Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
Infect Immun ; 89(9): e0018721, 2021 08 16.
Article in English | MEDLINE | ID: mdl-33941577

ABSTRACT

The gut microbiome orchestrates epithelial homeostasis and both local and remote immunological responses. Critical to these regulatory interactions are innate immune receptors termed Toll-like receptors (TLRs). Studies to date have implicated innate immunity and Toll-like receptors in shaping key features of the gut microbiome. However, a variety of biological and environmental variables are also implicated in determining gut microbiota composition. In this report, we hypothesized that cohousing and environment dominated the regulation of the gut microbiota in animal models independent of innate immunity. To determine the importance of these variables, innate immunity, or environment in shaping gut microbiota, we used a randomized cohousing strategy and transgenic TLR-deficient mice. We have found that mice cohoused together by genotype exhibited limited changes over time in the composition of the gut microbiota. However, for mice randomized to cage, we report extensive changes in the gut microbiota, independent of TLR function, whereby the fecal microbiota of TLR-deficient mice converges with that of wild-type mice. TLR5-deficient mice in these experiments exhibit greater susceptibility to comparative changes in the microbiota than other TLR-deficient mice and wild-type mice. Our work has broad implications for the study of innate immunity and host-microbiota interactions. Given the profound impact that gut dysbiosis may have on immunity, this report highlights the potential impact of cohousing on the gut microbiota and indices of inflammation as outcomes in biological models of infectious or inflammatory disease.


Subject(s)
Gastrointestinal Microbiome , Homeostasis , Host Microbial Interactions , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Toll-Like Receptors/metabolism , Animals , Disease Susceptibility , Dysbiosis , Immunity, Innate , Immunity, Mucosal , Mice , Models, Animal
2.
Proc Natl Acad Sci U S A ; 115(48): 12176-12181, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30442661

ABSTRACT

Class IA PI3Ks have many roles in health and disease. The rules that govern intersubunit and receptor associations, however, remain unclear. We engineered mouse lines in which individual endogenous class IA PI3K subunits were C-terminally tagged with 17aa that could be biotinylated in vivo. Using these tools we quantified PI3K subunits in streptavidin or PDGFR pull-downs and cell lysates. This revealed that p85α and ß bound equivalently to p110α or p110ß but p85α bound preferentially to p110δ. p85s were found in molar-excess over p110s in a number of contexts including MEFs (p85ß, 20%) and liver (p85α, 30%). In serum-starved MEFs, p110-free-p85s were preferentially, compared with heterodimeric p85s, bound to PDGFRs, consistent with in vitro assays that demonstrated they bound PDGFR-based tyrosine-phosphorylated peptides with higher affinity and co-operativity; suggesting they may act to tune a PI3K activation threshold. p110α-heterodimers were recruited 5-6× more efficiently than p110ß-heterodimers to activated PDGFRs in MEFs or to PDGFR-based tyrosine-phosphorylated peptides in MEF-lysates. This suggests that PI3Kα has a higher affinity for relevant tyrosine-phosphorylated motifs than PI3Kß. Nevertheless, PI3Kß contributes substantially to acute PDGF-stimulation of PIP3 and PKB in MEFs because it is synergistically, and possibly sequentially, activated by receptor-recruitment and small GTPases (Rac/CDC42) via its RBD, whereas parallel activation of PI3Kα is independent of its RBD. These results begin to provide molecular clarity to the rules of engagement between class IA PI3K subunits in vivo and past work describing "excess p85," p85α as a tumor suppressor, and differential receptor activation of PI3Kα and PI3Kß.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Protein Isoforms/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Class Ia Phosphatidylinositol 3-Kinase/genetics , Dimerization , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Protein Isoforms/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Signal Transduction
3.
Oncogene ; 35(25): 3324-34, 2016 06 23.
Article in English | MEDLINE | ID: mdl-26455320

ABSTRACT

Suppression of detachment-induced cell death, known as anoikis, is an essential step for cancer metastasis to occur. We report here that expression of KLF12, a member of the Kruppel-like family of transcription factors, is downregulated in lung cancer cell lines that have been selected to grow in the absence of cell adhesion. Knockdown of KLF12 in parental cells results in decreased apoptosis following cell detachment from matrix. KLF12 regulates anoikis by promoting the cell cycle transition through S phase and therefore cell proliferation. Reduced expression levels of KLF12 results in increased ability of lung cancer cells to form tumours in vivo and is associated with poorer survival in lung cancer patients. We therefore identify KLF12 as a novel metastasis-suppressor gene whose loss of function is associated with anoikis resistance through control of the cell cycle.


Subject(s)
Anoikis/genetics , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/genetics , Lung Neoplasms/genetics , A549 Cells , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation , Flow Cytometry , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Metastasis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics
4.
Br J Cancer ; 111(6): 1021-46, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25117809

ABSTRACT

Cell-line misidentification and contamination with microorganisms, such as mycoplasma, together with instability, both genetic and phenotypic, are among the problems that continue to affect cell culture. Many of these problems are avoidable with the necessary foresight, and these Guidelines have been prepared to provide those new to the field and others engaged in teaching and instruction with the information necessary to increase their awareness of the problems and to enable them to deal with them effectively. The Guidelines cover areas such as development, acquisition, authentication, cryopreservation, transfer of cell lines between laboratories, microbial contamination, characterisation, instability and misidentification. Advice is also given on complying with current legal and ethical requirements when deriving cell lines from human and animal tissues, the selection and maintenance of equipment and how to deal with problems that may arise.


Subject(s)
Biomedical Research/standards , Cell Line/microbiology , Equipment and Supplies/standards , Mycoplasma , Safety/standards , Animals , Biomedical Research/ethics , Cell Line/classification , Cryopreservation/standards , Culture Media/standards , Equipment Contamination/prevention & control , Genomic Instability , Humans , Mycoplasma/isolation & purification , Phenotype , Quality Control , Specimen Handling/methods , Specimen Handling/standards , United Kingdom
5.
Oncogene ; 30(29): 3222-33, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21358673

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) pathway is commonly activated in breast cancers due to frequent mutations in PIK3CA, loss of expression of PTEN or over-expression of receptor tyrosine kinases. PI3K pathway activation leads to stimulation of the key growth and proliferation regulatory kinase mammalian target of rapamycin (mTOR), which can be inhibited by rapamycin analogues and by kinase inhibitors; the effectiveness of these drugs in breast cancer treatment is currently being tested in clinical trials. To identify the molecular determinants of response to inhibitors that target mTOR via different mechanisms in breast cancer cells, we investigated the effects of pharmacological inhibition of mTOR using the allosteric mTORC1 inhibitor everolimus and the active-site mTORC1/mTORC2 kinase inhibitor PP242 on a panel of 31 breast cancer cell lines. We demonstrate here that breast cancer cells harbouring PIK3CA mutations are selectively sensitive to mTOR allosteric and kinase inhibitors. However, cells with PTEN loss of function are not sensitive to these drugs, suggesting that the functional consequences of these two mechanisms of activation of the mTOR pathway are quite distinct. In addition, a subset of HER2-amplified cell lines showed increased sensitivity to PP242, but not to everolimus, irrespective of the PIK3CA/PTEN status. These selective sensitivities were confirmed in more physiologically relevant three-dimensional cell culture models. Our findings provide a rationale to guide selection of breast cancer patients who may benefit from mTOR inhibitor therapy and highlight the importance of accurately assessing the expression of PTEN protein and not just its mutational status.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Drug Screening Assays, Antitumor , Everolimus , G1 Phase , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Mutation , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
6.
Oncogene ; 30(32): 3513-21, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21423205

ABSTRACT

We performed a kinome-wide siRNA screen and identified 70 kinases altering cell migration in A549 lung cancer cells. In particular, ribosomal S6 kinase 1 (RSK1) silencing increased, whereas RSK2 and RSK4 downregulation inhibited cell motility. In a secondary collagen-based three-dimensional invasion screen, 38 of our hits cross-validated, including RSK1 and RSK4. In two further lung cancer cell lines, RSK1 but not RSK4 silencing showed identical modulation of cell motility. We therefore selected RSK1 for further investigation. Bioinformatic analysis followed by co-immunoprecipitation-based validation revealed that the actin regulators VASP and Mena interact with RSK1. Moreover, RSK1 phosphorylated VASP on T278, a site regulating its binding to actin. In addition, silencing of RSK1 enhanced the metastatic potential of these cells in vivo using a zebrafish model. Finally, we investigated the relevance of this finding in human lung cancer samples. In isogenically matched tissue, RSK1 was reduced in metastatic versus primary lung cancer lesions. Moreover, patients with RSK1-negative lung tumours showed increased number of metastases. Our results suggest that the findings of our high-throughput in vitro screen can reliably identify relevant clinical targets and as a proof of principle, RSK1 may provide a biomarker for metastasis in lung cancer patients.


Subject(s)
Lung Neoplasms/genetics , RNA Interference , RNA, Small Interfering/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Animals , Binding Sites , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microscopy, Fluorescence , Neoplasm Metastasis , Neoplasm Transplantation , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Threonine/genetics , Threonine/metabolism , Transplantation, Heterologous , Zebrafish/embryology
7.
Oncogene ; 29(33): 4658-70, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20562906

ABSTRACT

Activating mutations in the KRAS gene are among the most prevalent genetic changes in human cancers. To identify synthetic lethal interactions in cancer cells harbouring mutant KRAS, we performed a large-scale screen in isogenic paired colon cancer cell lines that differ by a single allele of mutant KRAS using an inducible short hairpin RNA interference library. Snail2, a zinc finger transcriptional repressor encoded by the SNAI2 gene, was found to be selectively required for the long-term survival of cancer cells with mutant KRAS that have undergone epithelial-mesenchymal transition (EMT), a transdifferentiation event that is frequently seen in advanced tumours and is promoted by RAS activation. Snail2 expression is regulated by the RAS pathway and is required for EMT. Our findings support Snail2 as a possible target for the treatment of the broad spectrum of human cancers of epithelial origin with mutant RAS that have undergone EMT and are characterized by a high degree of chemoresistance and radioresistance.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/genetics , Genes, ras , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Zinc Fingers/genetics , ras Proteins/genetics , ras Proteins/metabolism
8.
Oncogene ; 29(7): 1003-16, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-19935711

ABSTRACT

The rapamycin-insensitive companion of mammalian target of rapamycin (mTOR) (Rictor) is a key member of mTOR complex-2 (mTORC2), which phosphorylates the AGC kinases Akt/PKB, PKC and SGK1 at a C-terminal hydrophobic motif. We identified several novel sites on Rictor that are phosphorylated, including Thr1135, which is conserved across all vertebrates. Phosphorylation of this site on Rictor is stimulated by amino acids and growth factors through a rapamycin-sensitive signaling cascade. We demonstrate here that Rictor is a direct target of the ribosomal protein S6 kinase-1 (S6K1). Rictor phosphorylation at Thr1135 does not lead to major changes in mTORC2-kinase activity. However, phosphorylation of this site turns over rapidly and mediates 14-3-3 binding to Rictor and mTORC2, providing possibility for altered interactions of the complex. These findings reveal an unexpected signaling input into mTORC2, which is regulated by amino acids, growth factors and rapamycin.


Subject(s)
Carrier Proteins/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , 14-3-3 Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cell Line , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Molecular Sequence Data , Phosphorylation , Rapamycin-Insensitive Companion of mTOR Protein , Rats , Threonine , Transcription Factors/metabolism
9.
Cell Death Differ ; 16(8): 1118-25, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19282869

ABSTRACT

High temperature requirement A2 (HtrA2/Omi) is a mitochondrial protease that exhibits proapoptotic and cell-protective properties and has been linked to Parkinson's disease (PD). Impaired mitochondrial function is a common trait in PD patients, and is likely to play a significant role in pathogenesis of parkinsonism, but the molecular mechanisms remain poorly understood. Genetic studies in Drosophila have provided valuable insight into the function of other PD-linked genes, in particular PINK1 and parkin, and their role in maintaining mitochondrial integrity. Recently, HtrA2 was shown to be phosphorylated in a PINK1-dependent manner, suggesting it might act in the PINK1 pathway. Here, we describe the characterization of mutations in Drosophila HtrA2, and genetic analysis of its function with PINK1 and parkin. Interestingly, we find HtrA2 appears to be dispensable for developmental or stress-induced apoptosis. In addition, we found HtrA2 mutants share some phenotypic similarities with parkin and PINK1 mutants, suggesting that it may function in maintaining mitochondrial integrity. Our genetic interaction studies, including analysis of double-mutant combinations and epistasis experiments, suggest HtrA2 acts downstream of PINK1 but in a pathway parallel to Parkin.


Subject(s)
Apoptosis , Drosophila Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Serine Endopeptidases/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Drosophila/metabolism , Drosophila Proteins/genetics , Female , Fertility/genetics , High-Temperature Requirement A Serine Peptidase 2 , Male , Mitochondria/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Phosphorylation , Serine Endopeptidases/genetics , Ubiquitin-Protein Ligases
10.
Cell Death Differ ; 16(3): 449-64, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19023330

ABSTRACT

Cellular stress responses can be activated following functional defects in organelles such as mitochondria and the endoplasmic reticulum. Mitochondrial dysfunction caused by loss of the serine protease HtrA2 leads to a progressive movement disorder in mice and has been linked to parkinsonian neurodegeneration in humans. Here, we demonstrate that loss of HtrA2 results in transcriptional upregulation of nuclear genes characteristic of the integrated stress response, including the transcription factor CHOP, selectively in the brain. We also show that loss of HtrA2 results in the accumulation of unfolded proteins in the mitochondria, defective mitochondrial respiration and enhanced production of reactive oxygen species that contribute to the induction of CHOP expression and to neuronal cell death. CHOP expression is also significantly increased in Parkinson's disease patients' brain tissue. We therefore propose that this brain-specific transcriptional response to stress may be important in the advance of neurodegenerative diseases.


Subject(s)
Brain/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Oxidative Stress , Serine Endopeptidases/metabolism , Transcription, Genetic , Animals , Antioxidants/metabolism , Cell Respiration/physiology , Corpus Striatum/metabolism , Corpus Striatum/pathology , High-Temperature Requirement A Serine Peptidase 2 , Humans , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Reactive Oxygen Species/metabolism , Serine Endopeptidases/genetics , Tissue Distribution , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
11.
Cell Death Differ ; 15(6): 997-1008, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18274554

ABSTRACT

Mitochondrial dysfunction mediated by Bax and Bak is a critical step in mammalian cell apoptosis. However, the molecular mechanism of Bax activation remains unknown and has been difficult to investigate due to its rapid and stochastic nature. It is currently unclear whether mitochondria play a passive role in the initiation of apoptosis, remaining unaffected by cell stresses until Bax and Bak are active, or whether they actively participate in Bax/Bak activation. Here, two viral proteins, E1B19K and BHRF1, are examined for their ability to block Bax activation at different steps and thereby reveal the timing of mitochondrial changes during apoptosis. We demonstrate that BHRF1 strongly inhibits Bax activation but not upstream apoptotic signaling events, while E1B19K permits initial stages of Bax activation but prevents the subsequent oligomerization of Bax that is required for mitochondrial dysfunction. In this defined system we show that changes in mitochondrial ultrastructure, characteristic of cells undergoing apoptosis, precede Bax activation and are not blocked by E1B19K and BHRF1. We suggest that the ability of mitochondria to respond to apoptotic stress prior to Bax activation indicates that these organelles may play a direct role in activating Bax.


Subject(s)
Apoptosis , Mitochondria/ultrastructure , Viral Proteins/metabolism , bcl-2-Associated X Protein/antagonists & inhibitors , Cell Line, Tumor , Cell Survival , Humans , Mitochondria/physiology , bcl-2-Associated X Protein/metabolism
12.
Oncogene ; 26(55): 7709-14, 2007 Dec 06.
Article in English | MEDLINE | ID: mdl-17603562

ABSTRACT

The mechanisms of signal transduction from cell surface receptors to the interior of the cell are fundamental to the understanding of the role that positive and negative growth factors play in cell physiology and in human diseases. Here, we show that a functional link between phosphatidylinositol-3-OH kinase (PI3K) and Ras is suppressed by the beta-galactoside binding protein (betaGBP) molecule, a cytokine and a negative cell-cycle regulator. Ras-mitogen-activated protein kinase (MAPK) signalling is blocked by betaGBP owing to its ability to inhibit the p110 catalytic subunit of PI3K, whose basal activity is required for Ras activation. Functional inhibition of p110 by betaGBP results in downregulation of PI3K activity, suppression of Ras-GTP loading, consequent loss of MAPK activation and block of cell proliferation. This study sheds light on the molecular mechanisms whereby betaGBP can control cell proliferation and, by extension, may potentially control tumorigenesis by controlling PI3K.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Galectin 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , ras Proteins/metabolism , Animals , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Galectin 1/genetics , Galectin 1/pharmacology , Humans , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
13.
Carcinogenesis ; 28(6): 1178-87, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17227781

ABSTRACT

Previously, we have identified the transcription factor CUTL1 as an important mediator of tumor invasion and target of tumor growth factor-beta. Using high-throughput approaches, we identified several putative downstream effectors of CUTL1, among them WNT5A, a secreted member of the Wnt multigene family. The aim of this study was to investigate the role of WNT5A as a novel target of CUTL1 in pancreatic cancer. CUTL1 and WNT5A were stably over-expressed as well as transiently and stably knocked down by RNA interference. Effects on proliferation, migration and invasiveness were investigated by thymidine incorporation, Boyden chamber experiments and time-lapse microscopy. Expression of WNT5A in pancreatic cancer tissues was analyzed by real-time polymerase chain reaction (RT-PCR) and immunohistochemistry. We found that CUTL1 transcriptionally up-regulated WNT5A on RNA, protein and promoter level. WNT5A significantly enhanced migration, proliferation and invasiveness, mediating the pro-invasive effects of CUTL1 to a major extent. WNT5A effects were accompanied by a marked modulation of marker genes associated with epithelial-mesenchymal transition. Using RT-PCR and immunohistochemistry, we found that WNT5A is up-regulated early during pancreatic cancerogenesis in pancreatic intraepithelial neoplasias lesions and in invasive pancreatic adenocarcinomas, as compared with normal pancreas tissues. These data identify WNT5A as important target of CUTL1 and as novel mediator of invasiveness and tumor progression in pancreatic cancer.


Subject(s)
Cell Movement/physiology , Homeodomain Proteins/physiology , Nuclear Proteins/physiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Wnt Proteins/physiology , Cell Line, Tumor , Disease Progression , Homeodomain Proteins/metabolism , Humans , Neoplasm Invasiveness , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , Repressor Proteins/metabolism , Transcription Factors , Up-Regulation/genetics , Wnt Proteins/biosynthesis , Wnt Proteins/genetics , Wnt-5a Protein
14.
Oncogene ; 26(7): 970-81, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-16909112

ABSTRACT

Transforming growth factor-beta (TGFbeta)-activated signalling pathways can lead to apoptosis, growth arrest or promotion of malignant behaviour, dependent on cellular context. The molecular mechanisms involved in TGFbeta-induced apoptosis remain controversial; although changes in gene expression are thought to be pivotal to the process, several different candidate apoptotic initiators and mediators have been proposed. Smad4, a critical component of the TGFbeta-induced transcriptional machinery, is shown here to be essential for induction of apoptosis. Gene expression analysis identified the proapoptotic Bcl-2 family members, Bmf and Bim, as induced by TGFbeta, dependent on both Smad4 and p38 function and the generation of reactive oxygen species. TGFbeta-induced Bmf and Bim localize to cellular membranes implicated in apoptosis. Inhibition of the TGFbeta-induced expression of both these proteins together provides significant protection of cells from apoptosis. The TGFbeta-triggered cell death programme thus involves induction of multiple BH3-only proteins during the induction of apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/biosynthesis , Membrane Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Transforming Growth Factor beta/physiology , Up-Regulation/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Bcl-2-Like Protein 11 , Cell Line , Cell Line, Tumor , Membrane Proteins/genetics , Mice , Proto-Oncogene Proteins/genetics , Rats , Up-Regulation/genetics
15.
Oncogene ; 25(54): 7117-30, 2006 Nov 16.
Article in English | MEDLINE | ID: mdl-16751808

ABSTRACT

Oncogenic Ras interferes with adhesive functions of epithelial cells, but requires tumor growth factor beta (TGFbeta) signaling to cause epithelial-mesenchymal transition (EMT) and tumor progression in model systems. To investigate the mechanisms by which Ras and TGFbeta pathways cooperate in EMT induction, we introduced a tamoxifen-inducible version of Raf-1 (RafER) into fully polarized, mammary epithelial cells (EpH4). EMT characterized by loss of E-cadherin expression and upregulation of invasiveness-promoting genes was induced by TGFbeta plus 4-hydroxytamoxifen (4HT) activation of RafER. Downregulation of E-cadherin by RafER plus TGFbeta was detectable in total cell lysates after 48 h and much earlier in detergent-insoluble fractions of E-cadherin. Both pathways cooperated to strongly enhance endocytosis of E-cadherin, mainly via the clathrin-dependent route. Pulse-chase experiments showed decreased E-cadherin protein stability in cells stimulated with TGFbeta and 4HT and increased E-cadherin half-life in the presence of monensin. Monensin and chloroquine prevented E-cadherin degradation to different extent, but only monensin effectively blocked the loss of E-cadherin from the junctional complexes. Both lysosome inhibitors caused accumulation of E-cadherin vesicles, some of which were positive for Cathepsin D and lysosome-associated membrane protein 1 (LAMP-1). In addition, TGFbeta and mitogen-activated protein kinase hyperactivation synergistically induced E-cadherin ubiquitination, suggesting that the cooperation of Raf and TGFbeta favors lysosomal degradation of E-cadherin instead of its recycling. Our data indicate that early stages of EMT involve cooperative, post-translational downregulation of E-cadherin, whereas loss of E-cadherin via transcriptional repression is a late event in EMT.


Subject(s)
Cadherins/metabolism , Cell Transformation, Neoplastic , Epithelial Cells/metabolism , Transforming Growth Factor beta/metabolism , raf Kinases/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Down-Regulation , Endocytosis , Epithelial Cells/pathology , Fluorescent Antibody Technique , Immunoprecipitation , Lysosomes/metabolism , Mice , Microscopy, Confocal , Protein Processing, Post-Translational , Recombinant Fusion Proteins
17.
Cell Death Differ ; 13(10): 1651-62, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16439990

ABSTRACT

Apoptosis represents an important cellular defence mechanism against viral pathogens by virtue of its ability to remove infected cells. Consequently, many viruses have developed numerous strategies to prevent or delay host cell apoptosis in order to achieve productive replication. Here we report that deletion of the F1L gene from the vaccinia genome results in increased apoptosis during infection. We demonstrate that F1L, which has no sequence homology to Bcl-2 family members, inhibits apoptosis at the level of mitochondria by binding to Bak. As a consequence, F1L prevents Bak activation, oligomerization and interaction with active Bax, all critical steps in the induction of apoptosis. We demonstrate that residues 64-84 of F1L interact directly with the Bcl-2 homology domain 3 (BH3) domain of Bak. This region of F1L has limited sequence similarity to known Bak-interacting BH3 domains. We also find that such additional BH3-like domains exist in the vaccinia genome. We conclude that F1L uses this specific, BH3-like domain to bind and inhibit Bak at the mitochondria.


Subject(s)
Apoptosis/physiology , Vaccinia virus/physiology , Vaccinia virus/pathogenicity , Viral Proteins/physiology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Amino Acid Sequence , Apoptosis/drug effects , Base Sequence , Binding Sites , Gene Deletion , Genes, Viral , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondria/virology , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Staurosporine/pharmacology , Vaccinia virus/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2 Homologous Antagonist-Killer Protein/chemistry , bcl-2 Homologous Antagonist-Killer Protein/genetics
18.
Oncogene ; 25(6): 877-87, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16170339

ABSTRACT

The impact of the 3-hydroxy-3methylglutaryl CoA reductase inhibitor simvastatin on human small-cell lung cancer (SCLC) cell growth and survival was investigated. Simvastatin profoundly impaired basal and growth factor-stimulated SCLC cell growth in vitro and induced apoptosis. SCLC cells treated with simvastatin were sensitized to the effects of the chemotherapeutic agent etoposide. Moreover, SCLC tumour growth in vivo was inhibited by simvastatin. These responses correlated with the inhibition of stem cell factor (SCF)-stimulated activation of extracellular signal-regulated kinase (Erk), protein kinase B (PKB) and ribosomal S6 kinase by simvastatin. Constitutive activation of the Erk pathway was sufficient to rescue SCLC cell from the effects of simvastatin. The drug did not directly affect activation of c-Kit or its localization to lipid rafts, but in addition to its ability to block Ras membrane localization, it selectively downregulated H-Ras protein levels at the post-translational level. Downregulation of either H- or K-Ras by RNA interference (RNAi) did not impair Erk activation by growth factors, whereas an RNAi specific for N-Ras inhibited activation of Erk, PKB and SCLC cell growth. Together our data demonstrate that inhibiting Ras signalling with simvastatin potently disrupts growth and survival in human SCLC cells.


Subject(s)
Carcinoma, Small Cell/drug therapy , Growth Substances/metabolism , Lung Neoplasms/drug therapy , Simvastatin/pharmacology , ras Proteins/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Base Sequence , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Cell Proliferation/drug effects , Etoposide/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Kinase Kinases/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Tumor Cells, Cultured , Up-Regulation , ras Proteins/drug effects , ras Proteins/genetics
19.
Z Gastroenterol ; 43(10): 1133-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16220453

ABSTRACT

The lipid kinase phosphoinositide 3-OH kinase (PI3K) and its downstream target Akt, also known as protein kinase B (PKB), are crucial effectors in oncogenic signaling induced by various receptor-tyrosine kinases. In recent years, data are accumulating that PI3K/Akt signaling components are frequently altered in a variety of human malignancies. This review summarizes the major effects of PI3K/Akt signaling on proliferation, survival and resistance to apoptosis, angiogenesis and cell motility in gastrointestinal cancers. In addition, activation of PI3K/Akt by various growth factors, the modulation of downstream targets by Akt-induced phosphorylation as well as novel treatment strategies targeting this pathway in gastrointestinal tumors are discussed.


Subject(s)
Gastrointestinal Neoplasms , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Animals , Apoptosis , Carcinoma, Hepatocellular , Colonic Neoplasms , DNA Repair , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/mortality , Humans , Immunohistochemistry , Leukemia, Experimental/metabolism , Liver Neoplasms , MAP Kinase Signaling System , Mice , Neoplasm Metastasis , Pancreatic Neoplasms , Phosphorylation , Stomach Neoplasms , Tumor Cells, Cultured
20.
Gut ; 54(1): 109-16, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15591514

ABSTRACT

BACKGROUND: Ras signalling is frequently aberrant in pancreatic cancer so that there is constitutive activation of the phosphatidylinositol 3-kinase (PI3K) and AKT/protein kinase B pathway, as well as the RAF/MEK/ERK pathway. AIMS: In the present study we investigated the role of the PI3K/AKT pathway in malignant transformation of pancreatic cancer cells. METHODS: A genetic approach was used to interfere with signal transduction in vitro and in vivo. RASN17, a dominant negative mutant of RAS, was applied to inhibit the PI3K/AKT pathway upstream of PI3K. The regulatory p85beta subunit of PI3K and the negative regulator PTEN were utilised to inhibit the pathway at the level of PI3K, and AAA-AKT, a dominant negative mutant of AKT was employed to interfere with PI3K/AKT signalling at the level of AKT. RESULTS: Antiproliferative, proapoptotic, and anticancer effects were documented, showing that inhibition of the PI3K pathway in these cell lines suppresses tumour cell growth in vitro and reduces growth in nude mice. CONCLUSIONS: The PI3K/AKT pathway represents a potential therapeutic target for pancreatic cancer, and gene therapy may be one approach to produce selective inhibition.


Subject(s)
Genetic Therapy/methods , Pancreatic Neoplasms/therapy , Phosphatidylinositol 3-Kinases/physiology , Adenoviridae/genetics , Animals , Apoptosis/genetics , Cell Division/genetics , Genetic Vectors , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Signal Transduction/genetics , Transduction, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...