Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Oncol ; 12: 1028959, 2022.
Article in English | MEDLINE | ID: mdl-36531051

ABSTRACT

The PDZ-LIM domain-containing protein 2 (PDLIM2) regulates cell polarity and the protein stability of key transcription factors in epithelial and hemopoietic cells. We previously reported that PDLIM2 is more highly expressed in Triple Negative Breast Cancer (TNBC) than in other breast cancer types or normal breast tissue. In the course of the TNBC study, it was noted that PDLIM2 was highly expressed in the stroma of PDLIM2-expressing tumours. Here, we investigated the phenotype of these stromal cells and whether any infiltrating immune population was linked to PDLIM2 expression. We found that high PDLIM2 expression in breast tumours was associated with higher levels of infiltrating M2 macrophages, but was not associated with infiltrating T cell sub-populations. We then tested whether PDLIM2 contributes to macrophage differentiation or function by using cultures of bone marrow-derived macrophages from wildtype and Pdlim2 knockout mice. This demonstrated that PDLIM2 is required for naïve macrophage migration and for the full adoption of IL-4-induced M2 polarization, including expression of M2 phenotypic markers, cell adhesion and cell migration. TLR4-, TLR3- or IFNγ-induced M1 macrophage activity was less dependent on PDLIM2. Finally, analysis of publicly available breast cancer datasets showed that high PDLIM2 expression is associated with increased M2 macrophage infiltration. We conclude that PDLIM2 expression influences the tumour associated stroma and, in particular, M2 macrophage infiltration that may contribute to the progression of TNBC or other subsets of breast cancer.

2.
J Oncol ; 2019: 3980273, 2019.
Article in English | MEDLINE | ID: mdl-31346333

ABSTRACT

Cathepsin S (CTSS) has previously been implicated in a number of cancer types, where it is associated with poor clinical features and outcome. To date, patient outcome in breast cancer has not been examined with respect to this protease. Here, we carried out immunohistochemical (IHC) staining of CTSS using a breast cancer tissue microarray in patients who received adjuvant therapy. We scored CTSS expression in the epithelial and stromal compartments and evaluated the association of CTSS expression with matched clinical outcome data. We observed differences in outcome based on CTSS expression, with stromal-derived CTSS expression correlating with a poor outcome and epithelial CTSS expression associated with an improved outcome. Further subtype characterisation revealed high epithelial CTSS expression in TNBC patients with improved outcome, which remained consistent across two independent TMA cohorts. Further in silico gene expression analysis, using both in-house and publicly available datasets, confirmed these observations and suggested high CTSS expression may also be beneficial to outcome in ER-/HER2+ cancer. Furthermore, high CTSS expression was associated with the BL1 Lehmann subgroup, which is characterised by defects in DNA damage repair pathways and correlates with improved outcome. Finally, analysis of matching IHC analysis reveals an increased M1 (tumour destructive) polarisation in macrophage in patients exhibiting high epithelial CTSS expression. In conclusion, our observations suggest epithelial CTSS expression may be prognostic of improved outcome in TNBC. Improved outcome observed with HER2+ at the gene expression level furthermore suggests CTSS may be prognostic of improved outcome in ER- cancers as a whole. Lastly, from the context of these patients receiving adjuvant therapy and as a result of its association with BL1 subgroup CTSS may be elevated in patients with defects in DNA damage repair pathways, indicating it may be predictive of tumour sensitivity to DNA damaging agents.

3.
Cancer Res ; 79(10): 2619-2633, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30885980

ABSTRACT

The PDLIM2 protein regulates stability of transcription factors including NF-κB and STATs in epithelial and hemopoietic cells. PDLIM2 is strongly expressed in certain cancer cell lines that exhibit an epithelial-to-mesenchymal phenotype, and its suppression is sufficient to reverse this phenotype. PDLIM2 supports the epithelial polarity of nontransformed breast cells, suggesting distinct roles in tumor suppression and oncogenesis. To better understand its overall function, we investigated PDLIM2 expression and activity in breast cancer. PDLIM2 protein was present in 60% of tumors diagnosed as triple-negative breast cancer (TNBC), and only 20% of other breast cancer subtypes. High PDLIM2 expression in TNBC was positively correlated with adhesion signaling and ß-catenin activity. Interestingly, PDLIM2 was restricted to the cytoplasm/membrane of TNBC cells and excluded from the nucleus. In breast cell lines, PDLIM2 retention in the cytoplasm was controlled by cell adhesion, and translocation to the nucleus was stimulated by insulin-like growth factor-1 or TGFß. Cytoplasmic PDLIM2 was associated with active ß-catenin and ectopic expression of PDLIM2 was sufficient to increase ß-catenin levels and its transcriptional activity in reporter assays. Suppression of PDLIM2 inhibited tumor growth in vivo, whereas overexpression of PDLIM2 disrupted growth in 3D cultures. These results suggest that PDLIM2 may serve as a predictive biomarker for a large subset of TNBC whose phenotype depends on adhesion-regulated ß-catenin activity and which may be amenable to therapies that target these pathways. SIGNIFICANCE: This study shows that PDLIM2 expression defines a subset of triple-negative breast cancer that may benefit from targeting the ß-catenin and adhesion signaling pathways. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/10/2619/F1.large.jpg.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Adhesion , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , beta Catenin/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , Female , HEK293 Cells , Humans
4.
Article in English | MEDLINE | ID: mdl-26191041

ABSTRACT

IGF-1R expression and activation levels generally cannot be correlated in cancer cells, suggesting that cellular proteins may modulate IGF-1R activity. Strong candidates for such modulation are found in cell-matrix and cell-cell adhesion signaling complexes. Activated IGF-1R is present at focal adhesions, where it can stabilize ß1 integrin and participate in signaling complexes that promote invasiveness associated with epithelial mesenchymal transition (EMT) and resistance to therapy. Whether IGF-1R contributes to EMT or to non-invasive tumor growth may be strongly influenced by the degree of extracellular matrix engagement and the presence or absence of key proteins in IGF-1R-cell adhesion complexes. One such protein is PDLIM2, which promotes both cell polarization and EMT by regulating the stability of transcription factors including NFκB, STATs, and beta catenin. PDLIM2 exhibits tumor suppressor activity, but is also highly expressed in certain invasive cancers. It is likely that distinct adhesion complex proteins modulate IGF-1R signaling during cancer progression or adaptive responses to therapy. Thus, identifying the key modulators will be important for developing effective therapeutic strategies and predictive biomarkers.

5.
Neoplasia ; 16(5): 422-31, 2014 May.
Article in English | MEDLINE | ID: mdl-24863845

ABSTRACT

PDLIM2 is a cytoskeletal and nuclear PDZ-LIM domain protein that regulates the stability of Nuclear Factor kappa-B (NFκB) and other transcription factors, and is required for polarized cell migration. PDLIM2 expression is suppressed by methylation in different cancers, but is strongly expressed in invasive breast cancer cells that have undergone an Epithelial Mesenchymal Transition (EMT). PDLIM2 is also expressed in non-transformed breast myoepithelial MCF10A cells and here we asked whether it is important for maintaining the polarized, epithelial phenotype of these cells. Suppression of PDLIM2 in MCF10A cells was sufficient to disrupt cell polarization and acini formation with increased proliferation and reduced apoptosis in the luminal space compared to control acini with hollow lumina. Spheroids with suppressed PDLIM2 exhibited increased expression of cell-cell and cell-matrix adhesion proteins including beta 1 (ß1) integrin. Interestingly, levels of the Insulin-like growth factor 1 receptor (IGF-1 R) and Receptor of activated protein kinase C 1 (RACK1), which scaffolds IGF-1R to ß1 integrin, were also increased, indicating a transformed phenotype. Focal Adhesion Kinase (FAK) and cofilin phosphorylation, and RhoA Guanosine Triphosphatase (GTPase) activity were all enhanced in these spheroids compared to control acini. Importantly, inhibition of either FAK or Rho Kinase (ROCK) was sufficient to rescue the polarity defect. We conclude that PDLIM2 expression is essential for feedback regulation of the ß1-integrin-RhoA signalling axis and integration of cellular microenvironment signals with gene expression to control the polarity of breast epithelial acini structures. This is a mechanism by which PDLIM2 could mediate tumour suppression in breast epithelium.


Subject(s)
Cell Polarity/physiology , Epithelial Cells/metabolism , Integrin beta1/metabolism , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Cell Culture Techniques/methods , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Feedback, Physiological , Fluorescent Antibody Technique , Humans , Microscopy, Confocal , Signal Transduction/physiology
6.
Mol Biol Cell ; 25(1): 184-95, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24196835

ABSTRACT

Epithelial cell differentiation and polarized migration associated with epithelial-to-mesenchymal transition (EMT) in cancer requires integration of gene expression with cytoskeletal dynamics. Here we show that the PDZ-LIM domain protein PDLIM2 (Mystique/SLIM), a known cytoskeletal protein and promoter of nuclear nuclear factor κB (NFκB) and signal transducer and activator of transcription (STAT) degradation, regulates transcription factor activity and gene expression through the COP9 signalosome (CSN). Although repressed in certain cancers, PDLIM2 is highly expressed in invasive cancer cells. Here we show that PDLIM2 suppression causes loss of directional migration, inability to polarize the cytoskeleton, and reversal of the EMT phenotype. This is accompanied by altered activity of several transcription factor families, including ß-catenin, Ap-1, NFκB, interferon regulatory factors, STATs, JUN, and p53. We also show that PDLIM2 associates with CSN5, and cells with suppressed PDLIM2 exhibit reduced nuclear accumulation and deneddylation activity of the CSN toward the cullin 1 and cullin 3 subunits of cullin-RING ubiquitin ligases. Thus PDLIM2 integrates cytoskeleton signaling with gene expression in epithelial differentiation by controlling the stability of key transcription factors and CSN activity.


Subject(s)
Epithelial-Mesenchymal Transition , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/physiology , Microfilament Proteins/physiology , COP9 Signalosome Complex , Cell Differentiation , Cell Movement , Cell Polarity , Epithelial Cells/physiology , Humans , MCF-7 Cells , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Peptide Hydrolases/metabolism , Protein Transport , beta Catenin
7.
Blood ; 115(13): 2709-18, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20124512

ABSTRACT

Inhibition of integrins alphavbeta3 and alphavbeta5 in human brain microvascular endothelial cells (HBMECs) by the function-blocking peptide RGDfV induces loss of spreading on vitronectin, cell detachment, and apoptosis. We demonstrate that cell detachment is not required for apoptosis because plating on bovine serum albumin-blocked poly-L-lysine (allows attachment, but not integrin ligation and cell spreading) also induced apoptosis. Latrunculin B (LatB), which inhibits F-actin polymerization, induced transient loss of HBMEC spreading on vitronectin, but not their detachment, and induced apoptosis despite recovery of cell spreading. However, LatB did not cause apoptosis in 5 tumor cell lines. In HBMECs, both LatB and RGDfV induced transient Y412 and Y245 phosphorylation of endogenous c-Abl, a nonreceptor tyrosine kinase that reciprocally regulates F-actin. LatB also induced nuclear translocation of c-Abl in HBMECs. STI-571 (imatinib), a targeted therapy for BCR-ABL1(+) leukemias and inhibitor of c-Abl, platelet-derived growth factor receptor, and c-Kit, decreased endothelial apoptosis. LatB-induced HBMEC apoptosis, and its inhibition by STI-571 also occurred in a 3-dimensional collagen model, supporting physiologic relevance. Last, siRNA to c-Abl (but not nonspecific siRNA) also inhibited RGDfV- and LatB-induced apoptosis. Thus, endogenous c-Abl mediates endothelial apoptosis induced by inhibition of integrins alphavbeta3/alphavbeta5 or by LatB-induced disruption of F-actin.


Subject(s)
Actins/antagonists & inhibitors , Apoptosis/physiology , Endothelial Cells/cytology , Integrin alphaVbeta3/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/physiology , Receptors, Vitronectin/antagonists & inhibitors , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cattle , Cell Adhesion , Cell Line, Tumor , Cell Shape/drug effects , Culture Media/pharmacology , Culture Media, Serum-Free , Endothelial Cells/drug effects , Fetal Blood , Genes, abl , Glioblastoma/pathology , Humans , Medulloblastoma/pathology , Membrane Potential, Mitochondrial , Neuroblastoma/pathology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Thiazolidines/pharmacology
8.
J Biol Chem ; 285(1): 381-91, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19875448

ABSTRACT

Endocytosis and trafficking of receptors and nutrient transporters are dependent on an acidic intra-endosomal pH that is maintained by the vacuolar H(+)-ATPase (V-ATPase) proton pump. V-ATPase activity has also been associated with cancer invasiveness. Here, we report on a new V-ATPase-associated protein, which we identified in insulin-like growth factor I (IGF-I) receptor-transformed cells, and which was separately identified in Caenorhabditis elegans as HRG-1, a member of a family of heme-regulated genes. We found that HRG-1 is present in endosomes but not in lysosomes, and it is trafficked to the plasma membrane upon nutrient withdrawal in mammalian cells. Suppression of HRG-1 with small interfering RNA causes impaired endocytosis of transferrin receptor, decreased cell motility, and decreased viability of HeLa cells. HRG-1 interacts with the c subunit of the V-ATPase and enhances V-ATPase activity in isolated yeast vacuoles. Endosomal acidity and V-ATPase assembly are decreased in cells with suppressed HRG-1, whereas transferrin receptor endocytosis is enhanced in cells that overexpress HRG-1. Cellular uptake of a fluorescent heme analogue is enhanced by HRG-1 in a V-ATPase-dependent manner. Our findings indicate that HRG-1 regulates V-ATPase activity, which is essential for endosomal acidification, heme binding, and receptor trafficking in mammalian cells. Thus, HRG-1 may facilitate tumor growth and cancer progression.


Subject(s)
Endosomes/drug effects , Endosomes/enzymology , Hemeproteins/metabolism , Insulin-Like Growth Factor I/pharmacology , Receptors, Transferrin/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Survival/drug effects , Endocytosis/drug effects , Gene Expression Regulation/drug effects , Hemeproteins/genetics , Humans , Hydrogen-Ion Concentration/drug effects , Mice , Protein Binding/drug effects , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Saccharomyces cerevisiae/metabolism
9.
Life Sci ; 77(26): 3287-305, 2005 Nov 12.
Article in English | MEDLINE | ID: mdl-16005026

ABSTRACT

This study investigates whether pregnancy or lactation affects microparticle uptake across the small intestinal mucosal barrier, since aspects of gastrointestinal physiology such as motility may be altered in these conditions. It also reports on validation of the model by several methods and discusses the findings in relation to possible mechanisms. Anaesthetised, pregnant, lactating, virgin female or male adult rats were gavaged with fluorescent latex microparticles. The small intestine was removed and fixed either 5 or 30 min later and successive segments of equal length were examined with fluorescence microscopy. Minor adjustments were made to experimental methods to explore details of the uptake mechanism. Control sections contained no particles. All experimental samples showed luminal and surface particles and also contained particles within the tissue, most associated with villous absorptive enterocytes. Particle uptake was greatest at the 30-min time-point, when maximum uptake was usually in the proximal jejunum; although in the early lactating group, this was shifted distally. Total tissue uptake was increased in pregnant and early lactating groups, mainly at villous absorptive and mucus-secreting cells. Accumulation and progression of particles was reflected in increased numbers in the lamina propria. These data were validated by several methods, including particle detection in the blood and mesenteric lymph nodes in some groups. At both time-points, uptake profiles for pregnancy and early lactation differed from those of other groups, implying possible links between particle uptake and hormone levels, surface mucus and tight junction patency.


Subject(s)
Jejunum/physiology , Microspheres , Pregnancy/physiology , Animals , Biological Transport/physiology , Blood/metabolism , Female , Intestinal Mucosa/physiology , Intestinal Mucosa/ultrastructure , Jejunum/ultrastructure , Lactation/physiology , Lymph Nodes/physiology , Male , Rats , Rats, Sprague-Dawley
10.
Blood ; 105(11): 4353-61, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15705795

ABSTRACT

Matrix ligation of integrins alphavbeta3/alphavbeta5 is critical for endothelial survival and angiogenesis. We have previously shown that ceramide, a proapoptotic lipid second messenger, increases during endothelial anoikis (detachment-induced apoptosis). We now show that RGDfV, an integrin alphavbeta3/alphavbeta5 cyclic function-blocking peptide, increased ceramide and decreased sphingomyelin in human brain microvascular endothelial cells (HBMECs) plated on vitronectin, suggesting that sphingomyelin hydrolysis contributes to RGDfV-induced ceramide increase. Desipramine and imipramine, inhibitors of acid sphingomyelinase (ASMase), suppressed RGDfV-induced ceramide increase. Importantly, desipramine, imipramine, and a third ASMase inhibitor, SR33557, but not inhibitors of neutral sphingomyelinase, suppressed RGDfV-induced apoptosis, suggesting that ASMase was required for integrin-mediated apoptosis. Myriocin, an inhibitor of de novo ceramide synthesis, had no effect on RGDfV-induced HBMEC apoptosis. Interestingly, ASMase inhibitors also suppressed the RGDfV-induced loss of spreading on vitronectin. RGDfV induced a similar increase in ceramide and apoptosis in HBMECs on poly-l-lysine or vitronectin, although cells detached only from vitronectin, indicating that cell detachment was not required for RGDfV-induced apoptosis. Our results suggest involvement of ASMase and ceramide in endothelial apoptosis induced by inhibition of integrins alphavbeta3/alphavbeta5, and propose a novel molecular mechanism for the antiangiogenic effect of RGDfV.


Subject(s)
Apoptosis , Ceramides/metabolism , Endothelium, Vascular/cytology , Integrin alphaVbeta3/antagonists & inhibitors , Integrins/antagonists & inhibitors , Receptors, Vitronectin/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Brain/blood supply , Brain/cytology , Humans , Integrin alphaVbeta3/physiology , Integrins/physiology , Oligopeptides/pharmacology , Receptors, Vitronectin/physiology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/physiology , Sphingomyelins/metabolism
11.
Mol Vis ; 9: 665-72, 2003 Dec 10.
Article in English | MEDLINE | ID: mdl-14685146

ABSTRACT

PURPOSE: Progressive dysfunction and death of vascular smooth muscle cells and pericytes is a pathophysiological hallmark of diabetic retinopathy, although the underlying mechanisms behind this process remain ill-defined. The multifunctional peptide platelet-derived growth factor (PDGF) is known to act as an important survival factor for both of these vascular cell-types at times of physiological stress. The retinal cell source(s) of PDGF remain unknown. It is important to understand how diabetes alters expression of this important growth factor. METHODS: Streptozotocin-diabetes was established in C57 mice. Following 8 weeks of sustained diabetes, the eyes were enucleated and in situ hybridization was used to localize expression of PDGF-A and PDGF-B chains in retina from both diabetic and non-diabetic controls. mRNA levels for both forms of PDGF, and their cognate PDGF-alpha and PDGF-beta receptors, were also quantified using real-time PCR. RESULTS: In situ hybridization demonstrated that PDGF-A and PDGF-B were predominantly expressed by the retinal ganglion cells/nerve fibre layer in both normal and diabetic mice, and this localization pattern did not alter in diabetes. PDGF-A receptor was expressed exclusively in the ganglion cell layer of the retina while PDGF-B receptor was mostly localized to the Muller cell end-feet at the internal limiting membrane with lesser immunoreactivity in the ganglion cells, inner plexiform layer, and inner nuclear layer. PDGF-A and PDGF-alpha receptor mRNA expression levels remained unaltered between treatment groups, although retinal immunolocalization patterns between both receptors was distinct. However, there was a significant decrease of PDGF-B mRNA levels in diabetic retina when compared to non-diabetic controls (p<0.001), although there was no significant difference in PDGF-alpha receptor(insert space) expression. CONCLUSIONS: Previous studies have shown PDGF expression in a range of cell-types during retinal development, but these results confirm ganglion cells as the principal PDGF source in mature retina. It may be significant that diabetes can reduce PDGF-B mRNA expression since this may have serious implications for vascular survival during diabetic retinopathy progression.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Nerve Fibers/metabolism , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins c-sis/genetics , Retinal Ganglion Cells/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/pathology , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis/metabolism , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...