Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Chem Theory Comput ; 20(14): 6278-6286, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38975986

RESUMEN

Small modifications in the chemical structure of ligands are known to dramatically change their ability to inhibit the activity of a protein. Unraveling the mechanisms that govern these dramatic changes requires scrutinizing the dynamics of protein-ligand binding and unbinding at the atomic level. As an exemplary case, we have studied Glycogen Synthase Kinase-3ß (GSK-3ß), a multifunctional kinase that has been implicated in a host of pathological processes. As such, there is a keen interest in identifying ligands that inhibit GSK-3ß activity. One family of compounds that are highly selective and potent inhibitors of GSK-3ß is exemplified by a molecule termed COB-187. COB-187 consists of a five-member heterocyclic ring with a thione at C2, a pyridine substituted methyl at N3, and a hydroxyl and phenyl at C4. We have studied the inhibition of GSK-3ß by COB-187-related ligands that differ in a single heavy atom from each other (either in the location of nitrogen in their pyridine ring, or with the pyridine ring replaced by a phenyl ring), or in the length of the alkyl group joining the pyridine and the N3. The inhibition experiments show a large range of half-maximal inhibitory concentration (IC50) values from 10 nM to 10 µM, implying that these ligands exhibit vastly different propensities to inhibit GSK-3ß. To explain these differences, we perform Markov State Modeling (MSM) using fully atomistic simulations. Our MSM results are in excellent agreement with the experiments in that they accurately capture differences in the binding propensities of the ligands. The simulations show that the binding propensities are related to the ligands' ability to attain a compact conformation where their two aromatic rings are spatially close. We rationalize this result by sampling numerous binding and unbinding events via funnel metadynamics simulations, which show that indeed while approaching the bound state, the ligands prefer to be in their compact conformation. We find that the presence of nitrogen in the aromatic ring increases the probability of attaining the compact conformation. Protein-ligand binding is understood to be dictated by the energetics of interactions and entropic factors, like the release of bound water from the binding pockets. This work shows that changes in the conformational distribution of ligands due to atom-level modifications in the structure play an important role in protein-ligand binding.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Humanos , Cadenas de Markov , Ligandos , Piridinas/química , Piridinas/farmacología , Termodinámica
2.
Sci Rep ; 14(1): 11179, 2024 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-38750069

RESUMEN

During a SARS-CoV-2 infection, macrophages recognize viral components resulting in cytokine production. While this response fuels virus elimination, overexpression of cytokines can lead to severe COVID-19. Previous studies suggest that the spike protein (S) of SARS-CoV-2 can elicit cytokine production via the transcription factor NF-κB and the toll-like receptors (TLRs). In this study, we found that: (i) S and the S2 subunit induce CXCL10, a chemokine implicated in severe COVID-19, gene expression by human macrophage cells (THP-1); (ii) a glycogen synthase kinase-3 inhibitor attenuates this induction; (iii) S and S2 do not activate NF-κB but do activate the transcription factor IRF; (iv) S and S2 do not require TLR2 to elicit CXCL10 production or activate IRF; and (v) S and S2 elicit CXCL10 production by peripheral blood mononuclear cells (PBMCs). We also discovered that the cellular response, or lack thereof, to S and S2 is a function of the recombinant S and S2 used. While such a finding raises the possibility of confounding LPS contamination, we offer evidence that potential contaminating LPS does not underly induced increases in CXCL10. Combined, these results provide insights into the complex immune response to SARS-CoV-2 and suggest possible therapeutic targets for severe COVID-19.


Asunto(s)
COVID-19 , Quimiocina CXCL10 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , Quimiocina CXCL10/metabolismo , COVID-19/virología , COVID-19/inmunología , COVID-19/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/virología , FN-kappa B/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/inmunología , Células THP-1
3.
J Inflamm Res ; 16: 5339-5366, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38026235

RESUMEN

Purpose: Non-alcoholic fatty liver disease (NAFLD), recently renamed metabolic (dysfunction) associated fatty liver disease (MAFLD), is the most common chronic liver disease in the United States. Presently, there is an intense and ongoing effort to identify and develop novel therapeutics for this disease. In this study, we explored the anti-inflammatory activity of a new compound, termed IOI-214, and its therapeutic potential to ameliorate NAFLD/MAFLD in male C57BL/6J mice fed a high fat (HF) diet. Methods: Murine macrophages and hepatocytes in culture were treated with lipopolysaccharide (LPS) ± IOI-214 or DMSO (vehicle), and RT-qPCR analyses of inflammatory cytokine gene expression were used to assess IOI-214's anti-inflammatory properties in vitro. Male C57BL/6J mice were also placed on a HF diet and treated once daily with IOI-214 or DMSO for 16 weeks. Tissues were collected and analyzed to determine the effects of IOI-214 on HF diet-induced NAFL D/MAFLD. Measurements such as weight, blood glucose, serum cholesterol, liver/serum triglyceride, insulin, and glucose tolerance tests, ELISAs, metabolomics, Western blots, histology, gut microbiome, and serum LPS binding protein analyses were conducted. Results: IOI-214 inhibited LPS-induced inflammation in macrophages and hepatocytes in culture and abrogated HF diet-induced mesenteric fat accumulation, hepatic inflammation and steatosis/hepatocellular ballooning, as well as fasting hyperglycemia without affecting insulin resistance or fasting insulin, cholesterol or TG levels despite overall obesity in vivo in male C57BL/6J mice. IOI-214 also decreased systemic inflammation in vivo and improved gut microbiota dysbiosis and leaky gut. Conclusion: Combined, these data indicate that IOI-214 works at multiple levels in parallel to inhibit the inflammation that drives HF diet-induced NAFLD/MAFLD, suggesting that it may have therapeutic potential for NAFLD/MAFLD.

4.
Biochem Biophys Res Commun ; 605: 171-176, 2022 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-35367865

RESUMEN

A key component of severe COVID-19 is a "cytokine storm" i.e., the excessive expression of unneeded cytokines. Previous studies suggest that SARS-CoV-2 proteins can induce macrophages to secrete pro-inflammatory cytokines; a process that may involve Toll-like receptors (TLRs). Glycogen synthase kinase-3 (GSK-3) has been implicated in TLR signal transduction and a selective GSK-3 inhibitor, termed COB-187, dramatically attenuates cytokine expression induced by the TLR ligand lipopolysaccharide (LPS). In the present study, we provide evidence that the SARS-CoV-2 spike protein (S) and the S2 subunit (S2) induce production of CXCL10 (a chemokine elevated in severe COVID-19) by a human macrophage cell line. Further, we report that two clinically relevant GSK-3 inhibitors and COB-187 attenuate S and S2 protein-induced CXCL10 production. Combined, our observations provide impetus for investigating GSK-3 inhibitors as potential therapeutics for severe COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Glucógeno Sintasa Quinasa 3 , Citocinas/metabolismo , Humanos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus
5.
Bioorg Med Chem ; 40: 116179, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33991821

RESUMEN

Glycogen synthase kinase-3 (GSK-3) has been implicated in numerous pathologies making GSK-3 an attractive therapeutic target. Our group has identified a compound termed COB-187 that is a potent and selective inhibitor of GSK-3. In this study, we probed the mechanism by which COB-187 inhibits GSK-3ß. Progress curves, generated via real-time monitoring of kinase activity, indicated that COB-187 inhibition of GSK-3ß is time-dependent and subsequent jump dilution assays revealed that COB-187 binding to GSK-3ß is reversible. Further, a plot of the kinetic constant (kobs) versus COB-187 concentration suggested that, within the range of concentrations studied, COB-187 binds to GSK-3ß via an induced-fit mechanism. There is a critical cysteine residue at the entry to the active site of GSK-3ß (Cys-199). We generated a mutant version of GSK-3ß wherein Cys-199 was substituted with an alanine. This mutation caused a dramatic decrease in the activity of COB-187; specifically, an IC50 in the nM range for wild type versus >100 µM for the mutant. A screen of COB-187 against 34 kinases that contain a conserved cysteine in their active site revealed that COB-187 is highly selective for GSK-3 indicating that COB-187's inhibition of GSK-3ß via Cys-199 is specific. Combined, these findings suggest that COB-187 inhibits GSK-3ß via a specific, reversible, time and Cys-199-dependent mechanism.


Asunto(s)
Cistina/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Sitios de Unión/efectos de los fármacos , Cistina/metabolismo , Relación Dosis-Respuesta a Droga , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Factores de Tiempo
6.
Leuk Res ; 99: 106464, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33130330

RESUMEN

The role of interferon-gamma (IFN-γ) in Chronic Myelogenous/Myeloid Leukemia (CML) and in the treatment of CML remains unclear; specifically, the effect of IFN-γ on apoptosis. There is reported interplay between IFN-γ and glycogen synthase kinase-3 (GSK-3), a kinase which has been implicated in both cell death and, conversely, cell survival. Thus, we utilized the CML-derived HAP1 cell line and a mutant HAP1 GSK-3ß knocked-down cell line (GSK-3ß 31bp) to investigate whether GSK-3 modulates IFN-γ's action on CML cells. Significantly less GSK-3ß 31bp cells, relative to HAP1 cells, were present after 48 h treatment with IFN-γ. IFN-γ treatment significantly decreased GSK-3ß 31bp substrate adhesiveness (relative to HAP1 cells); an observation often correlated with cell death. Fluorescence microscopy revealed that IFN-γ induces a modest level of apoptosis in the HAP1 cells and that IFN-γ induced apoptosis is significantly enhanced in GSK-3ß 31bp cells. Utilizing a complementary GSK-3ß knocked-down cell line (8bp) we found, via flow cytometric analysis, that IFN-γ induced apoptosis is significantly enhanced in GSK-3ß 8bp cells relative to HAP1 cells. Combined, our findings suggest that IFN-γ induces apoptosis of CML cells and that loss of GSK-3ß significantly augments IFN-γ-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Interferón gamma/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/farmacología , Sistemas CRISPR-Cas , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Codón sin Sentido , Interacciones Farmacológicas , Citometría de Flujo , Mutación del Sistema de Lectura , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Interferón gamma/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/farmacología , Espectrometría de Fluorescencia
7.
Eur J Pharmacol ; 883: 173340, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32634441

RESUMEN

Sepsis is a serious condition that can lead to long-term organ damage and death. At the molecular level, the hallmark of sepsis is the elevated expression of a multitude of potent cytokines, i.e. a cytokine storm. For sepsis involving gram-negative bacteria, macrophages recognize lipopolysaccharide (LPS) shed from the bacteria, activating Toll-like-receptor 4 (TLR4), and triggering a cytokine storm. Glycogen synthase kinase-3 (GSK-3) is a highly active kinase that has been implicated in LPS-induced cytokine production. Thus, compounds that inhibit GSK-3 could be potential therapeutics for sepsis. Our group has recently described a novel and highly selective inhibitor of GSK-3 termed COB-187. In the present study, using THP-1 macrophages, we evaluated the ability of COB-187 to attenuate LPS-induced cytokine production. We found that COB-187 significantly reduced, at the protein and mRNA levels, cytokines induced by LPS (e.g. IL-6, TNF-α, IL-1ß, CXCL10, and IFN-ß). Further, the data suggest that the inhibition could be due, at least in part, to COB-187 reducing NF-κB (p65/p50) DNA binding activity as well as reducing IRF-3 phosphorylation at Serine 396. Thus, COB-187 appears to be a potent inhibitor of the cytokine storm induced by LPS.


Asunto(s)
Antiinflamatorios/farmacología , Síndrome de Liberación de Citoquinas/prevención & control , Citocinas/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Síndrome de Liberación de Citoquinas/inducido químicamente , Síndrome de Liberación de Citoquinas/enzimología , Síndrome de Liberación de Citoquinas/genética , Citocinas/genética , Regulación hacia Abajo , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Factor 3 Regulador del Interferón/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos/enzimología , FN-kappa B/metabolismo , Fosforilación , Sepsis/inducido químicamente , Sepsis/enzimología , Sepsis/genética , Sepsis/prevención & control , Transducción de Señal , Células THP-1
8.
Cell Mol Bioeng ; 13(2): 113-124, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32175025

RESUMEN

Cancers of the digestive tract cause nearly one quarter of the cancer deaths worldwide, and nearly half of these are due to cancers of the esophagus and colon. Early detection of cancer significantly increases the rate of survival, and thus it is critical that cancer within these organs is detected early. In this regard, endoscopy is routinely used to screen for transforming/cancerous (i.e. dysplastic to fully cancerous) tissue. Numerous studies have revealed that the biochemistry of the luminal surface of such tissue within the colon and esophagus becomes altered throughout disease progression. Molecular endoscopic imaging (MEI), an emerging technology, seeks to exploit these changes for the early detection of cancer. The general approach for MEI is as follows: the luminal surface of an organ is exposed to molecular ligands, or particulate probes bearing a ligand, cognate to biochemistry unique to pre-cancerous/cancerous tissue. After a wash, the tissue is imaged to determine the presence of the probes. Detection of the probes post-washing suggests pathologic tissue. In the current review we provide a succinct, but extensive, review of ligands and target moieties that could be, or are currently being investigated, as possible cognate chemistries for MEI. This is followed by a review of the biophysics that determines, in large part, the success of a particular MEI design. The work draws an analogy between MEI and the well-advanced field of cell adhesion and provides a road map for engineering MEI to achieve assays that yield highly selective recognition of transforming/cancerous tissue in situ.

9.
Am J Physiol Cell Physiol ; 317(6): C1289-C1303, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31553649

RESUMEN

Glycogen synthase kinase-3 (GSK-3) is a multitasking protein kinase that regulates numerous critical cellular functions. Not surprisingly, elevated GSK-3 activity has been implicated in a host of diseases including pathological inflammation, diabetes, cancer, arthritis, asthma, bipolar disorder, and Alzheimer's. Therefore, reagents that inhibit GSK-3 activity provide a means to investigate the role of GSK-3 in cellular physiology and pathophysiology and could become valuable therapeutics. Finding a potent inhibitor of GSK-3 that can selectively target this kinase, among over 500 protein kinases in the human genome, is a significant challenge. Thus there remains a critical need for the identification of selective inhibitors of GSK-3. In this work, we introduce a novel small organic compound, namely COB-187, which exhibits potent and highly selective inhibition of GSK-3. Specifically, this study 1) utilized a molecular screen of 414 kinase assays, representing 404 unique kinases, to reveal that COB-187 is a highly potent and selective inhibitor of GSK-3; 2) utilized a cellular assay to reveal that COB-187 decreases the phosphorylation of canonical GSK-3 substrates indicating that COB-187 inhibits cellular GSK-3 activity; and 3) reveals that a close isomer of COB-187 is also a selective and potent inhibitor of GSK-3. Taken together, these results demonstrate that we have discovered a region of chemical design space that contains novel GSK-3 inhibitors. These inhibitors will help to elucidate the intricate function of GSK-3 and can serve as a starting point for the development of potential therapeutics for diseases that involve aberrant GSK-3 activity.


Asunto(s)
Compuestos de Bifenilo/farmacología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Animales , Compuestos de Bifenilo/síntesis química , Diseño de Fármacos , Pruebas de Enzimas , Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Fosforilación , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/genética , Células RAW 264.7 , Relación Estructura-Actividad , Especificidad por Sustrato , Células THP-1 , Acetato de Tetradecanoilforbol/farmacología , Tiadiazoles/química , Tiadiazoles/farmacología
10.
Sci Rep ; 9(1): 8511, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186472

RESUMEN

Cell adhesion mediated by selectins (expressed by activated endothelium, activated platelets, and leukocytes) binding to their resepective selectin ligands (expressed by cancer cells) may be involved in metastasis. Therefore, methods of characterizing selectin ligands expressed on human tissue may serve as valuable assays. Presented herein is an innovative method for detecting functional selectin ligands expressed on human tissue that uses a dynamic approach, which allows for control over the force applied to the bonds between the probe and target molecules. This new method of tissue interrogation, known as dynamic biochemical tissue analysis (DBTA), involves the perfusion of molecular probe-coated microspheres over tissues. DBTA using selectin-coated probes is able to detect functional selectin ligands expressed on tissue from multiple cancer types at both primary and metastatic sites.


Asunto(s)
Bioquímica/métodos , Neoplasias/metabolismo , Especificidad de Órganos , Selectinas/metabolismo , Animales , Adhesión Celular , Línea Celular Tumoral , Epítopos/metabolismo , Humanos , Ligandos , Ratones , Metástasis de la Neoplasia
11.
Integr Biol (Camb) ; 10(12): 747-757, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30398503

RESUMEN

Esophageal cancer has a 5 year survival rate of ∼20%. This dismal prognosis is due, in part, to the fact that esophageal cancer often presents at a late stage. Thus, there is a critical need for assays that enable the early detection of cancerous tissue within the esophagus. The luminal surface of the esophagus expresses signature molecule(s) at sites of transformation providing an avenue for the development of in situ assays that detect neoplastic growth within the esophagus. An attractive approach, receiving increased attention, is the endoscopic administration of particles conjugated with ligands to signature molecules present on transforming tissue. Detection of the particles within the esophagus, post-washing, would indicate the presence of the signature molecule and thus transforming tissue. In this work, we utilized cancerous and normal esophageal cells to provide in vitro proof of principle for this approach utilizing ligand-conjugated microspheres and demonstrate the need, and provide the framework for, engineering this technology. Specifically, the study (i) reveals selective increased expression of signature molecules on cancerous esophageal cells relative to normal cells; (ii) demonstrates selective binding of ligand-conjugated microspheres to cancerous esophageal cells relative to normal cells; (iii) demonstrates that the selective recognition of cancerous, relative to normal esophageal cells, is highly dependent on the biophysical design of the assay; and (iv) advocates utilizing the knowledge from the field of cell adhesion as a guide for the effective development of ligand-conjugated particle-based schemes that seek to detect esophageal oncogenesis in situ.


Asunto(s)
Adhesión Celular , Neoplasias Esofágicas/diagnóstico , Esófago/patología , Adenocarcinoma/diagnóstico , Línea Celular Tumoral , Transformación Celular Neoplásica , Selectina E/química , Endoscopía , Neoplasias Esofágicas/mortalidad , Citometría de Flujo , Fucosa/química , Humanos , Ligandos , Microesferas , Tamaño de la Partícula , Polisacáridos/química , Estrés Mecánico
12.
J Endocrinol ; 237(3): 337-351, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29666152

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of both metabolic and inflammatory diseases and has become the leading chronic liver disease worldwide. High-fat (HF) diets promote an increased uptake and storage of free fatty acids (FFAs) and triglycerides (TGs) in hepatocytes, which initiates steatosis and induces lipotoxicity, inflammation and insulin resistance. Activation and signaling of Toll-like receptor 4 (TLR4) by FFAs induces inflammation evident in NAFLD and insulin resistance. Currently, there are no effective treatments to specifically target inflammation associated with this disease. We have established the efficacy of phenylmethimazole (C10) to prevent lipopolysaccharide and palmitate-induced TLR4 signaling. Because TLR4 is a key mediator in pro-inflammatory responses, it is a potential therapeutic target for NAFLD. Here, we show that treatment with C10 inhibits HF diet-induced inflammation in both liver and mesenteric adipose tissue measured by a decrease in mRNA levels of pro-inflammatory cytokines. Additionally, C10 treatment improves glucose tolerance and hepatic steatosis despite the development of obesity due to HF diet feeding. Administration of C10 after 16 weeks of HF diet feeding reversed glucose intolerance, hepatic inflammation, and improved hepatic steatosis. Thus, our findings establish C10 as a potential therapeutic for the treatment of NAFLD.


Asunto(s)
Citoprotección/efectos de los fármacos , Dieta/efectos adversos , Intolerancia a la Glucosa/prevención & control , Hepatocitos/efectos de los fármacos , Inflamación/etiología , Inflamación/prevención & control , Hígado/efectos de los fármacos , Metimazol/análogos & derivados , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Tionas/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Células Cultivadas , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Metimazol/farmacología , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología , Triglicéridos/metabolismo
13.
PLoS One ; 12(3): e0173747, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28282455

RESUMEN

A growing body of evidence suggests that L-selectin ligands presented on circulating tumor cells facilitate metastasis by binding L-selectin presented on leukocytes. Commonly used methods for detecting L-selectin ligands on tissues, e.g., immunostaining, are performed under static, no-flow conditions. However, such analysis does not assay for functional L-selectin ligands, specifically those ligands that promote adhesion under shear flow conditions. Recently our lab developed a method, termed dynamic biochemical tissue analysis (DBTA), to detect functional selectin ligands in situ by probing tissues with L-selectin-coated microspheres under hemodynamic flow conditions. In this investigation, DBTA was used to probe human colon tissues for L-selectin ligand activity. The detection of L-selectin ligands using DBTA was highly specific. Furthermore, DBTA reproducibly detected functional L-selectin ligands on diseased, e.g., cancerous or inflamed, tissues but not on noncancerous tissues. In addition, DBTA revealed a heterogeneous distribution of functional L-selectin ligands on colon cancer tissues. Most notably, detection of L-selectin ligands by immunostaining using HECA-452 antibody only partially correlated with functional L-selectin ligands detected by DBTA. In summation, the results of this study demonstrate that DBTA detects functional selectin ligands to provide a unique characterization of pathological tissue.


Asunto(s)
Bioquímica/métodos , Neoplasias del Colon/metabolismo , Selectina L/metabolismo , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/patología , Adenocarcinoma Papilar/metabolismo , Adenocarcinoma Papilar/patología , Carcinoma de Células en Anillo de Sello/metabolismo , Carcinoma de Células en Anillo de Sello/patología , Neoplasias del Colon/patología , Formaldehído , Glicoconjugados/análisis , Glicoconjugados/metabolismo , Humanos , Ligandos , Microscopía Fluorescente , Microesferas , Fijación del Tejido/métodos
14.
Eur J Pharmacol ; 803: 130-137, 2017 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-28343970

RESUMEN

Inhibition of interleukin-6 (IL-6) holds significant promise as a therapeutic approach for triple negative breast cancer (TNBC). We previously reported that phenylmethimazole (C10) reduces IL-6 expression in several cancer cell lines. We have identified a more potent derivative of C10 termed COB-141. In the present work, we tested the hypothesis that C10 and COB-141 inhibit TNBC cell expressed IL-6 and investigated the potential for classical IL-6 pathway induced signaling within TNBC cells. A panel of TNBC cell lines (MDA-MB-231, Hs578T, MDA-MB-468) was used. Enzyme linked immunosorbent assays (ELISA) revealed that C10 and COB-141 inhibit MDA-MB-231 cell IL-6 secretion, with COB-141 being ~6.5 times more potent than C10. Therefore, the remainder of the study focused on COB-141 which inhibited IL-6 secretion, and was found, via quantitative real time polymerase chain reaction (QRT-PCR), to inhibit IL-6 mRNA in the TNBC panel. COB-141 had little, if any, effect on metabolic activity indicating that the IL-6 inhibition is not via a toxic effect. Flow cytometric analysis and QRT-PCR revealed that the TNBC cell lines do not express the IL-6 receptor (IL-6Rα). Trans-AM assays suggested that COB-141 exerts its inhibitory effect, at least in part, by reducing NF-κB (p65/p50) DNA binding. In summary, COB-141 is a potent inhibitor of TNBC cell expressed IL-6 and the inhibition does not appear to be due to non-specific toxicity. The TNBC cell lines do not have an intact classical IL-6 signaling pathway. COB-141's inhibitory effect may be due, at least in part, to reducing NF-κB (p65/p50) DNA binding.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interleucina-6/metabolismo , Metimazol/análogos & derivados , Tiazoles/química , Tionas/química , Tionas/farmacología , Neoplasias de la Mama Triple Negativas/patología , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Interleucina-8/metabolismo , Metimazol/química , Metimazol/farmacología , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo
15.
Eur J Pharmacol ; 751: 59-66, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25641748

RESUMEN

The expression of vascular cell adhesion molecule-1 (VCAM-1) on the vascular endothelium can be increased by pro-inflammatory cytokines [e.g. tumor necrosis factor-α (TNF-α)]. VCAM-1 contributes to leukocyte adhesion to, and emigration from, the vasculature which is a key aspect of pathological inflammation. As such, a promising therapeutic approach for pathological inflammation is to inhibit the expression of VCAM-1. Methimazole [3-methyl-1, 3 imidazole-2 thione (MMI)] is routinely used for the treatment of Graves׳ disease and patients treated with MMI have decreased levels of circulating VCAM-1. In this study we used cultured human umbilical vein endothelial cells (HUVEC) to investigate the effect of MMI structural modifications on TNF-α induced VCAM-1 expression. We found that addition of a phenyl ring at the 4-nitrogen of MMI yields a compound that is significantly more potent than MMI at inhibiting 24h TNF-α-induced VCAM-1 protein expression. Addition of a para methoxy to the appended phenyl group increases the inhibition while substitution of a thiazole ring for an imidazole ring in the phenyl derivatives yields no clear difference in inhibition. Addition of the phenyl ring to MMI appears to increase toxicity as does substitution of a thiazole ring for an imidazole ring in the phenyl MMI derivatives. Each of the compounds reduced TNF-α-induced VCAM-1 mRNA expression and had a functional inhibitory effect, i.e. each inhibited monocytic cell adhesion to 24h TNF-α-activated HUVEC under fluid flow conditions. Combined, these studies provide important insights into the design of MMI-related anti-inflammatory compounds.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Metimazol/química , Metimazol/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/genética , Fenómenos Biomecánicos/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular , Humanos , Imidazoles/química , Monocitos/citología , Monocitos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Relación Estructura-Actividad , Tiazoles/química
16.
Am J Physiol Cell Physiol ; 308(1): C68-78, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25339657

RESUMEN

Adhesion of circulating tumor cells to vascular endothelium is mediated by specialized molecules that are functional under shear forces exerted by hematogenous flow. Endothelial E-selectin binding to glycoforms of CD44 mediates shear-resistant cell adhesion in numerous physiological and pathological conditions. However, this pathway is poorly understood in breast cancer and is the focus of the present investigation. All breast cancer cell lines used in this study strongly expressed CD44. In particular, BT-20 cells expressed CD44s and multiple CD44v isoforms, whereas MDA-MB-231 cells predominantly expressed CD44s but weakly expressed CD44v isoforms. CD44 expressed by BT-20, but not MDA-MB-231, cells possessed E-selectin ligand activity as detected by Western blotting and antigen capture assays. Importantly, CD44 expressed by intact BT-20 cells were functional E-selectin ligands, regulating cell rolling and adhesion under physiological flow conditions, as found by shRNA-targeted silencing of CD44. Antigen capture assays strongly suggest greater shear-resistant E-selectin ligand activity of BT-20 cell CD44v isoforms than CD44s. Surprisingly, CD44 was not recognized by the HECA-452 MAb, which detects sialofucosylated epitopes traditionally expressed by selectin ligands, suggesting that BT-20 cells express a novel glycoform of CD44v as an E-selectin ligand. The activity of this glycoform was predominantly attributed to N-linked glycans. Furthermore, expression of CD44v as an E-selectin ligand correlated with high levels of fucosyltransferase-3 and -6 and epithelial, rather than mesenchymal, cell phenotype. Together, these data demonstrate that expression of CD44 as a functional E-selectin ligand may be important in breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Adhesión Celular , Selectina E/metabolismo , Receptores de Hialuranos/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Células CHO , Línea Celular Tumoral , Movimiento Celular , Cricetulus , Selectina E/genética , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Fucosiltransferasas/metabolismo , Glicosilación , Humanos , Receptores de Hialuranos/genética , Ligandos , Metástasis de la Neoplasia , Fenotipo , Isoformas de Proteínas , Interferencia de ARN , Flujo Sanguíneo Regional , Transfección
17.
Drug Dev Res ; 75(8): 497-509, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25408546

RESUMEN

Preclinical Research Phenylmethimazole (C10) is an inhibitor of Toll-like receptor (TLR3 and TLR4) expression and signaling. In this study, we carried out a detailed investigation of the effect of C10 on TLR4 and its molecular signaling products in RAW 264.7 macrophages using quantitative real-time polymerase chain reaction (PCR), ELISA and cell toxicity assays, a set of in vitro assays that may be used to screen future C10 analogs. C10 exhibited an inhibitory effect on TLR4 MyD88-dependent and MyD88-independent pathways. Within the TLR4 pathway, C10 inhibited the expression of cytokines, cytokine receptors, kinases, adapter molecules and transcription factors, suggesting a pathway-wide inhibitory effect. We also found that C10 dose-dependently inhibited the expression of TLR4 signaling products, specifically IL-6, inducible nitric oxide (NO) synthase and IFNß. Additionally, pre-treatment of RAW 264.7 cells with C10 resulted in protection from lipopolysaccharide (LPS) insults, suggesting C10 may be bound to the target thus exhibiting activity during/following LPS stimulation. Also, dimethyl sulfoxide, the solvent for C10 exhibited inhibitory effect on TLR4 signaling products independent from the effects of C10. Combined, this study enhances understanding of the actions of C10 on the TLR4 signaling pathway providing a path for the development of new C10 analogs for inhibiting TLR expression and signaling [corrected].


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Metimazol/análogos & derivados , Transducción de Señal/efectos de los fármacos , Tionas/farmacología , Receptor Toll-Like 4/genética , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/citología , Metimazol/farmacología , Ratones , Receptor Toll-Like 4/metabolismo
18.
Inflamm Res ; 63(4): 277-85, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24346141

RESUMEN

OBJECTIVE AND DESIGN: Atherosclerosis (ATH) is a chronic inflammatory disease that involves cascades of signaling events mediated by various effector proteins. Here we sought to determine if the expression of Wnt5a, a secreted glycoprotein, is altered in discrete regions of the arterial plaque. METHODS: Atherosclerotic plaque tissues from 14 human subjects undergoing elective carotid endarterectomy were used in this study. Immunohistochemistry and laser capture microdissection combined with quantitative real-time PCR were used to determine the expression of Wnt5a and Toll-like receptors (TLRs) in different sections of the arterial lesions. Atherosclerotic serum samples (n = 30) and serum from healthy subjects (n = 16) were quantified for Wnt5a using an enzyme-linked immunosorbent assay (ELISA). RESULTS: The data analysis revealed that Wnt5a transcripts and protein were elevated in advanced arterial lesions relative to less advanced arterial lesions; that Wnt5a expression correlated with the presence of TLR4 and TLR2 transcripts; and that the average amount of Wnt5a protein present in atherosclerotic patient serum was significantly higher compared to healthy controls. CONCLUSIONS: This study is the first to provide evidence that the expression of Wnt5a increases as the disease progresses to a more advanced stage, and that this expression is coincident with that of TLR2 and TLR4. In addition, we found that the average Wnt5a levels in the serum of atherosclerotic patients are elevated relative to healthy controls, which is consistent with the hypothesis that Wnt5a plays a role in ATH.


Asunto(s)
Aterosclerosis/genética , Proteínas Proto-Oncogénicas/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Proteínas Wnt/genética , Adulto , Anciano , Anciano de 80 o más Años , Aterosclerosis/sangre , Aterosclerosis/metabolismo , Aterosclerosis/patología , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/sangre , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/metabolismo , Proteínas Wnt/sangre , Proteínas Wnt/metabolismo , Proteína Wnt-5a
19.
Molecules ; 17(10): 12365-77, 2012 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-23090018

RESUMEN

Previous studies revealed that phenylmethimazole (C10) inhibits IRF3 signaling, preventing dsRNA-induction of type 1 interferon gene expression, production, and downstream signaling. In the present study, we investigated the molecular basis for C10 inhibition of dsRNA-stimulated IRF3 signaling. IRF-3 Trans-AM assays were used to measure C10 effects on dsRNA induction of IRF3 DNA binding. Green fluorescent protein-labeled IRF3 was used to measure C10 effects on dsRNA-induced IRF3 nuclear translocation. Native PAGE, SDS PAGE, and western blotting were used to identify effects of C10 on IRF3 homodimer formation and phosphorylation, respectively. There was a significant impairment of dsRNA-induced IRF3 DNA binding activity in human embryonic kidney and pancreatic cancer cells with C10 treatment. C10 also blocked dsRNA-induced IRF3 nuclear translocation and homodimer formation without blocking serine 396 phosphorylation of IRF3. Together, these results indicate that C10 interferes with IRF3 signaling by blocking dsRNA-induced IRF3 homodimer formation, a prerequisite for nuclear translocation and DNA binding activities.


Asunto(s)
Núcleo Celular/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Metimazol/análogos & derivados , Multimerización de Proteína/efectos de los fármacos , ARN Bicatenario/farmacología , Tionas/farmacología , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , ADN/metabolismo , Células HEK293 , Humanos , Metimazol/farmacología , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos
20.
Front Oncol ; 2: 103, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22934288

RESUMEN

Although significant progress has been made in the fight against cancer, successful treatment strategies have yet to be developed to combat those tumors that have metastasized to distant organs. Poor characterization of the molecular mechanisms of cancer spread is a major impediment to designing predictive diagnostics and effective clinical interventions against late stage disease. In hematogenous metastasis, it is widely suspected that circulating tumor cells (CTCs) express specific adhesion molecules that actively initiate contact with the vascular endothelium lining the vessel walls of the target organ. This "tethering" is mediated by ligands expressed by CTCs that bind to E-selectin expressed by endothelial cells. However, it is currently unknown whether expression of functional E-selectin ligands on CTCs is related to cancer stem cell regulatory or maintenance pathways, particularly epithelial-to-mesenchymal transition and the reverse, mesenchymal-to-epithelial transition. In this hypothesis and theory article, we explore the potential roles of these mechanisms on the dynamic regulation of selectin ligands mediating CTC trafficking during metastasis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA