple-negative breast cancer cell line than previously studied sulfonamide inhibitors. SLC-149 is also a better inhibitor of activity in cells expressing CAIX versus CAXII. However, SLC-149 has little effect on cytotoxicity, and high concentrations are required to inhibit cell growth, migration, and invasion. These data support the hypothesis that CAIX activity, shown to be important in regulating extracellular pH, does not underlie its ability to control cell growth.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/therapeutic use , Carbonic Anhydrase II/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Hydrogen-Ion Concentration , Models, Molecular , Triple Negative Breast Neoplasms/drug therapy
9.
Eur J Med Chem ; 212: 113035, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33303238

ABSTRACT

Here we report the synthesis of a series of taurine substituted sulfonamide derivatives 1-29 having the ureido moiety installed at the tail section as selective inhibitors of the tumor associated human (h) Carbonic Anhydrase (CA; EC 4.2.1.1) IX and XII. The series was deeply investigated for their kinetic features which demonstrated a strong dependence on the ureido moiety. High resolution X-ray crystallographic investigation on selected ligand adducts complexed with hCA II and hCA IX-mimic revealed a strong correlation between the ureido moiety and the amino acid residues Q92 and Q67 in both the hCA II and hCA IX-mimic, contributing to highly stabilized ligand-protein complex.


Subject(s)
Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship
10.
IUCrJ ; 7(Pt 6): 985-994, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-33209313

ABSTRACT

Enzymes are catalysts of biological processes. Significant insight into their catalytic mechanisms has been obtained by relating site-directed mutagenesis studies to kinetic activity assays. However, revealing the detailed relationship between structural modifications and functional changes remains challenging owing to the lack of information on reaction intermediates and of a systematic way of connecting them to the measured kinetic parameters. Here, a systematic approach to investigate the effect of an active-site-residue mutation on a model enzyme, human carbonic anhydrase II (CA II), is described. Firstly, structural analysis is performed on the crystallographic intermediate states of native CA II and its V143I variant. The structural comparison shows that the binding affinities and configurations of the substrate (CO2) and product (HCO3 -) are altered in the V143I variant and the water network in the water-replenishment pathway is restructured, while the proton-transfer pathway remains mostly unaffected. This structural information is then used to estimate the modifications of the reaction rate constants and the corresponding free-energy profiles of CA II catalysis. Finally, the obtained results are used to reveal the effect of the V143I mutation on the measured kinetic parameters (k cat and k cat/K m) at the atomic level. It is believed that the systematic approach outlined in this study may be used as a template to unravel the structure-function relationships of many other biologically important enzymes.

11.
J Med Chem ; 63(21): 13064-13075, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33085484

ABSTRACT

This study provides a structure-activity relationship study of a series of lipophilic carbonic anhydrase (CA) inhibitors with an acetazolamide backbone. The inhibitors were tested against the tumor-expressed CA isozyme IX (CA IX), and the cytosolic CA I, CA II, and membrane-bound CA IV. The study identified several low nanomolar potent inhibitors against CA IX, with lipophilicities spanning two log units. Very potent pan-inhibitors with nanomolar potency against CA IX and sub-nanomolar potency against CA II and CA IV, and with potency against CA I one order of magnitude better than the parent acetazolamide 1 were also identified in this study, together with compounds that displayed selectivity against membrane-bound CA IV. A comprehensive X-ray crystallographic study (12 crystal structures), involving both CA II and a soluble CA IX mimetic (CA IX-mimic), revealed the structural basis of this particular inhibition profile and laid the foundation for further developments toward more potent and selective inhibitors for the tumor-expressed CA IX.


Subject(s)
Acetazolamide/chemistry , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/chemistry , Acetazolamide/metabolism , Binding Sites , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase Inhibitors/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Dynamics Simulation , Neoplasms/enzymology , Neoplasms/pathology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Structure-Activity Relationship
12.
Nat Commun ; 11(1): 4557, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32917908

ABSTRACT

Why metalloenzymes often show dramatic changes in their catalytic activity when subjected to chemically similar but non-native metal substitutions is a long-standing puzzle. Here, we report on the catalytic roles of metal ions in a model metalloenzyme system, human carbonic anhydrase II (CA II). Through a comparative study on the intermediate states of the zinc-bound native CA II and non-native metal-substituted CA IIs, we demonstrate that the characteristic metal ion coordination geometries (tetrahedral for Zn2+, tetrahedral to octahedral conversion for Co2+, octahedral for Ni2+, and trigonal bipyramidal for Cu2+) directly modulate the catalytic efficacy. In addition, we reveal that the metal ions have a long-range (~10 Å) electrostatic effect on restructuring water network in the active site. Our study provides evidence that the metal ions in metalloenzymes have a crucial impact on the catalytic mechanism beyond their primary chemical properties.


Subject(s)
Carbonic Anhydrases/chemistry , Ions/chemistry , Metalloproteins/chemistry , Metals/chemistry , Binding Sites , Carbonic Anhydrase II/chemistry , Carbonic Anhydrase II/metabolism , Carbonic Anhydrases/metabolism , Catalysis , Catalytic Domain , Cobalt/chemistry , Copper/chemistry , Crystallography, X-Ray , Humans , Ions/metabolism , Kinetics , Metalloproteins/metabolism , Metals/metabolism , Models, Molecular , Nickel/chemistry , Protein Conformation , Structure-Activity Relationship , Substrate Specificity , Zinc/chemistry
13.
J Med Chem ; 63(13): 7422-7444, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32519851

ABSTRACT

The "tail approach" has become a milestone in human carbonic anhydrase inhibitor (hCAI) design for various therapeutics, including antiglaucoma agents. Besides the classical hydrophobic/hydrophilic division of hCAs active site, several subpockets have been identified at the middle/outer active sites rim, which could be targeted to increase the CAI isoform selectivity. This postulate is explored here by three-tailed benzenesulfonamide CAIs (TTI) to fully exploit such amino acid differences among hCAs. In this proof-of-concept study, an extensive structure-activity relationship (SAR) study was carried out with 32 such benzenesulfonamides differing in tails combination that were assayed for hCAs I, II, IV, and XII inhibition. A structural study was undertaken by X-ray crystallography and in silico tools to assess the ligand/target interaction mode. The most active and selective inhibitors against isoforms implicated in glaucoma were assessed in a rabbit model of the disease achieving an intraocular pressure-lowering action comparable to the clinically used dorzolamide.


Subject(s)
Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Sulfonamides/chemistry , Animals , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Computer Simulation , Crystallography, X-Ray , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Glaucoma/drug therapy , Humans , Intraocular Pressure/drug effects , Ligands , Male , Proof of Concept Study , Rabbits , Structure-Activity Relationship , Sulfonamides/pharmacology
14.
Biomolecules ; 10(4)2020 03 31.
Article in English | MEDLINE | ID: mdl-32244293

ABSTRACT

Carbonic anhydrase II (CAII) is a metalloenzyme that catalyzes the reversible hydration/dehydration of CO2/HCO3-. In addition, CAII is attributed to other catalytic reactions, including esterase activity. Aspirin (acetyl-salicylic acid), an everyday over-the-counter drug, has both ester and carboxylic acid moieties. Recently, compounds with a carboxylic acid group have been shown to inhibit CAII. Hence, we hypothesized that Aspirin could act as a substrate for esterase activity, and the product salicylic acid (SA), an inhibitor of CAII. Here, we present the crystal structure of CAII in complex with SA, a product of CAII crystals pre-soaked with Aspirin, to 1.35Å resolution. In addition, we provide kinetic data to support the observation that CAII converts Aspirin to its deacetylated form, SA. This data may also explain the short half-life of Aspirin, with CAII so abundant in blood, and that Aspirin could act as a suicide inhibitor of CAII.


Subject(s)
Aspirin/pharmacology , Carbonic Anhydrase II/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Carbonic Anhydrase II/chemistry , Catalytic Domain , Kinetics , Models, Molecular
15.
J Biol Chem ; 295(19): 6498-6508, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32238431

ABSTRACT

Arrestin-1 is the arrestin family member responsible for inactivation of the G protein-coupled receptor rhodopsin in photoreceptors. Arrestin-1 is also well-known to interact with additional protein partners and to affect other signaling cascades beyond phototransduction. In this study, we investigated one of these alternative arrestin-1 binding partners, the glycolysis enzyme enolase-1, to map the molecular contact sites between these two proteins and investigate how the binding of arrestin-1 affects the catalytic activity of enolase-1. Using fluorescence quench protection of strategically placed fluorophores on the arrestin-1 surface, we observed that arrestin-1 primarily engages enolase-1 along a surface that is opposite of the side of arrestin-1 that binds photoactivated rhodopsin. Using this information, we developed a molecular model of the arrestin-1-enolase-1 complex, which was validated by targeted substitutions of charge-pair interactions. Finally, we identified the likely source of arrestin's modulation of enolase-1 catalysis, showing that selective substitution of two amino acids in arrestin-1 can completely remove its effect on enolase-1 activity while still remaining bound to enolase-1. These findings open up opportunities for examining the functional effects of arrestin-1 on enolase-1 activity in photoreceptors and their surrounding cells.


Subject(s)
Arrestin/chemistry , Biomarkers, Tumor/chemistry , DNA-Binding Proteins/chemistry , Models, Molecular , Multienzyme Complexes/chemistry , Phosphopyruvate Hydratase/chemistry , Rhodopsin/chemistry , Tumor Suppressor Proteins/chemistry , Arrestin/genetics , Binding Sites , Biomarkers, Tumor/genetics , Catalysis , DNA-Binding Proteins/genetics , Humans , Multienzyme Complexes/genetics , Phosphopyruvate Hydratase/genetics , Rhodopsin/genetics , Tumor Suppressor Proteins/genetics
16.
IUCrJ ; 7(Pt 2): 287-293, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-32148856

ABSTRACT

Nitric oxide (NO) promotes vasodilation through the activation of guanylate cyclase, resulting in the relaxation of the smooth muscle vasculature and a subsequent decrease in blood pressure. Therefore, its regulation is of interest for the treatment and prevention of heart disease. An example is pulmonary hypertension which is treated by targeting this NO/vasodilation pathway. In bacteria, plants and fungi, nitrite (NO2 -) is utilized as a source of NO through enzymes known as nitrite reductases. These enzymes reduce NO2 - to NO through a catalytic metal ion, often copper. Recently, several studies have shown nitrite reductase activity of mammalian carbonic anhydrase II (CAII), yet the molecular basis for this activity is unknown. Here we report the crystal structure of copper-bound human CAII (Cu-CAII) in complex with NO2 - at 1.2 Šresolution. The structure exhibits Type 1 (T-1) and 2 (T-2) copper centers, analogous to bacterial nitrite reductases, both required for catalysis. The copper-substituted CAII active site is penta-coordinated with a 'side-on' bound NO2 -, resembling a T-2 center. At the N terminus, several residues that are normally disordered form a porphyrin ring-like configuration surrounding a second copper, acting as a T-1 center. A structural comparison with both apo- (without metal) and zinc-bound CAII (Zn-CAII) provides a mechanistic picture of how, in the presence of copper, CAII, with minimal conformational changes, can function as a nitrite reductase.

17.
Methods Enzymol ; 634: 281-309, 2020.
Article in English | MEDLINE | ID: mdl-32093837

ABSTRACT

The carbonic anhydrases (CAs; EC 4.2.1.1) are a family of metalloenzymes that catalyze the reversible hydration of carbon dioxide (CO2) and bicarbonate (HCO3-). Since their discovery in 1933, CAs have been at the forefront of scientific discovery: the understanding of enzymatic reactions, structural biology, molecular dynamics, drug discovery, and clinical medicine. These ubiquitous enzymes equilibrate the reaction between CO2, HCO3-, and protons. Hence, CAs have important roles in ion transport, acid-base regulation, gas exchange, photosynthesis, and CO2 fixation. In this chapter, we describe the protocols leading to, and the analysis of CA neutron crystal structures. This accumulation of structural knowledge adds to our understanding of the enzymatic mechanism and development of CA inhibitors.


Subject(s)
Carbonic Anhydrases , Bicarbonates , Carbon Dioxide , Carbonic Anhydrase Inhibitors , Neutrons
18.
Oncogene ; 39(8): 1710-1723, 2020 02.
Article in English | MEDLINE | ID: mdl-31723238

ABSTRACT

Tumor cells rely on glycolysis to meet their elevated demand for energy. Thereby they produce significant amounts of lactate and protons, which are exported via monocarboxylate transporters (MCTs), supporting the formation of an acidic microenvironment. The present study demonstrates that carbonic anhydrase IX (CAIX), one of the major acid/base regulators in cancer cells, forms a protein complex with MCT1 and MCT4 in tissue samples from human breast cancer patients, but not healthy breast tissue. Formation of this transport metabolon requires binding of CAIX to the Ig1 domain of the MCT1/4 chaperon CD147 and is required for CAIX-mediated facilitation of MCT1/4 activity. Application of an antibody, directed against the CD147-Ig1 domain, displaces CAIX from the transporter and suppresses CAIX-mediated facilitation of proton-coupled lactate transport. In cancer cells, this "metabolon disruption" results in a decrease in lactate transport, reduced glycolysis, and ultimately reduced cell proliferation. Taken together, the study shows that carbonic anhydrases form transport metabolons with acid/base transporters in human tumor tissue and that these interactions can be exploited to interfere with tumor metabolism and proliferation.


Subject(s)
Breast Neoplasms/pathology , Carbonic Anhydrase IX/metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Symporters/metabolism , Basigin/chemistry , Basigin/metabolism , Humans , MCF-7 Cells , Models, Molecular , Protein Domains
19.
FEBS J ; 287(12): 2597-2611, 2020 06.
Article in English | MEDLINE | ID: mdl-31782893

ABSTRACT

Cell surface-localized P1 adhesin (aka Antigen I/II or PAc) of the cariogenic bacterium Streptococcus mutans mediates sucrose-independent adhesion to tooth surfaces. Previous studies showed that P1's C-terminal segment (C123, AgII) is also liberated as a separate polypeptide, contributes to cellular adhesion, interacts specifically with intact P1 on the cell surface, and forms amyloid fibrils. Identifying how C123 specifically interacts with P1 at the atomic level is essential for understanding related virulence properties of S. mutans. However, with sizes of ~ 51 and ~ 185 kDa, respectively, C123 and full-length P1 are too large to achieve high-resolution data for full structural analysis by NMR. Here, we report on biologically relevant interactions of the individual C3 domain with A3VP1, a polypeptide that represents the apical head of P1 as it is projected on the cell surface. Also evaluated are C3's interaction with C12 and the adhesion-inhibiting monoclonal antibody (MAb) 6-8C. NMR titration experiments with 15 N-enriched C3 demonstrate its specific binding to A3VP1. Based on resolved C3 assignments, two binding sites, proximal and distal, are identified. Complementary NMR titration of A3VP1 with a C3/C12 complex suggests that binding of A3VP1 occurs on the distal C3 binding site, while the proximal site is occupied by C12. The MAb 6-8C binding interface to C3 overlaps with that of A3VP1 at the distal site. Together, these results identify a specific C3-A3VP1 interaction that serves as a foundation for understanding the interaction of C123 with P1 on the bacterial surface and the related biological processes that stem from this interaction. DATABASE: BMRB submission code: 27935.


Subject(s)
Adhesins, Bacterial/chemistry , Nuclear Magnetic Resonance, Biomolecular , Streptococcus mutans/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Binding
20.
J Med Chem ; 62(4): 2202-2212, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30721041

ABSTRACT

3,17ß-Bis-sulfamoyloxy-2-methoxyestra-1,3,5(10)-triene (STX140), a bis-sulfamate derivative of the endogenous steroid 2-methoxyestradiol, has shown promising anticancer potency both in vitro and in vivo, with excellent bioavailability. Its activity against taxane-resistant xenografts makes it a potential drug candidate against triple-negative breast cancer (TNBC). These properties are linked to the ability of STX140 to act in a multitargeting fashion in vivo as a microtubule disruptor, leading to cell cycle arrest and with both proapoptotic and anti-angiogenic activities. Carbonic anhydrase IX (CA IX) is a well-established biomarker for aggressive cancers, including TNBC. This study reports, for the first time, the inhibitory activities of a series of steroidal and nonsteroidal sulfamate derivatives against CA IX in comparison to the ubiquitous CA II, with some compounds demonstrating 100-200-fold selectivity for CA IX over CA II. X-ray crystallographic studies of four of the most promising compounds reveal that isoform-specific residue interactions are responsible for the high specificity.


Subject(s)
Antigens, Neoplasm/chemistry , Antineoplastic Agents/chemistry , Carbonic Anhydrase IX/chemistry , Carbonic Anhydrase Inhibitors/chemistry , Estrenes/chemistry , Antigens, Neoplasm/metabolism , Antineoplastic Agents/metabolism , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/metabolism , Catalytic Domain , Crystallography, X-Ray , Estrenes/metabolism , Humans , Kinetics , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL