Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 298(12): 102644, 2022 12.
Article in English | MEDLINE | ID: mdl-36309093

ABSTRACT

Idiopathic pulmonary fibrosis is a progressive and normally fatal disease with limited treatment options. The tyrosine kinase inhibitor nintedanib has recently been approved for the treatment of idiopathic pulmonary fibrosis, and its effectiveness has been linked to its ability to inhibit a number of receptor tyrosine kinases including the platelet-derived growth factor, vascular endothelial growth factor, and fibroblast growth factor receptors. We show here that nintedanib also inhibits salt-inducible kinase 2 (SIK2), with a similar IC50 to its reported tyrosine kinase targets. Nintedanib also inhibited the related kinases SIK1 and SIK3, although with 12-fold and 72-fold higher IC50s, respectively. To investigate if the inhibition of SIK2 may contribute to the effectiveness of nintedanib in treating lung fibrosis, mice with kinase-inactive knockin mutations were tested using a model of bleomycin-induced lung fibrosis. We found that loss of SIK2 activity protects against bleomycin-induced fibrosis, as judged by collagen deposition and histological scoring. Loss of both SIK1 and SIK2 activity had a similar effect to loss of SIK2 activity. Total SIK3 knockout mice have a developmental phenotype making them unsuitable for analysis in this model; however, we determined that conditional knockout of SIK3 in the immune system did not affect bleomycin-induced lung fibrosis. Together, these results suggest that SIK2 is a potential drug target for the treatment of lung fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Injury , Animals , Mice , Bleomycin , Fibrosis , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Lung/metabolism , Lung Injury/chemically induced , Lung Injury/genetics , Lung Injury/metabolism , Protein Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Disease Models, Animal
2.
Sci Rep ; 11(1): 21550, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732767

ABSTRACT

Salt Inducible Kinases (SIKs), of which there are 3 isoforms, are established to play roles in innate immunity, metabolic control and neuronal function, but their role in adaptive immunity is unknown. To address this gap, we used a combination of SIK knockout and kinase-inactive knock-in mice. The combined loss of SIK1 and SIK2 activity did not block T cell development. Conditional knockout of SIK3 in haemopoietic cells, driven by a Vav-iCre transgene, resulted in a moderate reduction in the numbers of peripheral T cells, but normal B cell numbers. Constitutive knockout of SIK2 combined with conditional knockout of SIK3 in the haemopoietic cells resulted in a severe reduction in peripheral T cells without reducing B cell number. A similar effect was seen when SIK3 deletion was driven via CD4-Cre transgene to delete at the DP stage of T cell development. Analysis of the SIK2/3 Vav-iCre mice showed that thymocyte number was greatly reduced, but development was not blocked completely as indicated by the presence of low numbers CD4 and CD8 single positive cells. SIK2 and SIK3 were not required for rearrangement of the TCRß locus, or for low level cell surface expression of the TCR complex on the surface of CD4/CD8 double positive thymocytes. In the absence of both SIK2 and SIK3, progression to mature single positive cells was greatly reduced, suggesting a defect in negative and/or positive selection in the thymus. In agreement with an effect on negative selection, increased apoptosis was seen in thymic TCRbeta high/CD5 positive cells from SIK2/3 knockout mice. Together, these results show an important role for SIK2 and SIK3 in thymic T cell development.


Subject(s)
Protein Serine-Threonine Kinases/genetics , T-Lymphocytes/cytology , Alleles , Animals , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Proliferation , Cell Separation , Exons , Female , Flow Cytometry , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Isoforms , Signal Transduction/physiology , Spleen/metabolism , Thymocytes/cytology , Transgenes
3.
Biochem J ; 478(7): 1377-1397, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33861845

ABSTRACT

The salt-inducible kinases, SIK1, SIK2 and SIK3, most closely resemble the AMP-activated protein kinase (AMPK) and other AMPK-related kinases, and like these family members they require phosphorylation by LKB1 to be catalytically active. However, unlike other AMPK-related kinases they are phosphorylated by cyclic AMP-dependent protein kinase (PKA), which promotes their binding to 14-3-3 proteins and inactivation. The most well-established substrates of the SIKs are the CREB-regulated transcriptional co-activators (CRTCs), and the Class 2a histone deacetylases (HDAC4/5/7/9). Phosphorylation by SIKs promotes the translocation of CRTCs and Class 2a HDACs to the cytoplasm and their binding to 14-3-3s, preventing them from regulating their nuclear binding partners, the transcription factors CREB and MEF2. This process is reversed by PKA-dependent inactivation of the SIKs leading to dephosphorylation of CRTCs and Class 2a HDACs and their re-entry into the nucleus. Through the reversible regulation of these substrates and others that have not yet been identified, the SIKs regulate many physiological processes ranging from innate immunity, circadian rhythms and bone formation, to skin pigmentation and metabolism. This review summarises current knowledge of the SIKs and the evidence underpinning these findings, and discusses the therapeutic potential of SIK inhibitors for the treatment of disease.


Subject(s)
Circadian Rhythm , Mental Disorders/drug therapy , Neoplasms/drug therapy , Osteoporosis/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Humans , Mental Disorders/enzymology , Mental Disorders/pathology , Neoplasms/enzymology , Neoplasms/pathology , Osteoporosis/enzymology , Osteoporosis/pathology
4.
Biochemistry ; 60(19): 1533-1551, 2021 05 18.
Article in English | MEDLINE | ID: mdl-33913706

ABSTRACT

There are three RhoGDIs in mammalian cells, which were initially defined as negative regulators of Rho family small GTPases. However, it is now accepted that RhoGDIs not only maintain small GTPases in their inactive GDP-bound form but also act as chaperones for small GTPases, targeting them to specific intracellular membranes and protecting them from degradation. Studies to date with RhoGDIs have usually focused on the interactions between the "typical" or "classical" small GTPases, such as the Rho, Rac, and Cdc42 subfamily members, and either the widely expressed RhoGDI-1 or the hematopoietic-specific RhoGDI-2. Less is known about the third member of the family, RhoGDI-3 and its interacting partners. RhoGDI-3 has a unique N-terminal extension and is found to localize in both the cytoplasm and the Golgi. RhoGDI-3 has been shown to target RhoB and RhoG to endomembranes. In order to facilitate a more thorough understanding of RhoGDI function, we undertook a systematic study to determine all possible Rho family small GTPases that interact with the RhoGDIs. RhoGDI-1 and RhoGDI-2 were found to have relatively restricted activity, mainly binding members of the Rho and Rac subfamilies. RhoGDI-3 displayed wider specificity, interacting with the members of Rho, Rac, and Cdc42 subfamilies but also forming complexes with "atypical" small Rho GTPases such as Wrch2/RhoV, Rnd2, Miro2, and RhoH. Levels of RhoA, RhoB, RhoC, Rac1, RhoH, and Wrch2/RhoV bound to GTP were found to decrease following coexpression with RhoGDI-3, confirming its role as a negative regulator of these small Rho GTPases.


Subject(s)
rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , HEK293 Cells , Humans , Monomeric GTP-Binding Proteins/metabolism , Protein Binding , rho GTP-Binding Proteins/chemistry , rho Guanine Nucleotide Dissociation Inhibitor alpha/physiology , rho Guanine Nucleotide Dissociation Inhibitor beta/physiology , rho Guanine Nucleotide Dissociation Inhibitor gamma/physiology , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/physiology
5.
J Biol Chem ; 296: 100428, 2021.
Article in English | MEDLINE | ID: mdl-33600797

ABSTRACT

Cytokines and chemokines are important regulators of airway hyper-responsiveness, immune cell infiltration, and inflammation and are produced when mast cells are stimulated with interleukin-33 (IL-33). Here, we establish that the salt-inducible kinases (SIKs) are required for the IL-33-stimulated transcription of il13, gm-csf and tnf and hence the production of these cytokines. The IL-33-stimulated secretion of IL-13, granulocyte-macrophage colony stimulating factor, and tumor necrosis factor was strongly reduced in fetal liver-derived mast cells from mice expressing a kinase-inactive mutant of SIK3 and abolished in cells expressing kinase-inactive mutants of SIK2 and SIK3. The IL-33-dependent secretion of these cytokines and several chemokines was also abolished in SIK2/3 double knock-out bone marrow-derived mast cells (BMMC), reduced in SIK3 KO cells but little affected in BMMC expressing kinase-inactive mutants of SIK1 and SIK2 or lacking SIK2 expression. In SIK2 knock-out BMMC, the expression of SIK3 was greatly increased. Our studies identify essential roles for SIK2 and SIK3 in producing inflammatory mediators that trigger airway inflammation. The effects of SIKs were independent of IκB kinase ß, IκB kinase ß-mediated NF-κB-dependent gene transcription, and activation of the mitogen-activated protein kinase family members p38α and c-jun N-terminal kinases. Our results suggest that dual inhibitors of SIK2 and SIK3 may have therapeutic potential for the treatment of mast cell-driven diseases.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Animals , Chemokines , Cytokines , Female , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , I-kappa B Kinase/metabolism , Inflammation Mediators/metabolism , Interleukin-33/genetics , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mast Cells/metabolism , Mast Cells/physiology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , NF-kappaB-Inducing Kinase
6.
Cell Death Dis ; 11(1): 49, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31969556

ABSTRACT

The signalling pathways initiated by members of the transforming growth factor-ß (TGFß) family of cytokines control many metazoan cellular processes, including proliferation and differentiation, epithelial-mesenchymal transition (EMT) and apoptosis. TGFß signalling is therefore strictly regulated to ensure appropriate context-dependent physiological responses. In an attempt to identify novel regulatory components of the TGFß signalling pathway, we performed a pharmacological screen by using a cell line engineered to report the endogenous transcription of the TGFß-responsive target gene PAI-1. The screen revealed that small molecule inhibitors of salt-inducible kinases (SIKs) attenuate TGFß-mediated transcription of PAI-1 without affecting receptor-mediated SMAD phosphorylation, SMAD complex formation or nuclear translocation. We provide evidence that genetic inactivation of SIK isoforms also attenuates TGFß-dependent transcriptional responses. Pharmacological inhibition of SIKs by using multiple small-molecule inhibitors potentiated apoptotic cell death induced by TGFß stimulation. Our data therefore provide evidence for a novel function of SIKs in modulating TGFß-mediated transcriptional and cellular responses.


Subject(s)
Apoptosis/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Serpin E2/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/drug effects , Cytoplasm/genetics , Cytoplasm/metabolism , Gene Knockout Techniques , Gene Silencing , Humans , Indans/pharmacology , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Pyrimidines/pharmacology , Serpin E2/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Smad Proteins/metabolism
7.
PLoS One ; 12(9): e0184907, 2017.
Article in English | MEDLINE | ID: mdl-28931068

ABSTRACT

Disruption of protein folding in the endoplasmic reticulum (ER) causes ER stress. Activation of the unfolded protein response (UPR) acts to restore protein homeostasis or, if ER stress is severe or persistent, drive apoptosis, which is thought to proceed through the cell intrinsic, mitochondrial pathway. Indeed, cells that lack the key executioner proteins BAX and BAK are protected from ER stress-induced apoptosis. Here we show that chronic ER stress causes the progressive inhibition of the extracellular signal-regulated kinase (ERK1/2) signalling pathway. This is causally related to ER stress since reactivation of ERK1/2 can protect cells from ER stress-induced apoptosis whilst ERK1/2 pathway inhibition sensitises cells to ER stress. Furthermore, cancer cell lines harbouring constitutively active BRAFV600E are addicted to ERK1/2 signalling for protection against ER stress-induced cell death. ERK1/2 signalling normally represses the pro-death proteins BIM, BMF and PUMA and it has been proposed that ER stress induces BIM-dependent cell death. We found no evidence that ER stress increased the expression of these proteins; furthermore, BIM was not required for ER stress-induced death. Rather, ER stress caused the PERK-dependent inhibition of cap-dependent mRNA translation and the progressive loss of pro-survival proteins including BCL2, BCLXL and MCL1. Despite these observations, neither ERK1/2 activation nor loss of BAX/BAK could confer long-term clonogenic survival to cells exposed to ER stress. Thus, ER stress induces cell death by at least two biochemically and genetically distinct pathways: a classical BAX/BAK-dependent apoptotic response that can be inhibited by ERK1/2 signalling and an alternative ERK1/2- and BAX/BAK-independent cell death pathway.


Subject(s)
Apoptosis , Colorectal Neoplasms/pathology , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Colony-Forming Units Assay , Colorectal Neoplasms/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mitochondria/metabolism , Mitochondria/pathology , Signal Transduction , Tumor Cells, Cultured , Unfolded Protein Response
8.
Biochem J ; 474(4): 521-537, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27920213

ABSTRACT

The salt-inducible kinases (SIKs) control a novel molecular switch regulating macrophage polarization. Pharmacological inhibition of the SIKs induces a macrophage phenotype characterized by the secretion of high levels of anti-inflammatory cytokines, including interleukin (IL)-10, and the secretion of very low levels of pro-inflammatory cytokines, such as tumour necrosis factor α. The SIKs, therefore, represent attractive new drug targets for the treatment of macrophage-driven diseases, but which of the three isoforms, SIK1, SIK2 or SIK3, would be appropriate to target remains unknown. To address this question, we developed knock-in (KI) mice for SIK1, SIK2 and SIK3, in which we introduced a mutation that renders the enzymes catalytically inactive. Characterization of primary macrophages from the single and double KI mice established that all three SIK isoforms, and in particular SIK2 and SIK3, contribute to macrophage polarization. Moreover, we discovered that inhibition of SIK2 and SIK3 during macrophage differentiation greatly enhanced the production of IL-10 compared with their inhibition in mature macrophages. Interestingly, macrophages differentiated in the presence of SIK inhibitors, MRT199665 and HG-9-91-01, still produced very large amounts of IL-10, but very low levels of pro-inflammatory cytokines, even after the SIKs had been reactivated by removal of the drugs. Our data highlight an integral role for SIK2 and SIK3 in innate immunity by preventing the differentiation of macrophages into a potent and stable anti-inflammatory phenotype.


Subject(s)
Immunity, Innate , Macrophages/immunology , Protein Kinases/immunology , Protein Serine-Threonine Kinases/immunology , Animals , Cell Differentiation/drug effects , Gene Expression , Gene Knock-In Techniques , Indans/pharmacology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-12 Subunit p40/biosynthesis , Interleukin-12 Subunit p40/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Phenylurea Compounds/pharmacology , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyrimidines/pharmacology , Transgenes , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
9.
Biochim Biophys Acta ; 1843(10): 2150-63, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24440275

ABSTRACT

Perturbations in endoplasmic reticulum (ER) homeostasis, including depletion of Ca(2+) or altered redox status, induce ER stress due to protein accumulation, misfolding and oxidation. This activates the unfolded protein response (UPR) to re-establish the balance between ER protein folding capacity and protein load, resulting in cell survival or, following chronic ER stress, promotes cell death. The mechanisms for the transition between adaptation to ER stress and ER stress-induced cell death are still being understood. However, the identification of numerous points of cross-talk between the UPR and mitogen-activated protein kinase (MAPK) signalling pathways may contribute to our understanding of the consequences of ER stress. Indeed, the MAPK signalling network is known to regulate cell cycle progression and cell survival or death responses following a variety of stresses. In this article, we review UPR signalling and the activation of MAPK signalling pathways in response to ER stress. In addition, we highlight components of the UPR that are modulated in response to MAPK signalling and the consequences of this cross-talk. We also describe several diseases, including cancer, type II diabetes and retinal degeneration, where activation of the UPR and MAPK signalling contribute to disease progression and highlight potential avenues for therapeutic intervention. This article is part of a Special Issue entitled: Calcium Signaling In Health and Disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Mitogen-Activated Protein Kinases/genetics , Signal Transduction , Unfolded Protein Response/genetics , Calcium/metabolism , Cell Cycle , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...