Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Hepatology ; 2023 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-37870288

RESUMEN

BACKGROUND AND AIMS: The liver has a remarkable capacity to regenerate, which is sustained by the ability of hepatocytes to act as facultative stem cells that, while normally quiescent, re-enter the cell cycle after injury. Growth factor signaling is indispensable in rodents, whereas Wnt/ß-catenin is not required for effective tissue repair. However, the molecular networks that control human liver regeneration remain unclear. METHODS: Organotypic 3D spheroid cultures of primary human or murine hepatocytes were used to identify the signaling network underlying cell cycle re-entry. Furthermore, we performed chemogenomic screening of a library enriched for epigenetic regulators and modulators of immune function to determine the importance of epigenomic control for human hepatocyte regeneration. RESULTS: Our results showed that, unlike in rodents, activation of Wnt/ß-catenin signaling is the major mitogenic cue for adult primary human hepatocytes. Furthermore, we identified TGFß inhibition and inflammatory signaling through NF-κB as essential steps for the quiescent-to-regenerative switch that allows Wnt/ß-catenin-induced proliferation of human cells. In contrast, growth factors, but not Wnt/ß-catenin signaling, triggered hyperplasia in murine hepatocytes. High-throughput screening in a human model confirmed the relevance of NFκB and revealed the critical roles of polycomb repressive complex 2, as well as of the bromodomain families I, II, and IV. CONCLUSIONS: This study revealed a network of NFκB, TGFß, and Wnt/ß-catenin that controls human hepatocyte regeneration in the absence of exogenous growth factors, identified novel regulators of hepatocyte proliferation, and highlighted the potential of organotypic culture systems for chemogenomic interrogation of complex physiological processes.

2.
Int J Mol Sci ; 23(23)2022 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-36499165

RESUMEN

The PCTAIRE subfamily belongs to the CDK (cyclin-dependent kinase) family and represents an understudied class of kinases of the dark kinome. They exhibit a highly conserved binding pocket and are activated by cyclin Y binding. CDK16 is targeted to the plasma membrane after binding to N-myristoylated cyclin Y and is highly expressed in post-mitotic tissues, such as the brain and testis. Dysregulation is associated with several diseases, including breast, prostate, and cervical cancer. Here, we used the N-(1H-pyrazol-3-yl)pyrimidin-4-amine moiety from the promiscuous inhibitor 1 to target CDK16, by varying different residues. Further optimization steps led to 43d, which exhibited high cellular potency for CDK16 (EC50 = 33 nM) and the other members of the PCTAIRE and PFTAIRE family with 20-120 nM and 50-180 nM, respectively. A DSF screen against a representative panel of approximately 100 kinases exhibited a selective inhibition over the other kinases. In a viability assessment, 43d decreased the cell count in a dose-dependent manner. A FUCCI cell cycle assay revealed a G2/M phase cell cycle arrest at all tested concentrations for 43d, caused by inhibition of CDK16.


Asunto(s)
Quinasas Ciclina-Dependientes , Ciclinas , Masculino , Humanos , Ciclinas/metabolismo , Secuencia de Aminoácidos , Quinasas Ciclina-Dependientes/metabolismo , Unión Proteica
3.
Molecules ; 27(4)2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35209227

RESUMEN

Phenotypical screening is a widely used approach in drug discovery for the identification of small molecules with cellular activities. However, functional annotation of identified hits often poses a challenge. The development of small molecules with narrow or exclusive target selectivity such as chemical probes and chemogenomic (CG) libraries, greatly diminishes this challenge, but non-specific effects caused by compound toxicity or interference with basic cellular functions still pose a problem to associate phenotypic readouts with molecular targets. Hence, each compound should ideally be comprehensively characterized regarding its effects on general cell functions. Here, we report an optimized live-cell multiplexed assay that classifies cells based on nuclear morphology, presenting an excellent indicator for cellular responses such as early apoptosis and necrosis. This basic readout in combination with the detection of other general cell damaging activities of small molecules such as changes in cytoskeletal morphology, cell cycle and mitochondrial health provides a comprehensive time-dependent characterization of the effect of small molecules on cellular health in a single experiment. The developed high-content assay offers multi-dimensional comprehensive characterization that can be used to delineate generic effects regarding cell functions and cell viability, allowing an assessment of compound suitability for subsequent detailed phenotypic and mechanistic studies.


Asunto(s)
Descubrimiento de Drogas/métodos , Genómica/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Imagen Molecular/métodos , Bibliotecas de Moléculas Pequeñas , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos/métodos , Humanos , Reproducibilidad de los Resultados , Coloración y Etiquetado
4.
Methods Mol Biol ; 2706: 59-73, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37558941

RESUMEN

The characterization of chemogenomic libraries with respect to their general effect on cellular health represents essential data for the annotation of phenotypic responses. Here, we describe a multidimensional high-content live cell assay that allows to examine cell viability in different cell lines, based on their nuclear morphology as well as modulation of small molecules of tubulin structure, mitochondrial health, and membrane integrity. The protocol monitors cells during a time course of 48 h using osteosarcoma cells, human embryonic kidney cells, and untransformed human fibroblasts as an example. The described protocol can be easily established and it can be adapted to other cell lines or other parameters important for cellular health.


Asunto(s)
Fibroblastos , Microscopía , Humanos , Línea Celular , Tubulina (Proteína) , Mitocondrias
5.
Methods Mol Biol ; 2706: 75-88, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37558942

RESUMEN

In recent years, the assembly and annotation of chemogenomic libraries have gained interest by the phenotypic screening community. Apart from basic annotations of the compound potency and selectivity, these compound libraries benefit in particular from annotation regarding the effect of the inhibitors on cellular viability to distinguish between on-target effects of a compound and unspecific cytotoxicity. Here, we provide a protocol to determine viability as a first determinant in compound quality control, using the Incucyte live-cell imaging system. The compounds are classified according to their calculated growth rate to determine a cytotoxic, cytostatic, or healthy outcome. All compounds affecting the growth rate can be further evaluated regarding their specific effects on cell health in a high-content live-cell multiplex assay, described in Chapter 5 .


Asunto(s)
Supervivencia Celular , Microscopía , Microscopía/métodos
6.
Cell Chem Biol ; 30(12): 1634-1651.e6, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-37797617

RESUMEN

Drug-induced phospholipidosis (DIPL), characterized by excessive accumulation of phospholipids in lysosomes, can lead to clinical adverse effects. It may also alter phenotypic responses in functional studies using chemical probes. Therefore, robust methods are needed to predict and quantify phospholipidosis (PL) early in drug discovery and in chemical probe characterization. Here, we present a versatile high-content live-cell imaging approach, which was used to evaluate a chemogenomic and a lysosomal modulation library. We trained and evaluated several machine learning models using the most comprehensive set of publicly available compounds and interpreted the best model using SHapley Additive exPlanations (SHAP). Analysis of high-quality chemical probes extracted from the Chemical Probes Portal using our algorithm revealed that closely related molecules, such as chemical probes and their matched negative controls can differ in their ability to induce PL, highlighting the importance of identifying PL for robust target validation in chemical biology.


Asunto(s)
Lipidosis , Enfermedades por Almacenamiento Lisosomal , Humanos , Lipidosis/inducido químicamente , Fosfolípidos , Aprendizaje Automático , Descubrimiento de Drogas
7.
ACS Chem Biol ; 18(4): 822-836, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-36944371

RESUMEN

Well-characterized small molecules are essential tools for studying the biology and therapeutic relevance of a target protein. However, many compounds reported in the literature and routinely studied in biomedical research lack the potency and selectivity required for mechanistic cellular studies on the function of a given protein. Furthermore, commercially available compounds often do not include useful tools developed by industry as part of their research and development efforts, as they frequently remain proprietary. The freely available donated chemical probe (DCP) library, fueled by generous donations of compounds from industry and academia, enables easy access to a steadily growing collection of these valuable and well-characterized tools. Here, we provide a systematic description of the current DCP library collection and their associated comprehensive characterization data, including a variety of in vitro and cellular assays. Of note, we characterized the set in relevant human primary models by employing hepatotoxicity screening in primary human liver spheroids and viability screening in patient-derived colorectal cancer organoids and matched normal-adjacent epithelium. Taken together, the DCP library represents a well-annotated, openly available collection of tool compounds for studying a wide range of targets, including kinases, G-protein-coupled receptors, and ion channels. As such, it represents a unique resource for the biomedical research community.


Asunto(s)
Sondas Moleculares , Neoplasias , Bibliotecas de Moléculas Pequeñas , Humanos , Hígado , Sistemas Microfisiológicos , Neoplasias/metabolismo , Organoides/metabolismo , Organoides/patología , Proteínas/metabolismo , Bibliotecas de Moléculas Pequeñas/clasificación , Sondas Moleculares/química , Sondas Moleculares/farmacología
8.
Eur J Med Chem ; 254: 115347, 2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37094449

RESUMEN

Salt-inducible kinases 1-3 (SIK1-3) are key regulators of the LKB1-AMPK pathway and play an important role in cellular homeostasis. Dysregulation of any of the three isoforms has been associated with tumorigenesis in liver, breast, and ovarian cancers. We have recently developed the dual pan-SIK/group I p21-activated kinase (PAK) chemical probe MRIA9. However, inhibition of p21-activated kinases has been associated with cardiotoxicity in vivo, which complicates the use of MRIA9 as a tool compound. Here, we present a structure-based approach involving the back-pocket and gatekeeper residues, for narrowing the selectivity of pyrido[2,3-d]pyrimidin-7(8H)-one-based inhibitors towards SIK kinases, eliminating PAK activity. Optimization was guided by high-resolution crystal structure analysis and computational methods, resulting in a pan-SIK inhibitor, MR22, which no longer exhibited activity on STE group kinases and displayed excellent selectivity in a representative kinase panel. MR22-dependent SIK inhibition led to centrosome dissociation and subsequent cell-cycle arrest in ovarian cancer cells, as observed with MRIA9, conclusively linking these phenotypic effects to SIK inhibition. Taken together, MR22 represents a valuable tool compound for studying SIK kinase function in cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Proteínas Serina-Treonina Quinasas , Proteínas Quinasas Activadas por AMP/metabolismo , Hígado/metabolismo , Isoformas de Proteínas , Inhibidores de Proteínas Quinasas/farmacología
9.
STAR Protoc ; 3(4): 101791, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36317177

RESUMEN

Well-characterized small molecules enable the study of cell processes and facilitate target validation. Here, we describe a high-content multiplex screen to investigate cell viability over 48 h, which can be combined with investigating phenotypic features, such as tubulin binding and mitochondrial content, as initial cellular quality control of diverse compounds. The protocol is on a live-cell basis and easily adaptable and scalable. It details cell preparation, compound handling, plate layout configuration, image acquisition with the CQ1, and data analysis using the CellPathfinder software. For complete details on the use and execution of this protocol, please refer to Tjaden et al. (2022).


Asunto(s)
Mitocondrias , Programas Informáticos , Supervivencia Celular
10.
J Med Chem ; 65(11): 7799-7817, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35608370

RESUMEN

Serine/threonine kinase 17A (death-associated protein kinase-related apoptosis-inducing protein kinase 1─DRAK1) is a part of the death-associated protein kinase (DAPK) family and belongs to the so-called dark kinome. Thus, the current state of knowledge of the cellular function of DRAK1 and its involvement in pathophysiological processes is very limited. Recently, DRAK1 has been implicated in tumorigenesis of glioblastoma multiforme (GBM) and other cancers, but no selective inhibitors of DRAK1 are available yet. To this end, we optimized a pyrazolo[1,5-a]pyrimidine-based macrocyclic scaffold. Structure-guided optimization of this macrocyclic scaffold led to the development of CK156 (34), which displayed high in vitro potency (KD = 21 nM) and selectivity in kinomewide screens. Crystal structures demonstrated that CK156 (34) acts as a type I inhibitor. However, contrary to studies using genetic knockdown of DRAK1, we have seen the inhibition of cell growth of glioma cells in 2D and 3D culture only at low micromolar concentrations.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Proteínas Serina-Treonina Quinasas , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Proteínas Quinasas Asociadas a Muerte Celular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Serina
11.
Biochem Pharmacol ; 204: 115191, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35907497

RESUMEN

Focused compound libraries are well-established tools for hit identification in drug discovery and chemical probe development. We present the compilation and application of a focused screening library of fatty acid mimetics (FAMs), which are compounds designed to bind the orthosteric site of proteins that endogenously accommodate natural fatty acids and lipid metabolites. This set complies with chemical properties of FAM and was found suitable for use also in cellular setting. Several hits were retrieved in screening the focused library against diverse fatty acid binding targets including the enzymes soluble epoxide hydrolase (sEH) and leukotriene A4 hydrolase (LTA4H), the nuclear receptors peroxisome proliferator-activated receptor γ (PPARγ) and retinoid X receptor α (RXRα), the carrier proteins fatty acid binding protein 4 and 5 (FABP4 and FABP5), as well as the G-protein coupled receptors leukotriene B4 receptor 1 (BLT1) and free-fatty acid receptor 1 (FFAR1). Thus, the focused FAM library is suitable to obtain chemical starting matter for fatty acid binding proteins and provides a valuable extension to available screening collections.


Asunto(s)
Epóxido Hidrolasas , Ácidos Grasos , Epóxido Hidrolasas/metabolismo , Proteínas de Unión a Ácidos Grasos , Ácidos Grasos/metabolismo , PPAR gamma/metabolismo , Receptores de Leucotrieno B4/metabolismo , Receptor alfa X Retinoide/metabolismo
12.
Cell Chem Biol ; 28(4): 546-558.e10, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33484635

RESUMEN

Building on the pyrazolopyrimidine CK2 (casein kinase 2) inhibitor scaffold, we designed a small targeted library. Through comprehensive evaluation of inhibitor selectivity, we identified inhibitor 24 (SGC-CK2-1) as a highly potent and cell-active CK2 chemical probe with exclusive selectivity for both human CK2 isoforms. Remarkably, despite years of research pointing to CK2 as a key driver in cancer, our chemical probe did not elicit a broad antiproliferative phenotype in >90% of >140 cell lines when tested in dose-response. While many publications have reported CK2 functions, CK2 biology is complex and an available high-quality chemical tool such as SGC-CK2-1 will be indispensable in deciphering the relationships between CK2 function and phenotypes.


Asunto(s)
Quinasa de la Caseína II/antagonistas & inhibidores , Desarrollo de Medicamentos , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirazoles/síntesis química , Pirazoles/química , Pirimidinas/síntesis química , Pirimidinas/química , Relación Estructura-Actividad
13.
J Med Chem ; 64(18): 13451-13474, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34506142

RESUMEN

Discoidin domain receptors 1 and 2 (DDR1/2) play a central role in fibrotic disorders, such as renal and pulmonary fibrosis, atherosclerosis, and various forms of cancer. Potent and selective inhibitors, so-called chemical probe compounds, have been developed to study DDR1/2 kinase signaling. However, these inhibitors showed undesired activity on other kinases such as the tyrosine protein kinase receptor TIE or tropomyosin receptor kinases, which are related to angiogenesis and neuronal toxicity. In this study, we optimized our recently published p38 mitogen-activated protein kinase inhibitor 7 toward a potent and cell-active dual DDR/p38 chemical probe and developed a structurally related negative control. The structure-guided design approach used provided insights into the P-loop folding process of p38 and how targeting of non-conserved amino acids modulates inhibitor selectivity. The developed and comprehensively characterized DDR/p38 probe, 30 (SR-302), is a valuable tool for studying the role of DDR kinase in normal physiology and in disease development.


Asunto(s)
Benzamidas/farmacología , Receptor con Dominio Discoidina 1/metabolismo , Receptor con Dominio Discoidina 2/metabolismo , Sulfonamidas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Sitio Alostérico , Animales , Benzamidas/síntesis química , Benzamidas/metabolismo , Línea Celular Tumoral , Receptor con Dominio Discoidina 1/química , Receptor con Dominio Discoidina 2/química , Perros , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Microsomas Hepáticos/metabolismo , Unión Proteica , Sulfonamidas/síntesis química , Sulfonamidas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/química
14.
Eur J Med Chem ; 208: 112770, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-32883634

RESUMEN

Casein kinase 2 (CK2) is a constitutively expressed serine/threonine kinase that has a large diversity of cellular substrates. Thus, CK2 has been associated with a plethora of regulatory functions and dysregulation of CK2 has been linked to disease development in particular to cancer. The broad implications in disease pathology makes CK2 an attractive target. To date, the most advanced CK2 inhibitor is silmitasertib, which has been investigated in clinical trials for treatment of various cancers, albeit several off-targets for silmitasertib have been described. To ascertain the role of CK2 inhibition in cancer, other disease and normal physiology the development of a selective CK2 inhibitor would be highly desirable. In this study we explored the pyrazolo [1,5-a]pyrimidine hinge-binding moiety for the development of selective CK2 inhibitors. Optimization of this scaffold, which included macrocyclization, led to IC20 (31) a compound that displayed high in vitro potency for CK2 (KD = 12 nM) and exclusive selectivity for CK2. X-ray analysis revealed a canonical type-I binding mode for IC20 (31). However, the polar carboxylic acid moiety that is shared by many CK2 inhibitors including silmitasertib was required for potency but limits the cellular activity of IC20 (31) and the cellular IC50 dropped to the low micromolar range. In summary, IC20 (31) represents a highly selective and potent inhibitor of CK2, which can be used as a tool compound to study CK2 biology and potential new applications for the treatment of diseases.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Sitios de Unión , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/química , Quinasa de la Caseína II/metabolismo , Cristalografía por Rayos X , Células HEK293 , Humanos , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Pirazoles/síntesis química , Pirazoles/metabolismo , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Relación Estructura-Actividad
15.
Commun Chem ; 3(1): 174, 2020 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-36703463

RESUMEN

Non-alcoholic steatohepatitis (NASH) - a hepatic manifestation of the metabolic syndrome - is a multifactorial disease with alarming global prevalence. It involves steatosis, inflammation and fibrosis in the liver, thus demanding multiple modes of action for robust therapeutic efficacy. Aiming to fuse complementary validated anti-NASH strategies in a single molecule, we have designed and systematically optimized a scaffold for triple activation of farnesoid X receptor (FXR), peroxisome proliferator-activated receptor (PPAR) α and PPARδ. Pilot profiling of the resulting triple modulator demonstrated target engagement in native cellular settings and in mice, rendering it a suitable tool to probe the triple modulator concept in vivo. In DIO NASH in mice, the triple agonist counteracted hepatic inflammation and reversed hepatic fibrosis highlighting the potential of designed polypharmacology in NASH.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA