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.
BMC Cancer ; 17(1): 555, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28830374

ABSTRACT

BACKGROUND: The Empty Spiracles Homeobox (EMX-) 2 gene has been associated with regulation of growth and differentiation in neuronal development. While recent studies provide evidence that EMX2 regulates tumorigenesis of various solid tumors, its role in colorectal cancer remains unknown. We aimed to assess the prognostic significance of EMX2 expression in stage III colorectal adenocarcinoma. METHODS: Expression levels of EMX2 in human colorectal cancer and adjacent mucosa were assessed by qRT-PCR technology, and results were correlated with clinical and survival data. siRNA-mediated knockdown and adenoviral delivery-mediated overexpression of EMX2 were performed in order to investigate its effects on the migration of colorectal cancer cells in vitro. RESULTS: Compared to corresponding healthy mucosa, colorectal tumor samples had decreased EMX2 expression levels. Furthermore, EMX2 down-regulation in colorectal cancer tissue was associated with distant metastasis (M1) and impaired overall patient survival. In vitro knockdown of EMX2 resulted in increased tumor cell migration. Conversely, overexpression of EMX2 led to an inhibition of tumor cell migration. CONCLUSIONS: EMX2 is frequently down-regulated in human colorectal cancer, and down-regulation of EMX2 is a prognostic marker for disease-free and overall survival. EMX2 might thus represent a promising therapeutic target in colorectal cancer.


Subject(s)
Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression , Homeodomain Proteins/genetics , Liver Neoplasms/diagnosis , Liver Neoplasms/secondary , Transcription Factors/genetics , Adenoviridae/genetics , Cell Line, Tumor , Cell Movement/genetics , Cohort Studies , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Female , Follow-Up Studies , Gene Transfer Techniques , Genetic Vectors/genetics , Homeodomain Proteins/metabolism , Humans , Liver Neoplasms/mortality , Liver Neoplasms/therapy , Male , Neoplasm Staging , Prognosis , Transcription Factors/metabolism , Transduction, Genetic
2.
MAbs ; 6(1): 130-42, 2014.
Article in English | MEDLINE | ID: mdl-24256717

ABSTRACT

The development of efficient strategies for generating fully human monoclonal antibodies with unique functional properties that are exploitable for tailored therapeutic interventions remains a major challenge in the antibody technology field. Here, we present a methodology for recovering such antibodies from antigen-encountered human B cell repertoires. As the source for variable antibody genes, we cloned immunoglobulin G (IgG)-derived B cell repertoires from lymph nodes of 20 individuals undergoing surgery for head and neck cancer. Sequence analysis of unselected "LYmph Node Derived Antibody Libraries" (LYNDAL) revealed a naturally occurring distribution pattern of rearranged antibody sequences, representing all known variable gene families and most functional germline sequences. To demonstrate the feasibility for selecting antibodies with therapeutic potential from these repertoires, seven LYNDAL from donors with high serum titers against herpes simplex virus (HSV) were panned on recombinant glycoprotein B of HSV-1. Screening for specific binders delivered 34 single-chain variable fragments (scFvs) with unique sequences. Sequence analysis revealed extensive somatic hypermutation of enriched clones as a result of affinity maturation. Binding of scFvs to common glycoprotein B variants from HSV-1 and HSV-2 strains was highly specific, and the majority of analyzed antibody fragments bound to the target antigen with nanomolar affinity. From eight scFvs with HSV-neutralizing capacity in vitro,the most potent antibody neutralized 50% HSV-2 at 4.5 nM as a dimeric (scFv)2. We anticipate our approach to be useful for recovering fully human antibodies with therapeutic potential.


Subject(s)
Antibodies, Viral , Cloning, Molecular , Gene Library , Herpesvirus 1, Human/immunology , Herpesvirus 2, Human/immunology , Immunoglobulin G , Immunoglobulin Variable Region , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Female , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Male , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
3.
Virus Genes ; 41(1): 81-5, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20480219

ABSTRACT

A herpesvirus of smelt (Osmerus eperlanus) was identified by thin section electron microscopy. Degenerated cells of skin lesions located on the back fin of smelt showed either intranucleic- or cytoplasmic herpesvirus-specific structures. In the nuclei "naked" virus capsids with a diameter of about 100 nm were observed. The diameter of the complete virion including its unilaterally extended envelope ranged from 200 to 350 nm. Remarkably, in complete virions the electron-opaque tegument is completely filling the region between nucleocapsid and envelope and as another unique feature the virion shows a "comet-shape" due to a long unilateral extension of its envelope. This kind of shape had been not reported for any of herpesviruses known so far. Consequently this virus was termed herpesvirus of Osmerus eperlanus (HVOE1) or Comet herpesvirus of smelt. Due to the long time storage at the nonstandard temperature of smelt virus the biological and genomic analysis of the HVOE1 was hampered. All attempts to study host range of HVOE1 failed as no virus replication was observed, indicating that infectivity was lost or the suitable cell culture was missing. The genomic DNA of HVOE1 was analyzed by DNA restriction endonucleases.


Subject(s)
Fish Diseases/virology , Herpesviridae Infections/veterinary , Herpesviridae/ultrastructure , Osmeriformes/virology , Animals , Genome, Viral , Herpesviridae/genetics , Herpesviridae Infections/virology , Microscopy, Electron , Virion/ultrastructure
4.
J Pharm Pharmacol ; 60(11): 1545-50, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18957177

ABSTRACT

The effect of anise oil, dwarf-pine oil and chamomile oil against different thymidine-kinase-positive (aciclovir-sensitive) and thymidine-kinase-negative (aciclovir-resistant) herpes simplex virus type 1 (HSV-1) strains was examined. Clinical HSV-1 isolates containing frameshift mutations in the thymidine kinase (TK) gene, an insertion or a deletion, yield a non-functional thymidine kinase enzyme resulting in phenotypical resistance against aciclovir. The inhibitory activity of three different essential oils against herpes simplex virus isolates was tested in-vitro using a plaque reduction assay. All essential oils exhibited high levels of antiviral activity against aciclovir-sensitive HSV strain KOS and aciclovir-resistant clinical HSV isolates as well as aciclovir-resistant strain Angelotti. At maximum noncytotoxic concentrations of the plant oils, plaque formation was significantly reduced by 96.6-99.9%, when herpesviruses were preincubated with drugs before attachment to host cells. No significant effect on viral infectivity could be achieved by adding these compounds during the replication phase. These results indicate that anise oil, dwarf-pine oil and chamomile oil affected the virus by interrupting adsorption of herpesviruses and in a different manner than aciclovir, which is effective after attachment inside the infected cells. Thus the investigated essential oils are capable of exerting a direct effect on HSV and might be useful in the treatment of drug-resistant viruses. Chamomile oil did not reveal any irritating potential on hen's egg chorioallantoic membrane, demonstrated the highest selectivity index among the oils tested and was highly active against clinically relevant aciclovir-resistant HSV-1 strains.


Subject(s)
Antiviral Agents/administration & dosage , Herpesvirus 1, Human/drug effects , Oils, Volatile/administration & dosage , Acyclovir/pharmacology , Animals , Antiviral Agents/isolation & purification , Antiviral Agents/toxicity , Cell Line , Chickens , Chlorocebus aethiops , Chorioallantoic Membrane/drug effects , Dogs , Dose-Response Relationship, Drug , Drug Resistance, Viral , Herpesvirus 1, Human/genetics , Illicium/chemistry , Matricaria/chemistry , Oils, Volatile/isolation & purification , Oils, Volatile/toxicity , Pinus/chemistry , Thymidine Kinase/genetics , Vero Cells , Viral Plaque Assay , Virus Replication/drug effects
5.
Virus Genes ; 30(2): 157-80, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15744574

ABSTRACT

In the last decades a significant number of so far unknown or underestimated pathogens have emerged as fundamental health hazards of the human population despite intensive research and exceptional efforts of modern medicine to embank and eradicate infectious diseases. Almost all incidents caused by such emerging pathogens could be ascribed to agents that are zoonotic or expanded their host range and crossed species barriers. Many different factors influence the status of a pathogen to remain unnoticed or evolves into a worldwide threat. The ability of an infectious agent to adapt to changing environmental conditions and variations in human behavior, population development, nutrition, education, social, and health status are relevant factors affecting the correlation between pathogen and host. Hantaviruses belong to the emerging pathogens having gained more and more attention in the last decades. These viruses are members of the family Bunyaviridae and are grouped into a separate genus known as Hantavirus. The serotypes Hantaan (HTN), Seoul (SEO), Puumala (PUU), and Dobrava (DOB) virus predominantly cause hemorrhagic fever with renal syndrome (HFRS), a disease characterized by renal failure, hemorrhages, and shock. In the recent past, many hantavirus isolates have been identified and classified in hitherto unaffected geographic regions in the New World (North, Middle, and South America) with characteristic features affecting the lungs of infected individuals and causing an acute pulmonary syndrome. Hantavirus outbreaks in the United States of America at the beginning of the 10th decade of the last century fundamentally changed our knowledge about the appearance of the hantavirus specific clinical picture, mortality, origin, and transmission route in human beings. The hantavirus pulmonary syndrome (HPS) was first recognized in 1993 in the Four Corners Region of the United States and had a lethality of more than 50%. Although the causative virus was first termed in connection with the geographic name of its outbreak region the analysis of the individual viruses indicate that the causing virus of HPS was a genetically distinct hantavirus and consequently termed as Sin Nombre virus. Hantaviruses are distributed worldwide and are assumed to share a long time period of co-evolution with specific rodent species as their natural reservoir. The degree of relatedness between virus serotypes normally coincides with the relatedness between their respective hosts. There are no known diseases that are associated with hantavirus infections in rodents underlining the amicable relationship between virus and host developed by mutual interaction in hundreds of thousands of years. Although rodents are the major reservoir, antibodies against hantaviruses are also present in domestic and wild animals like cats, dogs, pigs, cattle, and deer. Domestic animals and rodents live jointly in a similar habitat. Therefore the transmission of hantaviruses from rodents to domestic animals seems to be possible, if the target organs, tissues, and cell parenchyma of the co-habitat domestic animals possess adequate virus receptors and are suitable for hantavirus entry and replication. The most likely incidental infection of species other than rodents as for example humans turns hantaviruses from harmless to life-threatening pathogenic agents focusing the attention on this virus group, their ecology and evolution in order to prevent the human population from a serious health risk. Much more studies on the influence of non-natural hosts on the ecology of hantaviruses are needed to understand the directions that the hantavirus evolution could pursue. At least, domestic animals that share their environmental habitat with rodents and humans particularly in areas known as high endemic hantavirus regions have to be copiously screened. Each transfer of hantaviruses from their original natural hosts to other often incidental hosts is accompanied by a change of ecology, a change of environment, a modulation of numerous factors probably influencing the pathogenicity and virulence of the virus. The new environment exerts a modified evolutionary pressure on the virus forcing it to adapt and probably to adopt a form that is much more dangerous for other host species compared to the original one.


Subject(s)
Hantavirus Infections/transmission , Animals , Disease Outbreaks , Disease Reservoirs , Ecosystem , Orthohantavirus/classification , Orthohantavirus/isolation & purification , Orthohantavirus/pathogenicity , Hantavirus Infections/epidemiology , Hantavirus Infections/etiology , Hantavirus Infections/prevention & control , Hantavirus Pulmonary Syndrome/transmission , Hantavirus Pulmonary Syndrome/virology , Humans , Zoonoses/transmission , Zoonoses/virology
6.
Virus Genes ; 30(2): 279-96, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15744583

ABSTRACT

The objective of this study was to investigate the molecular mechanisms of neurobiological processes involved in the degeneration of the central nervous system. The bovine spongiform encephalopathy (BSE) was used as experimental model system for investigation of transmissible spongiform encephalopathy (TSE). The experimental strategy was to evaluate the possibility for protection of bovine PrP(C) transgenic mice against a bovine PrP(Sc) infection by DNA vaccination using the complete or partial cDNA sequences of the bovine prion protein. Three recombinant plasmids pCR3.1-EX-PrP-BSE-C20 (C20), pCR3.1-EX-PrP-BSE-90-235-C4 (C4), and pCR3.1-EX-PrP-BSE-106-131-C14 (C14) were constructed. These mammalian expression vectors harbor complete (C20) or partial (C4 and C14) cDNA sequences of the Bos taurus PrP(C) (BTPrP(C)) encoding for amino acid residues 1-264 (C20), 90-235 (C4), and 106-131 (C14) of the BTPrP(C). Transgenic mice harboring and expressing BTPrP(C) were generated using the donor strain C57/CBA, receptor strain Swiss mouse, and recombinant plasmid MoPrPXho-boPrP. Crossing of positive transgenic mice to bovine PrP and negative to murine PrP with 129/OLA (murine PrP-/-) and C57BL6x129/OLA (murine PrP+/-) mice was carried out to amplify the colony of transgenic mice termed bovine PrP(C) transgenic Swiss mice (BTPrP-TgM). The capabilities of C20, C4, and C14 to express the corresponding cDNA sequence of BTPrP(C) in vitro and in vivo were confirmed prior to DNA vaccination of the BTPrP-TgM using NIH 3T3 cells and BALB/c mice, respectively. In order to prove the capability of the constructed expression vectors to protect BTPrP-TgM in vivo against a BSE infection 80 female BTPrP-TgM were vaccinated intramuscularly and subcutaneously with DNA of the plasmids C20, C4, C14, and parental vector pCR3.1 (100 microg DNA corresponding to about 26-30 pmol DNA/animal and application) in four groups (each consists of 20 animals). DNA vaccination was followed by three additional boosters. The vaccinated animals (15 animals of each group) were challenged twice per oral with homogenates of brain material obtained from BSE cattle containing the infectious PrP(Sc) (100 microl/animal which corresponds to 15 mg of a 15% brain homogenate). The first and second challenge experiments were performed 76-83 and 181 days post DNA vaccination, respectively. A part of the vaccinated animals (3-5 animals of each group) that served as internal negative control were mock infected using the brain homogenate of healthy cattle or Phosphate saline buffer (PBS). A variety of symptoms and clinical pictures were observed during the monitoring of DNA vaccinated animals. However, the observed diseases seem to be similar in all experimental animal groups. After an observation period of 14 months post the second challenge experiment the remaining animals (some animals died or were sacrificed when moribund during the study) were sacrificed after expiration of the experimental schedule. The right hemisphere of the brain and a half of the spleen tissue of the individual animals were used for detection of PrP(Sc) by Western blot analysis. The misfolded bovine PrP(Sc) was not detected in the brain or spleen tissues of those animals that were vaccinated with DNA of C20, which was able to express the complete bovine PrP(C) protein in vitro and in vivo. In contrast, the bovine PrP(Sc) was detected in the brain or spleen tissues of animals that were DNA vaccinated with DNA of the parental vector pCR3.1, with DNA of C4, or with DNA of C14. The results of these studies underline that the constructed expression vector C20 possesses the protective capacity to inhibit the formation of misfolded bovine PrP(Sc) in BTPrP-TgM under the conditions used. A delay of occurrence of TSE-specific symptoms in the majority of the vaccinated animals seems to be due to the prolonged incubation time of BSE infection.


Subject(s)
PrPC Proteins/genetics , PrPC Proteins/immunology , PrPSc Proteins/pathogenicity , Prion Diseases/prevention & control , Vaccines, DNA/genetics , Vaccines, DNA/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Cattle , DNA, Complementary/genetics , Female , Gene Expression , Genetic Vectors , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , NIH 3T3 Cells , Plasmids/genetics , Prion Diseases/genetics , Prion Diseases/immunology
7.
Neurosci Lett ; 374(3): 197-202, 2005 Feb 21.
Article in English | MEDLINE | ID: mdl-15663962

ABSTRACT

Mortality and morbidity rates remain high among patients with herpes simplex virus encephalitis (HSVE). Chemokine-mediated recruitment and activation of leukocytes to focal areas of viral CNS infection are crucial steps in antiviral response and clearance. However, the inflammatory reaction and cellular antiviral response may enhance collateral damage to neurons and account for chronic progressive brain damage. We identified a specific mRNA expression of the interferon-gamma-inducible chemokines (CXCL9, CXCL10 and CXCL11), and RANTES (CCL5) in the acute course and long-term of experimental HSVE. This pattern was substantially altered by anti-viral and anti-inflammatory treatment. Our findings indicate a pivotal role of these chemokines in the immunopathogenesis of HSVE.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Chemokines/metabolism , Encephalitis, Herpes Simplex/metabolism , Gene Expression Regulation/drug effects , Acyclovir/therapeutic use , Animals , Chemokines/genetics , Disease Models, Animal , Drug Interactions , Drug Therapy, Combination , Encephalitis, Herpes Simplex/drug therapy , Female , Methylprednisolone/therapeutic use , Mice , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Viral Load/methods
8.
Neurosci Lett ; 368(3): 274-8, 2004 Sep 30.
Article in English | MEDLINE | ID: mdl-15364410

ABSTRACT

Herpes simplex virus encephalitis (HSVE) causes elevated morbidity and mortality despite antiviral treatment. Virus-independent mechanisms may perpetuate brain damage. Matrix metalloproteinases (MMPs) target extracellular matrix components. This study describes the protein and mRNA expression of MMP2 and MMP9 in experimental HSVE in the short and long term. Ten SJL-NBOM mice were infected with neurovirulent HSV-1 and compared with nine controls. The presence of MMP2 and MMP9 in brain tissue was analyzed with sodium-dodecyl-sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) gelatin zymography and mRNA expression of MMP2 and MMP9 with quantitative real-time PCR at days 7, 21 and 180 post-inoculation. Infected animals had a significantly elevated gelatinolytic activity of MMP2 at all time points, and of MMP9 at days 21 and 180. Increased presence of MMP2 and MMP9 in chronic HSVE may contribute to ongoing damage. Inhibition of MMP2 and MMP9 might be a suitable target for therapeutic intervention.


Subject(s)
Encephalitis, Herpes Simplex/enzymology , Herpesvirus 1, Human/enzymology , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Animals , Brain/enzymology , Brain/virology , Disease Models, Animal , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/pathology , Female , Herpesvirus 1, Human/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Mice , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Time Factors
9.
Neurosci Lett ; 318(1): 21-4, 2002 Jan 18.
Article in English | MEDLINE | ID: mdl-11786216

ABSTRACT

In the brain tissue of 36 mice infected with herpes simplex virus type 1, strain F, we determined the expression of inducible nitric oxide synthase (iNOS) with semiquantitative reverse transcription polymerase chain reaction. The viral burden was quantitated by polymerase chain reaction. Nitric oxide, induced by iNOS, may contribute to neuronal cell damage following virus infection. As the experimental therapeutic strategy in herpes simplex virus encephalitis (HSVE), we used: N-nitro-L-arginin (L-NA), a selective inhibitor of iNOS; and combination therapies of either methylprednisolone/acyclovir or L-NA/acyclovir. The viral burden peaked in acute disease, and then returned to a low baseline value, except in untreated controls. The expression of iNOS mRNA was suppressed by L-NA and by acyclovir/corticosteroids. INOS inhibition may provide an additional therapeutic strategy targeted specifically to suppress iNOS expression as a potential secondary mechanism of tissue damage in acute and chronic HSVE.


Subject(s)
Brain/enzymology , Encephalitis, Herpes Simplex/metabolism , Herpesvirus 1, Human , Nitric Oxide Synthase/genetics , Acyclovir/pharmacology , Animals , Antiviral Agents/pharmacology , Brain/virology , Drug Therapy, Combination , Encephalitis, Herpes Simplex/drug therapy , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic , Methylprednisolone/pharmacology , Mice , Mice, Inbred Strains , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Nitroarginine/pharmacology , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL