Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
Add more filters

Country/Region as subject
Publication year range
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.
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
4.
Nat Genet ; 10(3): 294-300, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7670467

ABSTRACT

Cellular Ras proteins are activated primarily by specific guanine-nucleotide releasing factors such as the Son of Sevenless (Sos) proteins. This activation event is thought to occur in response to plasma membrane localization of a complex containing Sos and a small adapter protein Grb2. We have isolated a dominant mutant allele of mSos1 which transforms Rat1 cells, yet is no longer able to bind Grb2. Biochemical experiments reveal that the subcellular distribution of this truncated Sos protein is not altered with respect to the wild type Sos protein. These data argue against a role for Grb2 in the direct recruitment of Sos proteins to the plasma membrane and suggest that Grb2 may function to overcome negative regulation of Sos by its C terminus.


Subject(s)
Adaptor Proteins, Signal Transducing , Proteins/genetics , Proteins/metabolism , Alleles , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , DNA/genetics , Frameshift Mutation , GRB2 Adaptor Protein , Guanine Nucleotide Exchange Factors , Models, Biological , Molecular Sequence Data , Polymerase Chain Reaction , Protein Binding , Rats , Signal Transduction , Transformation, Genetic , ras Guanine Nucleotide Exchange Factors
5.
Curr Opin Cell Biol ; 10(2): 262-7, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9561851

ABSTRACT

Protein kinase B (PKB)/Akt is a growth-factor-regulated serine/threonine kinase which contains a pleckstrin homology domain. Binding of phosphoinositide 3-OH kinase products to the pleckstrin homology domain results in translocation of PKB/Akt to the plasma membrane where it is activated by phosphorylation by upstream kinases including the phosphoinoside-dependent kinase 1 (PDK1). Activated PKB/Akt provides a survival signal that protects cells from apoptosis induced by various stresses, and also mediates a number of metabolic effects of insulin.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Animals , Enzyme Activation/physiology , Growth Substances/physiology , Humans , Insulin/physiology , Models, Biological , Proto-Oncogene Proteins c-akt
6.
Nat Cell Biol ; 3(8): E190-5, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11483980

ABSTRACT

Parallel quantification of large numbers of messenger RNA transcripts using microarray technology promises to provide detailed insight into cellular processes involved in the regulation of gene expression. This should allow new understanding of signalling networks that operate in the cell and of the molecular basis and classification of disease. But can the technology deliver such far-reaching promises?


Subject(s)
Gene Expression Regulation/physiology , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/metabolism , Animals , Cluster Analysis , Humans , Neoplasms/genetics , RNA, Messenger/genetics , Signal Transduction/genetics
7.
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
9.
J Cell Biol ; 139(4): 1017-23, 1997 Nov 17.
Article in English | MEDLINE | ID: mdl-9362519

ABSTRACT

Detachment of epithelial cells from the extracellular matrix leads to induction of programmed cell death, a process that has been termed "anoikis." It has been reported recently that detachment of MDCK cells from matrix results in activation of Jun-NH2-terminal kinases (JNKs) and speculated that these stress activated protein kinases play a causal role in the induction of anoikis (Frisch, S.M., K. Vuori, D. Kelaita, and S. Sicks. 1996. J. Cell Biol. 135:1377-1382). We report here that although JNK is activated by detachment of normal MDCK cells, study of cell lines expressing activated signaling proteins usually controlled by Ras shows that stimulation of JNK fails to correlate with induction of anoikis. Activated phosphoinositide 3-OH kinase and activated PKB/Akt protect MDCK cells from detachment-induced apoptosis without suppressing JNK activation. Conversely, activated Raf and dominant negative SEK1, a JNK kinase, attenuate detachment-induced JNK activation without protecting from apoptosis. zVAD-fmk, a peptide inhibitor of caspases, prevents MDCK cell anoikis without affecting JNK activation. p38, a related stress-activated kinase, is also stimulated by detachment from matrix, but inhibition of this kinase with SB 203580 does not protect from anoikis. It is therefore unlikely that either JNK or p38 play a direct role in detachment-induced programmed cell death in epithelial cells.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Epithelial Cells/cytology , Mitogen-Activated Protein Kinases , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Adhesion , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Dogs , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Proto-Oncogene Proteins p21(ras)/metabolism , Pyridines/pharmacology , Signal Transduction , p38 Mitogen-Activated Protein Kinases
10.
J Cell Biol ; 151(7): 1549-60, 2000 Dec 25.
Article in English | MEDLINE | ID: mdl-11134082

ABSTRACT

Attachment of many cell types to extracellular matrix proteins triggers cell spreading, a process that strengthens cell adhesion and is a prerequisite for many adhesion-dependent processes including cell migration, survival, and proliferation. Cell spreading requires integrins with intact beta cytoplasmic domains, presumably to connect integrins with the actin cytoskeleton and to activate signaling pathways that promote cell spreading. Several signaling proteins are known to regulate cell spreading, including R-Ras, PI 3-kinase, PKCepsilon and Rac1; however, it is not known whether they do so through a mechanism involving integrin beta cytoplasmic domains. To study the mechanisms whereby cell spreading is regulated by integrin beta cytoplasmic domains, we inhibited cell spreading on collagen I or fibrinogen by expressing tac-beta1, a dominant-negative inhibitor of integrin function, and examined whether cell spreading could be restored by the coexpression of either V38R-Ras, p110alpha-CAAX, myr-PKCepsilon, or L61Rac1. Each of these activated signaling proteins was able to restore cell spreading as assayed by an increase in the area of cells expressing tac-beta1. R-Ras and Rac1 rescued cell spreading in a GTP-dependent manner, whereas PKCstraightepsilon required an intact kinase domain. Importantly, each of these signaling proteins required intact beta cytoplasmic domains on the integrins mediating adhesion in order to restore cell spreading. In addition, the rescue of cell spreading by V38R-Ras was inhibited by LY294002, suggesting that PI 3-kinase activity is required for V38R-Ras to restore cell spreading. In contrast, L61Rac1 and myr-PKCstraightepsilon each increased cell spreading independent of PI 3-kinase activity. Additionally, the dominant-negative mutant of Rac1, N17Rac1, abrogated cell spreading and inhibited the ability of p110alpha-CAAX and myr-PKCstraightepsilon to increase cell spreading. These studies suggest that R-Ras, PI 3-kinase, Rac1 and PKCepsilon require the function of integrin beta cytoplasmic domains to regulate cell spreading and that Rac1 is downstream of PI 3-kinase and PKCepsilon in a pathway involving integrin beta cytoplasmic domain function in cell spreading.


Subject(s)
GTP Phosphohydrolases/metabolism , Integrin beta1/chemistry , Integrin beta1/metabolism , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , rac1 GTP-Binding Protein/metabolism , ras Proteins/metabolism , Amino Acid Substitution/genetics , Animals , CHO Cells , Cell Adhesion , Cell Size , Collagen/metabolism , Cricetinae , Cytoplasm/chemistry , Enzyme Activation , Fibroblasts , Fibronectins/metabolism , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , Genes, Dominant/genetics , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Models, Biological , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Kinase C-epsilon , Protein Structure, Tertiary , Protein Subunits , Signal Transduction , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/genetics , ras Proteins/chemistry , ras Proteins/genetics
11.
Trends Biochem Sci ; 15(12): 469-72, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2127644

ABSTRACT

Considerable advances have recently been made in understanding the structure and function of the proteins encoded by the ras proto-oncogenes. In addition, a large number of ras-related small GTP-binding proteins with very diverse activities have now been identified. This review explores developments in this rapidly expanding field.


Subject(s)
GTP-Binding Proteins/genetics , Oncogene Protein p21(ras) , Animals
12.
Trends Biochem Sci ; 22(9): 355-8, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9301337

ABSTRACT

PKB/Akt is a serine/threonine kinase that contains a pleckstrin-homology (PH) domain and is activated in response to growth-factor treatment of cells by a mechanism involving phosphoinositide 3-OH kinase. PKB/Akt provides a survival signal that protects cells from apoptosis induced by various stresses, perhaps explaining its discovery as a retroviral oncogene and its amplification in many human tumours.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Cell Survival/physiology , Humans , Models, Biological , Phosphatidylinositol 3-Kinases , Phosphotransferases/metabolism , Proto-Oncogene Proteins c-akt
13.
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
14.
Curr Opin Genet Dev ; 2(1): 13-8, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1633420

ABSTRACT

Ras proteins play a critical role in controlling normal cellular growth and, when activated by mutation, in causing malignant transformation. Regulation of p21ras is achieved by GTPase activating proteins, which control the rate of hydrolysis of GTP to GDP, and also by GDP dissociation stimulators, which catalyze the exchange of guanine nucleotides. Several such proteins have now been identified and their control mechanisms characterized.


Subject(s)
Proteins/physiology , Proto-Oncogene Proteins p21(ras) , Animals , GTPase-Activating Proteins , Gene Expression Regulation , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Neurofibromin 1 , ras GTPase-Activating Proteins
15.
Curr Opin Genet Dev ; 8(1): 49-54, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9529605

ABSTRACT

Activated Ras proteins have either positive or negative effects on the regulation of apoptosis depending on cell type and other factors. In part, this is due to the ability of Ras to control directly multiple effector pathways, including PI3-kinase, which provides a universal survival signal, and Raf, which can inhibit survival. The mechanisms remain partly unclear, however, especially with regard to Raf effects on apoptosis regulation. Recently Ras has been shown to be able to protect cells from apoptosis either through activation of PKB/Akt via PI3-kinase, or through activation of NF-kappa B.


Subject(s)
Apoptosis , Signal Transduction , ras Proteins/metabolism , Animals , Humans
16.
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.
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
18.
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
19.
Curr Biol ; 7(4): R258-60, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9162506

ABSTRACT

The Ras proteins are key mediators of the early cellular response to mitogens; the way in which they influence the later events in the cell cycle is beginning to fall into place.


Subject(s)
Cell Cycle , Signal Transduction , ras Proteins/physiology , 3T3 Cells , Animals , Cell Cycle/drug effects , Cells, Cultured , Cyclin-Dependent Kinases/metabolism , Cyclins/physiology , Humans , Mice , Mitogens/pharmacology , Models, Biological
20.
Curr Biol ; 9(5): R176-9, 1999 Mar 11.
Article in English | MEDLINE | ID: mdl-10074440

ABSTRACT

Programmed cell death has seemed to be regulated in quite different ways in mammals and Drosophila. Recent results on the way Ras and downstream pathways can influence cell-death induction suggest the regulatory pathways in these distinct organisms might be more similar than was at first sight apparent.


Subject(s)
Apoptosis , Drosophila Proteins , Insect Proteins/metabolism , Neuropeptides/metabolism , Peptides/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Drosophila/metabolism , Eye/metabolism , Humans , Insect Proteins/genetics , Neuropeptides/genetics , Peptides/genetics
SELECTION OF CITATIONS
SEARCH DETAIL