Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Appl Immunohistochem Mol Morphol ; 29(10): 734-740, 2021.
Article in English | MEDLINE | ID: mdl-34261974

ABSTRACT

The aim of our study was to analyze the expressions of nuclear factor of activated T cells (NFAT)-related substances including long noncoding RNA NRON which participates in pathophysiology of Sjögren's syndrome (SS), and to assess the histologic findings in individuals with SS. In this study, the expressions of NRON, NFATc1, CD3/CD4, and proviral integration site for Moloney murine leukemia virus (PIM)-1 were examined by in situ hybridization, immunohistochemical analysis, and immunofluorescence in labial salivary glands (LSGs) obtained from 16 patients with SS and five controls. The microcell count method has been applied to calculate the NFATc1-positive area/infiltrating cell area in LSGs, and we compared those results to the infiltrating cell area, focus score, serum immunoglobulin G, and the European League Against Rheumatism Sjögren's Syndrome Disease Activity Index. The NRON expression in the nuclei of cell-infiltration lesions of the SS patients were prominent. The NFATc1 expression was strong in the cytoplasm of infiltrating mononuclear cells and weak in ducts of both SS and controls. In SS, the NFATc1-positive area/infiltrating cell area was positively correlated with the infiltrating cell area and focus score. CD3/CD4 was expressed in infiltrating mononuclear cells, and PIM-1 colocalized with NFATc1 in the cytoplasm. These results suggest NRON along with NFATc1/PIM-1 in SS LSGs might participate in SS pathophysiology.


Subject(s)
Gene Expression Regulation , In Situ Hybridization , NFATC Transcription Factors/biosynthesis , Proto-Oncogene Proteins c-pim-1/biosynthesis , RNA, Long Noncoding/biosynthesis , Salivary Glands, Minor/metabolism , Sjogren's Syndrome/metabolism , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Salivary Glands, Minor/pathology , Sjogren's Syndrome/pathology
2.
Cells ; 9(9)2020 08 31.
Article in English | MEDLINE | ID: mdl-32878131

ABSTRACT

Enhancing cardiomyocyte survival is crucial to blunt deterioration of myocardial structure and function following pathological damage. PIM1 (Proviral Insertion site in Murine leukemia virus (PIM) kinase 1) is a cardioprotective serine threonine kinase that promotes cardiomyocyte survival and antagonizes senescence through multiple concurrent molecular signaling cascades. In hematopoietic stem cells, PIM1 interacts with the receptor tyrosine kinase c-Kit upstream of the ERK (Extracellular signal-Regulated Kinase) and Akt signaling pathways involved in cell proliferation and survival. The relationship between PIM1 and c-Kit activity has not been explored in the myocardial context. This study delineates the interaction between PIM1 and c-Kit leading to enhanced protection of cardiomyocytes from stress. Elevated c-Kit expression is induced in isolated cardiomyocytes from mice with cardiac-specific overexpression of PIM1. Co-immunoprecipitation and proximity ligation assay reveal protein-protein interaction between PIM1 and c-Kit. Following treatment with Stem Cell Factor, PIM1-overexpressing cardiomyocytes display elevated ERK activity consistent with c-Kit receptor activation. Functionally, elevated c-Kit expression confers enhanced protection against oxidative stress in vitro. This study identifies the mechanistic relationship between PIM1 and c-Kit in cardiomyocytes, demonstrating another facet of cardioprotection regulated by PIM1 kinase.


Subject(s)
Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Humans , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-pim-1/biosynthesis , Proto-Oncogene Proteins c-pim-1/genetics , Up-Regulation
3.
Neurosci Lett ; 709: 134375, 2019 09 14.
Article in English | MEDLINE | ID: mdl-31349016

ABSTRACT

Neuropathic pain induced by peripheral nerve injury is a complex and chronic state that is accompanied by poor quality of life. However, whether PIM1 (proviral integration site 1) contributes to the development of nociceptive hypersensitivity induced by nerve injury remains unknown. The present study was designed to investigate the effects of PIM1 on spinal nerve ligation (SNL) induced pain hypersensitivity. Here, we found that PIM1 positive neurons in the dorsal root ganglion (DRG) were colocalized with nociceptive neuronal markers CGRP, IB4 and substance P and were upregulated after SNL surgery. Knockdown PIM1 in the DRG by AAV5-shPIM1 alleviated SNL-induced pain hypersensitivity. In neuroblastoma cells (neuro-2a), PIM1 regulated the expression of CXCR4 phosphorylated at ser339 (pCXCR4) as well as the CXCL12/CXCR4 pathway. In the DRG tissues, we found that PIM1 was co-expressed with CXCR4, and knockdown of PIM1 attenuated pCXCR4 (ser339) protein expression but had little effect on total CXCR4 protein expression after SNL surgery. These findings suggest that PIM1 contributes to nerve injury-induced nociceptive hypersensitivity. Based on these findings and the characteristics of PIM1, we speculate that PIM1 might be a viable therapeutic target for the treatment of neuropathic pain in the near future.


Subject(s)
Ganglia, Spinal/metabolism , Neuralgia/metabolism , Peripheral Nerve Injuries/metabolism , Proto-Oncogene Proteins c-pim-1/biosynthesis , Animals , Cells, Cultured , Ganglia, Spinal/chemistry , Male , Mice , Mice, Inbred C57BL , Neuralgia/prevention & control , Peripheral Nerve Injuries/prevention & control , Proto-Oncogene Proteins c-pim-1/analysis
4.
Leuk Res ; 83: 106166, 2019 08.
Article in English | MEDLINE | ID: mdl-31203104

ABSTRACT

About ˜80% of mast cell neoplasm patients harbor the c-Kit activating mutation D816 V, which is associated with c-Kit inhibitor resistance and poor prognosis. However, the molecular basis for these effects is not fully known. To address this issue, in this study we screened molecules whose expression is altered by KIT D816 V mutation and found that Pim kinases were overexpressed in D816V-mutant neoplastic mast cells. This was accompanied by upregulation of signal transducer and activator of transcription (STAT) and mammalian target of rapamycin (mTOR) and downregulation of Akt and extracellular signal-regulated kinase (ERK1/2). Activated Pim kinases promoted the survival of D816 V cells by maintaining mTOR and p70S6K activation even under nutrient starvation. Conversely, cell proliferation was suppressed by inhibiting Pim kinases. The mRNA level of C-X-C chemokine receptor type 4 (CXCR4) was about 2-fold higher in D816 V cells; this was associated with a 2-fold increase in migratory capacity, which was modulated by Pim kinases. We also confirmed that upregulation of Pim kinases is a feature specific to cells with the D816 V mutation and is not observed in cells with the c-Kit activating N822 K mutation. These data suggest Pim kinases as a promising therapeutic target for the treatment of mast cell neoplasms with KIT D816 V mutation.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , Leukemia, Mast-Cell/enzymology , MAP Kinase Signaling System , Mutation, Missense , Proto-Oncogene Proteins c-kit/biosynthesis , Proto-Oncogene Proteins c-pim-1/biosynthesis , Up-Regulation , Amino Acid Substitution , Cell Line, Tumor , Cell Survival , Humans , Leukemia, Mast-Cell/genetics , Leukemia, Mast-Cell/pathology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-pim-1/genetics
5.
Anat Rec (Hoboken) ; 301(11): 1968-1976, 2018 11.
Article in English | MEDLINE | ID: mdl-30299595

ABSTRACT

Pim-1 is a proto-oncogene which has been discovered to involve in cell proliferation, differentiation, and survival. In this study, we observed the expression of Pim-1 in neonatal and adult rat retina and the changes in rat retina following optic nerve crush (ONC) in order to explore the relationship between Pim-1 and the survival of retinal ganglion cells (RGC). We discovered that Pim-1 was distributed mainly in retinal pigment epithelial cells (RPE) and retinal ganglion cell layer (GCL) in normal newborn rats, and it appeared in RPE, cone rod cell layer and GCL in normal adult rats by immunohistochemistry. Our double immunofluorescent staining of Pim-1 and γ-synuclein further confirmed that Pim-1 was localized in 80% of RGC. Moreover, we found that the amount of Pim-1 mRNA and protein in adult rat retina was transiently increased after ONC and then decreased 2 weeks after ONC, and the expression level was lower than that of neonatal rat retina under all conditions. We also discovered that Pim-1 expression in GCL detected by immunohistochemistry was upregulated at Day 1 and Day 3 after ONC, but downregulated at Day 14 after ONC when the survival of RGC was decreased and the apoptotic cells in GCL were increased by hematoxylin-eosin staining, immunohistochemistry, and TUNEL detection. We suggest that the overexpression of Pim-1 in the RGC is related to the optic nerve repair while the low expression of Pim-1 in RGC may be associated with apoptosis and weak intrinsic regeneration ability of RGC. Anat Rec, 301:1968-1976, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Nerve Crush/adverse effects , Optic Nerve Injuries/metabolism , Optic Nerve/metabolism , Proto-Oncogene Proteins c-pim-1/biosynthesis , Retina/metabolism , Animals , Animals, Newborn , Gene Expression , Male , Optic Nerve/chemistry , Optic Nerve Injuries/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Rats , Rats, Sprague-Dawley , Retina/chemistry , Retinal Ganglion Cells/chemistry , Retinal Ganglion Cells/metabolism
6.
J Biol Chem ; 292(20): 8207-8222, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28348080

ABSTRACT

Oxidative stress can induce cell dysfunction and lead to a broad range of degenerative alterations, including carcinogenesis, aging, and other oxidative stress-related conditions. To avoid undergoing carcinogenesis in response to oxidative stress, cells trigger a succession of checkpoint responses, including premature senescence and apoptosis. Increasing evidence indicates that H2O2, an important cause of oxidative stress, functions as an important physiological regulator of intracellular signaling pathways that participate in regulation of cell premature senescence and apoptosis. However, the precise mechanisms underlying this process remain to be studied extensively. In this study, we describe the importance of Pim-1 kinase in this checkpoint response to oxidative stress. Pim-1 binds to and phosphorylates the transcription factor high mobility group box transcription factor 1 (HBP1), activating it. H2O2 enhances the interaction between Pim-1 and HBP1 and promotes HBP1 accumulation. In turn, HBP1 rapidly and selectively up-regulates Pim-1 expression in H2O2-stimulated cells, thereby creating a Pim-1-HBP1 positive feedback loop that regulates H2O2-induced premature senescence and apoptosis. Furthermore, the Pim-1-HBP1 positive feedback loop exerts its effect by regulating the senescence markers DNMT1 and p16 and the apoptosis marker Bax. The Pim-1-HBP1 axis thus constitutes a novel checkpoint pathway critical for the inhibition of tumorigenesis.


Subject(s)
Apoptosis/drug effects , Cellular Senescence/drug effects , Gene Expression Regulation, Enzymologic/drug effects , High Mobility Group Proteins/metabolism , Hydrogen Peroxide/pharmacology , Proto-Oncogene Proteins c-pim-1/biosynthesis , Repressor Proteins/metabolism , Up-Regulation/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , HeLa Cells , High Mobility Group Proteins/genetics , Humans , Oxidative Stress/drug effects , Oxidative Stress/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Repressor Proteins/genetics
7.
Sci Rep ; 6: 38079, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27901106

ABSTRACT

The PIM family of serine/threonine kinases has three highly conserved isoforms (PIM1, PIM2 and PIM3). PIM proteins are regulated through transcription and stability by JAK/STAT pathways and are overexpressed in hematological malignancies and solid tumors. The PIM kinases possess weak oncogenic abilities, but enhance other genes or chemical carcinogens to induce tumors. We generated conditional transgenic mice that overexpress PIM1 or PIM2 in male reproductive organs and analyzed their contribution to tumorigenesis. We found an increase in alterations of sexual organs and hyperplasia in the transgenic mice correlating with inflammation. We also found that PIM1/2 are overexpressed in a subset of human male germ cells and prostate tumors correlating with inflammatory features and stem cell markers. Our data suggest that PIM1/2 kinase overexpression is a common feature of male reproductive organs tumors, which provoke tissue alterations and a large inflammatory response that may act synergistically during the process of tumorigenesis. There is also a correlation with markers of cancer stem cells, which may contribute to the therapy resistance found in tumors overexpressing PIM kinases.


Subject(s)
Biomarkers, Tumor , Prostatic Neoplasms , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-pim-1 , Proto-Oncogene Proteins , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Humans , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-pim-1/biosynthesis , Proto-Oncogene Proteins c-pim-1/genetics
8.
Oncotarget ; 7(41): 66525-66539, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27167190

ABSTRACT

P53 is frequently mutated in human tumors as a novel gain-of-function to promote tumor development. Although dimeric (M340Q/L344R) influences on tetramerisation on site-specific post-translational modifications of p53, it is not clear how dimeric (M340Q/L344R) plays a role during hepatocarcinogenesis. Herein, we reveal that P53 (N340Q/L344R) promotes hepatocarcinogenesis through upregulation of PKM2. Mechanistically, P53 (N340Q/L344R) forms complex with CUDR and the complex binds to the promoter regions of PKM2 which enhances the expression, phosphorylation of PKM2 and its polymer formation. Thereby, the polymer PKM2 (tetramer) binds to the eleventh threonine on histone H3 that increases the phosphorylation of the eleventh threonine on histone H3 (pH3T11). Furthermore, pH3T11 blocks HDAC3 binding to H3K9Ac that prevents H3K9Ac from deacetylation and stabilizes the H3K9Ac modification. On the other hand, it also decreased tri-methylation of histone H3 on the ninth lysine (H3K9me3) and increases one methylation of histone H3 on the ninth lysine (H3K9me1). Moreover, the combination of H3K9me1 and HP1 α forms more H3K9me3-HP1α complex which binds to the promoter region of Pim1, enhancing the expression of Pim1 that enhances the expression of TERT, oncogenic lncRNA HOTAIR and reduces the TERRA expression. Ultimately, P53 (N340Q/L344R) accerlerates the growth of liver cancer cells Hep3B by activating telomerase and prolonging telomere through the cascade of P53 (N340Q/L344R)-CUDR-PKM2-pH3T11- (H3K9me1-HP1α)-Pim1- (TERT-HOTAIR-TERRA). Understanding the novel functions of P53 (N340Q/L344R) will help in the development of new liver cancer therapeutic approaches that may be useful in a broad range of cancer types.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins c-pim-1/biosynthesis , RNA, Long Noncoding/genetics , Thyroid Hormones/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Chromobox Protein Homolog 5 , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Proto-Oncogene Proteins c-pim-1/genetics , Up-Regulation , Thyroid Hormone-Binding Proteins
9.
Cancer Sci ; 107(7): 899-907, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27088547

ABSTRACT

The PIM1 protein is an important regulator of cell proliferation, the cell cycle, apoptosis, and metabolism in various human cancers. MicroRNAs (miRNAs) are powerful post-transcriptional gene regulators that function through translational repression or transcript destabilization. Therefore, we aimed to identify whether a close relationship exists between PIM1 and miRNAs. PIM1 protein levels and mRNA levels were significantly upregulated in astrocytoma tissues, indicating the oncogenic role of PIM1 in astrocytoma. Further bioinformatics analysis indicated that miR-124-3p targeted the 3'-UTR of PIM1. We also observed an inverse correlation between the miR-124-3p levels and PIM1 protein or mRNA levels in astrocytoma samples. Next, we experimentally confirmed that miR-124-3p directly recognizes the 3'-UTR of the PIM1 transcript and regulates PIM1 expression at both the protein and mRNA levels. Furthermore, we examined the biological consequences of miR-124-3p targeting PIM1 in vitro. We showed that the repression of PIM1 in astrocytoma cancer cells by miR-124-3p suppressed proliferation, invasion, and aerobic glycolysis and promoted apoptosis. We observed that the restoration or inhibition of PIM1 activity resulted in effects that were similar to those induced by miR-124-3p inhibitors or mimics in cancer cells. Finally, overexpression of PIM1 rescued the inhibitory effects of miR-124-3p. In summary, these findings aid in understanding the tumor-suppressive role of miR-124-3p in astrocytoma pathogenesis through the inhibition of PIM1 translation.


Subject(s)
Apoptosis/genetics , Astrocytoma/genetics , Astrocytoma/pathology , Energy Metabolism/genetics , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , 3' Untranslated Regions/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Protein Biosynthesis , Proto-Oncogene Proteins c-pim-1/biosynthesis , Up-Regulation
10.
Appl Immunohistochem Mol Morphol ; 23(10): 717-23, 2015.
Article in English | MEDLINE | ID: mdl-26551340

ABSTRACT

BACKGROUND: The Provirus integrating site Moloney murine leukemia virus (Pim) family are proteins with serine/threonine kinase activity. Studies have demonstrated overexpression of Pims in cancer. To our knowledge, only a single study has examined Pim-1 in urothelial carcinoma. The aim of this investigation was to evaluate Pim-1, Pim-2, and Pim-3 in urothelial carcinoma and assess for expression that may contribute to disease progression and serve as a site for targeted therapy. METHODS: This retrospective study included 137 cases taken from specimens from the University of Utah, Department of Pathology (2008 to 2011). Tissue was stained with antibodies against Pim-1, Pim-2, and Pim-3. Cases were classified into 3 groups, based upon current World Health Organization criteria (invasive high-grade urothelial carcinoma [IHG] [n=84], noninvasive high-grade urothelial carcinoma/carcinoma in situ [n=32], and noninvasive low-grade urothelial carcinoma [NILG] [n=21]). Cases were scored and recorded as positive or negative on the basis of the percentage of cells with cytoplasmic and/or nuclear staining. RESULTS: NILG showed higher expression of Pim-1 (relative expression rate [RER]=2.28; 95% confidence interval [CI], 0.183-0.764) and Pim-3 (RER=3.06; 95% CI, 0.423-0.816) compared with other lesions. IHG had lower expression of Pim-1 (RER=0.31; 95% CI, 0.401-0.844) and Pim-3 (RER=0.354; 95% CI, 0.322-0.816) and noninvasive high-grade urothelial carcinoma (NIHG) demonstrated increased expression of Pim-1 and (RER=2.09; 95% CI, 0.124-0.739) and Pim-2 (RER=1.70; 95% CI, 0.151-0.591). At least 1 Pim kinase protein was expressed at the following rates: 49% in IHG, 66% in NIHG, and 76% in NILG. CONCLUSION: A high percentage of urothelial carcinomas express Pim kinases. Pim expression differs in NILG, NIHG, and IHG lesions.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-pim-1/biosynthesis , Urologic Neoplasms , Urothelium , Female , Humans , Male , Urologic Neoplasms/enzymology , Urologic Neoplasms/pathology , Urothelium/enzymology , Urothelium/pathology
11.
Cytokine ; 75(2): 365-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26142823

ABSTRACT

Cytokines play important roles in cardiac repair and regeneration. Recently, we demonstrated that interleukin (IL)-6 family cytokines induce the endothelial differentiation of Sca-1+ cardiac resident stem cells through STAT3/Pim-1 signaling pathway. In contrast, the biological functions of IL-12 family cytokines in heart remain to be elucidated, though they show structural homology with IL-6. In the present study, we examined the effects of IL-12 family cytokines on the transdifferentiation of cardiac Sca-1+ cells into cardiac cells. RT-PCR analyses revealed that IL-27 receptor α (IL-27Rα), but not IL-12R or IL-23R, was expressed in cardiac Sca-1+ cells. The transcript expression of IL-27 was elevated in murine hearts in cardiac injury models. Intriguingly, IL-27 stimulation for 14 days induced the endothelial cell (EC) marker genes, such as CD-31 and VE-cadherin. Immunoblot analyses clarified that IL-27 treatment rapidly phosphorylated STAT3. IL-27 upregulated the expression of Pim-1, but the overexpression of dominant negative STAT3 abrogated the induction of Pim-1 by IL-27. Finally, adenoviral transfection of dominant negative Pim-1 inhibited IL-27-induced EC differentiation of cardiac Sca-1+ cells. These findings demonstrated that IL-27 promoted the commitment of cardiac stem cells into the EC lineage, possibly leading to neovascularization as a novel biological function. IL-27 could not only regulate the inflammation but also contribute to the maintenance of the tissue homeostasis through stem cell differentiation at inflammatory sites.


Subject(s)
Interleukins/pharmacology , Myocardium/cytology , Proto-Oncogene Proteins c-pim-1/metabolism , STAT3 Transcription Factor/metabolism , Stem Cells/cytology , Animals , Cadherins/biosynthesis , Cell Differentiation/physiology , Cell Transdifferentiation/physiology , Cells, Cultured , Endothelial Cells/cytology , Heart Injuries/pathology , Interleukin-12/immunology , Interleukins/immunology , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Proto-Oncogene Proteins c-pim-1/biosynthesis , Receptors, Cytokine/biosynthesis , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-12/biosynthesis
12.
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
13.
PLoS Pathog ; 10(8): e1004304, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25121590

ABSTRACT

Epstein-Barr virus (EBV), a ubiquitous human herpesvirus, can latently infect the human population. EBV is associated with several types of malignancies originating from lymphoid and epithelial cell types. EBV latent antigen 3C (EBNA3C) is essential for EBV-induced immortalization of B-cells. The Moloney murine leukemia provirus integration site (PIM-1), which encodes an oncogenic serine/threonine kinase, is linked to several cellular functions involving cell survival, proliferation, differentiation, and apoptosis. Notably, enhanced expression of Pim-1 kinase is associated with numerous hematological and non-hematological malignancies. A higher expression level of Pim-1 kinase is associated with EBV infection, suggesting a crucial role for Pim-1 in EBV-induced tumorigenesis. We now demonstrate a molecular mechanism which reveals a direct role for EBNA3C in enhancing Pim-1 expression in EBV-infected primary B-cells. We also showed that EBNA3C is physically associated with Pim-1 through its amino-terminal domain, and also forms a molecular complex in B-cells. EBNA3C can stabilize Pim-1 through abrogation of the proteasome/Ubiquitin pathway. Our results demonstrate that EBNA3C enhances Pim-1 mediated phosphorylation of p21 at the Thr145 residue. EBNA3C also facilitated the nuclear localization of Pim-1, and promoted EBV transformed cell proliferation by altering Pim-1 mediated regulation of the activity of the cell-cycle inhibitor p21/WAF1. Our study demonstrated that EBNA3C significantly induces Pim-1 mediated proteosomal degradation of p21. A significant reduction in cell proliferation of EBV-transformed LCLs was observed upon stable knockdown of Pim-1. This study describes a critical role for the oncoprotein Pim-1 in EBV-mediated oncogenesis, as well as provides novel insights into oncogenic kinase-targeted therapeutic intervention of EBV-associated cancers.


Subject(s)
B-Lymphocytes/metabolism , Cell Transformation, Viral/physiology , Epstein-Barr Virus Nuclear Antigens/metabolism , Proto-Oncogene Proteins c-pim-1/biosynthesis , Blotting, Western , Cell Proliferation/physiology , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoprecipitation , Phosphorylation , Real-Time Polymerase Chain Reaction , Transfection
14.
Future Oncol ; 10(5): 865-76, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24799066

ABSTRACT

The PIM kinases represent a family of serine/threonine kinases, which is composed of three different members (PIM1, PIM2 and PIM3). Aberrant expression of PIM kinases is observed in variety of tumors, including pancreatic cancer. The PIM kinases play pivotal roles in the regulation of cell cycle, apoptosis, properties of stem cells, metabolism, autophagy, drug resistance and targeted therapy. The roles of PIM kinases in pancreatic cancer include the regulation of proliferation, apoptosis, cell cycle, formation, angiogenesis and prediction prognosis. Blocking the activities of PIM kinases could prevent pancreatic cancer development. PIM kinases may be a novel target for cancer therapy.


Subject(s)
Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins/genetics , Apoptosis/genetics , Cell Proliferation/genetics , Humans , Molecular Targeted Therapy , Neoplasm Staging , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , 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/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/biosynthesis
15.
Cardiovasc Diabetol ; 13: 68, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24685144

ABSTRACT

BACKGROUND: Diabetic women are five times more likely to develop congestive heart failure compared with two fold for men. The underlying mechanism for this gender difference is not known. Here we investigate the molecular mechanisms responsible for this female disadvantage and attempt safeguarding cardiomyocytes viability and function through restoration of pro-survival Pim-1. METHODS AND RESULTS: Diabetes was induced by injection of streptozotocin in CD1 mice of both genders. Functional and dimensional parameters measurement using echocardiography revealed diastolic dysfunction in female diabetic mice within 8 weeks after STZ-induced diabetes. This was associated with significant downregulation of pro-survival Pim-1 and upregulation of pro-apoptotic Caspase-3, microRNA-1 and microRNA-208a. Male diabetic mice did not show any significant changes at this time point (P < 0.05 vs. female diabetic). Further, the onset of ventricular remodelling was quicker in female diabetic mice showing marked left ventricular dilation, reduced ejection fraction and poor contractility (P < 0.05 vs. male diabetic at 12 and 16 weeks of STZ-induced diabetes). Molecular analysis of samples from human diabetic hearts confirmed the results of pre-clinical studies, showing marked downregulation of Pim-1 in the female diabetic heart (P < 0.05 vs. male diabetic). Finally, in vitro restoration of Pim-1 reversed the female disadvantage in diabetic cardiomyocytes. CONCLUSIONS: We provide novel insights into the molecular mechanisms behind the rapid onset of cardiomyopathy in female diabetics. These results suggest the requirement for the development of gender-specific treatments for diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , Down-Regulation/physiology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/biosynthesis , Sex Characteristics , Animals , Cell Survival/physiology , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/pathology , Female , Humans , Male , Mice , Proto-Oncogene Proteins c-pim-1/metabolism , Time Factors
16.
Mol Cell Biol ; 34(13): 2517-32, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24777602

ABSTRACT

MET, the receptor for hepatocyte growth factor (HGF), plays an important role in signaling normal and tumor cell migration and invasion. Here, we describe a previously unrecognized mechanism that promotes MET expression in multiple tumor cell types. The levels of the Pim-1 protein kinase show a positive correlation with the levels of MET protein in human tumor cell lines and patient-derived tumor materials. Using small interfering RNA (siRNA), Pim knockout mice, small-molecule inhibitors, and overexpression of Pim-1, we confirmed this correlation and found that Pim-1 kinase activity regulates HGF-induced tumor cell migration, invasion, and cell scattering. The novel biochemical mechanism for these effects involves the ability of Pim-1 to control the translation of MET by regulating the phosphorylation of eukaryotic initiation factor 4B (eIF4B) on S406. This targeted phosphorylation is required for the binding of eIF4B to the eIF3 translation initiation complex. Importantly, Pim-1 action was validated by the evaluation of patient blood and bone marrow from a phase I clinical trial of a Pim kinase inhibitor, AZD1208. These results suggest that Pim inhibitors may have an important role in the treatment of patients where MET is driving tumor biology.


Subject(s)
Eukaryotic Initiation Factors/genetics , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/biosynthesis , Proto-Oncogene Proteins c-pim-1/biosynthesis , Animals , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Movement , Eukaryotic Initiation Factor-3/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Neoplasm Invasiveness , Phosphorylation , Protein Binding , Protein Biosynthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction , Thiazolidines/pharmacology
18.
Cardiovasc Toxicol ; 13(4): 391-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23990052

ABSTRACT

Pim-1 is a serine/threonine kinase and involved in cell survival and proliferation. Recently, it has been shown that pim-1 signaling pathway plays an important role in cardiovascular protection and differentiation. In this study, we sought to explore the expression of pim-1 in human vascular endothelial cells (ECs) and its regulation by epigallocatechin-3-O-gallate (EGCG), a green tea polyphenol which has anti-oxidant, anti-inflammatory and vascular protective effects. By using quantitative reverse transcriptase PCR (qRT-PCR) and Western blotting, we showed that EGCG dose-dependently increased the expression of pim-1 in cultured umbilical vein endothelial cells. Next, we showed that EGCG activated a luciferase reporter driven by peroxisome proliferators-activated receptor (PPAR)-responsive elements. The induced expression of pim-1 was inhibited in ECs pretreated with GW9662, a specific antagonist of PPARγ. In addition, pim-1 was also up-regulated in endothelial cells treated with rosiglitazone, a specific agonist for PPARγ, or those infected with the adenovirus expressing a constitutively active PPARγ. Collectively, our results provided new evidence that pim-1 can be up-regulated by EGCG via a PPARγ-mediated mechanism and may mediate its vascular protective effects.


Subject(s)
Catechin/analogs & derivatives , Human Umbilical Vein Endothelial Cells/enzymology , PPAR gamma/physiology , Polyphenols/pharmacology , Proto-Oncogene Proteins c-pim-1/biosynthesis , Tea , Catechin/pharmacology , Gene Expression Regulation, Enzymologic , Human Umbilical Vein Endothelial Cells/drug effects , Humans
19.
Eur J Immunol ; 43(9): 2283-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712827

ABSTRACT

γ-Chain (γc) cytokine receptor signaling is required for the development of all lymphocytes. Why γc signaling plays such an essential role is not fully understood, but induction of the serine/threonine kinase Pim1 is considered a major downstream event of γc as Pim1 prevents apoptosis and increases metabolic activity. Consequently, we asked whether Pim1 overexpression would suffice to restore lymphocyte development in γc-deficient mice. By analyzing Pim1-transgenic γc-deficient mice (Pim1(Tg) γc(KO) ), we show that Pim1 promoted T-cell development and survival in the absence of γc. Interestingly, such effects were largely limited to CD4(+) lineage αß T cells as CD4(+) T-cell numbers improved to near normal levels but CD8(+) T cells remained severely lymphopenic. Notably, Pim1 over-expression failed to promote development and survival of any T-lineage cells other than αß T cells, as we observed complete lack of γδ, NKT, FoxP3(+) T regulatory cells and TCR-ß(+) CD8αα IELs in Pim1(Tg) γc(KO) mice. Collectively, these results uncover distinct requirements for γc signaling between CD4(+) αß T cells and all other T-lineage cells, and they identify Pim1 as a novel effector molecule sufficient to drive CD4(+) αß T-cell development and survival in the absence of γc cytokine receptor signaling.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chemokines, C/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Receptors, Cytokine/metabolism , Animals , CD8 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Cell Survival , Chemokines, C/deficiency , Forkhead Transcription Factors/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells , Proto-Oncogene Proteins c-pim-1/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction , T-Lymphocytes, Regulatory
20.
J Mol Cell Cardiol ; 53(5): 695-706, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23010478

ABSTRACT

Stem cell transplantation has been successfully used for amelioration of cardiomyopathic injury using adult cardiac progenitor cells (CPC). Engineering of mouse CPC with the human serine/threonine kinase Pim-1 (CPCeP) enhances regeneration and cell survival in vivo, but it is unknown if such apparent lineage commitment is associated with maturation of electrophysiological properties and excitation-contraction coupling. This study aims to determine electrophysiology and Ca(2+)-handling properties of CPCeP using neonatal rat cardiomyocyte (NRCM) co-culture to promote cardiomyocyte lineage commitment. Measurements of membrane capacitance, dye transfer, expression of connexin 43 (Cx43), and transmission of ionic currents (I(Ca), I(Na)) from one cell to the next suggest that a subset of co-cultured CPCeP and NRCM becomes connected via gap junctions. Unlike NRCM, CPCeP had no significant I(Na), but expressed nifedipine-sensitive I(Ca) that could be measured more consistently with Ba(2+) as permeant ion using ramp-clamp protocols than with Ca(2+) and step-depolarization protocols. The magnitude of I(Ca) in CPCeP increased during culture (4-7 days vs. 1-3 days) and was larger in co-cultures with NRCM and with NRCM-conditioned medium, than in mono-cultured CPCeP. I(Ca) was virtually absent in CPC without engineered expression of Pim-1. Caffeine and KCl-activated Ca(2+)-transients were significantly present in co-cultured CPCeP, but smaller than in NRCM. Conversely, ATP-induced (IP(3)-mediated) Ca(2+) transients were larger in CPCeP than in NRCM. I(NCX) and I(ATP) were expressed in equivalent densities in CPCeP and NRCM. These in vitro studies suggest that CPCeP in co-culture with NRCM: a) develop I(Ca) current and Ca(2+) signaling consistent with cardiac lineage, b) form electrical connections via Cx43 gap junctions, and c) respond to paracrine signals from NRCM. These properties may be essential for durable and functional myocardial regeneration under in vivo conditions.


Subject(s)
Adult Stem Cells/physiology , Cell Differentiation , Myocytes, Cardiac/physiology , Proto-Oncogene Proteins c-pim-1/genetics , Recombinant Fusion Proteins/genetics , Adenosine Triphosphate/physiology , Adult Stem Cells/metabolism , Animals , Calcium Signaling , Cell Communication , Cell Shape , Cells, Cultured , Coculture Techniques , Connexin 43/metabolism , Culture Media, Conditioned , Gap Junctions/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Membrane Potentials , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Phenotype , Proto-Oncogene Proteins c-pim-1/biosynthesis , Rats , Recombinant Fusion Proteins/biosynthesis , Sodium-Calcium Exchanger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...