Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Protein Sci ; 32(11): e4794, 2023 11.
Article in English | MEDLINE | ID: mdl-37800277

ABSTRACT

The enzyme Thiosulfate sulfurtransferase (TST, EC 2.8.1.1), is a positive genetic predictor of diabetes type 2 and obesity. As increased TST activity protects against the development of diabetic symptoms in mice, an activating compound for TST may provide therapeutic benefits in diabetes and obesity. We identified a small molecule activator of human TST through screening of an inhouse small molecule library. Kinetic studies in vitro suggest that two distinct isomers of the compound are required for full activation as well as an allosteric mode of activation. Additionally, we studied the effect of TST protein and the activator on TST activity through mitochondrial respiration. Molecular docking and molecular dynamics (MD) approaches supports an allosteric site for the binding of the activator, which is supported by the lack of activation in the Escherichia coli. mercaptopyruvate sulfurtransferase. Finally, we show that increasing TST activity in isolated mitochondria increases mitochondrial oxygen consumption.


Subject(s)
Diabetes Mellitus , Thiosulfate Sulfurtransferase , Mice , Humans , Animals , Thiosulfate Sulfurtransferase/chemistry , Thiosulfate Sulfurtransferase/genetics , Thiosulfate Sulfurtransferase/metabolism , Molecular Docking Simulation , Kinetics , Mitochondria/metabolism , Diabetes Mellitus/metabolism , Respiration , Obesity/metabolism
2.
Angew Chem Int Ed Engl ; 62(42): e202310059, 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37638390

ABSTRACT

Macrophage polarization plays a crucial role in inflammatory processes. The histone deacetylase 3 (HDAC3) has a deacetylase-independent function that can activate pro-inflammatory gene expression in lipopolysaccharide-stimulated M1-like macrophages and cannot be blocked by traditional small-molecule HDAC3 inhibitors. Here we employed the proteolysis targeting chimera (PROTAC) technology to target the deacetylase-independent function of HDAC3. We developed a potent and selective HDAC3-directed PROTAC, P7, which induces nearly complete HDAC3 degradation at low micromolar concentrations in both THP-1 cells and human primary macrophages. P7 increases the anti-inflammatory cytokine secretion in THP-1-derived M1-like macrophages. Importantly, P7 decreases the secretion of pro-inflammatory cytokines in M1-like macrophages derived from human primary macrophages. This can be explained by the observed inhibition of macrophage polarization from M0-like into M1-like macrophage. In conclusion, we demonstrate that the HDAC3-directed PROTAC P7 has anti-inflammatory activity and blocks macrophage polarization, demonstrating that this molecular mechanism can be targeted with small molecule therapeutics.

3.
Curr Protoc ; 3(6): e805, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37338240

ABSTRACT

Symmetrical deposition of parental and newly synthesized chromatin proteins over both sister chromatids is important for the maintenance of epigenetic integrity. However, the mechanisms to maintain equal distribution of parental and newly synthesized chromatid proteins over sister chromatids remains largely unknown. Here, we describe the protocol for the recently developed double-click seq method that enables mapping of asymmetry in the deposition of parental and newly synthesized chromatin proteins on both sister chromatids in DNA replication. The method involved metabolic labeling of new chromatin proteins with l-Azidohomoalanine (AHA) and newly synthesized DNA with Ethynyl-2'-deoxyuridine (EdU) followed by two subsequent click reactions for biotinylation and subsequently by corresponding separation steps. This enables isolation of parental DNA that was bound to nucleosomes containing new chromatin proteins. Sequencing of these DNA samples and mapping around origins of replication in the cellular DNA enables estimation of the asymmetry in deposition of chromatin proteins over the leading and lagging strand in DNA replication. Altogether, this method contributes to the toolbox to understand histone deposition in DNA replication. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Metabolic labeling with AHA and EdU and isolation of nuclei Basic Protocol 2: First click reaction, MNase digestion and streptavidin enrichment of labeled nucleosomes Basic Protocol 3: Second click reaction, Replication-Enriched Nucleosome Sequencing (RENS) Protocol.


Subject(s)
DNA , Nucleosomes , Nucleosomes/genetics , DNA/genetics , DNA/metabolism , Histones/genetics , Histones/metabolism , Nucleoproteins/genetics , Nucleoproteins/metabolism , DNA Replication
4.
J Med Chem ; 66(13): 8767-8781, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37352470

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine and essential signaling protein associated with inflammation and cancers. One of the newly described roles of MIF is binding to apoptosis-inducing factor (AIF) that "brings" cells to death in pathological conditions. The interaction between MIF and AIF and their nuclear translocation stands as a central event in parthanatos. However, classical competitive MIF tautomerase inhibitors do not interfere with MIF functions in parthanatos. In this study, we employed a pharmacophore-switch to provide allosteric MIF tautomerase inhibitors that interfere with the MIF/AIF co-localization. Synthesis and screening of a focused compound collection around the 1,2,3-triazole core enabled identification of the allosteric tautomerase MIF inhibitor 6y with low micromolar potency (IC50 = 1.7 ± 0.1 µM). This inhibitor prevented MIF/AIF nuclear translocation and protects cells from parthanatos. These findings indicate that alternative modes to target MIF hold promise to investigate MIF function in parthanatos-mediated diseases.


Subject(s)
Macrophage Migration-Inhibitory Factors , Parthanatos , Humans , Macrophage Migration-Inhibitory Factors/metabolism , Apoptosis Inducing Factor , Inflammation/metabolism , Intramolecular Oxidoreductases/metabolism
5.
J Med Chem ; 66(10): 6591-6616, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37155735

ABSTRACT

KAT8 is a lysine acetyltransferase primarily catalyzing the acetylation of Lys16 of histone H4 (H4K16). KAT8 dysregulation is linked to the development and metastatization of many cancer types, including non-small cell lung cancer (NSCLC) and acute myeloid leukemia (AML). Few KAT8 inhibitors have been reported so far, none of which displaying selective activity. Based on the KAT3B/KDAC inhibitor C646, we developed a series of N-phenyl-5-pyrazolone derivatives and identified compounds 19 and 34 as low-micromolar KAT8 inhibitors selective over a panel of KATs and KDACs. Western blot, immunofluorescence, and CETSA experiments demonstrated that both inhibitors selectively target KAT8 in cells. Moreover, 19 and 34 exhibited mid-micromolar antiproliferative activity in different cancer cell lines, including NSCLC and AML, without impacting the viability of nontransformed cells. Overall, these compounds are valuable tools for elucidating KAT8 biology, and their simple structures make them promising candidates for future optimization studies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Leukemia, Myeloid, Acute , Lung Neoplasms , Lysine Acetyltransferases , Humans , Lysine Acetyltransferases/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Histones/metabolism , Acetylation , Histone Acetyltransferases/metabolism
6.
Angew Chem Int Ed Engl ; 62(30): e202300681, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37026576

ABSTRACT

Photopharmacology is an attractive approach for achieving targeted drug action with the use of light. In photopharmacology, molecular photoswitches are introduced into the structure of biologically active small molecules to allow for the optical control of their potency. Going beyond trial and error, photopharmacology has progressively applied rational drug design methodologies to devise light-controlled bioactive ligands. In this review, we categorize photopharmacological efforts from the standpoint of medicinal chemistry strategies, focusing on diffusible photochromic ligands modified with photoswitches that operate through E-Z bond isomerization. In the vast majority of cases, photoswitchable ligands are designed as analogs of existing compounds, through a variety of approaches. By analyzing in detail a comprehensive list of instructive examples, we describe the state of the art and discuss future opportunities for rational design in photopharmacology.


Subject(s)
Chemistry, Pharmaceutical , Drug Design , Ligands
7.
Bioorg Chem ; 136: 106546, 2023 07.
Article in English | MEDLINE | ID: mdl-37098288

ABSTRACT

Various diseases are deeply associated with aberrations in HDAC8 functions. These aberrations can be assigned to either structural functions or catalytic functions of HDAC8. Therefore, development of HDAC8 degradation inducers might be more promising than HDAC8 inhibitors. We employed the proteolysis targeting chimera (PROTAC) strategy to develop a selective and potent HDAC8 degradation inducer CT-4 with single-digit nanomolar DC50 values and over 95% Dmax in both triple-negative breast cancer MDA-MB-231 cells and T-cell leukemia cells. Notably, CT-4 demonstrated potent anti-migration activity and limited anti-proliferative activity in MDA-MB-231 cells. In contrast, CT-4 effectively induced apototic cell death in Jurkat cells, as assessed by a caspase 3/7 activity assay and flow cytometry. Our findings suggest that the development of HDAC8 degradation inducers holds great potential for the treatment of HDAC8-related diseases.


Subject(s)
Proteolysis Targeting Chimera , Repressor Proteins , Humans , Cell Line, Tumor , Histone Deacetylases/metabolism , Jurkat Cells , Proteolysis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry
8.
ACS Pharmacol Transl Sci ; 5(9): 710-723, 2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36110375

ABSTRACT

Despite the success of drug discovery over the past decades, many potential drug targets still remain intractable for small molecule modulation. The development of proteolysis targeting chimeras (PROTACs) that trigger degradation of the target proteins provides a conceptually novel approach to address drug targets that remained previously elusive. Currently, the main challenge of PROTAC development is the identification of efficient, tissue- and cell-selective PROTAC molecules with good drug-likeness and favorable safety profiles. This review focuses on strategies to enhance the effectiveness and selectivity of PROTACs. We provide a comprehensive summary of recently reported PROTAC design strategies and discuss the advantages and disadvantages of these strategies. Future perspectives for PROTAC design will also be discussed.

9.
Microbes Infect ; 24(8): 105017, 2022.
Article in English | MEDLINE | ID: mdl-35709935

ABSTRACT

The acquisition of iron is a crucial mechanism for the survival of pathogenic bacteria such as Pseudomonas aeruginosa in eukaryotic hosts. The key iron chelator in this organism is the siderophore pyoverdine, which was shown to be crucial for iron homeostasis. Pyoverdine is a non-ribosomal peptide with several maturation steps in the cytoplasm and others in the periplasmatic space. A key enzyme for its maturation is the acylase PvdQ. The inhibition of PvdQ stops the maturation of pyoverdine causing a significant imbalance in the iron homeostasis and hence can negatively influence the survival of P. aeruginosa. In this work, we successfully synthesized chromene-derived inhibitory molecules targeting PvdQ in a low micromolar range. In silico modeling as well as kinetic evaluations of the inhibitors suggest a competitive inhibition of the PvdQ function. Further, we evaluated the inhibitor in vivo on P. aeruginosa cells and report a dose-dependent reduction of pyoverdine formation. The compound also showed a protecting effect in a Galleria mellonella infection model.


Subject(s)
Benzopyrans , Pseudomonas aeruginosa , Benzopyrans/pharmacology , Amidohydrolases/chemistry , Siderophores , Iron , Bacterial Proteins/chemistry
10.
J Med Chem ; 65(3): 2059-2077, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35041425

ABSTRACT

The homologous cytokines macrophage migration inhibitory factor (MIF) and d-dopachrome tautomerase (d-DT or MIF2) play key roles in cancers. Molecules binding to the MIF tautomerase active site interfere with its biological activity. In contrast, the lack of potent MIF2 inhibitors hinders the exploration of MIF2 as a drug target. In this work, screening of a focused compound collection enabled the identification of a MIF2 tautomerase inhibitor R110. Subsequent optimization provided inhibitor 5d with an IC50 of 1.0 µM for MIF2 tautomerase activity and a high selectivity over MIF. 5d suppressed the proliferation of non-small cell lung cancer cells in two-dimensional (2D) and three-dimensional (3D) cell cultures, which can be explained by the induction of cell cycle arrest via deactivation of the mitogen-activated protein kinase (MAPK) pathway. Thus, we discovered and characterized MIF2 inhibitors (5d) with improved antiproliferative activity in cellular models systems, which indicates the potential of targeting MIF2 in cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Intramolecular Oxidoreductases/metabolism , Pyrimidinones/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Drug Design , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Kinetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/metabolism , Molecular Dynamics Simulation , Phosphorylation/drug effects , Pyrimidinones/metabolism , Pyrimidinones/pharmacology , Structure-Activity Relationship
11.
Cell Mol Life Sci ; 79(2): 105, 2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35091838

ABSTRACT

The family of macrophage migration inhibitory factor (MIF) proteins in humans consist of MIF, its functional homolog D-dopachrome tautomerase (D-DT, also known as MIF-2) and the relatively unknown protein named DDT-like (DDTL). MIF is a pleiotropic cytokine with multiple properties in tissue homeostasis and pathology. MIF was initially found to associate with inflammatory responses and therefore established a reputation as a pro-inflammatory cytokine. However, increasing evidence demonstrates that MIF influences many different intra- and extracellular molecular processes important for the maintenance of cellular homeostasis, such as promotion of cellular survival, antioxidant signaling, and wound repair. In contrast, studies on D-DT are scarce and on DDTL almost nonexistent and their functions remain to be further investigated as it is yet unclear how similar they are compared to MIF. Importantly, the many and sometimes opposing functions of MIF suggest that targeting MIF therapeutically should be considered carefully, taking into account timing and severity of tissue injury. In this review, we focus on the latest discoveries regarding the role of MIF family members in tissue injury, inflammation and repair, and highlight the possibilities of interventions with therapeutics targeting or mimicking MIF family proteins.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Signal Transduction/physiology , Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Survival/physiology , Histocompatibility Antigens Class II/metabolism , Homeostasis/physiology , Humans , Protein Binding
12.
Chemistry ; 28(1): e202103030, 2022 Jan 03.
Article in English | MEDLINE | ID: mdl-34724273

ABSTRACT

Macrophage migration inhibitory factor (MIF) and its homolog MIF2 (also known as D-dopachrome tautomerase or DDT) play key roles in cell growth and immune responses. MIF and MIF2 expression is dysregulated in cancers and neurodegenerative diseases. Accurate and convenient detection of MIF and MIF2 will facilitate research on their roles in cancer and other diseases. Herein, we report the development and application of a 4-iodopyrimidine based probe 8 for the selective labeling of MIF and MIF2. Probe 8 incorporates a fluorophore that allows in situ imaging of these two proteins. This enabled visualization of the translocation of MIF2 from the cytoplasm to the nucleus upon methylnitronitrosoguanidine stimulation of HeLa cells. This observation, combined with literature on nuclease activity for MIF, enabled the identification of nuclease activity for MIF2 on human genomic DNA.


Subject(s)
Macrophage Migration-Inhibitory Factors , HeLa Cells , Humans , Intramolecular Oxidoreductases
13.
J Proteome Res ; 20(11): 5218-5221, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34669399

ABSTRACT

Affinity ligands such as antibodies are widely used in (bio)medical research for purifying proteins from complex biological samples. These ligands are generally immobilized onto solid supports which facilitate the separation of a captured protein from the sample matrix. Adsorptive microtiter plates are commonly used as solid supports prior to immunochemical detection (e.g., immunoassays) but hardly ever prior to liquid chromatography-mass spectrometry (LC-MS-)-based detection. Here, we describe the use of adsorptive microtiter plates for protein enrichment prior to LC-MS detection, and we discuss opportunities and challenges of corresponding workflows, based on examples of targeted (i.e., soluble receptor for advanced glycation end-products (sRAGE) in human serum) and discovery-based workflows (i.e., transcription factor p65 (NF-κB) in lysed murine RAW 264.7 macrophages and peptidyl-prolyl cis-trans isomerase FKBP5 (FKBP5) in lysed human A549 alveolar basal epithelial cells). Thereby, we aim to highlight the potential usefulness of adsorptive microtiter plates in affinity purification workflows prior to LC-MS detection, which could increase their usage in mass spectrometry-based protein research.


Subject(s)
Workflow , Animals , Chromatography, Affinity , Chromatography, Liquid/methods , Humans , Mass Spectrometry/methods , Mice , Receptor for Advanced Glycation End Products
14.
Bioorg Chem ; 117: 105396, 2021 12.
Article in English | MEDLINE | ID: mdl-34649152

ABSTRACT

Non-small-cell lung carcinoma (NSCLC) is one of the most common forms of lung cancer, and a leading cause of cancer death among human beings. There is an urgent demand for novel therapeutics for the treatment of NSCLC to enhance the efficacy of the currently applied Tyrosine kinase inhibitors (TKIs) therapy and to overcome therapy-resistance. Here, we report a novel small-molecule inhibitor that simultaneously targets histone deacetylase (HDAC) and macrophage migration inhibitory factor (MIF). The HDAC/MIF dual inhibitor proved to be toxic for EGFR mutated (H1650, TKI-resistant) or knock out (A549 EGFR-/-) NSCLC cell lines. Further experiments showed that HDAC inhibition inhibits cell survival and proliferation, while MIF inhibition downregulates pAKT or AKT expression level, which both interfere with cell survival. Furthermore, the combination treatment of TKI and HDAC/MIF dual inhibitor showed that the dual inhibitor enhanced TKI inhibitory efficacy, highlighting the advantages of HDAC/MIF dual inhibitor for more effective treatment of NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Lung Neoplasms/drug therapy , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , A549 Cells , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Survival/drug effects , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Humans , Intramolecular Oxidoreductases/metabolism , Lung Neoplasms/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
15.
ACS Chem Biol ; 16(11): 2193-2201, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34592816

ABSTRACT

Following DNA replication, equal amounts of chromatin proteins are distributed over sister chromatids by re-deposition of parental chromatin proteins and deposition of newly synthesized chromatin proteins. Molecular mechanisms balancing the allocation of new and old chromatin proteins remain largely unknown. Here, we studied the genome-wide distribution of new chromatin proteins relative to parental DNA template strands and replication initiation zones using the double-click-seq. Under control conditions, new chromatin proteins were preferentially found on DNA replicated by the lagging strand machinery. Strikingly, replication stress induced by hydroxyurea or curaxin treatment and inhibition of ataxia telangiectasia and Rad3-related protein (ATR) or p53 inactivation inverted the observed chromatin protein deposition bias to the strand replicated by the leading strand polymerase in line with previously reported effects on replication protein A occupancy. We propose that asymmetric deposition of newly synthesized chromatin proteins onto sister chromatids reflects differences in the processivity of leading and lagging strand synthesis.


Subject(s)
Chromatin/metabolism , DNA Replication/physiology , Hydroxyurea/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Chromatin/chemistry , Gene Expression Regulation/drug effects , Humans , Stress, Physiological , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Anal Chem ; 93(38): 12872-12880, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34519498

ABSTRACT

Histone acetylation is an important, reversible post-translational protein modification and a hallmark of epigenetic regulation. However, little is known about the dynamics of this process, due to the lack of analytical methods that can capture site-specific acetylation and deacetylation reactions. We present a new approach that combines metabolic and chemical labeling (CoMetChem) using uniformly 13C-labeled glucose and stable isotope-labeled acetic anhydride. Thereby, chemically equivalent, fully acetylated histone species are generated, enabling accurate relative quantification of site-specific lysine acetylation dynamics in tryptic peptides using high-resolution mass spectrometry. We show that CoMetChem enables site-specific quantification of the incorporation or loss of lysine acetylation over time, allowing the determination of reaction rates for acetylation and deacetylation. Thus, the CoMetChem methodology provides a comprehensive description of site-specific acetylation dynamics.


Subject(s)
Epigenesis, Genetic , Histones , Acetylation , Chromatography, Liquid , Histones/metabolism , Isotopes , Protein Processing, Post-Translational , Tandem Mass Spectrometry
17.
Angew Chem Int Ed Engl ; 60(40): 21875-21883, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34388301

ABSTRACT

Lipoxygenase (LOX) activity provides oxidative lipid metabolites, which are involved in inflammatory disorders and tumorigenesis. Activity-based probes to detect the activity of LOX enzymes in their cellular context provide opportunities to explore LOX biology and LOX inhibition. Here, we developed Labelox B as a potent covalent LOX inhibitor for one-step activity-based labeling of proteins with LOX activity. Labelox B was used to establish an ELISA-based assay for affinity capture and antibody-based detection of specific LOX isoenzymes. Moreover, Labelox B enabled efficient activity-based labeling of endogenous LOXs in living cells. LOX proved to localize in the nucleus, which was rationalized by identification of a functional bromodomain-like consensus motif in 15-LOX-1. This indicates that 15-LOX-1 is not only involved in oxidative lipid metabolism, but also in chromatin binding, which suggests a potential role in chromatin modifications.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Humans , Molecular Conformation
18.
Angew Chem Int Ed Engl ; 60(32): 17514-17521, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34018657

ABSTRACT

Macrophage migration inhibitory factor (MIF) is involved in protein-protein interactions that play key roles in inflammation and cancer. Current strategies to develop small molecule modulators of MIF functions are mainly restricted to the MIF tautomerase active site. Here, we use this site to develop proteolysis targeting chimera (PROTAC) in order to eliminate MIF from its protein-protein interaction network. We report the first potent MIF-directed PROTAC, denoted MD13, which induced almost complete MIF degradation at low micromolar concentrations with a DC50 around 100 nM in A549 cells. MD13 suppresses the proliferation of A549 cells, which can be explained by deactivation of the MAPK pathway and subsequent induction of cell cycle arrest at the G2/M phase. MD13 also exhibits antiproliferative effect in a 3D tumor spheroid model. In conclusion, we describe the first MIF-directed PROTAC (MD13) as a research tool, which also demonstrates the potential of PROTACs in cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoxazines/pharmacology , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Phthalimides/pharmacology , A549 Cells , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/chemical synthesis , Benzoxazines/chemical synthesis , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Intramolecular Oxidoreductases/chemistry , MAP Kinase Signaling System/drug effects , Macrophage Migration-Inhibitory Factors/chemistry , Phthalimides/chemical synthesis , Proteolysis/drug effects , Spheroids, Cellular/drug effects , Ubiquitin-Protein Ligases/metabolism
19.
Chem Commun (Camb) ; 57(34): 4126-4129, 2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33908493

ABSTRACT

Biaryl sulfonamides are excellent candidates for the azologization approach that yields photoswitchable drugs more active in their metastable cis state, compared to the stable trans state. Here we present the scope and limitations of this strategy for rational design in photopharmacology.

20.
Drug Discov Today ; 26(7): 1728-1734, 2021 07.
Article in English | MEDLINE | ID: mdl-33746067

ABSTRACT

Epigenetic mechanisms are important for the regular development and maintenance of the tissue-specific expression of cytokine genes. One of the crucial cytokines involved in cancer and inflammation is macrophage migration inhibitory factor (MIF), which triggers the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling pathways by binding to CD74 and other receptors. Altered expression of this cytokine and altered activity states of the connected pathways are linked to inflammatory disease and cancer. Therapeutic strategies based on epigenetic mechanisms have the potential to regulate MIF-mediated signaling in cancer and inflammation.


Subject(s)
Inflammation/genetics , Macrophage Migration-Inhibitory Factors/genetics , Neoplasms/genetics , Animals , Epigenesis, Genetic , Humans , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...