Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 19(1): 68, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34193159

ABSTRACT

BACKGROUND: The oncogenic PIM kinases and the tumor-suppressive LKB1 kinase have both been implicated in the regulation of cell growth and metabolism, albeit in opposite directions. Here we investigated whether these kinases interact with each other to influence AMPK activation and tumorigenic growth of prostate and breast cancer cells. METHODS: We first determined how PIM and LKB1 kinases affect AMPK phosphorylation levels. We then used in vitro kinase assays to demonstrate that LKB1 is phosphorylated by PIM kinases, and site-directed mutagenesis to identify the PIM target sites in LKB1. The cellular functions of PIM and LKB1 kinases were evaluated using either pan-PIM inhibitors or CRISPR/Cas9 genomic editing, with which all three PIM family members and/or LKB1 were knocked out from PC3 prostate and MCF7 breast cancer cell lines. In addition to cell proliferation assays, we examined the effects of PIM and/or LKB1 loss on tumor growth using the chick embryo chorioallantoic membrane (CAM) xenograft model. RESULTS: We provide both genetic and pharmacological evidence to demonstrate that inhibition of PIM expression or activity increases phosphorylation of AMPK at Thr172 in both PC3 and MCF7 cells, but not in their derivatives lacking LKB1. This is explained by our observation that all three PIM family kinases can phosphorylate LKB1 at Ser334. Wild-type LKB1, but not its phosphodeficient derivative, can restore PIM inhibitor-induced AMPK phosphorylation in LKB1 knock-out cells. In the CAM model, loss of LKB1 enhances tumorigenicity of PC3 xenografts, while cells lacking both LKB1 and PIMs exhibit slower proliferation rates and form smaller tumors. CONCLUSION: PIM kinases are novel negative regulators of LKB1 that affect AMPK activity in an LKB1-dependent fashion. The impairment of cell proliferation and tumor growth in cells lacking both LKB1 and PIMs indicates that these kinases possess a shared signaling role in the context of cancer. These data also suggest that PIM inhibitors may be a rational therapeutic option for LKB1-deficient tumors. Video Abstract.


Subject(s)
AMP-Activated Protein Kinase Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Proto-Oncogene Proteins c-pim-1/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Humans , Phosphorylation , Protein Binding , Substrate Specificity
2.
J Biol Chem ; 296: 100593, 2021.
Article in English | MEDLINE | ID: mdl-33775697

ABSTRACT

Dysregulation of the developmentally important Notch signaling pathway is implicated in several types of cancer, including breast cancer. However, the specific roles and regulation of the four different Notch receptors have remained elusive. We have previously reported that the oncogenic PIM kinases phosphorylate Notch1 and Notch3. Phosphorylation of Notch1 within the second nuclear localization sequence of its intracellular domain (ICD) enhances its transcriptional activity and tumorigenicity. In this study, we analyzed Notch3 phosphorylation and its functional impact. Unexpectedly, we observed that the PIM target sites are not conserved between Notch1 and Notch3. Notch3 ICD (N3ICD) is phosphorylated within a domain, which is essential for formation of a transcriptionally active complex with the DNA-binding protein CSL. Through molecular modeling, X-ray crystallography, and isothermal titration calorimetry, we demonstrate that phosphorylation of N3ICD sterically hinders its interaction with CSL and thereby inhibits its CSL-dependent transcriptional activity. Surprisingly however, phosphorylated N3ICD still maintains tumorigenic potential in breast cancer cells under estrogenic conditions, which support PIM expression. Taken together, our data indicate that PIM kinases modulate the signaling output of different Notch paralogs by targeting distinct protein domains and thereby promote breast cancer tumorigenesis via both CSL-dependent and CSL-independent mechanisms.


Subject(s)
Breast Neoplasms/pathology , Carcinogenesis , Proto-Oncogene Proteins c-pim-1/metabolism , Receptor, Notch3/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Models, Molecular , Muscle Proteins/metabolism , Phosphorylation , Protein Domains , Receptor, Notch3/chemistry
3.
Cell Commun Signal ; 18(1): 121, 2020 08 08.
Article in English | MEDLINE | ID: mdl-32771000

ABSTRACT

BACKGROUND: The PIM family kinases promote cancer cell survival and motility as well as metastatic growth in various types of cancer. We have previously identified several PIM substrates, which support cancer cell migration and invasiveness. However, none of them are known to regulate cellular movements by directly interacting with the actin cytoskeleton. Here we have studied the phosphorylation-dependent effects of PIM1 on actin capping proteins, which bind as heterodimers to the fast-growing actin filament ends and stabilize them. METHODS: Based on a phosphoproteomics screen for novel PIM substrates, we have used kinase assays and fluorescence-based imaging techniques to validate actin capping proteins as PIM1 substrates and interaction partners. We have analysed the functional consequences of capping protein phosphorylation on cell migration and adhesion by using wound healing and real-time impedance-based assays. We have also investigated phosphorylation-dependent effects on actin polymerization by analysing the protective role of capping protein phosphomutants in actin disassembly assays. RESULTS: We have identified capping proteins CAPZA1 and CAPZB2 as PIM1 substrates, and shown that phosphorylation of either of them leads to increased adhesion and migration of human prostate cancer cells. Phosphorylation also reduces the ability of the capping proteins to protect polymerized actin from disassembly. CONCLUSIONS: Our data suggest that PIM kinases are able to induce changes in actin dynamics to support cell adhesion and movement. Thus, we have identified a novel mechanism through which PIM kinases enhance motility and metastatic behaviour of cancer cells. Video abstract.


Subject(s)
Actin Capping Proteins/metabolism , Cell Movement , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-pim-1/metabolism , Actins/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Surface Extensions/metabolism , Cytoplasm/metabolism , Humans , Male , Mice , Phosphorylation , Protein Multimerization , Protein Subunits/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors
4.
Cell Commun Signal ; 17(1): 148, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31730483

ABSTRACT

BACKGROUND: Progression of prostate cancer from benign local tumors to metastatic carcinomas is a multistep process. Here we have investigated the signaling pathways that support migration and invasion of prostate cancer cells, focusing on the role of the NFATC1 transcription factor and its post-translational modifications. We have previously identified NFATC1 as a substrate for the PIM1 kinase and shown that PIM1-dependent phosphorylation increases NFATC1 activity without affecting its subcellular localization. Both PIM kinases and NFATC1 have been reported to promote cancer cell migration, invasion and angiogenesis, but it has remained unclear whether the effects of NFATC1 are phosphorylation-dependent and which downstream targets are involved. METHODS: We used mass spectrometry to identify PIM1 phosphorylation target sites in NFATC1, and analysed their functional roles in three prostate cancer cell lines by comparing phosphodeficient mutants to wild-type NFATC1. We used luciferase assays to determine effects of phosphorylation on NFAT-dependent transcriptional activity, and migration and invasion assays to evaluate effects on cell motility. We also performed a microarray analysis to identify novel PIM1/NFATC1 targets, and validated one of them with both cellular expression analyses and in silico in clinical prostate cancer data sets. RESULTS: Here we have identified ten PIM1 target sites in NFATC1 and found that prevention of their phosphorylation significantly decreases the transcriptional activity as well as the pro-migratory and pro-invasive effects of NFATC1 in prostate cancer cells. We observed that also PIM2 and PIM3 can phosphorylate NFATC1, and identified several novel putative PIM1/NFATC1 target genes. These include the ITGA5 integrin, which is differentially expressed in the presence of wild-type versus phosphorylation-deficient NFATC1, and which is coexpressed with PIM1 and NFATC1 in clinical prostate cancer specimens. CONCLUSIONS: Based on our data, phosphorylation of PIM1 target sites stimulates NFATC1 activity and enhances its ability to promote prostate cancer cell migration and invasion. Therefore, inhibition of the interplay between PIM kinases and NFATC1 may have therapeutic implications for patients with metastatic forms of cancer.


Subject(s)
Cell Movement , NFATC Transcription Factors/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Cell Proliferation , Humans , Male , Mass Spectrometry , PC-3 Cells , Phosphorylation , Prostatic Neoplasms/pathology , Signal Transduction , Tumor Cells, Cultured
5.
eNeuro ; 6(4)2019.
Article in English | MEDLINE | ID: mdl-31387876

ABSTRACT

The mammalian PIM family of serine/threonine kinases regulate several cellular functions, such as cell survival and motility. Because PIM expression is observed in sensory organs, such as olfactory epithelium, we now wanted to explore the physiological roles of PIM kinases there. As our model organism, we used the Caenorhabditis elegans nematodes, which express two PIM-related kinases, PRK-1 and PRK-2. We demonstrated PRKs to be true PIM orthologs with similar substrate specificity as well as sensitivity to PIM-inhibitory compounds. When we analyzed the effects of pan-PIM inhibitors on C. elegans sensory functions, we observed that PRK activity is selectively required to support olfactory sensations to volatile repellents and attractants sensed by AWB and AWCON neurons, respectively, but is dispensable for gustatory sensations. Analyses of prk-deficient mutant strains confirmed these findings and suggested that PRK-1, but not PRK-2 is responsible for the observed effects on olfaction. This regulatory role of PRK-1 is further supported by its observed expression in the head and tail neurons, including AWB and AWC neurons. Based on the evolutionary conservation of PIM-related kinases, our data may have implications in regulation of also mammalian olfaction.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Olfactory Receptor Neurons/enzymology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Smell/physiology , Amino Acid Sequence , Animals , Evolution, Molecular , Odorants , Species Specificity
6.
Int J Biochem Cell Biol ; 93: 74-85, 2017 12.
Article in English | MEDLINE | ID: mdl-29108877

ABSTRACT

PIM kinases are oncogenic serine/threonine kinases, the expression and activities of which are tightly regulated in normal tissues, but upregulated in many types of human malignancies, including both hematological and solid cancers. Since high PIM expression levels have been connected to cancer progression and poor patient survival, PIM kinases have become attractive targets for drug development. Many downstream targets have also been identified, through which PIM kinases promote cell survival, proliferation and metabolism. More recently, PIM kinases have been implicated in regulation of cell motility, which also plays an important role in tumor growth and cancer progression. This review summarizes effects of PIM kinases and their substrates especially on cancer cell migration, invasion and metastatic growth, based on data from cell-based assays, animal experiments and patients.


Subject(s)
Cell Movement , Cell Proliferation , Neoplasms/enzymology , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy , Proto-Oncogene Proteins c-pim-1/genetics
7.
Oncotarget ; 7(28): 43220-43238, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27281612

ABSTRACT

Tumorigenesis is a multistep process involving co-operation between several deregulated oncoproteins. In this study, we unravel previously unrecognized interactions and crosstalk between Pim kinases and the Notch signaling pathway, with implications for both breast and prostate cancer. We identify Notch1 and Notch3, but not Notch2, as novel Pim substrates and demonstrate that for Notch1, the serine residue 2152 is phosphorylated by all three Pim family kinases. This target site is located in the second nuclear localization sequence (NLS) of the Notch1 intracellular domain (N1ICD), and is shown to be important for both nuclear localization and transcriptional activity of N1ICD. Phosphorylation-dependent stimulation of Notch1 signaling promotes migration of prostate cancer cells, balances glucose metabolism in breast cancer cells, and supports in vivo growth of both types of cancer cells on chick embryo chorioallantoic membranes. Furthermore, Pim-induced growth of orthotopic prostate xenografts in mice is associated with enhanced nuclear Notch1 activity. Finally, simultaneous inhibition of Pim and Notch abrogates the cellular responses more efficiently than individual treatments, opening up new vistas for combinatorial cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Carcinogenesis/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-pim-1/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cell Movement , Chick Embryo , Female , Humans , MCF-7 Cells , Male , Mice , Phosphorylation , Receptor, Notch2/metabolism , Receptor, Notch3/metabolism , Serine/metabolism , Xenograft Model Antitumor Assays
8.
Exp Cell Res ; 342(2): 113-24, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26934497

ABSTRACT

The ability of cells to migrate and form metastases is one of the fatal hallmarks of cancer that can be conquered only with better understanding of the molecules and regulatory mechanisms involved. The oncogenic PIM kinases have been shown to support cancer cell survival and motility, but the PIM-regulated pathways stimulating cell migration and invasion are less well characterized than those affecting cell survival. Here we have identified the glycogen synthase kinase 3ß (GSK3B) and the forkhead box P3 (FOXP3) transcription factor as direct PIM targets, whose tumour-suppressive effects in prostate cancer cells are inhibited by PIM-induced phosphorylation, resulting in increased cell migration. Targeting GSK3B is also essential for the observed PIM-enhanced expression of the prostaglandin-endoperoxide synthase 2 (PTGS2), which is an important regulator of both cell migration and adhesion. Accordingly, selective inhibition of PIM activity not only reduces cell migration, but also affects integrin-mediated cell adhesion. Taken together, these data provide novel mechanistic insights on how and why patients with metastatic prostate cancer may benefit from therapies targeting PIM kinases, and how such approaches may also be applicable to inflammatory conditions.


Subject(s)
Adenocarcinoma/enzymology , Cell Movement , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-pim-1/physiology , Adenocarcinoma/pathology , Amino Acid Sequence , Cell Line, Tumor , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Male , Phosphorylation , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational , Protein Transport , Signal Transduction
9.
PLoS One ; 10(6): e0130340, 2015.
Article in English | MEDLINE | ID: mdl-26075720

ABSTRACT

BACKGROUND AND METHODS: Pim family proteins are oncogenic kinases implicated in several types of cancer and involved in regulation of cell proliferation, survival as well as motility. Here we have investigated the ability of Pim kinases to promote metastatic growth of prostate cancer cells in two xenograft models for human prostate cancer. We have also evaluated the efficacy of Pim-selective inhibitors to antagonize these effects. RESULTS: We show here that tumorigenic growth of both subcutaneously and orthotopically inoculated prostate cancer xenografts is enhanced by stable overexpression of either Pim-1 or Pim-3. Moreover, Pim-overexpressing orthotopic prostate tumors are highly invasive and able to migrate not only to the nearby prostate-draining lymph nodes, but also into the lungs to form metastases. When the xenografted mice are daily treated with the Pim-selective inhibitor DHPCC-9, both the volumes as well as the metastatic capacity of the tumors are drastically decreased. Interestingly, the Pim-promoted metastatic growth of the orthotopic xenografts is associated with enhanced angiogenesis and lymphangiogenesis. Furthermore, forced Pim expression also increases phosphorylation of the CXCR4 chemokine receptor, which may enable the tumor cells to migrate towards tissues such as the lungs that express the CXCL12 chemokine ligand. CONCLUSIONS: Our results indicate that Pim overexpression enhances the invasive properties of prostate cancer cells in vivo. These effects can be reduced by the Pim-selective inhibitor DHPCC-9, which can reach tumor tissues without serious side effects. Thus, Pim-targeting therapies with DHPCC-9-like compounds may help to prevent progression of local prostate carcinomas to fatally metastatic malignancies.


Subject(s)
Neoplasm Metastasis/pathology , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemokine CXCL12/metabolism , Heterografts , Humans , Lymphangiogenesis/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-pim-1/biosynthesis , Receptors, CXCR4/metabolism , Transplantation, Heterologous , Zebrafish
10.
PLoS One ; 8(2): e55409, 2013.
Article in English | MEDLINE | ID: mdl-23405147

ABSTRACT

Oncogenic Pim family kinases are often overexpressed in human hematopoietic malignancies as well as in solid tumours. These kinases contribute to tumorigenesis by promoting cell survival and by enhancing resistance against chemotherapy and radiation therapy. Furthermore, we have recently shown that they increase the metastatic potential of adherent cancer cells. Here we describe identification of tricyclic benzo[cd]azulenes and their derivatives as effective and selective inhibitors of Pim kinases. These compounds inhibit Pim autophosphorylation and abrogate the anti-apoptotic effects of Pim kinases. They also reduce cancer cell motility and suppress proliferation of lymphoblastoid cell lines infected and immortalized by the Epstein-Barr virus. Thus, these novel Pim-selective inhibitors provide promising compounds for both research and therapeutic purposes.


Subject(s)
Azulenes/pharmacology , Cell Transformation, Viral/drug effects , Herpesvirus 4, Human/drug effects , Myeloid Cells/drug effects , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Carcinogenesis/drug effects , Cell Line, Transformed , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Herpesvirus 4, Human/metabolism , Humans , Mice , Myeloid Cells/virology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-pim-1/metabolism
11.
Eur J Med Chem ; 50: 304-10, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22386260

ABSTRACT

We have previously demonstrated that pyrrolo[2,3-a]carbazole-3-carbaldehydes are potent Pim kinase inhibitors with in vitro antiproliferative activities. In the present study, we report the synthesis of new pyrrolocarbazoles substituted at the N-10 position. When their ability to inhibit Pim kinase activities were evaluated in in vitro assays, we observed that this nitrogen atom can be substituted without loss of Pim-1 and Pim-3 inhibitory potencies. Moreover, when we added a fluorescent dansyl group (compound 13), we were able to show that 13 penetrates the plasma membrane and enters the cytoplasm.


Subject(s)
Alkynes/chemistry , Azides/chemistry , Carbazoles/chemical synthesis , Copper/pharmacology , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Carbazoles/pharmacology , Catalysis , Cell Membrane/metabolism , Cytoplasm/metabolism , Humans , Male , Molecular Structure , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/metabolism , Structure-Activity Relationship , Subcellular Fractions , Tumor Cells, Cultured
12.
Mol Cancer ; 9: 279, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20958956

ABSTRACT

BACKGROUND: Pim family kinases are small constitutively active serine/threonine-specific kinases, elevated levels of which have been detected in human hematopoietic malignancies as well as in solid tumours. While we and others have previously shown that the oncogenic Pim kinases stimulate survival of hematopoietic cells, we now examined their putative role in regulating motility of adherent cancer cells. For this purpose, we inhibited Pim kinase activity using a small molecule compound, 1,10-dihydropyrrolo[2,3-a]carbazole-3-carbaldehyde (DHPCC-9), which we had recently identified as a potent and selective inhibitor for all Pim family members. RESULTS: We now demonstrate that the Pim kinase inhibitor DHPCC-9 is very effective also in cell-based assays. DHPCC-9 impairs the anti-apoptotic effects of Pim-1 in cytokine-deprived myeloid cells and inhibits intracellular phosphorylation of Pim substrates such as Bad. Moreover, DHPCC-9 slows down migration and invasion of cancer cells derived from either prostate cancer or squamocellular carcinoma patients. Silencing of Pim expression reduces cell motility, while Pim overexpression enhances it, strongly suggesting that the observed effects of DHPCC-9 are dependent on Pim kinase activity. Interestingly, DHPCC-9 also abrogates NFATc-dependent migration of cancer cells, implying that NFATc factors mediate at least part of the pro-migratory effects of Pim kinases. CONCLUSIONS: Altogether, our data indicate that DHPCC-9 is not only a powerful tool to investigate physiological effects of the oncogenic Pim family kinases, but also an attractive molecule for drug development to inhibit invasiveness of Pim-overexpressing cancer cells.


Subject(s)
Cell Movement/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/genetics , Humans , Mice , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...