Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Publication year range
1.
J Cell Sci ; 132(11)2019 05 31.
Article in English | MEDLINE | ID: mdl-31152052

ABSTRACT

Cancer cells are softer than the normal cells, and metastatic cells are even softer. These changes in biomechanical properties contribute to cancer progression by facilitating cell movement through physically constraining environments. To identify properties that enabled passage through physical constraints, cells that were more efficient at moving through narrow membrane micropores were selected from established cell lines. By examining micropore-selected human MDA MB 231 breast cancer and MDA MB 435 melanoma cancer cells, membrane fluidity and nuclear elasticity were excluded as primary contributors. Instead, reduced actin cytoskeleton anisotropy, focal adhesion density and cell stiffness were characteristics associated with efficient passage through constraints. By comparing transcriptomic profiles between the parental and selected populations, increased Ras/MAPK signalling was linked with cytoskeleton rearrangements and cell softening. MEK inhibitor treatment reversed the transcriptional, cytoskeleton, focal adhesion and elasticity changes. Conversely, expression of oncogenic KRas in parental MDA MB 231 cells, or oncogenic BRaf in parental MDA MB 435 cells, significantly reduced cell stiffness. These results reveal that MAPK signalling, in addition to tumour cell proliferation, has a significant role in regulating cell biomechanics.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Actin Cytoskeleton/physiology , Biomechanical Phenomena/physiology , Cell Movement/physiology , MAP Kinase Signaling System/physiology , Melanoma/physiopathology , Anisotropy , Cell Line, Tumor , Cell Plasticity/physiology , Cell Proliferation , Focal Adhesions/physiology , Humans , Micropore Filters , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism
2.
Gastroenterology ; 159(1): 183-199, 2020 07.
Article in English | MEDLINE | ID: mdl-32179094

ABSTRACT

BACKGROUND & AIMS: Intestinal epithelial homeostasis depends on a tightly regulated balance between intestinal epithelial cell (IEC) death and proliferation. While the disruption of several IEC death regulating factors result in intestinal inflammation, the loss of the anti-apoptotic BCL2 family members BCL2 and BCL2L1 has no effect on intestinal homeostasis in mice. We investigated the functions of the antiapoptotic protein MCL1, another member of the BCL2 family, in intestinal homeostasis in mice. METHODS: We generated mice with IEC-specific disruption of Mcl1 (Mcl1ΔIEC mice) or tamoxifen-inducible IEC-specific disruption of Mcl1 (i-Mcl1ΔIEC mice); these mice and mice with full-length Mcl1 (controls) were raised under normal or germ-free conditions. Mice were analyzed by endoscopy and for intestinal epithelial barrier permeability. Intestinal tissues were analyzed by histology, in situ hybridization, proliferation assays, and immunoblots. Levels of calprotectin, a marker of intestinal inflammation, were measured in intestinal tissues and feces. RESULTS: Mcl1ΔIEC mice spontaneously developed apoptotic enterocolopathy, characterized by increased IEC apoptosis, hyperproliferative crypts, epithelial barrier dysfunction, and chronic inflammation. Loss of MCL1 retained intestinal crypts in a hyperproliferated state and prevented the differentiation of intestinal stem cells. Proliferation of intestinal stem cells in MCL1-deficient mice required WNT signaling and was associated with DNA damage accumulation. By 1 year of age, Mcl1ΔIEC mice developed intestinal tumors with morphologic and genetic features of human adenomas and carcinomas. Germ-free housing of Mcl1ΔIEC mice reduced markers of microbiota-induced intestinal inflammation but not tumor development. CONCLUSION: The antiapoptotic protein MCL1, a member of the BCL2 family, is required for maintenance of intestinal homeostasis and prevention of carcinogenesis in mice. Loss of MCL1 results in development of intestinal carcinomas, even under germ-free conditions, and therefore does not involve microbe-induced chronic inflammation. Mcl1ΔIEC mice might be used to study apoptotic enterocolopathy and inflammatory bowel diseases.


Subject(s)
Carcinoma/pathology , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinoma/diagnosis , Carcinoma/genetics , Disease Models, Animal , Endoscopy , Epithelial Cells/pathology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/diagnostic imaging , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/genetics , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/genetics
3.
PLoS Genet ; 13(7): e1006870, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28708826

ABSTRACT

Wnt/ß-catenin signal transduction directs intestinal stem cell (ISC) proliferation during homeostasis. Hyperactivation of Wnt signaling initiates colorectal cancer, which most frequently results from truncation of the tumor suppressor Adenomatous polyposis coli (APC). The ß-catenin-TCF transcription complex activates both the physiological expression of Wnt target genes in the normal intestinal epithelium and their aberrantly increased expression in colorectal tumors. Whether mechanistic differences in the Wnt transcription machinery drive these distinct levels of target gene activation in physiological versus pathological states remains uncertain, but is relevant for the design of new therapeutic strategies. Here, using a Drosophila model, we demonstrate that two evolutionarily conserved transcription cofactors, Earthbound (Ebd) and Erect wing (Ewg), are essential for all major consequences of Apc1 inactivation in the intestine: the hyperactivation of Wnt target gene expression, excess number of ISCs, and hyperplasia of the epithelium. In contrast, only Ebd, but not Ewg, mediates the Wnt-dependent regulation of ISC proliferation during homeostasis. Therefore, in the adult intestine, Ebd acts independently of Ewg in physiological Wnt signaling, but cooperates with Ewg to induce the hyperactivation of Wnt target gene expression following Apc1 loss. These findings have relevance for human tumorigenesis, as Jerky (JRK/JH8), the human Ebd homolog, promotes Wnt pathway hyperactivation and is overexpressed in colorectal, breast, and ovarian cancers. Together, our findings reveal distinct requirements for Ebd and Ewg in physiological Wnt pathway activation versus oncogenic Wnt pathway hyperactivation following Apc1 loss. Such differentially utilized transcription cofactors may offer new opportunities for the selective targeting of Wnt-driven cancers.


Subject(s)
Centromere Protein B/genetics , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Neoplasms/genetics , Neuropeptides/genetics , Nuclear Proteins/biosynthesis , Transcription Factors/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Centromere Protein B/biosynthesis , DNA-Binding Proteins , Disease Models, Animal , Drosophila Proteins/biosynthesis , Epithelium/growth & development , Epithelium/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Hyperplasia/genetics , Hyperplasia/pathology , Intestines/growth & development , Neoplasms/pathology , Neuropeptides/biosynthesis , Nuclear Proteins/genetics , RNA-Binding Proteins , Stem Cells/metabolism , Transcription Factors/biosynthesis , Wnt Signaling Pathway/genetics
4.
EMBO J ; 34(18): 2321-33, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26240067

ABSTRACT

Wnt pathway deregulation is a common characteristic of many cancers. Only colorectal cancer predominantly harbours mutations in APC, whereas other cancer types (hepatocellular carcinoma, solid pseudopapillary tumours of the pancreas) have activating mutations in ß-catenin (CTNNB1). We have compared the dynamics and the potency of ß-catenin mutations in vivo. Within the murine small intestine (SI), an activating mutation of ß-catenin took much longer to achieve Wnt deregulation and acquire a crypt-progenitor cell (CPC) phenotype than Apc or Gsk3 loss. Within the colon, a single activating mutation of ß-catenin was unable to drive Wnt deregulation or induce the CPC phenotype. This ability of ß-catenin mutation to differentially transform the SI versus the colon correlated with higher expression of E-cadherin and a higher number of E-cadherin:ß-catenin complexes at the membrane. Reduction in E-cadherin synergised with an activating mutation of ß-catenin resulting in a rapid CPC phenotype within the SI and colon. Thus, there is a threshold of ß-catenin that is required to drive transformation, and E-cadherin can act as a buffer to sequester mutated ß-catenin.


Subject(s)
Cadherins/metabolism , Cell Transformation, Neoplastic , Colonic Neoplasms , Mutation , Neoplasm Proteins , Wnt Signaling Pathway , beta Catenin , Animals , Cadherins/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
5.
PLoS Biol ; 12(10): e1001966, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25313567

ABSTRACT

The high mortality of melanoma is caused by rapid spread of cancer cells, which occurs unusually early in tumour evolution. Unlike most solid tumours, thickness rather than cytological markers or differentiation is the best guide to metastatic potential. Multiple stimuli that drive melanoma cell migration have been described, but it is not clear which are responsible for invasion, nor if chemotactic gradients exist in real tumours. In a chamber-based assay for melanoma dispersal, we find that cells migrate efficiently away from one another, even in initially homogeneous medium. This dispersal is driven by positive chemotaxis rather than chemorepulsion or contact inhibition. The principal chemoattractant, unexpectedly active across all tumour stages, is the lipid agonist lysophosphatidic acid (LPA) acting through the LPA receptor LPAR1. LPA induces chemotaxis of remarkable accuracy, and is both necessary and sufficient for chemotaxis and invasion in 2-D and 3-D assays. Growth factors, often described as tumour attractants, cause negligible chemotaxis themselves, but potentiate chemotaxis to LPA. Cells rapidly break down LPA present at substantial levels in culture medium and normal skin to generate outward-facing gradients. We measure LPA gradients across the margins of melanomas in vivo, confirming the physiological importance of our results. We conclude that LPA chemotaxis provides a strong drive for melanoma cells to invade outwards. Cells create their own gradients by acting as a sink, breaking down locally present LPA, and thus forming a gradient that is low in the tumour and high in the surrounding areas. The key step is not acquisition of sensitivity to the chemoattractant, but rather the tumour growing to break down enough LPA to form a gradient. Thus the stimulus that drives cell dispersal is not the presence of LPA itself, but the self-generated, outward-directed gradient.


Subject(s)
Cell Movement , Chemotaxis , Lysophospholipids/metabolism , Melanoma/metabolism , Neoplasm Metastasis , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Mice
6.
Nature ; 477(7363): 225-8, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849978

ABSTRACT

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid cycle (TCA cycle) that catalyses the hydration of fumarate into malate. Germline mutations of FH are responsible for hereditary leiomyomatosis and renal-cell cancer (HLRCC). It has previously been demonstrated that the absence of FH leads to the accumulation of fumarate, which activates hypoxia-inducible factors (HIFs) at normal oxygen tensions. However, so far no mechanism that explains the ability of cells to survive without a functional TCA cycle has been provided. Here we use newly characterized genetically modified kidney mouse cells in which Fh1 has been deleted, and apply a newly developed computer model of the metabolism of these cells to predict and experimentally validate a linear metabolic pathway beginning with glutamine uptake and ending with bilirubin excretion from Fh1-deficient cells. This pathway, which involves the biosynthesis and degradation of haem, enables Fh1-deficient cells to use the accumulated TCA cycle metabolites and permits partial mitochondrial NADH production. We predicted and confirmed that targeting this pathway would render Fh1-deficient cells non-viable, while sparing wild-type Fh1-containing cells. This work goes beyond identifying a metabolic pathway that is induced in Fh1-deficient cells to demonstrate that inhibition of haem oxygenation is synthetically lethal when combined with Fh1 deficiency, providing a new potential target for treating HLRCC patients.


Subject(s)
Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Genes, Lethal/genetics , Genes, Tumor Suppressor , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Mutation/genetics , Animals , Bilirubin/metabolism , Cell Line , Cells, Cultured , Citric Acid Cycle , Computer Simulation , Fumarate Hydratase/deficiency , Fumarates/metabolism , Glutamine/metabolism , Heme/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Leiomyomatosis/congenital , Leiomyomatosis/drug therapy , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Mice , Mitochondria/metabolism , NAD/metabolism , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms
7.
J Cell Sci ; 127(Pt 18): 3893-901, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25015290

ABSTRACT

Chloride intracellular channel 3 (CLIC3) drives invasiveness of pancreatic and ovarian cancer by acting in concert with Rab25 to regulate the recycling of α5ß1 integrin from late endosomes to the plasma membrane. Here, we show that in two estrogen receptor (ER)-negative breast cancer cell lines, CLIC3 has little influence on integrin recycling, but controls trafficking of the pro-invasive matrix metalloproteinase MT1-MMP (also known as MMP14). In MDA-MB-231 cells, MT1-MMP and CLIC3 are localized primarily to late endosomal/lysosomal compartments located above the plane of adhesion and near the nucleus. MT1-MMP is transferred from these late endosomes to sites of cell-matrix adhesion in a CLIC3-dependent fashion. Correspondingly, CLIC3-knockdown opposes MT1-MMP-dependent invasive processes. These include the disruption of the basement membrane as acini formed from MCF10DCIS.com cells acquire invasive characteristics in 3D culture, and the invasion of MDA-MB-231 cells into Matrigel or organotypic plugs of type I collagen. Consistent with this, expression of CLIC3 predicts poor prognosis in ER-negative breast cancer. The identification of MT1-MMP as a cargo of a CLIC3-regulated pathway that drives invasion highlights the importance of late endosomal sorting and trafficking in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chloride Channels/metabolism , Endosomes/metabolism , Matrix Metalloproteinase 14/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Movement , Chloride Channels/genetics , Female , Humans , Matrix Metalloproteinase 14/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Transport
8.
BMC Cancer ; 15: 9, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25592066

ABSTRACT

BACKGROUND: Androgens drive the onset and progression of prostate cancer (PCa) via androgen receptor (AR) signalling. The principal treatment for PCa is androgen deprivation therapy, although the majority of patients eventually develop a lethal castrate-resistant form of the disease, where despite low serum testosterone levels AR signalling persists. Advanced PCa often has hyper-activated RAS/ERK1/2 signalling thought to be due to loss of function of key negative regulators of the pathway, the details of which are not fully understood. METHODS: We recently carried out a genome-wide study and identified a subset of 226 novel androgen-regulated genes (PLOS ONE 6:e29088, 2011). In this study we have meta-analysed this dataset with genes and pathways frequently mutated in PCa to identify androgen-responsive regulators of the RAS/ERK1/2 pathway. RESULTS: We find the PTGER4 and TSPYL2 genes are up-regulated by androgen stimulation and the ADCY1, OPKR1, TRIB1, SPRY1 and PTPRR are down-regulated by androgens. Further characterisation of PTPRR protein in LNCaP cells revealed it is an early and direct target of the androgen receptor which negatively regulates the RAS/ERK1/2 pathway and reduces cell proliferation in response to androgens. CONCLUSION: Our data suggest that loss of PTPRR in clinical PCa is one factor that might contribute to activation of the RAS/ERK1/2 pathway.


Subject(s)
Androgens/pharmacology , MAP Kinase Signaling System/drug effects , Prostatic Neoplasms/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 7/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Databases, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostatic Neoplasms/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 7/metabolism , Receptors, Androgen/metabolism
9.
J Clin Periodontol ; 42(1): 1-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25360483

ABSTRACT

AIM: To investigate the chemotactic accuracy of peripheral blood neutrophils from patients with chronic periodontitis compared with matched healthy controls, before and after non-surgical periodontal therapy. MATERIAL & METHODS: Neutrophils were isolated from patients and controls (n = 18) by density centrifugation. Using the Insall chamber and video microscopy, neutrophils were analysed for directional chemotaxis towards N-formyl-methionyl-leucyl-phenylalanine [fMLP (10 nM), or CXCL8 (200 ng/ml)]. Circular statistics were utilized for the analysis of cell movement. RESULTS: Prior to treatment, neutrophils from patients with chronic periodontitis had significantly reduced speed, velocity and chemotactic accuracy compared to healthy controls for both chemoattractants. Following periodontal treatment, patient neutrophils continued to display reduced speed in response to both chemoattractants. However, velocity and accuracy were normalized for the weak chemoattractant CXCL8 while they remained significantly reduced for fMLP. CONCLUSIONS: Chronic periodontitis is associated with reduced neutrophil chemotaxis, and this is only partially restored by successful treatment. Dysfunctional neutrophil chemotaxis may predispose patients with periodontitis to their disease by increasing tissue transit times, thus exacerbating neutrophil-mediated collateral host tissue damage.


Subject(s)
Chemotaxis, Leukocyte/physiology , Chronic Periodontitis/pathology , Neutrophils/physiology , Periodontal Debridement/methods , Adult , Case-Control Studies , Chemotactic Factors/pharmacology , Chronic Periodontitis/therapy , Dental Plaque Index , Dental Scaling/methods , Female , Follow-Up Studies , Humans , Interleukin-8/pharmacology , Male , Middle Aged , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Periodontal Index , Root Planing/methods
10.
Gut ; 63(9): 1481-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24717934

ABSTRACT

OBJECTIVE: Pancreatic cancer is a leading cause of cancer-related death in the Western world. Current chemotherapy regimens have modest survival benefit. Thus, novel, effective therapies are required for treatment of this disease. DESIGN: Activating KRAS mutation almost always drives pancreatic tumour initiation, however, deregulation of other potentially druggable pathways promotes tumour progression. PTEN loss leads to acceleration of Kras(G12D)-driven pancreatic ductal adenocarcinoma (PDAC) in mice and these tumours have high levels of mammalian target of rapamycin (mTOR) signalling. To test whether these KRAS PTEN pancreatic tumours show mTOR dependence, we compared response to mTOR inhibition in this model, to the response in another established model of pancreatic cancer, KRAS P53. We also assessed whether there was a subset of pancreatic cancer patients who may respond to mTOR inhibition. RESULTS: We found that tumours in KRAS PTEN mice exhibit a remarkable dependence on mTOR signalling. In these tumours, mTOR inhibition leads to proliferative arrest and even tumour regression. Further, we could measure response using clinically applicable positron emission tomography imaging. Importantly, pancreatic tumours driven by activated KRAS and mutant p53 did not respond to treatment. In human tumours, approximately 20% of cases demonstrated low PTEN expression and a gene expression signature that overlaps with murine KRAS PTEN tumours. CONCLUSIONS: KRAS PTEN tumours are uniquely responsive to mTOR inhibition. Targeted anti-mTOR therapies may offer clinical benefit in subsets of human PDAC selected based on genotype, that are dependent on mTOR signalling. Thus, the genetic signatures of human tumours could be used to direct pancreatic cancer treatment in the future.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Drug Administration Schedule , Gene Expression Regulation, Neoplastic , Humans , Injections, Intraperitoneal , Mice , Mice, Mutant Strains , Mutation , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Positron-Emission Tomography , Proto-Oncogene Proteins p21(ras)/deficiency , Proto-Oncogene Proteins p21(ras)/genetics , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
11.
J Cell Sci ; 125(Pt 6): 1465-77, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22328529

ABSTRACT

Upregulation of the extracellular signal-regulated kinase (ERK) pathway has been shown to contribute to tumour invasion and progression. Because the two predominant ERK isoforms (ERK1 and ERK2, also known as MAPK3 and MAPK1, respectively) are highly homologous and have indistinguishable kinase activities in vitro, both enzymes were believed to be redundant and interchangeable. To challenge this view, we show that ERK2 silencing inhibits invasive migration of MDA-MB-231 cells, and re-expression of ERK2 but not ERK1 restores the normal invasive phenotype. A detailed quantitative analysis of cell movement on 3D matrices indicates that ERK2 knockdown impairs cellular motility by decreasing the migration velocity as well as increasing the time that cells spend not moving. Using gene expression arrays we found that the expression of the genes for Rab17 and liprin-ß2 was increased by knockdown of ERK2 and restored to normal levels following re-expression of ERK2, but not ERK1. Both play inhibitory roles in the invasive behaviour of three independent cancer cell lines. Importantly, knockdown of either Rab17 or liprin-ß2 restores invasiveness of ERK2-depleted cells, indicating that ERK2 drives invasion of MDA-MB-231 cells by suppressing expression of these genes.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/antagonists & inhibitors , Cell Movement/physiology , Membrane Proteins/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Tumor Microenvironment/physiology , rab GTP-Binding Proteins/antagonists & inhibitors , Breast Neoplasms/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , rab GTP-Binding Proteins/biosynthesis , rab GTP-Binding Proteins/genetics
12.
BMC Cancer ; 14: 977, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25519703

ABSTRACT

BACKGROUND: Although chemotherapy for prostate cancer (PCa) can improve patient survival, some tumours are chemo-resistant. Tumour molecular profiles may help identify the mechanisms of drug action and identify potential prognostic biomarkers. We performed in vivo transcriptome profiling of pre- and post-treatment prostatic biopsies from patients with advanced hormone-naive prostate cancer treated with docetaxel chemotherapy and androgen deprivation therapy (ADT) with an aim to identify the mechanisms of drug action and identify prognostic biomarkers. METHODS: RNA sequencing (RNA-Seq) was performed on biopsies from four patients before and ~22 weeks after docetaxel and ADT initiation. Gene fusion products and differentially-regulated genes between treatment pairs were identified using TopHat and pathway enrichment analyses undertaken. Publically available datasets were interrogated to perform survival analyses on the gene signatures identified using cBioportal. RESULTS: A number of genomic rearrangements were identified including the TMPRSS2/ERG fusion and 3 novel gene fusions involving the ETS family of transcription factors in patients, both pre and post chemotherapy. In total, gene expression analyses showed differential expression of at least 2 fold in 575 genes in post-chemotherapy biopsies. Of these, pathway analyses identified a panel of 7 genes (ADAM7, FAM72B, BUB1B, CCNB1, CCNB2, TTK, CDK1), including a cell cycle-related geneset, that were differentially-regulated following treatment with docetaxel and ADT. Using cBioportal to interrogate the MSKCC-Prostate Oncogenome Project dataset we observed a statistically-significant reduction in disease-free survival of patients with tumours exhibiting alterations in gene expression of the above panel of 7 genes (p = 0.015). CONCLUSIONS: Here we report on the first "real-time" in vivo RNA-Seq-based transcriptome analysis of clinical PCa from pre- and post-treatment TRUSS-guided biopsies of patients treated with docetaxel chemotherapy plus ADT. We identify a chemotherapy-driven PCa transcriptome profile which includes the down-regulation of important positive regulators of cell cycle progression. A 7 gene signature biomarker panel has also been identified in high-risk prostate cancer patients to be of prognostic value. Future prospective study is warranted to evaluate the clinical value of this panel.


Subject(s)
Gene Expression Profiling , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Transcriptome , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Computational Biology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Male , Neoplasm Grading , Neoplasm Staging , Prognosis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy
13.
Eukaryot Cell ; 12(11): 1509-16, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24036345

ABSTRACT

The SCAR/WAVE complex drives actin-based protrusion, cell migration, and cell separation during cytokinesis. However, the contribution of the individual complex members to the activity of the whole remains a mystery. This is primarily because complex members depend on one another for stability, which limits the scope for experimental manipulation. Several studies suggest that Abi, a relatively small complex member, connects signaling to SCAR/WAVE complex localization and activation through its polyproline C-terminal tail. We generated a deletion series of the Dictyostelium discoideum Abi to investigate its exact role in regulation of the SCAR complex and identified a minimal fragment that would stabilize the complex. Surprisingly, loss of either the N terminus of Abi or the C-terminal polyproline tail conferred no detectable defect in complex recruitment to the leading edge or the formation of pseudopods. A fragment containing approximately 20% Abi--and none of the sites that couple to known signaling pathways--allowed the SCAR complex to function with normal localization and kinetics. However, expression of N-terminal Abi deletions exacerbated the cytokinesis defect of the Dictyostelium abi mutant, which was earlier shown to be caused by the inappropriate activation of SCAR. This demonstrates, unexpectedly, that Abi does not mediate the SCAR complex's ability to make pseudopods, beyond its role in complex stability. Instead, we propose that Abi has a modulatory role when the SCAR complex is activated through other mechanisms.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dictyostelium/metabolism , Protozoan Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Cell Movement , Cytokinesis , Dictyostelium/cytology , Dictyostelium/genetics , Dictyostelium/physiology , Gene Deletion , Molecular Sequence Data , Peptides/chemistry , Protein Binding , Protein Stability , Protein Structure, Tertiary , Protein Transport , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Pseudopodia/metabolism
14.
Small GTPases ; 12(4): 294-310, 2021 07.
Article in English | MEDLINE | ID: mdl-32569510

ABSTRACT

As normal cells become cancer cells, and progress towards malignancy, they become progressively softer. Advantages of this change are that tumour cells become more deformable, and better able to move through narrow constraints. We designed a positive selection strategy that enriched for cells which could move through narrow diameter micropores to identify cell phenotypes that enabled constrained migration. Using human MDA MB 231 breast cancer and MDA MB 435 melanoma cancer cells, we found that micropore selection favoured cells with relatively higher Ras/Raf/MEK/ERK mitogen-activated protein kinase (MAPK) signalling, which affected actin cytoskeleton organization, focal adhesion density and cell elasticity. In this follow-up study, we provide further evidence that selection through micropores enriched for cells with altered cell morphology and adhesion. Additional analysis of RNA sequencing data revealed a set of transcripts associated with small cell size that was independent of constrained migration. Gene set enrichment analysis identified the 'matrisome' as the most significantly altered gene set linked with small size. When grown as orthotopic xenograft tumours in immunocompromised mice, micropore selected cells grew significantly faster than Parent or Flow-Sorted cells. Using mathematical modelling, we determined that there is an interaction between 1) the cell to gap size ratio; 2) the bending rigidity of the cell, which enable movement through narrow gaps. These results extend our previous conclusion that Ras/Raf/MEK/ERK MAPK signalling has a significant role in regulating cell biomechanics by showing that the selective pressure of movement through narrow gaps also enriches for increased tumour growth in vivo.


Subject(s)
Breast Neoplasms/pathology , Melanoma/pathology , Micropore Filters , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Melanoma/genetics , Melanoma/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/genetics , Proto-Oncogene Proteins c-raf/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , ras Proteins/genetics
15.
Sci Adv ; 5(1): eaau7314, 2019 01.
Article in English | MEDLINE | ID: mdl-30613774

ABSTRACT

Currently available cell culture media may not reproduce the in vivo metabolic environment of tumors. To demonstrate this, we compared the effects of a new physiological medium, Plasmax, with commercial media. We prove that the disproportionate nutrient composition of commercial media imposes metabolic artifacts on cancer cells. Their supraphysiological concentrations of pyruvate stabilize hypoxia-inducible factor 1α in normoxia, thereby inducing a pseudohypoxic transcriptional program. In addition, their arginine concentrations reverse the urea cycle reaction catalyzed by argininosuccinate lyase, an effect not observed in vivo, and prevented by Plasmax in vitro. The capacity of cancer cells to form colonies in commercial media was impaired by lipid peroxidation and ferroptosis and was rescued by selenium present in Plasmax. Last, an untargeted metabolic comparison revealed that breast cancer spheroids grown in Plasmax approximate the metabolic profile of mammary tumors better. In conclusion, a physiological medium improves the metabolic fidelity and biological relevance of in vitro cancer models.


Subject(s)
Culture Media , Models, Biological , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/physiology , Arginine/metabolism , Argininosuccinate Lyase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Ferroptosis/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipid Peroxidation/drug effects , Pyruvic Acid/metabolism , Sodium Selenite/pharmacology , Spheroids, Cellular/metabolism , Urea/metabolism
16.
Dev Cell ; 51(4): 431-445.e7, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31668663

ABSTRACT

Pancreatic ductal adenocarcinoma is one of the most invasive and metastatic cancers and has a dismal 5-year survival rate. We show that N-WASP drives pancreatic cancer metastasis, with roles in both chemotaxis and matrix remodeling. lysophosphatidic acid, a signaling lipid abundant in blood and ascites fluid, is both a mitogen and chemoattractant for cancer cells. Pancreatic cancer cells break lysophosphatidic acid down as they respond to it, setting up a self-generated gradient driving tumor egress. N-WASP-depleted cells do not recognize lysophosphatidic acid gradients, leading to altered RhoA activation, decreased contractility and traction forces, and reduced metastasis. We describe a signaling loop whereby N-WASP and the endocytic adapter SNX18 promote lysophosphatidic acid-induced RhoA-mediated contractility and force generation by controlling lysophosphatidic acid receptor recycling and preventing degradation. This chemotactic loop drives collagen remodeling, tumor invasion, and metastasis and could be an important target against pancreatic cancer spread.


Subject(s)
Lysophospholipids/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, Lysophosphatidic Acid/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement/physiology , Chemotaxis , Female , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Transport , Rats , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/isolation & purification , Signal Transduction , Sorting Nexins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , rhoA GTP-Binding Protein/metabolism
17.
Cancer Res ; 66(15): 7405-13, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885335

ABSTRACT

Most head and neck squamous cell carcinoma (HNSCC) patients present with late-stage cancers, which are difficult to treat. Therefore, early diagnosis of high-risk premalignant lesions and incipient cancers is important. HNSCC is currently perceived as a single progression mechanism, resulting in immortal invasive cancers. However, we have found that approximately 40% of primary oral SCCs are mortal in culture, and these have a better prognosis. About 60% of oral premalignancies (dysplasias) are also mortal. The mortal and immortal tumors are generated in vivo as judged by p53 mutations and loss of p16(INK4A) expression being found only in the original tumors from which the immortal cultures were derived. To investigate the relationships of dysplasias to SCCs, we did microarray analysis of primary cultures of 4 normal oral mucosa biopsies, 19 dysplasias, and 16 SCCs. Spectral clustering using the singular value decomposition and other bioinformatic techniques showed that development of mortal and immortal SCCs involves distinct transcriptional changes. Both SCC classes share most of the transcriptional changes found in their respective dysplasias but have additional changes. Moreover, high-risk dysplasias that subsequently progress to SCCs more closely resemble SCCs than nonprogressing dysplasias. This indicates for the first time that there are divergent mortal and immortal pathways for oral SCC development via intermediate dysplasias. We believe that this new information may lead to new ways of classifying HNSCC in relation to prognosis.


Subject(s)
Carcinoma, Squamous Cell/genetics , Mouth Neoplasms/genetics , Precancerous Conditions/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Gene Expression Profiling , Humans , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Transcription, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
18.
Cell Death Dis ; 9(2): 19, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29339815

ABSTRACT

Analysis of publicly available genomic and gene expression data demonstrates that MCL1 expression is frequently elevated in breast cancer. Distinct from other pro-survival Bcl-2 family members, the short half-life of MCL-1 protein led us to investigate MCL-1 protein expression in a breast cancer tissue microarray and correlate this with clinical data. Here, we report associations between high MCL-1 and poor prognosis in specific subtypes of breast cancer including triple-negative breast cancer, an aggressive form that lacks targeted treatment options. Deletion of MCL-1 in the mammary epithelium of genetically engineered mice revealed an absolute requirement for MCL-1 in breast tumorigenesis. The clinical applicability of these findings was tested through a combination of approaches including knock-down or inhibition of MCL-1 to show triple-negative breast cancer cell line dependence on MCL-1 in vitro and in vivo. Our data demonstrate that high MCL-1 protein expression is associated with poor outcome in breast cancer and support the therapeutic targeting of MCL-1 in this disease.


Subject(s)
Breast Neoplasms/metabolism , Molecular Targeted Therapy , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Breast Neoplasms/classification , Breast Neoplasms/pathology , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Treatment Outcome , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
Cancer Res ; 78(17): 5060-5071, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29976574

ABSTRACT

Glioblastoma (GBM) is a lethal primary brain tumor characterized by treatment resistance and inevitable tumor recurrence, both of which are driven by a subpopulation of GBM cancer stem-like cells (GSC) with tumorigenic and self-renewal properties. Despite having broad implications for understanding GSC phenotype, the determinants of upregulated DNA-damage response (DDR) and subsequent radiation resistance in GSC are unknown and represent a significant barrier to developing effective GBM treatments. In this study, we show that constitutive DDR activation and radiation resistance are driven by high levels of DNA replication stress (RS). CD133+ GSC exhibited reduced DNA replication velocity and a higher frequency of stalled replication forks than CD133- non-GSC in vitro; immunofluorescence studies confirmed these observations in a panel of orthotopic xenografts and human GBM specimens. Exposure of non-GSC to low-level exogenous RS generated radiation resistance in vitro, confirming RS as a novel determinant of radiation resistance in tumor cells. GSC exhibited DNA double-strand breaks, which colocalized with "replication factories" and RNA: DNA hybrids. GSC also demonstrated increased expression of long neural genes (>1 Mbp) containing common fragile sites, supporting the hypothesis that replication/transcription collisions are the likely cause of RS in GSC. Targeting RS by combined inhibition of ATR and PARP (CAiPi) provided GSC-specific cytotoxicity and complete abrogation of GSC radiation resistance in vitro These data identify RS as a cancer stem cell-specific target with significant clinical potential.Significance: These findings shed new light on cancer stem cell biology and reveal novel therapeutics with the potential to improve clinical outcomes by overcoming inherent radioresistance in GBM. Cancer Res; 78(17); 5060-71. ©2018 AACR.


Subject(s)
Carcinogenesis , Glioblastoma/genetics , Neoplasm Recurrence, Local/genetics , Neoplastic Stem Cells , Radiation Tolerance/genetics , AC133 Antigen/genetics , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Replication/drug effects , DNA Replication/radiation effects , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
20.
Cancer Discov ; 8(5): 632-647, 2018 05.
Article in English | MEDLINE | ID: mdl-29500295

ABSTRACT

Exploiting oxidative stress has recently emerged as a plausible strategy for treatment of human cancer, and antioxidant defenses are implicated in resistance to chemotherapy and radiotherapy. Targeted suppression of antioxidant defenses could thus broadly improve therapeutic outcomes. Here, we identify the AMPK-related kinase NUAK1 as a key component of the antioxidant stress response pathway and reveal a specific requirement for this role of NUAK1 in colorectal cancer. We show that NUAK1 is activated by oxidative stress and that this activation is required to facilitate nuclear import of the antioxidant master regulator NRF2: Activation of NUAK1 coordinates PP1ß inhibition with AKT activation in order to suppress GSK3ß-dependent inhibition of NRF2 nuclear import. Deletion of NUAK1 suppresses formation of colorectal tumors, whereas acute depletion of NUAK1 induces regression of preexisting autochthonous tumors. Importantly, elevated expression of NUAK1 in human colorectal cancer is associated with more aggressive disease and reduced overall survival.Significance: This work identifies NUAK1 as a key facilitator of the adaptive antioxidant response that is associated with aggressive disease and worse outcome in human colorectal cancer. Our data suggest that transient NUAK1 inhibition may provide a safe and effective means for treatment of human colorectal cancer via disruption of intrinsic antioxidant defenses. Cancer Discov; 8(5); 632-47. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 517.


Subject(s)
Colorectal Neoplasms/metabolism , Oxidative Stress , Protein Kinases/metabolism , Repressor Proteins/metabolism , Animals , Binding Sites , Biomarkers , Colonic Polyps/genetics , Colonic Polyps/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Models, Animal , Disease Progression , Gene Expression , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Lymph Nodes/pathology , Mice , Models, Biological , NF-E2-Related Factor 2/metabolism , Nucleotide Motifs , Prognosis , Protein Binding , Protein Kinases/genetics , Protein Transport , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL