Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Cell Adh Migr ; 18(1): 1-3, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38860721

ABSTRACT

In September 2023 members of the cell adhesion and cell migration research community came together to share their latest research and consider how our work might be translated for clinical practice. Alongside invited speakers, selected speakers and poster presentations, the meeting also included a round table discussion of how we might overcome the challenges associated with research translation. This meeting report seeks to highlight the key outcomes of that discussion and spark interest in the cell adhesions and cell migration research community to cross the perceived valley of death and translate our work into therapeutic benefit.


Subject(s)
Cell Adhesion , Cell Movement , Humans , Translational Research, Biomedical , Animals , Neoplasms/pathology , Neoplasms/therapy
2.
Cell Signal ; 121: 111233, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38763182

ABSTRACT

Breast cancer is a heterogeneous disease that remains the most common malignancy among women worldwide. During genomic analysis of breast tumours, mRNA levels of IQGAP3 were found to be upregulated in triple negative tumours. IQGAP3 was subsequently found to be expressed across a panel of triple negative breast cancer (TNBC) cell lines. Depleting expression levels of IQGAP3 delivered elongated cells, disrupted cell migration, and inhibited the ability of cells to form specialised invasive adhesion structures, termed invadopodia. The morphological changes induced by IQGAP3 depletion were found to be dependent on RhoA. Indeed, reduced expression of IQGAP3 disrupted RhoA activity and actomyosin contractility. Interestingly, IQGAP3 was also found to interact with p-21 activated kinase 6 (PAK6); a protein already associated with the regulation of cell morphology. Moreover, PAK6 depletion phenocopied IQGAP3 depletion in these cells. Whereas PAK6 overexpression rescued the IQGAP3 depletion phenotype. Our work points to an important PAK6-IQGAP3-RhoA pathway that drives the cellular contractility of breast cancer cells promoting both cell migration and adhesive invasion of these cells. As this phenotype is relevant to the process of metastasis and re-seeding of metastasis, the pharmacological targeting of PAK6 could lead to clinical benefit in TNBC patients.

3.
Sci Adv ; 9(42): eadi0244, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37851808

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a very poor prognosis because of its high propensity to metastasize and its immunosuppressive microenvironment. Using a panel of pancreatic cancer cell lines, three-dimensional (3D) invasion systems, microarray gene signatures, microfluidic devices, mouse models, and intravital imaging, we demonstrate that ROCK-Myosin II activity in PDAC cells supports a transcriptional program conferring amoeboid invasive and immunosuppressive traits and in vivo metastatic abilities. Moreover, we find that immune checkpoint CD73 is highly expressed in amoeboid PDAC cells and drives their invasive, metastatic, and immunomodulatory traits. Mechanistically, CD73 activates RhoA-ROCK-Myosin II downstream of PI3K. Tissue microarrays of human PDAC biopsies combined with bioinformatic analysis reveal that rounded-amoeboid invasive cells with high CD73-ROCK-Myosin II activity and their immunosuppressive microenvironment confer poor prognosis to patients. We propose targeting amoeboid PDAC cells as a therapeutic strategy.


Subject(s)
Adenocarcinoma , Amoeba , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Humans , Mice , Adenocarcinoma/pathology , Amoeba/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cytoskeletal Proteins , Immunosuppression Therapy , Myosin Type II/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment
4.
Biochem J ; 479(16): 1709-1725, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35969127

ABSTRACT

The protein kinases PAK4, PAK5 and PAK6 comprise a family of ohnologues. In multiple cancers including melanomas PAK5 most frequently carries non-synonymous mutations; PAK6 and PAK4 have fewer; and PAK4 is often amplified. To help interpret these genomic data, initially we compared the cellular regulation of the sister kinases and their roles in melanoma cells. In common with many ohnologue protein kinases, PAK4, PAK5 and PAK6 each have two 14-3-3-binding phosphosites of which phosphoSer99 is conserved. PAK4 localises to the leading edge of cells in response to phorbol ester-stimulated binding of 14-3-3 to phosphoSer99 and phosphoSer181, which are phosphorylated by two different PKCs or PKDs. These phosphorylations of PAK4 are essential for its phorbol ester-stimulated phosphorylation of downstream substrates. In contrast, 14-3-3 interacts with PAK5 in response to phorbol ester-stimulated phosphorylation of Ser99 and epidermal growth factor-stimulated phosphorylation of Ser288; whereas PAK6 docks onto 14-3-3 and is prevented from localising to cell-cell junctions when Ser133 is phosphorylated in response to cAMP-elevating agents via PKA and insulin-like growth factor 1 via PKB/Akt. Silencing of PAK4 impairs viability, migration and invasive behaviour of melanoma cells carrying BRAFV600E or NRASQ61K mutations. These defects are rescued by ectopic expression of PAK4, more so by a 14-3-3-binding deficient PAK4, and barely by PAK5 or PAK6. Together these genomic, biochemical and cellular data suggest that the oncogenic properties of PAK4 are regulated by PKC-PKD signalling in melanoma, while PAK5 and PAK6 are dispensable in this cancer.


Subject(s)
Melanoma , Protein Kinases , Humans , Melanoma/genetics , Phorbol Esters , Phosphorylation , Protein Kinases/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
5.
Int J Biochem Cell Biol ; 146: 106207, 2022 05.
Article in English | MEDLINE | ID: mdl-35385780

ABSTRACT

Metastatic Breast Cancer has a poor 25% survival rate and currently there are no clinical therapeutics which target metastasis. 'Migrastatics' are a new drug class which target migration pathway effector proteins in order to inhibit cancer cell invasion and metastasis. The p21-activated kinases (PAKs) are essential drivers of breast cancer cell migration and invasion through their regulation of actin cytoskeletal dynamics. Therefore, the PAKs present as attractive migrastatic candidates. Here we review how PAKs regulate distinct aspects of breast cancer actin dynamics focussing on cytoskeletal reorganisation, cell:matrix adhesion, actomyosin contractility and degradative invasion. Lastly, we discuss the introduction of PAK migrastatics into the well-honed breast cancer clinical pipeline.


Subject(s)
Actins , Breast Neoplasms , Actins/metabolism , Breast Neoplasms/metabolism , Cytoskeleton/metabolism , Female , Humans , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
6.
BMC Cancer ; 22(1): 386, 2022 Apr 09.
Article in English | MEDLINE | ID: mdl-35397545

ABSTRACT

BACKGROUND: Invadopodia, actin-rich structures that release metallo-proteases at the interface with extra-cellular matrix, in a punctate manner are thought to be important drivers of tumour invasion. Invadopodia formation has been observed in-vitro and in-vivo in numerous metastatic cell lines derived from multiple tumour types. However, prostate cancer cell lines have not been routinely reported to generate invadopodia and the few instances have always required external stimulation. METHODS: In this study, the invasive potential of primary prostate adenocarcinoma cell lines, which have never been fully characterised before, was investigated both in-vitro invadopodia assays and in-vivo zebrafish dissemination assay. Subsequently, circulating tumour cells from prostate cancer patients were isolated and tested in the invadopodia assay. RESULTS: Retention of E-cadherin and N-cadherin expression indicated a transitional state of EMT progression, consistent with the idea of partial EMT that has been frequently observed in aggressive prostate cancer. All cell lines tested were capable of spontaneous invadopodia formation and possess a significant degradative ability in-vitro under basal conditions. These cell lines were invasive in-vivo and produced visible metastasis in the zebrafish dissemination assay. Importantly we have proceeded to demonstrate that circulating tumour cells isolated from prostate cancer patients exhibit invadopodia-like structures and degrade matrix with visible puncta. This work supports a role for invadopodia activity as one of the mechanisms of dissemination employed by prostate cancer cells. CONCLUSION: The combination of studies presented here provide clear evidence that invadopodia activity can play a role in prostate cancer progression.


Subject(s)
Neoplastic Cells, Circulating , Podosomes , Prostatic Neoplasms , Animals , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Male , Neoplasm Invasiveness/pathology , Neoplastic Cells, Circulating/metabolism , Podosomes/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Zebrafish
8.
Small GTPases ; 12(4): 265-272, 2021 07.
Article in English | MEDLINE | ID: mdl-33043786

ABSTRACT

Fatty acid synthase (FASN) is commonly overexpressed in prostate cancer and associated with tumour progression. FASN is responsible for de novo synthesis of the fatty acid palmitate; the building block for protein palmitoylation. A functional role for FASN in regulating cell proliferation is widely accepted. We recently reported that FASN activity can also mediate prostate cancer HGF-mediated cell motility. Moreover, we found that modulation of FASN expression specifically impacts on the palmitoylation of RhoU. Findings we will describe here. We now report that loss of FASN expression also impairs HGF-mediated cell dissociation responses. Taken together our results provide compelling evidence that FASN activity directly promotes cell migration and supports FASN as a potential therapeutic target in metastatic prostate cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Fatty Acid Synthase, Type I/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/pharmacology , Prostatic Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Fatty Acid Synthase, Type I/genetics , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Tumor Cells, Cultured
9.
Front Cell Dev Biol ; 9: 759259, 2021.
Article in English | MEDLINE | ID: mdl-35111748

ABSTRACT

p21-Activated kinase-1 (Pak1) is frequently overexpressed and/or amplified in human breast cancer and is necessary for transformation of mammary epithelial cells. Here, we show that Pak1 interacts with and phosphorylates the Calcium/Calmodulin-dependent Protein Kinase II (CaMKII), and that pharmacological inhibition or depletion of Pak1 leads to diminished activity of CaMKII. We found a strong correlation between Pak1 and CaMKII expression in human breast cancer samples, and combined inhibition of Pak1 and CaMKII with small-molecule inhibitors was synergistic and induced apoptosis more potently in Her2 positive and triple negative breast cancer (TNBC) cells. Co-adminstration of Pak and CaMKII small-molecule inhibitors resulted in a dramatic reduction of proliferation and an increase in apoptosis in a 3D cell culture setting, as well as an impairment in migration and invasion of TNBC cells. Finally, mice bearing xenografts of TNBC cells showed a significant delay in tumor growth when treated with small-molecule inhibitors of Pak and CaMKII. These data delineate a signaling pathway from Pak1 to CaMKII that is required for efficient proliferation, migration and invasion of mammary epithelial cells, and suggest new therapeutic strategies in breast cancer.

10.
Biomaterials ; 264: 120369, 2021 01.
Article in English | MEDLINE | ID: mdl-32977209

ABSTRACT

With a dismal survival rate, pancreatic cancer (PC) remains one of the most aggressive and devastating malignancies, predominantly due to the absence of a valid biomarker for diagnosis and limited therapeutic options for advanced diseases. Exosomes (Exo) as cell-derived vesicles, are widely used as natural nanocarriers for drug delivery. P21-activated kinase 4 (PAK4) is oncogenic when overexpressed, promoting cell survival, migration and anchorage-independent growth. Herein we validated PAK4 as a therapeutic target in an in vivo PC tumour mouse model using Exo-mediated RNAi following intra-tumoural administration. PC derived Exo were firstly isolated by ultracentrifugation on sucrose cushion and characterised for their surface marker expression, size, number, purity and morphology. SiRNA was encapsulated into Exo via electroporation and dual uptake of Exo and siRNA was investigated by flow cytometry and confocal microscopy. In vitro siPAK4 silencing in PC cells following uptake was assessed by flow cytometry, western blotting, and in vitro scratch assay. In vivo efficacy (tumour growth delay and mouse survival) of siPAK4 was evaluated in PC bearing NSG mouse model. Ex vivo tumours were examined using Haematoxylin and eosin (H&E) staining and immunohistochemistry. Results showed high quality PC-derived PANC-1 Exo were obtained. SiRNA was incorporated in Exo with 16.5% encapsulation efficiency. In vitro imaging confirmed Exo and siRNA co-localisation in cells. PAK4 knockdown was successful with 30 nM Exo-siPAK4 at 24 h post incubation in vitro. Intra-tumoural administration of Exo-siPAK4 (0.03 mg/kg siPAK4 and 6.1 × 1011 Exo, each dose, two doses) reduced PC tumour growth in vivo and enhanced mice survival (p < 0.001), with minimal toxicity observed compared to polyethylenimine (PEI) used as a commercial transfection reagent. H&E staining of tumours showed significant tissue apoptosis in siPAK4 treated groups. PAK4 knockdown prolongs survival of PC-bearing mice suggesting its potential as a new therapeutic target for PC. PANC-1 Exo demonstrated comparable efficacy but safer profile than PEI as in vivo RNAi transfection reagent.


Subject(s)
Exosomes , Pancreatic Neoplasms , Animals , Cell Line, Tumor , Exosomes/metabolism , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , RNA Interference , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
12.
Oncogene ; 39(18): 3666-3679, 2020 04.
Article in English | MEDLINE | ID: mdl-32139877

ABSTRACT

Fatty acid synthase (FASN) is commonly overexpressed in prostate cancer and associated with tumour progression. FASN is responsible for de novo synthesis of the fatty acid palmitate; the building block for protein palmitoylation. Recent work has suggested that alongside its established role in promoting cell proliferation FASN may also promote invasion. We now find depletion of FASN expression increases prostate cancer cell adhesiveness, impairs HGF-mediated cell migration and reduces 3D invasion. These changes in motility suggest that FASN can mediate actin cytoskeletal remodelling; a process known to be downstream of Rho family GTPases. Here, we demonstrate that modulation of FASN expression specifically impacts on the palmitoylation of the atypical GTPase RhoU. Impaired RhoU activity in FASN depleted cells leads to reduced adhesion turnover downstream of paxillin serine phosphorylation, which is rescued by addition of exogenous palmitate. Moreover, canonical Cdc42 expression is dependent on the palmitoylation status of RhoU. Thus we uncover a novel relationship between FASN, RhoU and Cdc42 that directly influences cell migration potential. These results provide compelling evidence that FASN activity directly promotes cell migration and supports FASN as a potential therapeutic target in metastatic prostate cancer.


Subject(s)
Fatty Acid Synthase, Type I/genetics , Lipogenesis/genetics , Prostatic Neoplasms/genetics , cdc42 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Phosphorylation/genetics , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction/genetics
13.
BMC Cancer ; 19(1): 1214, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31836008

ABSTRACT

BACKGROUND: Monocytes are a major component of the tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC). However, the complex interactions between tumor cells and monocytes and their role in tumor invasion have not been fully established. METHODS: To specifically test the impact of interaction on invasive potential two PDAC cell lines PaTu8902 and CFPAC-1 were selected on their ability to form invasive adhesions, otherwise known as invadopodia and invade in a spheroid invasion assay. RESULTS: Interestingly when the PDAC cells were co-cultured with undifferentiated THP1 monocyte-like cells invadopodia formation was significantly suppressed. Moreover, conditioned media of THP1 cells (CM) was also able to suppress invadopodia formation. Further investigation revealed that both tissue inhibitor of metalloproteinase (TIMP) 1 and 2 were present in the CM. However, suppression of invadopodia formation was found that was specific to TIMP2 activity. CONCLUSIONS: Our findings indicate that TIMP2 levels in the tumour microenvironment may have prognostic value in patients with PDAC. Furthermore, activation of TIMP2 expressing monocytes in the primary tumour could present a potential therapeutic opportunity to suppress cell invasion in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Communication/physiology , Monocytes/metabolism , Pancreatic Neoplasms/metabolism , Podosomes/pathology , Tissue Inhibitor of Metalloproteinase-2/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/physiology , Coculture Techniques , Humans , Monocytes/pathology , Pancreatic Neoplasms/pathology , Podosomes/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , THP-1 Cells , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tumor Microenvironment
14.
Cells ; 9(1)2019 12 18.
Article in English | MEDLINE | ID: mdl-31861264

ABSTRACT

Androgen receptor (AR) and glucocorticoid receptor (GR) are nuclear receptors whose function depends on their entry into the nucleus where they activate transcription of an overlapping set of genes. Both AR and GR have a role in resistance to androgen deprivation therapy (ADT), the mainstay of treatment for late stage prostate cancer. PlexinB1, a receptor for semaphorins, has been implicated in various cancers including prostate cancer and has a role in resistance to ADT. We show here that activation of PlexinB1 by Sema4D and Sema3C results in translocation of endogenous GR to the nucleus in prostate cancer cells, and that this effect is dependent on PlexinB1 expression. Sema4D/Sema3C promotes the translocation of GR-GFP to the nucleus and mutation of the nuclear localization sequence (NLS1) of GR abrogates this response. These findings implicate the importin α/ß system in the Sema4D/Sema3C-mediated nuclear import of GR. Knockdown of PlexinB1 in prostate cancer cells decreases the levels of glucocorticoid-responsive gene products and antagonizes the decrease in cell motility and cell area of prostate cancer cells upon dexamethasone treatment, demonstrating the functional significance of these findings. These results show that PlexinB1 activation has a role in the trafficking and activation of the nuclear receptor GR and thus may have a role in resistance to androgen deprivation therapy in late stage prostate cancer.


Subject(s)
Cell Nucleus/metabolism , Nerve Tissue Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Cell Surface/genetics , Receptors, Glucocorticoid/metabolism , Antigens, CD/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Dexamethasone/pharmacology , HeLa Cells , Humans , Male , Mutation , Nerve Tissue Proteins/metabolism , Nuclear Localization Signals , PC-3 Cells , Prostatic Neoplasms/metabolism , Protein Transport , Receptors, Cell Surface/metabolism , Receptors, Glucocorticoid/genetics , Semaphorins/metabolism , Transcriptional Activation
15.
Cell Rep ; 29(11): 3385-3393.e6, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31825823

ABSTRACT

p21-Activated kinase 4 (PAK4), a serine/threonine kinase, is purported to localize to podosomes: transient adhesive structures that degrade the extracellular matrix to facilitate rapid myeloid cell migration. We find that treatment of transforming growth factor ß (TGF-ß)-differentiated monocytic (THP-1) cells with a PAK4-targeted inhibitor significantly reduces podosome formation and induces the formation of focal adhesions. This switch in adhesions confers a diminution of matrix degradation and reduced cell migration. Furthermore, reduced PAK4 expression causes a significant reduction in podosome number that cannot be rescued by kinase-dead PAK4, supporting a kinase-dependent role. Concomitant with PAK4 depletion, phosphorylation of Akt is perturbed, whereas a specific phospho-Akt signal is detected within the podosomes. Using superresolution analysis, we find that PAK4 specifically localizes in the podosome ring, nearer to the actin core than other ring proteins. We propose PAK4 kinase activity intersects with the Akt pathway at the podosome ring:core interface to drive regulation of macrophage podosome turnover.


Subject(s)
Myeloid Cells/metabolism , Podosomes/metabolism , p21-Activated Kinases/metabolism , Cells, Cultured , Disulfides/pharmacology , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , HEK293 Cells , Humans , Myeloid Cells/drug effects , Myeloid Cells/ultrastructure , Naphthols/pharmacology , Phosphorylation , Podosomes/ultrastructure , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , THP-1 Cells , p21-Activated Kinases/antagonists & inhibitors
16.
Sci Total Environ ; 692: 966-974, 2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31540000

ABSTRACT

The Chernobyl Exclusion Zone (CEZ) represents a unique natural laboratory that received significant 129I contamination across a range of soils and land-use types in a short time period in 1986. Data are presented on 129I and 127I in soil samples collected from highly contaminated areas in the CEZ in 2015. The geometric mean (GM) total concentration of stable iodine (127I) was 6.7 × 10-7 g g-1 and the (GM) total concentration of 129I was 2.39 × 10-13 g g-1, equivalent to 1.56 mBq kg-1. GM total 127I concentration is below the European average soil concentration of 3.94 × 10-6 g g-1, while 129I is significantly higher than the pre-Chernobyl activity concentration for 129I of 0.094 mBq kg-1. Significant differences were found in the extractability of native, stable 127I and 129I almost 30 years after the introduction of 129I to the soils. Both 127I and 129I were predominantly associated with alkaline-extractable soil organic matter, established using a three-step sequential extraction procedure. Whereas 127I was significantly correlated with gross soil organic matter (measured by loss on ignition), however, 129I was not. The ratio of 129I/127I was significantly lower in extracts of soil organic matter than in more labile (soluble and adsorbed) fractions, indicating incomplete equilibration of 129I with native 127I in soil humic substances after 29 years residence time in the CEZ soils. The initial physico-chemical form of 129I in the CEZ soils is unknown, but the widespread presence of uranium oxide fuel particles is unlikely to have influenced the environmental behaviour of 129I. Our findings have implications for long-term radiation dose from 129I in contaminated soils and the use of native, stable 127I as a proxy for the long-term fate of 129I.

17.
Cell Death Dis ; 10(3): 180, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30792401

ABSTRACT

Potassium ion channels are critical in the regulation of cell motility. The acquisition of cell motility is an essential parameter of cancer metastasis. However, the role of K+ channels in cancer metastasis has been poorly studied. High expression of the hG1 gene, which encodes for Kv11.1 channel associates with good prognosis in estrogen receptor-negative breast cancer (BC). We evaluated the efficacy of the Kv11.1 activator NS1643 in arresting metastasis in a triple negative breast cancer (TNBC) mouse model. NS1643 significantly reduces the metastatic spread of breast tumors in vivo by inhibiting cell motility, reprogramming epithelial-mesenchymal transition via attenuation of Wnt/ß-catenin signaling and suppressing cancer cell stemness. Our findings provide important information regarding the clinical relevance of potassium ion channel expression in breast tumors and the mechanisms by which potassium channel activity can modulate tumor biology. Findings suggest that Kv11.1 activators may represent a novel therapeutic approach for the treatment of metastatic estrogen receptor-negative BC. Ion channels are critical factor for cell motility but little is known about their role in metastasis. Stimulation of the Kv11.1 channel suppress the metastatic phenotype in TNBC. This work could represent a paradigm-shifting approach to reducing mortality by targeting a pathway that is central to the development of metastases.


Subject(s)
ERG1 Potassium Channel/metabolism , Epithelial-Mesenchymal Transition , Triple Negative Breast Neoplasms/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cresols/pharmacology , Cresols/therapeutic use , ERG1 Potassium Channel/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , MCF-7 Cells , Mice , Neoplasm Metastasis , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Transplantation, Heterologous , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
18.
Small GTPases ; 10(4): 289-295, 2019 07.
Article in English | MEDLINE | ID: mdl-28301299

ABSTRACT

PAK1 and PAK4 are members of the p-21 activated kinase family of serine/threonine kinases. PAK1 has previously been implicated in both the formation and disassembly of invasive cell protrusions, termed invadopodia. We recently reported a novel role for PAK4 during invadopodia maturation and confirmed a specific role for PAK1 in invadopodia formation; findings we will review here. Moreover, we found that PAK4 induction of maturation is delivered via interaction with the RhoA regulator PDZ-RhoGEF. We can now reveal that loss of PAK4 expression leads to changes in invadopodia dynamics. Ultimately we propose that PAK4 but not PAK1 is a key mediator of RhoA activity and provide further evidence that modulation of PAK4 expression levels leads to changes in RhoA activity.


Subject(s)
Melanoma/metabolism , Podosomes/metabolism , p21-Activated Kinases/metabolism , Cell Line , Humans , Melanoma/genetics , Neoplasm Invasiveness , Rho Guanine Nucleotide Exchange Factors/metabolism , p21-Activated Kinases/genetics , rhoA GTP-Binding Protein/metabolism
19.
J Environ Radioact ; 192: 128-142, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29929171

ABSTRACT

Many of the freshwater Kd values required for quantifying radionuclide transfer in the environment (e.g. ERICA Tool, Symbiose modelling platform) are either poorly reported in the literature or not available. To partially address this deficiency, Working Group 4 of the IAEA program MODARIA (2012-2015) has completed an update of the freshwater Kd databases and Kd distributions given in TRS 472 (IAEA, 2010). Over 2300 new values for 27 new elements were added to the dataset and 270 new Kd values were added for the 25 elements already included in TRS 472 (IAEA, 2010). For 49 chemical elements, the Kd values have been classified according to three solid-liquid exchange conditions (adsorption, desorption and field) as was previously carried out in TRS 472. Additionally, the Kd values were classified into two environmental components (suspended and deposited sediments). Each combination (radionuclide x component x condition) was associated with log-normal distributions when there was at least ten Kd values in the dataset and to a geometric mean when there was less than ten values. The enhanced Kd dataset shows that Kd values for suspended sediments are significantly higher than for deposited sediments and that the variability of Kd distributions are higher for deposited than for suspended sediments. For suspended sediments in field conditions, the variability of Kd distributions can be significantly reduced as a function of the suspended load that explains more than 50% of the variability of the Kd datasets of U, Si, Mo, Pb, S, Se, Cd, Ca, B, K, Ra and Po. The distinction between adsorption and desorption conditions is justified for deterministic calculations because the geometric means are systematically greater in desorption conditions. Conversely, this distinction is less relevant for probabilistic calculations due to systematic overlapping between the Kd distributions of these two conditions.


Subject(s)
Fresh Water/chemistry , Radiation Monitoring/methods , Water Pollutants, Radioactive/analysis , Adsorption , Geologic Sediments , Radiation Monitoring/standards , Radioisotopes/analysis , Water Pollutants, Radioactive/standards
20.
Front Syst Neurosci ; 12: 7, 2018.
Article in English | MEDLINE | ID: mdl-29765309

ABSTRACT

The rat has arguably the most widely studied brain among all animals, with numerous reference atlases for rat brain having been published since 1946. For example, many neuroscientists have used the atlases of Paxinos and Watson (PW, first published in 1982) or Swanson (S, first published in 1992) as guides to probe or map specific rat brain structures and their connections. Despite nearly three decades of contemporaneous publication, no independent attempt has been made to establish a basic framework that allows data mapped in PW to be placed in register with S, or vice versa. Such data migration would allow scientists to accurately contextualize neuroanatomical data mapped exclusively in only one atlas with data mapped in the other. Here, we provide a tool that allows levels from any of the seven published editions of atlases comprising three distinct PW reference spaces to be aligned to atlas levels from any of the four published editions representing S reference space. This alignment is based on registration of the anteroposterior stereotaxic coordinate (z) measured from the skull landmark, Bregma (ß). Atlas level alignments performed along the z axis using one-dimensional Cleveland dot plots were in general agreement with alignments obtained independently using a custom-made computer vision application that utilized the scale-invariant feature transform (SIFT) and Random Sample Consensus (RANSAC) operation to compare regions of interest in photomicrographs of Nissl-stained tissue sections from the PW and S reference spaces. We show that z-aligned point source data (unpublished hypothalamic microinjection sites) can be migrated from PW to S space to a first-order approximation in the mediolateral and dorsoventral dimensions using anisotropic scaling of the vector-formatted atlas templates, together with expert-guided relocation of obvious outliers in the migrated datasets. The migrated data can be contextualized with other datasets mapped in S space, including neuronal cell bodies, axons, and chemoarchitecture; to generate data-constrained hypotheses difficult to formulate otherwise. The alignment strategies provided in this study constitute a basic starting point for first-order, user-guided data migration between PW and S reference spaces along three dimensions that is potentially extensible to other spatial reference systems for the rat brain.

SELECTION OF CITATIONS
SEARCH DETAIL
...