Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Biol Chem ; 288(1): 455-65, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23148218

ABSTRACT

Neuregulin (NRG; heregulin) is overexpressed in ∼30% of breast cancers and mediates various processes involved in tumor progression, including tumor cell migration and invasion. Here, we show that NRG mediates its effects on tumor cell migration via PKD1. Downstream of RhoA, PKD1 can prevent directed cell migration through phosphorylation of its substrate SSH1L. NRG exerts its inhibitory effects on PKD1 through Rac1/NADPH oxidase, leading to decreased PKD1 activation loop phosphorylation and decreased activity toward SSH1L. The consequence of PKD1 inhibition by NRG is decreased binding of 14-3-3 to SSH1L, localization of SSH1L to F-actin at the leading edge, and increased cofilin activity, resulting in increased reorganization of the actin cytoskeleton and cell motility. Our data provide a mechanism through which the Rho GTPase Rac1 cross-talks with PKD1 signaling pathways to facilitate directed cell migration.


Subject(s)
Actins/metabolism , Neuregulin-1/metabolism , Protein Kinase C/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Breast Neoplasms/metabolism , Cell Movement , Chemotaxis , Disease Progression , Female , Humans , Mice , Microscopy, Fluorescence/methods , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Small Interfering/metabolism , Signal Transduction , Wound Healing
2.
J Biol Chem ; 288(34): 24382-93, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23846685

ABSTRACT

Enabled/Vasodilator-stimulated phosphoprotein (Ena/VASP) protein family members link actin dynamics and cellular signaling pathways. VASP localizes to regions of dynamic actin reorganization such as the focal adhesion contacts, the leading edge or filopodia, where it contributes to F-actin filament elongation. Here we identify VASP as a novel substrate for protein kinase D1 (PKD1). We show that PKD1 directly phosphorylates VASP at two serine residues, Ser-157 and Ser-322. These phosphorylations occur in response to RhoA activation and mediate VASP re-localization from focal contacts to the leading edge region. The net result of this PKD1-mediated phosphorylation switch in VASP is increased filopodia formation and length at the leading edge. However, such signaling when persistent induced membrane ruffling and decreased cell motility.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Actins/genetics , Actins/metabolism , Cell Adhesion Molecules/genetics , Focal Adhesions/genetics , Focal Adhesions/metabolism , HeLa Cells , Humans , Microfilament Proteins/genetics , Phosphoproteins/genetics , Phosphorylation/physiology , Protein Kinase C/genetics , Protein Transport/physiology , Pseudopodia/genetics , Pseudopodia/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
3.
J Cell Sci ; 125(Pt 18): 4253-63, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22718346

ABSTRACT

The Forkhead transcription factor, FoxO3a, is a known suppressor of primary tumor growth through transcriptional regulation of key genes regulating cell cycle arrest and apoptosis. In many types of cancer, in response to growth factor signaling, FoxO3a is phosphorylated by Akt, resulting in its exclusion from the nucleus. Here we show that FoxO3a remains nuclear in anaplastic thyroid carcinoma (ATC). This correlates with lack of Akt phosphorylation at serine473 in ATC cell lines and tissues of ATC patients, providing a potential explanation for nuclear FoxO3a. Mechanistically, nuclear FoxO3a promotes cell cycle progression by transcriptional upregulation of cyclin A1, promoting proliferation of human ATC cells. Silencing FoxO3a with a reverse genetics approach leads to downregulation of CCNA1 mRNA and protein. These combined data suggest an entirely novel function for FoxO3a in ATC promotion by enhancing cell cycle progression and tumor growth through transcriptional upregulation of cyclin A1. This is clinically relevant since we detected highly elevated CCNA1 mRNA and protein levels in tumor tissues of ATC patients. Our data indicate therapeutic inactivation of FoxO3a may lead to attenuation of tumor expansion in ATC. This new paradigm also suggests caution in relation to current dogma focused upon reactivation of FoxO3a as a therapeutic strategy against cancers harboring active PI3-K and Akt signaling pathways.


Subject(s)
Cyclin A1/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Transcription, Genetic , Base Sequence , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cyclin A1/metabolism , Forkhead Box Protein O3 , Gene Silencing , HEK293 Cells , Humans , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/therapy
4.
Breast Cancer Res Treat ; 144(1): 79-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24510012

ABSTRACT

The treatment of patients with invasive breast cancer remains a major issue because of the acquisition of drug resistance to conventional chemotherapy. Here we propose a new therapeutic strategy by combining DNA methyltransferase inhibitors (DMTIs) with suramin. Cytotoxic effects of suramin or combination treatment with DMTIs were determined in highly invasive breast cancer cell lines MDA-MB-231, BT-20 and HCC1954, or control cells. In addition, effects on cell invasion were determined in 3-dimensional cell culture assays. DMTI-mediated upregulation of Protein Kinase D1 (PKD1) expression was shown by Western blotting. Effects of suramin on PKD1 activity was determined in vitro and in cells. The importance of PKD1 in mediating the effects of such combination treatment in cell invasion was demonstrated using 3D cell culture assays. A proof of principal animal experiment was performed showing that PKD1 is critical for breast cancer growth. We show that when used in combination, suramin and DMTIs impair the invasive phenotype of breast cancer cells. We show that PKD1, a kinase that previously has been described as a suppressor of tumor cell invasion, is an interface for both FDA-approved drugs, since the additive effects observed are due to DMTI-mediated re-expression and suramin-induced activation of PKD1. Our data reveal a mechanism of how a combination treatment with non-toxic doses of suramin and DMTIs may be of therapeutic benefit for patients with aggressive, multi-drug resistant breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Cell Movement/drug effects , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Animals , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Cell Line, Tumor , Cell Proliferation/drug effects , Decitabine , Female , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Phthalimides/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Suramin/administration & dosage , Tryptophan/administration & dosage , Tryptophan/analogs & derivatives , Xenograft Model Antitumor Assays
5.
Biochem J ; 455(2): 251-60, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23841590

ABSTRACT

PAKs (p21-activated kinases) are effectors of RhoGTPases. PAK4 contributes to regulation of cofilin at the leading edge of migrating cells through activation of LIMK (Lin-11/Isl-1/Mec-3 kinase). PAK4 activity is regulated by an autoinhibitory domain that is released upon RhoGTPase binding as well as phosphorylation at Ser474 in the activation loop of the kinase domain. In the present study, we add another level of complexity to PAK4 regulation by showing that phosphorylation at Ser99 is required for its targeting to the leading edge. This phosphorylation is mediated by PKD1 (protein kinase D1). Phosphorylation of PAK4 at Ser99 also mediates binding to 14-3-3 protein, and is required for the formation of a PAK4-LIMK-PKD1 complex that regulates cofilin activity and directed cell migration.


Subject(s)
Protein Kinase C/metabolism , Serine/genetics , p21-Activated Kinases/analysis , p21-Activated Kinases/metabolism , 14-3-3 Proteins/metabolism , Cell Movement , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Serine/metabolism , Signal Transduction , Transfection
6.
Front Immunol ; 15: 1278807, 2024.
Article in English | MEDLINE | ID: mdl-38576613

ABSTRACT

Pancreatic inflammation is a risk factor for the development of pancreatic cancer. Increased presence of inflammatory macrophages can be found in response to a KRAS mutation in acinar cells or in response to experimentally-induced pancreatitis. Inflammatory macrophages induce pancreatic acinar cells to undergo dedifferentiation to a duct-like progenitor stage, a process called acinar-to-ductal metaplasia (ADM). Occurrence of ADM lesions are believed to be the initiating event in tumorigenesis. Here we will discuss how macrophage-induced oxidative stress contributes to ADM and how ADM cells shape the fibrotic stroma needed for further progression.


Subject(s)
Pancreatic Neoplasms , Pancreatitis , Humans , Reactive Oxygen Species , Signal Transduction/genetics , Pancreatic Neoplasms/pathology , Pancreatitis/pathology , Macrophages/pathology
7.
Breast Cancer Res ; 15(2): R66, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23971832

ABSTRACT

INTRODUCTION: DNA methylation-induced silencing of genes encoding tumor suppressors is common in many types of cancer, but little is known about how such epigenetic silencing can contribute to tumor metastasis. The PRKD1 gene encodes protein kinase D1 (PKD1), a serine/threonine kinase that is expressed in cells of the normal mammary gland, where it maintains the epithelial phenotype by preventing epithelial-to-mesenchymal transition. METHODS: The status of PRKD1 promoter methylation was analyzed by reduced representation bisulfite deep sequencing, methylation-specific PCR (MSP-PCR) and in situ MSP-PCR in invasive and noninvasive breast cancer lines, as well as in humans in 34 cases of "normal" tissue, 22 cases of ductal carcinoma in situ, 22 cases of estrogen receptor positive, HER2-negative (ER+/HER2-) invasive lobular carcinoma, 43 cases of ER+/HER2- invasive ductal carcinoma (IDC), 93 cases of HER2+ IDC and 96 cases of triple-negative IDC. A reexpression strategy using the DNA methyltransferase inhibitor decitabine was used in vitro in MDA-MB-231 cells as well as in vivo in a tumor xenograft model and measured by RT-PCR, immunoblotting and immunohistochemistry. The effect of PKD1 reexpression on cell invasion was analyzed in vitro by transwell invasion assay. Tumor growth and metastasis were monitored in vivo using the IVIS Spectrum Pre-clinical In Vivo Imaging System. RESULTS: Herein we show that the gene promoter of PRKD1 is aberrantly methylated and silenced in its expression in invasive breast cancer cells and during breast tumor progression, increasing with the aggressiveness of tumors. Using an animal model, we show that reversion of PRKD1 promoter methylation with the DNA methyltransferase inhibitor decitabine restores PKD1 expression and blocks tumor spread and metastasis to the lung in a PKD1-dependent fashion. CONCLUSIONS: Our data suggest that the status of epigenetic regulation of the PRKD1 promoter can provide valid information on the invasiveness of breast tumors and therefore could serve as an early diagnostic marker. Moreover, targeted upregulation of PKD1 expression may be used as a therapeutic approach to reverse the invasive phenotype of breast cancer cells.


Subject(s)
Azacitidine/analogs & derivatives , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Epigenesis, Genetic/drug effects , Gene Silencing , Promoter Regions, Genetic/genetics , Protein Kinase C/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Azacitidine/pharmacology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/secondary , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/secondary , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/genetics , Carcinoma, Lobular/secondary , Cell Movement , Cell Proliferation , DNA Methylation/drug effects , Decitabine , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Protein Kinase C/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
iScience ; 26(6): 106820, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37250781

ABSTRACT

The innate immune system has a key role in pancreatic cancer initiation, but the specific contribution of different macrophage populations is still ill-defined. While inflammatory (M1) macrophages have been shown to drive acinar-to-ductal metaplasia (ADM), a cancer initiating event, alternatively activated (M2) macrophages have been attributed to lesion growth and fibrosis. Here, we determined cytokines and chemokines secreted by both macrophage subtypes. Then, we analyzed their role in ADM initiation and lesion growth, finding that while M1 secrete TNF, CCL5, and IL-6 to drive ADM, M2 induce this dedifferentiation process via CCL2, but the effects are not additive. This is because CCL2 induces ADM by generating ROS and upregulating EGFR signaling, thus using the same mechanism as cytokines from inflammatory macrophages. Therefore, while effects on ADM are not additive between macrophage polarization types, both act synergistically on the growth of low-grade lesions by activating different MAPK pathways.

9.
J Biol Chem ; 286(39): 34254-61, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21832093

ABSTRACT

Dynamic reorganization of the actin cytoskeleton at the leading edge is required for directed cell migration. Cofilin, a small actin-binding protein with F-actin severing activities, is a key enzyme initiating such actin remodeling processes. Cofilin activity is tightly regulated by phosphorylation and dephosphorylation events that are mediated by LIM kinase (LIMK) and the phosphatase slingshot (SSH), respectively. Protein kinase D (PKD) is a serine/threonine kinase that inhibits actin-driven directed cell migration by phosphorylation and inactivation of SSH. Here, we show that PKD can also regulate LIMK through direct phosphorylation and activation of its upstream kinase p21-activated kinase 4 (PAK4). Therefore, active PKD increases the net amount of phosphorylated inactive cofilin in cells through both pathways. The regulation of cofilin activity at multiple levels may explain the inhibitory effects of PKD on barbed end formation as well as on directed cell migration.


Subject(s)
Actin Depolymerizing Factors/metabolism , Actins/metabolism , Cell Movement/physiology , Protein Kinase C/metabolism , p21-Activated Kinases/metabolism , Actin Depolymerizing Factors/genetics , Actins/genetics , HeLa Cells , Humans , Lim Kinases/genetics , Lim Kinases/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Kinase C/genetics , p21-Activated Kinases/genetics
10.
Antioxidants (Basel) ; 11(1)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35052641

ABSTRACT

Pancreatic acinar-to-ductal metaplasia (ADM) is a reversible process that occurs after pancreatic injury, but becomes permanent and leads to pancreatic lesions in the presence of an oncogenic mutation in KRAS,. While inflammatory macrophage-secreted chemokines, growth factors that activate epidermal growth factor receptor (EGFR) and oncogenic KRAS have been implicated in the induction of ADM, it is currently unclear whether a common underlying signaling mechanism exists that drives this process. In this study, we show that different inducers of ADM increase levels of hydrogen peroxide, most likely generated at the mitochondria, and upregulate the expression of Protein Kinase D1 (PKD1), a kinase that can be activated by hydrogen peroxide. PKD1 expression in acinar cells affects their survival and mediates ADM, which is in part due to the PKD1 target NF-κB. Overall, our data implicate ROS-PKD1 signaling as a common feature of different inducers of pancreatic ADM.

11.
iScience ; 25(5): 104327, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35602933

ABSTRACT

Desmoplasia around pancreatic lesions is a barrier for immune cells and a hallmark of developing and established pancreatic cancer. However, the contribution of the innate immune system to this process is ill-defined. Using the KC mouse model and primary cells in vitro, we show that alternatively activated macrophages (AAM) crosstalk with pancreatic lesion cells and pancreatic stellate cells (PSCs) to mediate fibrosis and progression of lesions. TGFß1 secreted by AAM not only drives activation of quiescent PSCs but also in activated PSCs upregulates expression of TIMP1, a factor previously shown as crucial in fibrosis. Once activated, PSCs auto-stimulate proliferation via CXCL12. Furthermore, we found that TIMP1/CD63 signaling mediates PanIN lesion growth and TGFß1 contributes to a cadherin switch and drives structural collapse of lesions, indicating a potential progression step. Taken together, our data indicate TGFß1 produced by Ym1+ AAM as a major driver of processes that initiate the development of pancreatic cancer.

12.
iScience ; 24(1): 102019, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33521594

ABSTRACT

Doublecortin-like kinase 1 (DCLK1)-positive pancreatic cancer stem cells develop at a precancerous stage and may contribute to the lack of efficacy of pancreatic cancer therapy. Although PanIN cells express oncogenic KRas and have an increased activity of epidermal growth factor receptor (EGFR), we demonstrate that, in DCLK1+ PanIN cells, EGFR signaling is not propagated to the nucleus. Mimicking blockage of EGFR with erlotinib in PanIN organoid culture or in p48cre;KrasG12D mice led to a significant increase in DCLK1+ PanIN cells. As a mechanism of how EGFR inhibition leads to formation of DCLK1+ cells, we identify an increase in hydrogen peroxide contributing to activation of Protein Kinase D1 (PKD1). Active PKD1 then drives stemness and abundance of DCLK1+ cells in lesions. Our data suggest a signaling mechanism that leads to the development of DCLK1+ pancreatic cancer stem cells, which can be exploited to target this population in potential therapeutic approaches.

13.
J Cell Biochem ; 106(4): 714-28, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19173301

ABSTRACT

Cell-cell contacts mediated by cadherins are known to inhibit the small Rho-GTPase RhoA. We here show that in epithelial cells the disruption of these cell-cell contacts as mediated by a calcium switch leads to actin re-organization and the activation of RhoA. We identified the serine/threonine kinase protein kinase D1 (PKD1) as a downstream target for RhoA in this pathway. After disruption of cell-cell contacts, PKD1 relayed RhoA activation to the induction of the transcription factor NF-kappaB. We found that a signaling complex composed of the kinases ROCK, novel protein kinase C (nPKC), and Src family kinases (SFKs) is upstream of PKD1 and crucial for RhoA-mediated NF-kappaB activation. In conclusion, our data suggest a previously undescribed signaling pathway of how RhoA is activated by loss of cell-cell adhesions and by which it mediates the activation of NF-kappaB. We propose that this pathway is of relevance for epithelial tumor cell biology, where loss of cell-cell contacts has been implicated in regulating cell survival and motility.


Subject(s)
Cell Communication , NF-kappa B/genetics , Protein Kinase C/metabolism , Transcriptional Activation , rhoA GTP-Binding Protein/metabolism , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Multiprotein Complexes , Signal Transduction
14.
Breast Cancer Res ; 11(1): R13, 2009.
Article in English | MEDLINE | ID: mdl-19243594

ABSTRACT

INTRODUCTION: The biological and molecular events that regulate the invasiveness of breast tumour cells need to be further revealed to develop effective therapies that stop breast cancer from expanding and metastasising. METHODS: Human tissue samples of invasive breast cancer and normal breast, as well as breast cancer cell lines, were evaluated for protein kinase D (PKD) expression, to test if altered expression could serve as a marker for invasive breast cancer. We further utilised specific PKD1-shRNA and a system to inducibly-express PKD1 to analyse the role of PKD1 in the invasive behaviour of breast cancer cell lines in two-dimensional (2D) and three-dimensional (3D) culture. Invasive behaviour in breast cancer cell lines has been linked to matrix metalloproteinases (MMPs), so we also determined if PKD1 regulates the expression and activity of these enzymes. RESULTS: We found that the serine/threonine kinase, PKD1, is highly expressed in ductal epithelial cells of normal human breast tissue, but is reduced in its expression in more than 95% of all analysed samples of human invasive breast tumours. Additionally, PKD1 is not expressed in highly invasive breast cancer cell lines, whereas non-invasive or very low-invasive breast cancer cell lines express PKD1. Our results further implicate that in MDA-MB-231 cells PKD1 expression is blocked by epigenetic silencing via DNA methylation. The re-expression of constitutively-active PKD1 in MDA-MB-231 cells drastically reduced their ability to invade in 2D and 3D cell culture. Moreover, MCF-7 cells acquired the ability to invade in 2D and 3D cell culture when PKD1 expression was knocked-down by shRNA. PKD1 also regulated the expression of breast cancer cell MMPs, MMP-2, MMP-7, MMP-9, MMP-10, MMP-11, MMP-13, MMP-14 and MMP-15, providing a potential mechanism for PKD1 mediation of the invasive phenotype. CONCLUSIONS: Our results identify decreased expression of the PKD1 as a marker for invasive breast cancer. They further suggest that the loss of PKD1 expression increases the malignant potential of breast cancer cells. This may be due to the function of PKD1 as a negative regulator of MMP expression. Our data suggest re-expression of PKD1 as a potential therapeutic strategy.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Matrix Metalloproteinases/metabolism , Protein Kinase C/physiology , Blotting, Western , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/secondary , Cell Adhesion , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lymphatic Metastasis , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular
15.
Sci Rep ; 9(1): 16588, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31719634

ABSTRACT

Current treatment options for patients with pancreatic cancer are suboptimal, resulting in a five year survival rate of about 9%. Difficulties with treatment are due to an immunosuppressive, fibrotic tumor microenvironment that prevents drugs from reaching tumor cells, but also to the limited efficacy of existing FDA-approved chemotherapeutic compounds. We here show that the nucleoside analog Sangivamycin and its closely-related compound Toyocamycin target PDA cell lines, and are significantly more efficient than Gemcitabine. Using KINOMEscan screening, we identified the kinase Haspin, which is overexpressed in PDA cell lines and human PDA samples, as a main target for both compounds. Inhibition of Haspin leads to a decrease in Histone H3 phosphorylation and prevents Histone H3 binding to survivin, thus providing mechanistic insight of how Sangivamycin targets cell proliferation, mitosis and induces apoptotic cell death. In orthotopically implanted tumors in mice, Sangivamycin was efficient in decreasing the growth of established tumors. In summary, we show that Sangivamycin and derivatives can be an efficient new option for treatment of PDA.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic/drug effects , Histones/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidine Nucleosides/pharmacology , Survivin/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/pharmacology , Biomarkers, Tumor , Cell Proliferation , Histones/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Phosphorylation , Prognosis , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Survivin/genetics , Survivin/metabolism , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays
16.
Mol Cell Biol ; 25(19): 8520-30, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16166634

ABSTRACT

Efficient elimination of mitochondrial reactive oxygen species (mROS) correlates with increased cellular survival and organism life span. Detoxification of mitochondrial ROS is regulated by induction of the nuclear SOD2 gene, which encodes the manganese-dependent superoxide dismutase (MnSOD). However, the mechanisms by which mitochondrial oxidative stress activates cellular signaling pathways leading to induction of nuclear genes are not known. Here we demonstrate that release of mROS activates a signal relay pathway in which the serine/threonine protein kinase D (PKD) activates the NF-kappaB transcription factor, leading to induction of SOD2. Conversely, the FOXO3a transcription factor is dispensable for mROS-induced SOD2 induction. PKD-mediated MnSOD expression promotes increased survival of cells upon release of mROS, suggesting that mitochondrion-to-nucleus signaling is necessary for efficient detoxification mechanisms and cellular viability.


Subject(s)
Cell Nucleus/metabolism , Mitochondria/metabolism , Protein Kinase C/physiology , Reactive Oxygen Species , Alleles , Cell Survival , Cytosol/metabolism , Dose-Response Relationship, Drug , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Genes, Reporter , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Immunoprecipitation , Microscopy, Fluorescence , Models, Biological , Models, Genetic , NF-kappa B/metabolism , Oxidative Stress , Plasmids/metabolism , Protein Kinase C/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Superoxide Dismutase/metabolism , Transfection
17.
Oncotarget ; 9(91): 36358-36370, 2018 Nov 20.
Article in English | MEDLINE | ID: mdl-30555634

ABSTRACT

Phosphatidylinositol-4-phosphate 5-kinase type-1C (PIP5K1C) is a lipid kinase that regulates focal adhesion dynamics and cell attachment through site-specific formation of phosphatidylinositol-4,5-bisphosphate (PI4,5P2). By comparing normal breast tissue to carcinoma in situ and invasive ductal carcinoma subtypes, we here show that the phosphorylation status of PIP5K1C at serine residue 448 (S448) can be predictive for breast cancer progression to an aggressive phenotype, while PIP5K1C expression levels are not indicative for this event. PIP5K1C phosphorylation at S448 is downregulated in invasive ductal carcinoma, and similarly, the expression levels of PKD1, the kinase that phosphorylates PIP5K1C at this site, are decreased. Overall, since PKD1 is a negative regulator of cell migration and invasion in breast cancer, the phosphorylation status of this residue may serve as an indicator of aggressiveness of breast tumors.

18.
Mol Cell Biol ; 24(7): 2614-26, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15024053

ABSTRACT

Protein kinase D (PKD) participates in activation of the transcription factor NF-kappaB (nuclear factor kappaB) in cells exposed to oxidative stress, leading to increased cellular survival. We previously demonstrated that phosphorylation of PKD at Tyr463 in the PH (pleckstrin homology) domain is mediated by the Src-Abl pathway and that it is necessary for PKD activation and subsequent NF-kappaB induction. Here we show that activation of PKD in response to oxidative stress requires two sequential signaling events, i.e., phosphorylation of Tyr463 by Abl, which in turn promotes a second step, phosphorylation of the PKD activation loop (Ser738/Ser742). We show that this is mediated by PKCdelta (protein kinase Cdelta), a kinase that is activated by Src in response to oxidative stress. We also show that other PKCs, including PKCepsilon and PKCzeta, do not participate in PKD activation or NF-kappaB induction. We propose a model in which two coordinated signaling events are required for PKD activation. Tyrosine phosphorylation in the PH domain at Tyr463, mediated by the Src-Abl pathway, which in turn facilitates the phosphorylation of Ser738/Ser742 in the activation loop, mediated by the Src-PKCdelta pathway. Once active, the signal is relayed to the activation of NF-kappaB in oxidative stress responses.


Subject(s)
NF-kappa B/metabolism , Oxidative Stress , Protein Kinase C/metabolism , Signal Transduction/physiology , Enzyme Activation , Enzyme Inhibitors/metabolism , Genes, Reporter , HeLa Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , NF-kappa B/genetics , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C-delta , Proto-Oncogene Proteins c-abl/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , src-Family Kinases/metabolism
19.
Front Oncol ; 7: 41, 2017.
Article in English | MEDLINE | ID: mdl-28361035

ABSTRACT

Due to alterations in their metabolic activity and decreased mitochondrial efficiency, cancer cells often show increased generation of reactive oxygen species (ROS), but at the same time, to avoid cytotoxic signaling and to facilitate tumorigenic signaling, have mechanism in place that keep ROS in check. This requires signaling molecules that convey increases in oxidative stress to signal to the nucleus to upregulate antioxidant genes. Protein kinase D1 (PKD1), the serine/threonine kinase, is one of these ROS sensors. In this mini-review, we highlight the mechanisms of how PKD1 is activated in response to oxidative stress, so far known downstream effectors, as well as the importance of PKD1-initiated signaling for development and progression of pancreatic cancer.

20.
Sci Rep ; 7(1): 9524, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28842658

ABSTRACT

Dependent on their cellular localization, Protein Kinase D (PKD) enzymes regulate different processes including Golgi transport, cell signaling and response to oxidative stress. The localization of PKD within cells is mediated by interaction with different lipid or protein binding partners. With the example of PKD2, we here show that phosphorylation events can also contribute to localization of subcellular pools of this kinase. Specifically, in the present study, we show that tyrosine phosphorylation of PKD2 at residue Y87 defines its localization to the focal adhesions and leads to activation. This phosphorylation occurs downstream of RhoA signaling and is mediated via Src. Moreover, mutation of this residue blocks PKD2's interaction with Focal Adhesion Kinase (FAK). The presence and regulation of PKD2 at focal adhesions identifies a novel function for this kinase as a modulator of cell adhesion and migration.


Subject(s)
Cell Adhesion , Focal Adhesions , Protein Kinases/metabolism , src-Family Kinases/metabolism , Cell Movement , Fluorescent Antibody Technique , Immunohistochemistry , Phosphorylation , Protein Kinase D2 , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL