Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Pathol ; 252(1): 77-87, 2020 09.
Article in English | MEDLINE | ID: mdl-32558936

ABSTRACT

Atypical teratoid rhabdoid tumor (ATRT) is a fatal pediatric malignancy of the central neural system lacking effective treatment options. It belongs to the rhabdoid tumor family and is usually caused by biallelic inactivation of SMARCB1, encoding a key subunit of SWI/SNF chromatin remodeling complexes. Previous studies proposed that SMARCB1 loss drives rhabdoid tumor by promoting cell cycle through activating transcription of cyclin D1 while suppressing p16. However, low cyclin D1 protein expression is observed in most ATRT patient tumors. The underlying mechanism and therapeutic implication of this molecular trait remain unknown. Here, we show that SMARCB1 loss in ATRT leads to the reduction of cyclin D1 expression by upregulating MIR17HG, a microRNA (miRNA) cluster known to generate multiple miRNAs targeting CCND1. Furthermore, we find that this cyclin D1 deficiency in ATRT results in marked in vitro and in vivo sensitivity to the CDK4/6 inhibitor palbociclib as a single agent. Our study identifies a novel genetic interaction between SMARCB1 and MIR17HG in regulating cyclin D1 in ATRT and suggests a rationale to treat ATRT patients with FDA-approved CDK4/6 inhibitors. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Proteins/genetics , Rhabdoid Tumor/genetics , SMARCB1 Protein/genetics , Teratoma/genetics , Cell Line, Tumor , Cell Survival , Cyclin D1/metabolism , Humans , Proteins/metabolism , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , SMARCB1 Protein/metabolism , Teratoma/metabolism , Teratoma/pathology , Up-Regulation
2.
Mol Microbiol ; 111(4): 918-937, 2019 04.
Article in English | MEDLINE | ID: mdl-30556355

ABSTRACT

In bacteria, disulfide bonds confer stability on many proteins exported to the cell envelope or beyond, including bacterial virulence factors. Thus, proteins involved in disulfide bond formation represent good targets for the development of inhibitors that can act as antibiotics or anti-virulence agents, resulting in the simultaneous inactivation of several types of virulence factors. Here, we present evidence that the disulfide bond forming enzymes, DsbB and VKOR, are required for Pseudomonas aeruginosa pathogenicity and Mycobacterium tuberculosis survival respectively. We also report the results of a HTS of 216,767 compounds tested against P. aeruginosa DsbB1 and M. tuberculosis VKOR using Escherichia coli cells. Since both P. aeruginosa DsbB1 and M. tuberculosis VKOR complement an E. coli dsbB knockout, we screened simultaneously for inhibitors of each complemented E. coli strain expressing a disulfide-bond sensitive ß-galactosidase reported previously. The properties of several inhibitors obtained from these screens suggest they are a starting point for chemical modifications with potential for future antibacterial development.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Disulfides/metabolism , Membrane Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Pseudomonas aeruginosa/enzymology , Animals , Bacterial Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Female , High-Throughput Screening Assays , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/pathogenicity , Pseudomonas aeruginosa/pathogenicity , Small Molecule Libraries/pharmacology , Virulence , Virulence Factors/metabolism
3.
Mol Cell Proteomics ; 17(10): 1948-1964, 2018 10.
Article in English | MEDLINE | ID: mdl-30006486

ABSTRACT

Glioblastoma multiforme (GBM) is a highly aggressive and heterogeneous form of primary brain tumors, driven by a complex repertoire of oncogenic alterations, including the constitutively active epidermal growth factor receptor (EGFRvIII). EGFRvIII impacts both cell-intrinsic and non-cell autonomous aspects of GBM progression, including cell invasion, angiogenesis and modulation of the tumor microenvironment. This is, at least in part, attributable to the release and intercellular trafficking of extracellular vesicles (EVs), heterogeneous membrane structures containing multiple bioactive macromolecules. Here we analyzed the impact of EGFRvIII on the profile of glioma EVs using isogenic tumor cell lines, in which this oncogene exhibits a strong transforming activity. We observed that EGFRvIII expression alters the expression of EV-regulating genes (vesiculome) and EV properties, including their protein composition. Using mass spectrometry, quantitative proteomic analysis and Gene Ontology terms filters, we observed that EVs released by EGFRvIII-transformed cells were enriched for extracellular exosome and focal adhesion related proteins. Among them, we validated the association of pro-invasive proteins (CD44, BSG, CD151) with EVs of EGFRvIII expressing glioma cells, and downregulation of exosomal markers (CD81 and CD82) relative to EVs of EGFRvIII-negative cells. Nano-flow cytometry revealed that the EV output from individual glioma cell lines was highly heterogeneous, such that only a fraction of vesicles contained specific proteins (including EGFRvIII). Notably, cells expressing EGFRvIII released EVs double positive for CD44/BSG, and these proteins also colocalized in cellular filopodia. We also detected the expression of homophilic adhesion molecules and increased homologous EV uptake by EGFRvIII-positive glioma cells. These results suggest that oncogenic EGFRvIII reprograms the proteome and uptake of GBM-related EVs, a notion with considerable implications for their biological activity and properties relevant for the development of EV-based cancer biomarkers.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Extracellular Vesicles/metabolism , Glioblastoma/metabolism , Oncogenes , Proteome/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Extracellular Vesicles/ultrastructure , Female , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Mice , Neoplasm Invasiveness , Neoplasm Proteins/metabolism
4.
J Biol Chem ; 292(16): 6529-6541, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28232484

ABSTRACT

Disulfide bonds contribute to protein stability, activity, and folding in a variety of proteins, including many involved in bacterial virulence such as toxins, adhesins, flagella, and pili, among others. Therefore, inhibitors of disulfide bond formation enzymes could have profound effects on pathogen virulence. In the Escherichia coli disulfide bond formation pathway, the periplasmic protein DsbA introduces disulfide bonds into substrates, and then the cytoplasmic membrane protein DsbB reoxidizes DsbA's cysteines regenerating its activity. Thus, DsbB generates a protein disulfide bond de novo by transferring electrons to the quinone pool. We previously identified an effective pyridazinone-related inhibitor of DsbB enzymes from several Gram-negative bacteria. To map the protein residues that are important for the interaction with this inhibitor, we randomly mutagenized by error-prone PCR the E. coli dsbB gene and selected dsbB mutants that confer resistance to this drug using two approaches. We characterized in vivo and in vitro some of these mutants that map to two areas in the structure of DsbB, one located between the two first transmembrane segments where the quinone ring binds and the other located in the second periplasmic loop of DsbB, which interacts with DsbA. In addition, we show that a mutant version of a protein involved in lipopolysaccharide assembly, lptD4213, is synthetically lethal with the deletion of dsbB as well as with DsbB inhibitors. This finding suggests that drugs decreasing LptD assembly may be synthetically lethal with inhibitors of the Dsb pathway, potentiating the antibiotic effects.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Proteins/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Membrane Proteins/chemistry , Mutation , Anti-Bacterial Agents/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Disulfides/chemistry , Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Gene Library , Kinetics , Lipopolysaccharides/chemistry , Membrane Proteins/genetics , Mutagenesis , Polymerase Chain Reaction , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/genetics , Pyridazines/chemistry , Quinones/chemistry , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Temperature , Virulence
5.
Mol Microbiol ; 103(3): 413-422, 2017 02.
Article in English | MEDLINE | ID: mdl-27785850

ABSTRACT

Disulfide bonds are found in many proteins associated with the cell wall of Escherichia coli, and for some of these proteins the disulfide bond is critical to their stability and function. One protein found to contain a disulfide bond is the essential cell division protein FtsN, but the importance of this bond to the protein's structural integrity is unclear. While it evidently plays a role in the proper folding of the SPOR domain of FtsN, this domain is non-essential, suggesting that the disulfide bond might also be dispensable. However, we find that FtsN mutants lacking cysteines give rise to filamentous growth. Furthermore, FtsN protein levels in strains expressing these mutants were significantly lower than in a strain expressing the wild-type allele, as were FtsN levels in strains incapable of making disulfide bonds (dsb- ) exposed to anaerobic conditions. These results strongly suggest that FtsN lacking a disulfide bond is unstable, thereby making this disulfide critical for function. We have previously found that dsb- strains fail to grow anaerobically, and the results presented here suggest that this growth defect may be due in part to misfolded FtsN. Thus, proper cell division in E. coli is dependent upon disulfide bond formation.


Subject(s)
Escherichia coli Proteins/metabolism , Membrane Proteins/metabolism , Cell Division/physiology , Cell Wall/metabolism , Disulfides , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Membrane Proteins/genetics , Membrane Proteins/physiology , Protein Binding , Protein Domains , Structure-Activity Relationship
6.
J Bacteriol ; 199(16)2017 08 15.
Article in English | MEDLINE | ID: mdl-28559299

ABSTRACT

Disulfide bonds are critical to the stability and function of many bacterial proteins. In the periplasm of Escherichia coli, intramolecular disulfide bond formation is catalyzed by the two-component disulfide bond forming (DSB) system. Inactivation of the DSB pathway has been shown to lead to a number of pleotropic effects, although cells remain viable under standard laboratory conditions. However, we show here that dsb strains of E. coli reversibly filament under aerobic conditions and fail to grow anaerobically unless a strong oxidant is provided in the growth medium. These findings demonstrate that the background disulfide bond formation necessary to maintain the viability of dsb strains is oxygen dependent. LptD, a key component of the lipopolysaccharide transport system, fails to fold properly in dsb strains exposed to anaerobic conditions, suggesting that these mutants may have defects in outer membrane assembly. We also show that anaerobic growth of dsb mutants can be restored by suppressor mutations in the disulfide bond isomerization system. Overall, our results underscore the importance of proper disulfide bond formation to pathways critical to E. coli viability under conditions where oxygen is limited.IMPORTANCE While the disulfide bond formation (DSB) system of E. coli has been studied for decades and has been shown to play an important role in the proper folding of many proteins, including some associated with virulence, it was considered dispensable for growth under most laboratory conditions. This work represents the first attempt to study the effects of the DSB system under strictly anaerobic conditions, simulating the environment encountered by pathogenic E. coli strains in the human intestinal tract. By demonstrating that the DSB system is essential for growth under such conditions, this work suggests that compounds inhibiting Dsb enzymes might act not only as antivirulents but also as true antibiotics.


Subject(s)
Disulfides/metabolism , Escherichia coli/growth & development , Escherichia coli/metabolism , Anaerobiosis , Culture Media/chemistry , Escherichia coli/cytology , Gene Deletion , Microbial Viability , Oxidants/metabolism
7.
Am J Pathol ; 186(2): 446-59, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26687818

ABSTRACT

Pediatric embryonal brain tumors with multilayered rosettes demonstrate a unique oncogenic amplification of the chromosome 19 miRNA cluster, C19MC. Because oncogenic lesions often cause deregulation of vascular effectors, including procoagulant tissue factor (TF), this study explores whether there is a link between C19MC oncogenic miRNAs (oncomirs) and the coagulant properties of cancer cells, a question previously not studied. In a pediatric embryonal brain tumor tissue microarray, we observed an association between C19MC amplification and reduced fibrin content and TF expression, indicative of reduced procoagulant activity. In medulloblastoma cell lines (DAOY and UW228) engineered to express miR-520g, a biologically active constituent of the C19MC cluster, we observed reduced TF expression, procoagulant and TF signaling activities (responses to factor VIIa stimulation), and diminished TF emission as cargo of extracellular vesicles. Antimir and luciferase reporter assays revealed a specific and direct effect of miR-520g on the TF 3' untranslated region. Although the endogenous MIR520G locus is methylated in differentiated cells, exposure of DAOY cells to 5-aza-2'-deoxycytidine or their growth as stem cell-like spheres up-regulated endogenous miR-520g with a coincident reduction in TF expression. We propose that the properties of tumors harboring oncomirs may include unique alterations of the vascular microenvironment, including deregulation of TF, with a possible impact on the biology, therapy, and hemostatic adverse effects of both disease progression and treatment.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Tumor Microenvironment/genetics , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Differentiation/genetics , Humans , Medulloblastoma/genetics , Signal Transduction/genetics , Stem Cells/cytology , Thromboplastin/metabolism
8.
Nat Chem Biol ; 11(4): 292-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25686372

ABSTRACT

In bacteria, disulfide bonds confer stability on many proteins exported to the cell envelope or beyond. These proteins include numerous bacterial virulence factors, and thus bacterial enzymes that promote disulfide bond formation represent targets for compounds inhibiting bacterial virulence. Here, we describe a new target- and cell-based screening methodology for identifying compounds that inhibit the disulfide bond-forming enzymes Escherichia coli DsbB (EcDsbB) or Mycobacterium tuberculosis VKOR (MtbVKOR), which can replace EcDsbB, although the two are not homologs. Initial screening of 51,487 compounds yielded six specifically inhibiting EcDsbB. These compounds share a structural motif and do not inhibit MtbVKOR. A medicinal chemistry approach led us to select related compounds, some of which are much more effective DsbB inhibitors than those found in the screen. These compounds inhibit purified DsbB and prevent anaerobic growth of E. coli. Furthermore, these compounds inhibit all but one of the DsbBs of nine other Gram-negative pathogenic bacteria tested.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Escherichia coli/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Mycobacterium tuberculosis/metabolism , Agar/chemistry , Anti-Bacterial Agents/chemistry , Catalytic Domain , Chemistry, Pharmaceutical/methods , Combinatorial Chemistry Techniques , Disulfides , Dose-Response Relationship, Drug , Drug Design , Electron Transport , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Mass Spectrometry , Microbial Sensitivity Tests , Mycobacterium smegmatis/metabolism , Protein Conformation , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/chemistry , Pseudomonas aeruginosa/metabolism
9.
Proc Natl Acad Sci U S A ; 111(9): 3544-9, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24520174

ABSTRACT

The coagulation system links immediate (hemostatic) and late (inflammatory, angiogenic) tissue responses to injury, a continuum that often is subverted in cancer. Here we provide evidence that tumor dormancy is influenced by tissue factor (TF), the cancer cell-associated initiator of the coagulation system and a signaling receptor. Thus, indolent human glioma cells deficient for TF remain viable but permanently dormant at the injection site for nearly a year, whereas the expression of TF leads to a step-wise transition to latent and overt tumor growth phases, a process that is preceded by recruitment of vascular (CD105(+)) and myeloid (CD11b(+) and F4/80(+)) cells. Importantly, the microenvironment orchestrated by TF expression drives permanent changes in the phenotype, gene-expression profile, DNA copy number, and DNA methylation state of the tumor cells that escape from dormancy. We postulate that procoagulant events in the tissue microenvironment (niche) may affect the fate of occult tumor cells, including their biological and genetic progression to initiate a full-blown malignancy.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Glioma/physiopathology , Neoplastic Processes , Thromboplastin/metabolism , Tumor Microenvironment/genetics , Animals , Cell Line, Tumor , DNA Copy Number Variations , DNA Methylation , Gene Expression Profiling , Gene Silencing , Glioma/metabolism , Humans , Mice , Mutation/genetics , Statistics, Nonparametric
10.
J Biol Chem ; 290(40): 24534-46, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26272609

ABSTRACT

Cancer cells emit extracellular vesicles (EVs) containing unique molecular signatures. Here, we report that the oncogenic EGF receptor (EGFR) and its inhibitors reprogram phosphoproteomes and cargo of tumor cell-derived EVs. Thus, phosphorylated EGFR (P-EGFR) and several other receptor tyrosine kinases can be detected in EVs purified from plasma of tumor-bearing mice and from conditioned media of cultured cancer cells. Treatment of EGFR-driven tumor cells with second generation EGFR kinase inhibitors (EKIs), including CI-1033 and PF-00299804 but not with anti-EGFR antibody (Cetuximab) or etoposide, triggers a burst in emission of exosome-like EVs containing EGFR, P-EGFR, and genomic DNA (exo-gDNA). The EV release can be attenuated by treatment with inhibitors of exosome biogenesis (GW4869) and caspase pathways (ZVAD). The content of P-EGFR isoforms (Tyr-845, Tyr-1068, and Tyr-1173), ERK, and AKT varies between cells and their corresponding EVs and as a function of EKI treatment. Immunocapture experiments reveal the presence of EGFR and exo-gDNA within the same EV population following EKI treatment. These findings suggest that targeted agents may induce cancer cells to change the EV emission profiles reflective of drug-related therapeutic stress. We suggest that EV-based assays may serve as companion diagnostics for targeted anticancer agents.


Subject(s)
DNA/chemistry , ErbB Receptors/metabolism , Exosomes/metabolism , Extracellular Vesicles/metabolism , Phosphoproteins/metabolism , Animals , Antineoplastic Agents/chemistry , Biomarkers/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Cetuximab/chemistry , Culture Media, Conditioned/chemistry , Etoposide/chemistry , Glioma/metabolism , Humans , Mice , Mice, SCID , Morpholines/chemistry , Neoplasm Transplantation , Neoplasms/metabolism , Phosphorylation , Proteomics , Quinazolinones/chemistry , Transfection
11.
Angiogenesis ; 19(1): 25-38, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26374632

ABSTRACT

Oncogenic transformation is believed to impact the vascular phenotype and microenvironment in cancer, at least in part, through mechanisms involving extracellular vesicles (EVs). We explored these questions in the context of acute promyelocytic leukemia cells (NB4) expressing oncogenic fusion protein, PML-RARa and exquisitely sensitive to its clinically used antagonist, the all-trans retinoic acid (ATRA). We report that NB4 cells produce considerable numbers of EVs, which are readily taken up by cultured endothelial cells triggering their increased survival. NB4 EVs contain PML-RARa transcript, but no detectable protein, which is also absent in endothelial cells upon the vesicle uptake, thereby precluding an active intercellular trafficking of this oncogene in this setting. ATRA treatment changes the emission profile of NB4-related EVs resulting in preponderance of smaller vesicles, an effect that occurs in parallel with the onset of cellular differentiation. ATRA also increases IL-8 mRNA and protein content in NB4 cells and their EVs, while decreasing the levels of VEGF and tissue factor (TF). Endothelial cell uptake of NB4-derived EVs renders these cells more TF-positive and procoagulant, and this effect is diminished by pre-treatment of EV donor cells with ATRA. Profiling angiogenesis-related transcripts in intact and ATRA-treated APL cells and their EVs reveals multiple differences attributable to cellular responses and EV molecular packaging. These observations point to the potential significance of changes in the angiogenic signature and activity associated with EVs released from tumor cells subjected to targeted therapy.


Subject(s)
Extracellular Vesicles/pathology , Leukemia, Promyelocytic, Acute/pathology , Neovascularization, Pathologic/pathology , Oncogene Proteins, Fusion/metabolism , Blood Coagulation/drug effects , Cell Line, Tumor , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Gene Expression Regulation, Leukemic/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Neovascularization, Pathologic/genetics , Oncogene Proteins, Fusion/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thromboplastin/metabolism , Tretinoin/pharmacology
12.
Virol J ; 12: 18, 2015 Feb 08.
Article in English | MEDLINE | ID: mdl-25889293

ABSTRACT

BACKGROUND: Variant high pathogenicity avian influenza (HPAI) H5 viruses have recently emerged as a result of reassortment of the H5 haemagglutinin (HA) gene with different neuraminidase (NA) genes, including NA1, NA2, NA5, NA6 and NA8. These viruses form a newly proposed HA clade 2.3.4.4 (previously provisionally referred to as clade 2.3.4.6), and have been implicated in disease outbreaks in poultry in China, South Korea, Laos, Japan and Vietnam and a human fatality in China. There is real concern that this new clade may be wide spread and not readily identified using existing diagnostic algorithms. FINDINGS: Fluorescent probe based reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR) assays were developed to facilitate the identification of novel clade 2.3.4.4 viruses of H5N6 subtype emerging in Asia. Assays were aimed at the haemagglutinin (HA) gene for clade identification and at the NA gene to identify N6. The HA assay employing a minor groove binder (MGB) probe was able to detect and differentiate A/duck/Laos/XBY004/2014(H5N6) and related influenza A(H5N6) virus isolates belonging to the proposed clade 2.3.4.4 from other H5 HPAI viruses. In addition, an Eurasian N6 assay was able to differentiate N6 from other NA subtypes. CONCLUSIONS: Laos influenza A(H5N6) virus representative of proposed clade 2.3.4.4, was detected and differentiated from viruses in other H5N1 clades using a clade-specific HA RT-qPCR assay whereas the N6-NA subtype was determined by an Eurasian N6 RT-qPCR assay. Such a clade-specific assay would be of particular value for surveillance and in diagnostic laboratories where sequencing is not readily available.


Subject(s)
Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Influenza in Birds/diagnosis , Influenza in Birds/virology , Molecular Diagnostic Techniques/methods , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Asia , Birds , Poultry
13.
Proc Natl Acad Sci U S A ; 109(30): 12153-8, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22778399

ABSTRACT

Bacteroides fragilis can replicate in atmospheres containing ≤0.05% oxygen, but higher concentrations arrest growth by an unknown mechanism. Here we show that inactivation of a single gene, oxe (i.e., oxygen enabled) in B. fragilis allows for growth in concentrations as high as 2% oxygen while increasing the tolerance of this organism to room air. Known components of the oxidative stress response including the ahpC, kat, batA-E, and tpx genes were not individually important for microaerobic growth. However, a Δoxe strain scavenged H(2)O(2) at a faster rate than WT, indicating that reactive oxygen species may play a critical role in limiting growth of this organism to low-oxygen environments. Clinical isolates of B. fragilis displayed a greater capacity for growth under microaerobic conditions than fecal isolates, with some encoding polymorphisms in oxe. Additionally, isolation of oxygen-enabled mutants of Bacteroides thetaiotaomicron suggests that Oxe may mediate growth arrest of other anaerobes in oxygenated environments.


Subject(s)
Bacteroides fragilis/growth & development , Bacteroides fragilis/genetics , Gene Silencing , Genes, Bacterial/genetics , Oxidative Stress/genetics , Oxygen/metabolism , Anaerobiosis , Bacteroides fragilis/metabolism , DNA Primers/genetics , Hydrogen Peroxide/metabolism , Plasmids/genetics , Reactive Oxygen Species , Sequence Analysis, DNA , Species Specificity
14.
Emerg Infect Dis ; 20(6): 1040-3, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24856477

ABSTRACT

Recently discovered tick-borne phleboviruses have been associated with severe disease and death among persons in Asia and the United States. We report the discovery of a novel tick phlebovirus in Tasmania State, Australia, that is closely related to those zoonotic viruses found in Asia and North America.


Subject(s)
Bird Diseases/epidemiology , Disease Outbreaks , Genome, Viral , Phlebotomus Fever/veterinary , Phlebovirus/genetics , RNA, Viral/genetics , Ticks/virology , Animals , Bird Diseases/virology , Birds , Disease Vectors , High-Throughput Nucleotide Sequencing , Humans , Phlebotomus Fever/epidemiology , Phlebotomus Fever/virology , Phlebovirus/classification , Phlebovirus/isolation & purification , Phylogeny , Tasmania
15.
Biochem Biophys Res Commun ; 451(2): 295-301, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25086355

ABSTRACT

Cell free DNA is often regarded as a source of genetic cancer biomarkers, but the related mechanisms of DNA release, composition and biological activity remain unclear. Here we show that rat epithelial cell transformation by the human H-ras oncogene leads to an increase in production of small, exosomal-like extracellular vesicles by viable cancer cells. These EVs contain chromatin-associated double-stranded DNA fragments covering the entire host genome, including full-length H-ras. Oncogenic N-ras and SV40LT sequences were also found in EVs emitted from spontaneous mouse brain tumor cells. Disruption of acidic sphingomyelinase and the p53/Rb pathway did not block emission of EV-related oncogenic DNA. Exposure of non-transformed RAT-1 cells to EVs containing mutant H-ras DNA led to the uptake and retention of this material for an extended (30days) but transient period of time, and stimulated cell proliferation. Thus, our study suggests that H-ras-mediated transformation stimulates vesicular emission of this histone-bound oncogene, which may interact with non-transformed cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA, Neoplasm/metabolism , DNA/metabolism , Genes, ras , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA/genetics , DNA, Neoplasm/genetics , Exosomes/genetics , Exosomes/metabolism , Exosomes/ultrastructure , Genome, Human , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Mice , Microscopy, Electron, Transmission , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism
16.
Biochem Biophys Res Commun ; 454(2): 262-8, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25450387

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive form of glial brain tumors, associated with angiogenesis, thrombosis, and upregulation of tissue factor (TF), the key cellular trigger of coagulation and signaling. Since TF is upregulated by oncogenic mutations occurring in different subsets of human brain tumors we investigated whether TF contributes to tumourigenesis driven by oncogenic activation of EGFR (EGFRvIII) and RAS pathways in the brain. Here we show that TF expression correlates with poor prognosis in glioma, but not in GBM. In situ, the TF protein expression is heterogeneously expressed in adult and pediatric gliomas. GBM cells harboring EGFRvIII (U373vIII) grow aggressively as xenografts in SCID mice and their progression is delayed by administration of monoclonal antibodies blocking coagulant (CNTO 859) and signaling (10H10) effects of TF in vivo. Mice in which TF gene is disrupted in the neuroectodermal lineage exhibit delayed progression of spontaneous brain tumors driven by oncogenic N-ras and SV40 large T antigen (SV40LT) expressed under the control of sleeping beauty transposase. Reduced host TF levels in low-TF/SCID hypomorphic mice mitigated growth of glioma subcutaneously but not in the brain. Thus, we suggest that tumor-associated TF may serve as therapeutic target in the context of oncogene-driven disease progression in a subset of glioma.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioma/genetics , Oncogenes , Thromboplastin/genetics , Adolescent , Adult , Animals , Brain/metabolism , Brain/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , ErbB Receptors/metabolism , Gene Deletion , Glioblastoma/diagnosis , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/diagnosis , Glioma/metabolism , Glioma/pathology , Humans , Mice , Mice, SCID , Prognosis , Thromboplastin/metabolism
17.
Semin Thromb Hemost ; 40(3): 284-95, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24599437

ABSTRACT

Genetically altered cancer cells both provoke and respond to changes in their microenvironment, stroma, and vasculature. This includes local and systemic activation of the coagulation system, which is a part of the functional continuum involving inflammation, angiogenesis, and tissue repair programs, often reactivated in cancer. These responses coevolve with, and contribute to, the malignant process. Cancer coagulopathy is not only a source of comorbidity and mortality in cancer patients, but it also affects the disease biology including processes of tumor growth, initiation, dormancy, invasion, angiogenesis, metastasis, and therapeutic responsiveness. Notably, genetic and cellular differences between different cancer types are paralleled by a degree of diversity in the related coagulation system perturbations. Although some of these differences may be unspecific, iatrogenic, or indirect in nature, others are affected by oncogenic pathways (RAS, EGFR, HER2, MET, PTEN, and TP53) activated in cancer cells due to driver mutations of critical genes. Such mutations cooperate with hypoxia, cellular differentiation, and other influences to alter the expression of tissue factor, protease-activated receptors (e.g., PAR-1 and PAR-2), coagulation factors (FII and FVII), and other molecules related to the hemostatic system. Oncogenic pathways also control secretion of some of these entities from cancer cells, either as soluble proteins, or as cargo of extracellular vesicles/microparticles. Moreover, emerging evidence suggests that the expression profiles of coagulation-related genes differ between molecularly and genetically distinct subgroups of specific malignancies such as glioblastoma multiforme and medulloblastoma. Certain hereditary thrombophilias may also affect cancer pathogenesis. We suggest that mechanisms of cancer coagulopathy may be more diverse and genetically modulated than hitherto realized. If so, a possibility may exist to deliver more personalized, biologically based, anticoagulation, and thereby improve patient survival.


Subject(s)
Neoplasms/complications , Neoplasms/genetics , Thrombosis/complications , Thrombosis/genetics , Animals , Humans , Neoplasms/blood
18.
Exp Cell Res ; 319(17): 2747-57, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23954818

ABSTRACT

Transitions of the cancer cell phenotype between epithelial and mesenchymal states (EMT) are likely to alter the patterns of intercellular communication. In this regard we have previously documented that EMT-like changes trigger quantitative rearrangements in exosomal vesicle emission in A431 cancer cells driven by oncogenic epidermal growth factor receptor (EGFR). Here we report that extracellular vesicles (EVs) produced by these cancer cells in their epithelial and mesenchymal states exhibit profound qualitative differences in their proteome. Thus, induction of the EMT-like state through blockade of E-cadherin and EGFR stimulation provoked a mesenchymal shift in cellular morphology and enrichment in the CD44-high/CD24-low immunophenotype, often linked to cellular stemness. This change also resulted in reprogramming of the EV-related proteome (distinct from that of corresponding cells), which contained 30 unique protein signals, and revealed enrichment in pathways related to cellular growth, cell-to-cell signaling, and cell movement. Some of the most prominent EV-related proteins were validated, including integrin α2 and tetraspanin CD9. We propose that changes in cellular differentiation status translate into unique qualitative rearrangements in the cargo of EVs, a process that may have implications for intercellular communication and could serve as source of new biomarkers to detect EMT-like processes in cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Exosomes/metabolism , Proteome/genetics , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cadherins/drug effects , Cadherins/metabolism , Cell Line, Tumor , Cellular Reprogramming , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Integrin alpha2/genetics , Integrin alpha2/metabolism , Neoplastic Stem Cells/metabolism , Proteome/metabolism , Tetraspanin 29/genetics , Tetraspanin 29/metabolism
19.
Eval Program Plann ; 107: 102490, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39216425

ABSTRACT

This paper reviews and evaluates four U.S. P3 toll highways, identifying their performance and common challenges. Contrary to the conventional wisdom that federal and state governments should fund highways, this paper argues that users should bear the cost for both efficiency and equity considerations. The shift from government investment and operation of highways to Public-Private Partnerships (P3) was driven by the lack of public funding, more productive competitive business practices, and the government's desire for upfront payments to fund other unrelated public projects. However, these justifiable public benefits were accompanied by significant problems. Highways were often leased for 35 to 75 years, depending on the total equity and loans of the private partner and various risk factors.

20.
Nat Commun ; 15(1): 2865, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38570528

ABSTRACT

Targeting neovascularization in glioblastoma (GBM) is hampered by poor understanding of the underlying mechanisms and unclear linkages to tumour molecular landscapes. Here we report that different molecular subtypes of human glioma stem cells (GSC) trigger distinct endothelial responses involving either angiogenic or circumferential vascular growth (vasectasia). The latter process is selectively triggered by mesenchymal (but not proneural) GSCs and is mediated by a subset of extracellular vesicles (EVs) able to transfer EGFR/EGFRvIII transcript to endothelial cells. Inhibition of the expression and phosphorylation of EGFR in endothelial cells, either pharmacologically (Dacomitinib) or genetically (gene editing), abolishes their EV responses in vitro and disrupts vasectasia in vivo. Therapeutic inhibition of EGFR markedly extends anticancer effects of VEGF blockade in mice, coupled with abrogation of vasectasia and prolonged survival. Thus, vasectasia driven by intercellular transfer of oncogenic EGFR may represent a new therapeutic target in a subset of GBMs.


Subject(s)
Brain Neoplasms , Extracellular Vesicles , Glioblastoma , Glioma , Humans , Animals , Mice , Endothelial Cells/metabolism , Glioma/metabolism , Glioblastoma/metabolism , ErbB Receptors/metabolism , Extracellular Vesicles/metabolism , Neoplastic Stem Cells/metabolism , Brain Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL