Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Endocr Soc ; 6(12): bvac158, 2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36381014

ABSTRACT

Due to the complexity of estrogen signaling mediated by estrogen receptors (ERs) in a variety of biological environments, there is great interest in the identification and optimization of selective estrogen receptor ligands. Prodrugs that can be activated in specific environments allow for tissue selectivity. Therefore, there have been recent advances in the development of prodrugs for ERs that can be released through enzymatic reactions, chemical reactions (eg, oxidation by reactive oxygen species or reduction by ascorbic acid), or light-mediated processes. In addition, researchers have linked ER ligands to additional drugs for selective cellular targeting. In this review, we highlight the compounds that have been generated and their potential uses in disease states such as breast cancer, inflammation, and menopause.

2.
J Inorg Biochem ; 235: 111930, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35841722

ABSTRACT

Photoactivated chemotherapy (PACT) has emerged as a promising strategy to selectively target cancer cells by using light irradiation to generate cytotoxic complexes in situ through a mechanism involving ligand-loss. Due to their rich optical properties and excited state chemistry, Ru polypyridyl complexes have attracted significant attention for PACT. However, studying PACT is complicated by the fact that many of these Ru complexes can also undergo excited-state electron transfer to generate 1O2 species. In order to deconvolute the biological roles of possible photo-decomposition products without the added complication of excited-state electron transfer chemistry, we have developed a methodology to systematically investigate each product individually, and assess the structure-function relationship. Here, we synthesized a series of eight distinct Ru polypyridyl complexes: Ru-Xa ([Ru(NN)3]2+), Ru-Xb ([Ru(NN)2py2]2+), and Ru-Xc ([Ru(NN)(OH2)2]2+) where NN = 2,2'-bipyridine, 4,4'-dimethyl-2,2'-bipyridine, or dimethyl 2,2'-bipyridine-4,4'-dicarboxylate and py = pyridine. The cytotoxicity of these complexes was investigated in two cell lines amenable to PACT: H23 (breast cancer) and T47D (lung cancer). We confirmed that light irradiation of Ru-Xa and Ru-Xb complexes generate Ru-Xc complexes through UV-visible spectroscopy, and observed that the Ru-Xc complexes are the most toxic against the cancer cell lines. In addition, we have shown that ligand release and biological activity including bovine serum albumin (BSA) binding, lipophilicity, and DNA interaction are altered when different groups are appended to the bipyridine ligands. We believe that the methodology presented here will enhance the development of more potent and selective PACT agents moving forward.


Subject(s)
Antineoplastic Agents , Ruthenium , 2,2'-Dipyridyl , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , DNA/chemistry , Ligands , Ruthenium/chemistry , Ruthenium/pharmacology
3.
ACS Med Chem Lett ; 13(3): 483-491, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35295086

ABSTRACT

Antibiotic-resistant infections are a pressing global concern, causing millions of deaths each year. Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of nosocomial infections in healthcare settings and is increasingly responsible for community-acquired infections that are often more difficult to treat. Antibiotic adjuvants are small molecules that potentiate antibiotics through nontoxic mechanisms and show excellent promise as novel therapeutics. Screening of low-molecular-weight compounds was employed to identify novel antibiotic adjuvant scaffolds for further elaboration. Brominated carbazoles emerged from this screening as lead compounds for further evaluation. Lead carbazoles were able to potentiate several ß-lactam antibiotics in three medically relevant strains of MRSA. Gene expression studies determined that these carbazoles were dampening the transcription of key genes that modulate ß-lactam resistance in MRSA. The lead brominated carbazoles represent novel scaffolds for elaboration as antibiotic adjuvants.

4.
Eur J Med Chem Rep ; 4: 100034, 2022 Apr.
Article in English | MEDLINE | ID: mdl-37519829

ABSTRACT

COVID-19 is a global pandemic caused by infection with the SARS-CoV-2 virus. Remdesivir, a SARS-CoV-2 RNA polymerase inhibitor, is the only drug to have received widespread approval for treatment of COVID-19. The SARS-CoV-2 main protease enzyme (MPro), essential for viral replication and transcription, remains an active target in the search for new treatments. In this study, the ability of novel thiazolyl-indazole derivatives to inhibit MPro is evaluated. These compounds were synthesized via the heterocyclization of phenacyl bromide with (R)-carvone, (R)-pulegone and (R)-menthone thiosemicarbazones. The binding affinity and binding interactions of each compound were evaluated through Schrödinger Glide docking, AMBER molecular dynamics simulations, and MM-GBSA free energy estimation, and these results were compared with similar calculations of MPro binding various 5-mer substrates (VKLQA, VKLQS, VKLQG) and a previously identified MPro tight-binder X77. From these simulations, we can see that binding is driven by residue specific interactions such as π-stacking with His41, and S/π interactions with Met49 and Met165. The compounds were also experimentally evaluated in a MPro biochemical assay and the most potent compound containing a phenylthiazole moiety inhibited protease activity with an IC50 of 92.9 â€‹µM. This suggests that the phenylthiazole scaffold is a promising candidate for the development of future MPro inhibitors.

5.
ChemMedChem ; 16(7): 1163-1171, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33332774

ABSTRACT

Phosphorylation-dependent protein-protein interactions play a significant role in biological signaling pathways; therefore, small molecules that are capable of influencing these interactions can be valuable research tools and have potential as pharmaceutical agents. MEMO1 (mediator of ErbB2-cell driven motility) is a phosphotyrosine-binding protein that interacts with a variety of protein partners and has been found to be upregulated in breast cancer patients. Herein, we report the first small-molecule inhibitors of MEMO1 interactions identified through a virtual screening platform and validated in a competitive fluorescence polarization assay. Initial structure-activity relationships have been investigated for these phenazine-core inhibitors and the binding sites have been postulated using molecular dynamics simulations. The most potent biochemical inhibitor is capable of disrupting the large protein interface with a KI of 2.7 µm. In addition, the most promising phenazine core compounds slow the migration of breast cancer cell lines in a scratch assay.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Phenazines/pharmacology , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Female , Fluorescence Polarization , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Dynamics Simulation , Molecular Structure , Phenazines/chemical synthesis , Phenazines/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
6.
Sensors (Basel) ; 19(11)2019 Jun 06.
Article in English | MEDLINE | ID: mdl-31174353

ABSTRACT

Biosensing strategies that employ readily adaptable materials for different analytes, can be miniaturized into needle electrode form, and function in bodily fluids represent a significant step toward the development of clinically relevant in vitro and in vivo sensors. In this work, a general scheme for 1st generation amperometric biosensors involving layer-by-layer electrode modification with enzyme-doped xerogels, electrochemically-deposited polymer, and polyurethane semi-permeable membranes is shown to achieve these goals. With minor modifications to these materials, sensors representing potential point-of-care medical tools are demonstrated to be sensitive and selective for a number of conditions. The potential for bedside measurements or continuous monitoring of analytes may offer faster and more accurate clinical diagnoses for diseases such as diabetes (glucose), preeclampsia (uric acid), galactosemia (galactose), xanthinuria (xanthine), and sepsis (lactate). For the specific diagnostic application, the sensing schemes have been miniaturized to wire electrodes and/or demonstrated as functional in synthetic urine or blood serum. Signal enhancement through the incorporation of platinum nanoparticle film in the scheme offers additional design control within the sensing scheme. The presented sensing strategy has the potential to be applied to any disease that has a related biomolecule and corresponding oxidase enzyme and represents rare, adaptable, sensing capabilities.

7.
Bioorg Med Chem ; 27(10): 2075-2082, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30967304

ABSTRACT

The development and evaluation of selective estrogen receptor modulators (SERMs) is of interest because of the complex and significant role of estrogen receptors in normal tissues as well as disease states. In neurodegenerative disorders such as Alzheimer's disease and multiple sclerosis, estrogen receptor beta (ERß) seems to provide a protective anti-inflammatory response. Due to the increase in reactive oxygen species (ROS) in these diseases, we have masked ERß ligands, including diarylpropionitrile (DPN), as boronate esters that release the active estrogen in the presence of H2O2. Here we demonstrate their synthesis, decreased binding affinities, kinetics of release, and selectivity toward ROS. The most promising ligand can be unmasked in the presence of pathological H2O2 to modulate ERß transcription in cells.


Subject(s)
Estrogen Receptor beta/metabolism , Hydrogen Peroxide/metabolism , Ligands , Boronic Acids/chemistry , Drug Design , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/genetics , Humans , Hydrogen Peroxide/pharmacology , Nitriles/chemistry , Nitriles/metabolism , Nitriles/pharmacology , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/pharmacology , Propionates/chemistry , Propionates/metabolism , Propionates/pharmacology , Protein Binding , Reactive Oxygen Species/metabolism , Transcriptional Activation/drug effects
8.
Bioelectrochemistry ; 125: 116-126, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30449323

ABSTRACT

Modified electrodes featuring specific adsorption platforms able to access the electrochemistry of the copper containing enzyme galactose oxidase (GaOx) were explored, including interfaces featuring nanomaterials such as nanoparticles and carbon nanotubes (CNTs). Electrodes modified with various self-assembled monolayers (SAMs) including those with attached nanoparticles or amide-coupled functionalized CNTs were examined for their ability to effectively immobilize GaOx and study the redox activity related to its copper core. While stable GaOx electrochemistry has been notoriously difficult to achieve at modified electrodes, strategically designed functionalized CNT-based interfaces, cysteamine SAM-modified electrode subsequently amide-coupled to carboxylic acid functionalized single wall CNTs, were significantly more effective with high GaOx surface adsorption along with well-defined, more reversible, stable (≥ 8 days) voltammetry and an average ET rate constant of 0.74 s-1 in spite of increased ET distance - a result attributed to effective electronic coupling at the GaOx active site. Both amperometric and fluorescence assay results suggest embedded GaOx remains active. Fundamental ET properties of GaOx may be relevant to biosensor development targeting galactosemia while the use functionalized CNT platforms for adsorption/electrochemistry of electroactive enzymes/proteins may present an approach for fundamental protein electrochemistry and their future use in both direct and indirect biosensor schemes.


Subject(s)
Enzymes, Immobilized/chemistry , Fungi/enzymology , Galactose Oxidase/chemistry , Nanotubes, Carbon/chemistry , Adsorption , Biosensing Techniques , Electrochemical Techniques , Electron Transport , Kinetics , Models, Molecular , Surface Properties
9.
ChemMedChem ; 13(20): 2208-2216, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30117269

ABSTRACT

The immune system uses members of the toll-like receptor (TLR) family to recognize a variety of pathogen- and host-derived molecules in order to initiate immune responses. Although TLR-mediated, pro-inflammatory immune responses are essential for host defense, prolonged and exaggerated activation can result in inflammation pathology that manifests in a variety of diseases. Therefore, small-molecule inhibitors of the TLR signaling pathway might have promise as anti-inflammatory drugs. We previously identified a class of triaryl pyrazole compounds that inhibit TLR signaling by modulation of the protein-protein interactions essential to the pathway. We have now systematically examined the structural features essential for inhibition of this pathway, revealing characteristics of compounds that inhibited all TLRs tested (pan-TLR signaling inhibitors) as well as compounds that selectively inhibited certain TLRs. These findings reveal interesting classes of compounds that could be optimized for particular inflammatory diseases governed by different TLRs.


Subject(s)
Pyrazoles/chemistry , Pyrazoles/pharmacology , Signal Transduction/drug effects , Toll-Like Receptors/metabolism , Animals , Humans , Mice , Molecular Structure , RAW 264.7 Cells , Receptors, Estrogen/metabolism , Structure-Activity Relationship
10.
Sci Signal ; 11(543)2018 08 14.
Article in English | MEDLINE | ID: mdl-30108181

ABSTRACT

Toll-like receptors (TLRs) recognize various pathogen- and host tissue-derived molecules and initiate inflammatory immune responses. Exaggerated or prolonged TLR activation, however, can lead to etiologically diverse diseases, such as bacterial sepsis, metabolic and autoimmune diseases, or stroke. Despite the apparent medical need, no small-molecule drugs against TLR pathways are clinically available. This may be because of the complex signaling mechanisms of TLRs, which are governed by a series of protein-protein interactions initiated by Toll/interleukin-1 receptor homology domains (TIR) found in TLRs and the cytoplasmic adaptor proteins TIRAP and MyD88. Oligomerization of TLRs with MyD88 or TIRAP leads to the recruitment of members of the IRAK family of kinases and the E3 ubiquitin ligase TRAF6. We developed a phenotypic drug screening system based on the inducible homodimerization of either TIRAP, MyD88, or TRAF6, that ranked hits according to their hierarchy of action. From a bioactive compound library, we identified methyl-piperidino-pyrazole (MPP) as a TLR-specific inhibitor. Structure-activity relationship analysis, quantitative proteomics, protein-protein interaction assays, and cellular thermal shift assays suggested that MPP targets the TIR domain of MyD88. Chemical evolution of the original MPP scaffold generated compounds with selectivity for distinct TLRs that interfered with specific TIR interactions. Administration of an MPP analog to mice protected them from TLR4-dependent inflammation. These results validate this phenotypic screening approach and suggest that the MPP scaffold could serve as a starting point for the development of anti-inflammatory drugs.


Subject(s)
Piperidines/pharmacology , Pyrazoles/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Toll-Like Receptors/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , HEK293 Cells , Humans , Inhibitory Concentration 50 , Mice , Protein Binding/drug effects , Protein Interaction Maps/drug effects , RAW 264.7 Cells , Toll-Like Receptors/metabolism
11.
Biochemistry ; 57(34): 5169-5181, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30067338

ABSTRACT

ErbB2 signaling pathways are linked to breast cancer formation, growth, and aggression; therefore, understanding the behavior of proteins associated with these pathways as well as regulatory factors that influence ErbB2 function is essential. MEMO1 is a redox active protein that is shown to associate with phosphorylated ErbB2 and mediate cell motility. We have developed a fluorescence polarization assay to probe the interaction between MEMO1 and an ErbB2-derived peptide containing a phosphorylated tyrosine residue. This interaction is shown to be pH-dependent and stronger with longer peptides as would be expected for protein-protein interactions. We have quantitatively mapped the binding interface of MEMO1 to the peptide using the fluorescence polarization assay and molecular dynamics simulations. We have confirmed that phosphorylation of the peptide is essential for binding and through mutagenesis have identified residues that contribute to favorable interactions. Our results highlight the importance of the protein-protein interactions of MEMO1 that complement the oxidase activity. In the future, these studies will provide a method for screening for selective modulators of MEMO1, which will allow for additional biological investigations.


Subject(s)
Fluorescence Polarization , Molecular Dynamics Simulation , Nonheme Iron Proteins/metabolism , Receptor, ErbB-2/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mutagenesis, Site-Directed , Nonheme Iron Proteins/chemistry , Nonheme Iron Proteins/genetics , Phosphorylation , Protein Binding , Protein Conformation , Protein Stability , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics
12.
Nanomaterials (Basel) ; 9(1)2018 Dec 29.
Article in English | MEDLINE | ID: mdl-30597967

ABSTRACT

A first-generation amperometric galactose biosensor has been systematically developed utilizing layer-by-layer (LbL) construction of xerogels, polymers, and carbon nanotubes toward a greater fundamental understanding of sensor design with these materials and the potential development of a more efficient galactosemia diagnostic tool for clinical application. The effect of several parameters (xerogel silane precursor, buffer pH, enzyme concentration, drying time and the inclusion of a polyurethane (PU) outer layer) on galactose sensitivity were investigated with the critical nature of xerogel selection being demonstrated. Xerogels formed from silanes with medium, aliphatic side chains were shown to exhibit significant enhancements in sensitivity with the addition of PU due to decreased enzyme leaching. Semi-permeable membranes of diaminobenzene and resorcinol copolymer and Nafion were used for selective discrimination against interferent species and the accompanying loss of sensitivity with adding layers was countered using functionalized, single-walled carbon nanotubes (CNTs). Optimized sensor performance included effective galactose sensitivity (0.037 µA/mM) across a useful diagnostic concentration range (0.5 mM to 7 mM), fast response time (~30 s), and low limits of detection (~80 µM) comparable to literature reports on galactose sensors. Additional modification with anionic polymer layers and/or nanoparticles allowed for galactose detection in blood serum samples and additional selectivity effectiveness.

13.
Horm Cancer ; 8(2): 69-77, 2017 04.
Article in English | MEDLINE | ID: mdl-28194662

ABSTRACT

Triple-negative breast cancer (TNBC) has a faster rate of metastasis compared to other breast cancer subtypes, and no effective targeted therapies are currently FDA-approved. Recent data indicate that the androgen receptor (AR) promotes tumor survival and may serve as a potential therapeutic target in TNBC. Studies of AR in disease progression and the systemic effects of anti-androgens have been hindered by the lack of an AR-positive (AR+) immunocompetent preclinical model. In this study, we identified the transgenic MMTV-PyMT (mouse mammary tumor virus-polyoma middle tumor-antigen) mouse mammary gland carcinoma model of breast cancer and Met-1 cells derived from this model as tools to study the role of AR in breast cancer progression. AR protein expression was examined in late-stage primary tumors and lung metastases from MMTV-PyMT mice as well as in Met-1 cells by immunohistochemistry (IHC). Sensitivity of Met-1 cells to the AR agonist dihydrotestosterone (DHT) and anti-androgen therapy was examined using cell viability, migration/invasion, and anchorage-independent growth assays. Late-stage primary tumors and lung metastases from MMTV-PyMT mice and Met-1 cells expressed abundant nuclear AR protein, while negative for estrogen and progesterone receptors. Met-1 sensitivity to DHT and AR antagonists demonstrated a reliance on AR for survival, and AR antagonists inhibited invasion and anchorage-independent growth. These data suggest that the MMTV-PyMT model and Met-1 cells may serve as valuable tools for mechanistic studies of the role of AR in disease progression and how anti-androgens affect the tumor microenvironment.


Subject(s)
Lung Neoplasms/pathology , Mammary Neoplasms, Experimental/metabolism , Receptors, Androgen/metabolism , Triple Negative Breast Neoplasms/metabolism , Androgen Antagonists/administration & dosage , Androgen Antagonists/pharmacology , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Dihydrotestosterone/administration & dosage , Dihydrotestosterone/pharmacology , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/secondary , Mammary Tumor Virus, Mouse/physiology , Mice , Mice, Transgenic
14.
Nat Chem Biol ; 12(10): 795-801, 2016 10.
Article in English | MEDLINE | ID: mdl-27501397

ABSTRACT

Clinical resistance to the second-generation antiandrogen enzalutamide in castration-resistant prostate cancer (CRPC), despite persistent androgen receptor (AR) activity in tumors, highlights an unmet medical need for next-generation antagonists. We have identified and characterized tetra-aryl cyclobutanes (CBs) as a new class of competitive AR antagonists that exhibit a unique mechanism of action. These CBs are structurally distinct from current antiandrogens (hydroxyflutamide, bicalutamide, and enzalutamide) and inhibit AR-mediated gene expression, cell proliferation, and tumor growth in several models of CRPC. Conformational profiling revealed that CBs stabilize an AR conformation resembling an unliganded receptor. Using a variety of techniques, it was determined that the AR-CB complex was not recruited to AR-regulated promoters and, like apo AR, remains sequestered in the cytoplasm, bound to heat shock proteins. Thus, we have identified third-generation AR antagonists whose unique mechanism of action suggests that they may have therapeutic potential in CRPC.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology , Structure-Activity Relationship
15.
Tetrahedron Lett ; 56(44): 6097-6099, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26516292

ABSTRACT

The development of a reagent for the efficient synthesis of 5- and 6-membered azoles at room temperature is proposed. A variety of substituted 2-aminobenzimidazoles are synthesized in good to excellent yields. The ability to incorporate various protecting groups makes the imidoyl dichloride reagent amenable to a large number of syntheses. The reagent is applied to the total synthesis of the 2-aminobenzimidazole containing carcinogen, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), from 2-chloro-3-nitropyridine in >60 % yield in 6 steps.

16.
Elife ; 3: e02057, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24771768

ABSTRACT

Resveratrol has beneficial effects on aging, inflammation and metabolism, which are thought to result from activation of the lysine deacetylase, sirtuin 1 (SIRT1), the cAMP pathway, or AMP-activated protein kinase. In this study, we report that resveratrol acts as a pathway-selective estrogen receptor-α (ERα) ligand to modulate the inflammatory response but not cell proliferation. A crystal structure of the ERα ligand-binding domain (LBD) as a complex with resveratrol revealed a unique perturbation of the coactivator-binding surface, consistent with an altered coregulator recruitment profile. Gene expression analyses revealed significant overlap of TNFα genes modulated by resveratrol and estradiol. Furthermore, the ability of resveratrol to suppress interleukin-6 transcription was shown to require ERα and several ERα coregulators, suggesting that ERα functions as a primary conduit for resveratrol activity.DOI: http://dx.doi.org/10.7554/eLife.02057.001.


Subject(s)
Estrogen Receptor alpha/metabolism , Inflammation/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Adenylate Kinase/metabolism , Cyclic AMP/metabolism , Estrogen Receptor alpha/chemistry , Female , Humans , Interleukin-6/genetics , Ligands , MCF-7 Cells , Promoter Regions, Genetic , Protein Conformation , Resveratrol , Tumor Necrosis Factor-alpha/metabolism
17.
Bioconjug Chem ; 24(3): 408-18, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23350694

ABSTRACT

Recombinant human thyroid stimulating hormone (rhTSH or Thyrogen) has been approved for thyroid cancer diagnostics and treatment under a multidose regimen due to its short circulating half-life. To reduce dosing frequency, PEGylation strategies were explored to increase the duration of action of rhTSH. Lysine and N-terminal PEGylation resulted in heterogeneous product profiles with 40% or lower reaction yields of monoPEGylated products. Eleven cysteine mutants were designed based on a structure model of the TSH-TSH receptor (TSHR) complex to create unique conjugation sites on both α and ß subunits for site-specific conjugation. Sequential screening of mutant expression level, oligomerization tendency, and conjugation efficiency resulted in the identification of the αG22C rhTSH mutant for stable expression and scale-up PEGylation. The introduced cysteine in the αG22C rhTSH mutant was partially blocked when isolated from conditioned media and could only be effectively PEGylated after mild reduction with cysteine. This produced a higher reaction yield, ~85%, for the monoPEGylated product. Although the mutation had no effect on receptor binding, PEGylation of αG22C rhTSH led to a PEG size-dependent decrease in receptor binding. Nevertheless, the 40 kDa PEG αG22C rhTSH showed a prolonged duration of action compared to rhTSH in a rat pharmacodynamics model. Reverse-phase HPLC and N-terminal sequencing experiments confirmed site-specific modification at the engineered Cys 22 position on the α-subunit. This work is another demonstration of successful PEGylation of a cysteine-knot protein by an engineered cysteine mutation.


Subject(s)
Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Thyrotropin/administration & dosage , Thyrotropin/chemistry , Amino Acid Sequence , Animals , Binding Sites/drug effects , Binding Sites/physiology , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Female , Humans , Male , Molecular Sequence Data , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Thyrotropin/genetics , Time Factors
18.
ACS Chem Biol ; 7(7): 1221-31, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22533360

ABSTRACT

Lysine specific demethylase 1 (LSD1, also known as KDM1) is a histone modifying enzyme that regulates the expression of many genes important in cancer progression and proliferation. It is present in various transcriptional complexes including those containing the estrogen receptor (ER). Indeed, inhibition of LSD1 activity and or expression has been shown to attenuate estrogen signaling in breast cancer cells in vitro, implicating this protein in the pathogenesis of cancer. Herein we describe experiments that utilize small molecule inhibitors, phenylcyclopropylamines, along with small interfering RNA to probe the role of LSD1 in breast cancer proliferation and in estrogen-dependent gene transcription. Surprisingly, whereas we have confirmed that inhibition of LSD1 strongly inhibits proliferation of breast cancer cells, we have determined that the cytostatic actions of LSD1 inhibition are not impacted by ER status. These data suggest that LSD1 may be a useful therapeutic target in several types of breast cancer; most notably, inhibitors of LSD1 may have utility in the treatment of ER-negative cancers for which there are minimal therapeutic options.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Estrogen Receptor alpha/physiology , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Pargyline/pharmacology , Pargyline/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...