Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Data ; 9(1): 139, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35361787

ABSTRACT

The abnormal activation of signal transducer and activator of transcription (STAT) protein family is recognized as cause or driving force behind multiple diseases progression. Therefore, searching for potential treatment strategy is pursued by multiple scientific groups. We consider that providing comprehensive, integrated and unified dataset for STAT inhibitory compounds may serve as important tool for other researchers. We developed SINBAD (STAT INhbitor Biology And Drug-ability) in response to our experience with inhibitory compound research, knowing that gathering detailed information is crucial for effective experiment design and also for finding potential solutions in case of obtaining inconclusive results. SINBAD is a curated database of STAT inhibitors which have been published and described in scientific articles providing prove of their inhibitory properties. It is a tool allowing easy analysis of experimental conditions and provides detailed information about known STAT inhibitory compounds.


Subject(s)
Intracellular Signaling Peptides and Proteins , Pharmaceutical Preparations , Transcription Factors , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Signal Transduction , Transcription Factors/antagonists & inhibitors
2.
Front Immunol ; 10: 1176, 2019.
Article in English | MEDLINE | ID: mdl-31178872

ABSTRACT

Interferon regulatory factors (IRFs) are a family of homologous proteins that regulate the transcription of interferons (IFNs) and IFN-induced gene expression. As such they are important modulating proteins in the Toll-like receptor (TLR) and IFN signaling pathways, which are vital elements of the innate immune system. IRFs have a multi-domain structure, with the N-terminal part acting as a DNA binding domain (DBD) that recognizes a DNA-binding motif similar to the IFN-stimulated response element (ISRE). The C-terminal part contains the IRF-association domain (IAD), with which they can self-associate, bind to IRF family members or interact with other transcription factors. This complex formation is crucial for DNA binding and the commencing of target-gene expression. IRFs bind DNA and exert their activating potential as homo or heterodimers with other IRFs. Moreover, they can form complexes (e.g., with Signal transducers and activators of transcription, STATs) and collaborate with other co-acting transcription factors such as Nuclear factor-κB (NF-κB) and PU.1. In time, more of these IRF co-activating mechanisms have been discovered, which may play a key role in the pathogenesis of many diseases, such as acute and chronic inflammation, autoimmune diseases, and cancer. Detailed knowledge of IRFs structure and activating mechanisms predisposes IRFs as potential targets for inhibition in therapeutic strategies connected to numerous immune system-originated diseases. Until now only indirect IRF modulation has been studied in terms of antiviral response regulation and cancer treatment, using mainly antisense oligonucleotides and siRNA knockdown strategies. However, none of these approaches so far entered clinical trials. Moreover, no direct IRF-inhibitory strategies have been reported. In this review, we summarize current knowledge of the different IRF-mediated transcriptional regulatory mechanisms and how they reflect the diverse functions of IRFs in homeostasis and in TLR and IFN signaling. Moreover, we present IRFs as promising inhibitory targets and propose a novel direct IRF-modulating strategy employing a pipeline approach that combines comparative in silico docking to the IRF-DBD with in vitro validation of IRF inhibition. We hypothesize that our methodology will enable the efficient identification of IRF-specific and pan-IRF inhibitors that can be used for the treatment of IRF-dependent disorders and malignancies.


Subject(s)
Disease Susceptibility , Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Transcription, Genetic , Animals , Binding Sites , Biomarkers , Humans , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/genetics , Interferons/metabolism , Molecular Diagnostic Techniques , Molecular Targeted Therapy , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Signal Transduction , Structure-Activity Relationship , Toll-Like Receptors/metabolism , Trans-Activators/metabolism
3.
Front Immunol ; 9: 2141, 2018.
Article in English | MEDLINE | ID: mdl-30283459

ABSTRACT

Cardiovascular diseases (CVDs), including atherosclerosis, are globally the leading cause of death. Key factors contributing to onset and progression of atherosclerosis include the pro-inflammatory cytokines Interferon (IFN)α and IFNγ and the Pattern Recognition Receptor (PRR) Toll-like receptor 4 (TLR4). Together, they trigger activation of Signal Transducer and Activator of Transcription (STAT)s. Searches for compounds targeting the pTyr-SH2 interaction area of STAT3, yielded many small molecules, including STATTIC and STX-0119. However, many of these inhibitors do not seem STAT3-specific. We hypothesized that multi-STAT-inhibitors that simultaneously block STAT1, STAT2, and STAT3 activity and pro-inflammatory target gene expression may be a promising strategy to treat CVDs. Using comparative in silico docking of multiple STAT-SH2 models on multi-million compound libraries, we identified the novel multi-STAT inhibitor, C01L_F03. This compound targets the SH2 domain of STAT1, STAT2, and STAT3 with the same affinity and simultaneously blocks their activity and expression of multiple STAT-target genes in HMECs in response to IFNα. The same in silico and in vitro multi-STAT inhibiting capacity was shown for STATTIC and STX-0119. Moreover, C01L_F03, STATTIC and STX-0119 were also able to affect genome-wide interactions between IFNγ and TLR4 by commonly inhibiting pro-inflammatory and pro-atherogenic gene expression directed by cooperative involvement of STATs with IRFs and/or NF-κB. Moreover, we observed that multi-STAT inhibitors could be used to inhibit IFNγ+LPS-induced HMECs migration, leukocyte adhesion to ECs as well as impairment of mesenteric artery contractility. Together, this implicates that application of a multi-STAT inhibitory strategy could provide great promise for the treatment of CVDs.


Subject(s)
Atherosclerosis/genetics , Cardiovascular Diseases/genetics , Gene Expression Profiling/methods , Gene Expression/genetics , STAT Transcription Factors/genetics , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cell Line , Cells, Cultured , Cyclic S-Oxides/chemistry , Cyclic S-Oxides/pharmacology , Gene Expression/drug effects , Genome-Wide Association Study/methods , Humans , Male , Mice, Inbred C57BL , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , src Homology Domains
4.
Oncotarget ; 7(30): 48788-48812, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27166190

ABSTRACT

Key factors contributing to early stages of atherosclerosis and plaque development include the pro-inflammatory cytokines Interferon (IFN)α, IFNγ and Interleukin (IL)-6 and Toll-like receptor 4 (TLR4) stimuli. Together, they trigger activation of Signal Transducer and Activator of Transcription (STAT) and Interferon Regulatory Factor (IRF) families. In particular, STAT1, 2 and 3; IRF1 and 8 have recently been recognized as prominent modulators of inflammation, especially in immune and vascular cells during atherosclerosis. Moreover, inflammation-mediated activation of these STATs and IRFs coordinates a platform for synergistic amplification leading to pro-atherogenic responses.Searches for STAT3-targeting compounds, exploring the pTyr-SH2 interaction area of STAT3, yielded many small molecules including natural products. Only a few inhibitors for other STATs, but none for IRFs, are described. Promising results for several STAT3 inhibitors in recent clinical trials predicts STAT3-inhibiting strategies may find their way to the clinic. However, many of these inhibitors do not seem STAT-specific, display toxicity and are not very potent. This illustrates the need for better models, and screening and validation tools for novel STAT and IRF inhibitors.This review presents a summary of these findings. It postulates STAT1, STAT2 and STAT3 and IRF1 and IRF8 as interesting therapeutic targets and targeted inhibition could be a potential treatment strategy in CVDs. In addition, it proposes a pipeline approach that combines comparative in silico docking of STAT-SH2 and IRF-DBD models with in vitro STAT and IRF activation inhibition validation, as a novel tool to screen multi-million compound libraries and identify specific inhibitors for STATs and IRFs.


Subject(s)
Anticholesteremic Agents/therapeutic use , Atherosclerosis/drug therapy , Interferon Regulatory Factors/antagonists & inhibitors , Molecular Targeted Therapy/methods , STAT Transcription Factors/antagonists & inhibitors , Signal Transduction/drug effects , Atherosclerosis/pathology , Clinical Trials as Topic , Drug Evaluation, Preclinical/methods , Humans , Inflammation/drug therapy , Inflammation/pathology , Interferon Regulatory Factors/metabolism , Interferon-alpha/metabolism , Interferon-gamma/metabolism , Interleukin-6/metabolism , Molecular Docking Simulation , Molecular Targeted Therapy/trends , Phosphorylation , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , STAT Transcription Factors/metabolism , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...