Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Microbes Infect ; 14(11): 951-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22542841

ABSTRACT

The ability of opportunistic bacterial pathogens to grow in biofilms is decisive in the pathogenesis of chronic infectious diseases. Growth within biofilms does not only protect the bacteria against the host immune system but also from the killing by antimicrobial agents. Here, we introduce a mouse model in which intravenously administered planktonic Pseudomonas aeruginosa bacteria are enriched in transplantable subcutaneous mouse tumors. Electron microscopy images provide evidence that such bacteria reside in the tumor tissue within biofilm structures. Immunohistology furthermore demonstrated that infection of the tumor tissue elicits a host response characterized by strong neutrophilic influx. Interestingly, the biofilm defective PA14 pqsA transposon mutant formed less biofilm in vivo and was more susceptible to clearance by intravenous ciprofloxacin treatment as compared to the wild-type control. In conclusion, we have established an experimentally tractable model that may serve to identify novel bacterial and host factors important for in vivo biofilm formation and to re-evaluate bactericidal and anti-biofilm effects of currently used and novel antibacterial compounds.


Subject(s)
Biofilms , Neoplasms, Experimental/microbiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , 4-Quinolones/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Ciprofloxacin/pharmacology , Colony Count, Microbial , Disease Models, Animal , Drug Resistance, Bacterial , Female , Immunohistochemistry , Luminescent Measurements , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Neoplasm Transplantation , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development
2.
Cell Microbiol ; 13(8): 1223-33, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21507181

ABSTRACT

Systemic administration of Salmonella enterica serovar Typhimurium to tumour bearing mice results in preferential colonization of the tumours and retardation of tumour growth. Although the bacteria are able to invade the tumour cells in vitro, in tumours they were never detected intracellularly. Ultrastructural analysis of Salmonella-colonized tumours revealed that the bacteria had formed biofilms. Interestingly, depletion of neutrophilic granulocytes drastically reduced biofilm formation. Obviously, bacteria form biofilms in response to the immune reactions of the host. Importantly, we tested Salmonella mutants that were no longer able to form biofilms by deleting central regulators of biofilm formation. Such bacteria could be observed intracellularly in immune cells of the host or in tumour cells. Thus, tumour colonizing S. typhimurium might form biofilms as protection against phagocytosis. Since other bacteria are behaving similarly, solid murine tumours might represent a unique model to study biofilm formation in vivo.


Subject(s)
Biofilms/growth & development , Host-Pathogen Interactions , Neoplasms/microbiology , Salmonella typhimurium/growth & development , Salmonella typhimurium/physiology , Animals , Antibiosis , Disease Models, Animal , Mice , Microscopy, Electron , Neutrophils/microbiology , Phagocytosis , Salmonella Infections, Animal , Salmonella typhimurium/immunology
3.
J Clin Invest ; 120(4): 1151-64, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20237412

ABSTRACT

Angiogenesis is a hallmark of malignant neoplasias, as the formation of new blood vessels is required for tumors to acquire oxygen and nutrients essential for their continued growth and metastasis. However, the signaling pathways leading to tumor vascularization are not fully understood. Here, using a transplantable mouse tumor model, we have demonstrated that endogenous IFN-beta inhibits tumor angiogenesis through repression of genes encoding proangiogenic and homing factors in tumor-infiltrating neutrophils. We determined that IFN-beta-deficient mice injected with B16F10 melanoma or MCA205 fibrosarcoma cells developed faster-growing tumors with better-developed blood vessels than did syngeneic control mice. These tumors displayed enhanced infiltration by CD11b+Gr1+ neutrophils expressing elevated levels of the genes encoding the proangiogenic factors VEGF and MMP9 and the homing receptor CXCR4. They also expressed higher levels of the transcription factors c-myc and STAT3, known regulators of VEGF, MMP9, and CXCR4. In vitro, treatment of these tumor-infiltrating neutrophils with low levels of IFN-beta restored expression of proangiogenic factors to control levels. Moreover, depletion of these neutrophils inhibited tumor growth in both control and IFN-beta-deficient mice. We therefore suggest that constitutively produced endogenous IFN-beta is an important mediator of innate tumor surveillance. Further, we believe our data help to explain the therapeutic effect of IFN treatment during the early stages of cancer development.


Subject(s)
Interferon-beta/physiology , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/etiology , Neutrophils/physiology , Animals , CD11b Antigen/analysis , Female , Genes, myc , Killer Cells, Natural/physiology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Radiation Tolerance , STAT3 Transcription Factor/genetics
4.
PLoS One ; 4(8): e6692, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19693266

ABSTRACT

BACKGROUND: Several facultative anaerobic bacteria with potential therapeutic abilities are known to preferentially colonize solid tumors after systemic administration. How they efficiently find and invade the tumors is still unclear. However, this is an important issue to be clarified when bacteria should be tailored for application in cancer therapy. METHODOLOGY/PRINCIPAL FINDINGS: We describe the initial events of colonization of an ectopic transplantable tumor by Salmonella enterica serovar Typhimurium. Initially, after intravenous administration, bacteria were found in blood, spleen, and liver. Low numbers were also detected in tumors associated with blood vessels as could be observed by immunohistochemistry. A rapid increase of TNF-alpha in blood was observed at that time, in addition to other pro-inflammatory cytokines. This induced a tremendous influx of blood into the tumors by vascular disruption that could be visualized in H&E stainings and quantified by hemoglobin measurements of tumor homogenate. Most likely, together with the blood, bacteria were flushed into the tumor. In addition, blood influx was followed by necrosis formation, bacterial growth, and infiltration of neutrophilic granulocytes. Depletion of TNF-alpha retarded blood influx and delayed bacterial tumor-colonization. CONCLUSION: Our findings emphasize similarities between Gram-negative tumor-colonizing bacteria and tumor vascular disrupting agents and show the involvement of TNF-alpha in the initial phase of tumor-colonization by bacteria.


Subject(s)
Hemorrhage/microbiology , Neoplasms, Experimental/microbiology , Salmonella enterica/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Base Sequence , DNA Primers , Female , Hemorrhage/chemically induced , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction
5.
Microbes Infect ; 11(14-15): 1097-105, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19665575

ABSTRACT

The probiotic bacterium Escherichia coli Nissle 1917 (EcN) constitutes a prospective vector for delivering heterologous therapeutic molecules to treat several human disorders. To add versatility to this carrier system, bacteria should be equipped with expression modules that can be regulated deliberately in a temporal and quantitative manner. This approach is called in vivo remote control (IVRC) of bacterial vectors. Here, we have evaluated promoters P(araBAD), P(rhaBAD) and P(tet), which can be induced with L-arabinose, L-rhamnose or anhydrotetracycline, respectively. EcN harboring promoter constructs with luciferase as reporter gene were administered either orally to healthy mice or intravenously to tumor bearing animals. Subsequent to bacterial colonization of tissues, inducer substances were administered via the oral or systemic route. By use of in vivo bioluminescence imaging, the time course of reporter gene expression was analyzed. Each promoter displayed a specific in vivo induction profile depending on the niche of bacterial residence and the route of inducer administration. Importantly, we also observed colonization of gall bladders of mice when EcN was administered systemically at high doses. Bacteria in this anatomical compartment remained accessible to remote control of bacterial gene expression.


Subject(s)
Escherichia coli/drug effects , Gallbladder/microbiology , Gene Expression Regulation, Bacterial/drug effects , Intestines/microbiology , Neoplasms, Experimental/microbiology , Probiotics , Animals , Arabinose/administration & dosage , Arabinose/pharmacology , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Female , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , Rhamnose/administration & dosage , Rhamnose/pharmacology , Skin Neoplasms/microbiology , Tetracyclines/administration & dosage , Tetracyclines/pharmacology , Tissue Distribution
6.
Cancer Res ; 68(8): 2952-60, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413765

ABSTRACT

Administration of facultative anaerobic bacteria like Salmonella typhimurium, Shigella flexneri, and Escherichia coli to tumor-bearing mice leads to a preferential accumulation and proliferation of the microorganisms within the solid tumor. Until now, all known tumor-targeting bacteria have shown poor dissemination inside the tumors. They accumulate almost exclusively in large necrotic areas and spare a rim of viable tumor cells. Interestingly, the bacteria-containing necrotic region is separated from viable tumor cells by a barrier of host neutrophils that have immigrated into the tumor. We here report that depletion of host neutrophils results in a noticeably higher total number of bacteria in the tumor and that bacteria were now also able to migrate into vital tumor tissue. Most remarkably, an increase in the size of the necrosis was observed, and complete eradication of established tumors could be observed under these conditions. Thus, bacteria-mediated tumor therapy can be amplified by depletion of host neutrophils.


Subject(s)
Bacteria, Anaerobic/growth & development , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Neutrophils/microbiology , Animals , Bacteria, Anaerobic/isolation & purification , Cell Division , Escherichia coli/growth & development , Escherichia coli/isolation & purification , Female , Mice , Mice, Inbred BALB C , Necrosis , Salmonella typhimurium/growth & development , Salmonella typhimurium/isolation & purification , Shigella flexneri/growth & development , Shigella flexneri/isolation & purification
7.
Int J Med Microbiol ; 298(1-2): 21-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17702649

ABSTRACT

Live attenuated bacteria are well established as vaccines. Thus, their use as carriers for prophylactic and therapeutic macromolecules is a logical consequence. Here we describe several experimental applications of bacteria to carry heterologous macromolecules into the murine host. First, Listeria monocytogenes are described that are able to transfer eukaryotic expression plasmids into host cells for gene therapy. High multiplicities of infection are still required for efficient gene transfer and we point out some of the bottlenecks that counteract a more efficient transfer and application in vivo. Then, we describe Salmonella enterica serovar Typhimurium (S. typhimurium) as an expression plasmid transfer vehicle for oral DNA vaccination of mice. We demonstrate that the stabilization of the plasmid transformants results in an improved immune response. Stabilization was achieved by replacing the origin of replication of the original high-copy-number plasmid by a low-copy-number origin. Finally, we describe Salmonella carriers for the improved expression of heterologous proteins. We introduce a system in which the plasmid is carried as a single copy during cultivation but is amplified several fold upon infection of the host. Using the same in vivo inducible promoter for both protein expression and plasmid amplification, a substantial increase in antigen expression in vivo can be achieved. A modification of this approach is the introduction of inducible gene expression in vivo with a low-molecular-weight compound. Using P(BAD) promoter and L-arabinose as inducer we were able to deliberately activate genes in the bacterial carrier. No background activity could be observed with P(BAD) such that an inducible suicide gene could be introduced. This is adding an important safety feature to such live attenuated carrier bacteria.


Subject(s)
Bacterial Vaccines/immunology , Listeria monocytogenes/immunology , Salmonella typhimurium/immunology , Vaccines, DNA/immunology , Animals , Arabinose/pharmacology , Mice , Plasmids/immunology , Transformation, Genetic/immunology , Vaccines, Attenuated/immunology
8.
Cell Microbiol ; 9(8): 2008-21, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17419718

ABSTRACT

Cholesterol-dependent cytolysins (CDCs) represent a large family of conserved pore-forming toxins produced by several Gram-positive bacteria such as Listeria monocytogenes, Streptococcus pyrogenes and Bacillus anthracis. These toxins trigger a broad range of cellular responses that greatly influence pathogenesis. Using mast cells, we demonstrate that listeriolysin O (LLO), a prototype of CDCs produced by L. monocytogenes, triggers cellular responses such as degranulation and cytokine synthesis in a Ca(2+)-dependent manner. Ca(2+) signalling by LLO is due to Ca(2+) influx from extracellular milieu and release of from intracellular stores. We show that LLO-induced release of Ca(2+) from intracellular stores occurs via at least two mechanisms: (i) activation of intracellular Ca(2+) channels and (ii) a Ca(2+) channels independent mechanism. The former involves PLC-IP(3)R operated Ca(2+) channels activated via G-proteins and protein tyrosine kinases. For the latter, we propose a novel mechanism of intracellular Ca(2+) release involving injury of intracellular Ca(2+) stores such as the endoplasmic reticulum. In addition to Ca(2+) signalling, the discovery that LLO causes damage to an intracellular organelle provides a new perspective in our understanding of how CDCs affect target cells during infection by the respective bacterial pathogens.


Subject(s)
Calcium Channels/physiology , Calcium Signaling/physiology , Cholesterol/physiology , Heat-Shock Proteins/physiology , Hemolysin Proteins/physiology , Listeria monocytogenes/physiology , Animals , Bacterial Toxins/pharmacology , Calcium/metabolism , Cell Line , Cell Survival , Cytokines/metabolism , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , GTP-Binding Proteins/physiology , Heat-Shock Proteins/pharmacology , Hemolysin Proteins/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Intracellular Space/metabolism , Ion Channel Gating , Mast Cells/metabolism , Mast Cells/microbiology , Mast Cells/physiology , Phosphorylation , Protein-Tyrosine Kinases/physiology , Rats , Type C Phospholipases/metabolism
9.
Cell Microbiol ; 9(6): 1529-37, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17298393

ABSTRACT

We have used Salmonella enterica serovar Typhimurium (S. typhimurium) which are able to colonize tumours besides spleen and liver. Bacteria were equipped with constructs encoding green fluorescent protein or luciferase as reporters under control of the promoter PBAD that is inducible with L-arabinose. Reporter genes could be induced in culture but also when the bacteria resided within the mouse macrophages J774A.1. More important, strong expression of reporters by the bacteria could be detected in mice after administration of L-arabinose. This was especially pronounced in bacteria colonizing tumours. Histology demonstrated that the bacteria had accumulated in and close to necrotic areas of tumours. Bacterial gene induction was observed in both regions. PBAD is tightly controlled also in vivo because gene E of bacteriophage PhiX174 could be introduced as inducible suicide gene. The possibility to deliberately induce genes in bacterial carriers within the host should render them extremely powerful tools for tumour therapy.


Subject(s)
Arabinose/metabolism , Gene Expression Regulation, Bacterial , Neoplasms/microbiology , Neoplasms/therapy , Salmonella typhimurium/genetics , Animals , Arabinose/administration & dosage , Bacteriolysis , Cell Line, Tumor , Genes, Reporter , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , Salmonella typhimurium/metabolism , Transcription, Genetic , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...