Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Sci Adv ; 6(51)2020 12.
Article in English | MEDLINE | ID: mdl-33328224

ABSTRACT

The assortment of cellular microRNAs ("microRNAome") is a vital readout of cellular homeostasis, but the mechanisms that regulate the microRNAome are poorly understood. The microRNAome of glioblastoma is substantially down-regulated in comparison to the normal brain. Here, we find malfunction of the posttranscriptional maturation of the glioblastoma microRNAome and link it to aberrant nuclear localization of DICER, the major enzymatic complex responsible for microRNA maturation. Analysis of DICER's nuclear interactome reveals the presence of an RNA binding protein, RBM3, and of a circular RNA, circ2082, within the complex. Targeting of this complex by knockdown of circ2082 results in the restoration of cytosolic localization of DICER and widespread derepression of the microRNAome, leading to transcriptome-wide rearrangements that mitigate the tumorigenicity of glioblastoma cells in vitro and in vivo with correlation to favorable outcomes in patients with glioblastoma. These findings uncover the mechanistic foundation of microRNAome deregulation in malignant cells.


Subject(s)
Glioblastoma , MicroRNAs , Glioblastoma/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular , RNA-Binding Proteins/genetics
2.
Immunity ; 47(1): 159-170.e10, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723548

ABSTRACT

Clearance of pathogens or tumor cells by antibodies traditionally requires both Fab and Fc domains of IgG. Here, we show the Fc domain of IgG alone mediates recognition and clearance of herpes simplex virus (HSV1)-infected cells. The human natural killer (NK) cell surface is naturally coated with IgG bound by its Fc domain to the Fcγ receptor CD16a. NK cells utilize the Fc domain of bound IgG to recognize gE, an HSV1-encoded glycoprotein that also binds the Fc domain of IgG but at a site distinct from CD16a. The bridge formed by the Fc domain between the HSV1-infected cell and the NK cell results in NK cell activation and lysis of the HSV1-infected cell in the absence of HSV1-specific antibody in vitro and prevents fatal HSV1 infection in vivo. This mechanism also explains how bacterial IgG-binding proteins regulate NK cell function and may be broadly applicable to Fcγ-receptor-bearing cells.


Subject(s)
Antibodies, Viral/metabolism , Herpes Simplex/immunology , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/metabolism , Killer Cells, Natural/immunology , Simplexvirus/immunology , Animals , Antibodies, Viral/immunology , Cells, Cultured , Cytotoxicity, Immunologic , Female , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Binding , Receptor Aggregation , Receptors, IgG/metabolism , Signal Transduction , Viral Proteins/immunology
3.
Gene Ther ; 22(12): 947-59, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26196249

ABSTRACT

Oncolytic viruses (OV) have broad potential as an adjuvant for the treatment of solid tumors. The present study addresses the feasibility of clinically applicable drugs to enhance the oncolytic potential of the OV Delta24-RGD in glioblastoma. In total, 446 drugs were screened for their viral sensitizing properties in glioblastoma stem-like cells (GSCs) in vitro. Validation was done for 10 drugs to determine synergy based on the Chou Talalay assay. Mechanistic studies were undertaken to assess viability, replication efficacy, viral infection enhancement and cell death pathway induction in a selected panel of drugs. Four viral sensitizers (fluphenazine, indirubin, lofepramine and ranolazine) were demonstrated to reproducibly synergize with Delta24-RGD in multiple assays. After validation, we underscored general applicability by testing candidate drugs in a broader context of a panel of different GSCs, various solid tumor models and multiple OVs. Overall, this study identified four viral sensitizers, which synergize with Delta24-RGD and two other strains of OVs. The viral sensitizers interact with infection, replication and cell death pathways to enhance efficacy of the OV.


Subject(s)
Glioblastoma/therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Brain Neoplasms/virology , Cell Line, Tumor , Drug Evaluation, Preclinical , Fluphenazine/pharmacology , Glioblastoma/drug therapy , Glioblastoma/virology , HCT116 Cells , Humans , Indoles/pharmacology , Oncolytic Viruses/physiology , Virus Replication/drug effects
4.
Cancer Gene Ther ; 20(8): 437-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23827879

ABSTRACT

Oncolytic viral (OV) therapy has been considered as a promising treatment modality for brain tumors. Vasculostatin, the fragment of brain-specific angiogenesis inhibitor-1, shows anti-angiogenic activity against malignant gliomas. Previously, a vasculostatin-expressing oncolytic herpes simplex virus-1, Rapid Antiangiogenesis Mediated By Oncolytic virus (RAMBO), was reported to have a potent antitumor effect. Here, we investigated the therapeutic efficacy of RAMBO and cilengitide, an integrin inhibitor, combination therapy for malignant glioma. In vitro, tube formation was significantly decreased in RAMBO and cilengitide combination treatment compared with RAMBO or cilengitide monotherapy. Moreover, combination treatment induced a synergistic suppressive effect on endothelial cell migration compared with the control virus. RAMBO, combined with cilengitide, induced synergistic cytotoxicity on glioma cells. In the caspase-8 and -9 assays, the relative absorption of U87ΔEGFR cell clusters treated with cilengitide and with RAMBO was significantly higher than that of those treated with control. In addition, the activity of caspase 3/7 was significantly increased with combination therapy. In vivo, there was a significant increase in the survival of mice treated with combination therapy compared with RAMBO or cilengitide monotherapy. These results indicate that cilengitide enhanced vasculostatin-expressing OV therapy for malignant glioma and provide a rationale for designing future clinical trials combining these two agents.


Subject(s)
Angiogenic Proteins/biosynthesis , Brain Neoplasms/therapy , Brain Neoplasms/virology , Glioma/therapy , Glioma/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Snake Venoms/pharmacology , Angiogenic Proteins/genetics , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Chlorocebus aethiops , Combined Modality Therapy , Glioma/drug therapy , Glioma/pathology , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Receptors, G-Protein-Coupled , Vero Cells , Xenograft Model Antitumor Assays
5.
Gene Ther ; 20(7): 742-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23235561

ABSTRACT

Peripheral neuropathic pain is one of the most common and debilitating complications of diabetes. Several genes have been shown to be effective in reducing neuropathic pain in animal models of diabetes after transfer to the dorsal root ganglion using replication-defective herpes simplex virus (HSV)1-based vectors, yet there has never been a comparative analysis of their efficacy. We compared four different HSV1-based vectors engineered to produce one of two opioid receptor agonists (enkephalin or endomorphin), or one of two isoforms of glutamic acid decarboxylase (GAD65 or GAD67), alone and in combination, in the streptozotocin-induced diabetic rat and mouse models. Our results indicate that a single subcutaneous hindpaw inoculation of vectors expressing GAD65 or GAD67 reduced diabetes-induced mechanical allodynia to a degree that was greater than daily injections of gabapentin in rats. Diabetic mice that developed thermal hyperalgesia also responded to GAD65 or endomorphin gene delivery. The results suggest that either GAD65 or GAD67 vectors are the most effective in the treatment of diabetic pain. The vector combinations, GAD67+endomorphin, GAD67+enkephalin or endomorphin+enkephalin also produced a significant antinociceptive effect but the combination did not appear to be superior to single gene treatment. These findings provide further justification for the clinical development of antinociceptive gene therapies for the treatment of diabetic peripheral neuropathies.


Subject(s)
Diabetes Mellitus/therapy , Diabetic Neuropathies/therapy , Genetic Therapy , Simplexvirus/genetics , Animals , Diabetes Complications , Diabetes Mellitus/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/therapy , Diabetic Neuropathies/genetics , Disease Models, Animal , Ganglia, Spinal/physiopathology , Gene Transfer Techniques , Genetic Vectors , Humans , Mice , Rats
6.
Gene Ther ; 20(7): 761-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23254370

ABSTRACT

Epstein-Barr virus (EBV)-associated B-cell lymphoproliferative disease (LPD) after hematopoietic stem cell or solid organ transplantation remains a life-threatening complication. Expression of the virus-encoded gene product, EBER, has been shown to prevent apoptosis via blockade of PKR activation. As PKR is a major cellular defense against Herpes simplex virus (HSV), and oncolytic HSV-1 (oHSV) mutants have shown promising antitumor efficacy in preclinical models, we sought to determine whether EBV-LPD cells are susceptible to infection by oHSVs. We tested three primary EBV-infected lymphocyte cell cultures from neuroblastoma (NB) patients as models of naturally acquired EBV-LPD. NB12 was the most susceptible, NB122R was intermediate and NB88R2 was essentially resistant. Despite EBER expression, PKR was activated by oHSV infection. Susceptibility to oHSV correlated with the expression of the HSV receptor, nectin-1. The resistance of NB88R2 was reversed by exogenous nectin-1 expression, whereas downregulation of nectin-1 on NB12 decreased viral entry. Xenografts derived from the EBV-LPDs exhibited only mild (NB12) or no (NB88R2) response to oHSV injection, compared with a NB cell line that showed a significant response. We conclude that EBV-LPDs are relatively resistant to oHSV virotherapy, in some cases, due to low virus receptor expression but also due to intact antiviral PKR signaling.


Subject(s)
Herpesvirus 1, Human/genetics , Herpesvirus 4, Human/genetics , Lymphoproliferative Disorders/genetics , Oncolytic Viruses/genetics , Apoptosis/genetics , Cell Adhesion Molecules/metabolism , DNA, Viral/genetics , Herpesvirus 1, Human/immunology , Herpesvirus 4, Human/immunology , Humans , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/virology , Nectins , Oncolytic Virotherapy , Primary Cell Culture , Receptors, Virus/genetics
7.
Front Oncol ; 2: 19, 2012.
Article in English | MEDLINE | ID: mdl-22649779

ABSTRACT

PURPOSE: Family history is associated with gliomas, but this association has not been established for benign brain tumors. Using information from newly diagnosed primary brain tumor patients, we describe patterns of family cancer histories in patients with benign brain tumors and compare those to patients with gliomas. METHODS: Newly diagnosed primary brain tumor patients were identified as part of the Ohio Brain Tumor Study. Each patient was asked to participate in a telephone interview about personal medical history, family history of cancer, and other exposures. Information was available from 33 acoustic neuroma (65%), 78 meningioma (65%), 49 pituitary adenoma (73.1%), and 152 glioma patients (58.2%). The association between family history of cancer and each subtype was compared with gliomas using unconditional logistic regression models generating odds ratios (ORs) and 95% confidence intervals. RESULTS: There was no significant difference in family history of cancer between patients with glioma and benign subtypes. CONCLUSION: The results suggest that benign brain tumor may have an association with family history of cancer. More studies are warranted to disentangle the potential genetic and/or environmental causes for these diseases.

8.
Nat Cell Biol ; 14(2): 159-67, 2011 Dec 18.
Article in English | MEDLINE | ID: mdl-22179046

ABSTRACT

PTEN (Phosphatase and tensin homolog deleted on chromosome 10) expression in stromal fibroblasts suppresses epithelial mammary tumours, but the underlying molecular mechanisms remain unknown. Using proteomic and expression profiling, we show that Pten loss from mammary stromal fibroblasts activates an oncogenic secretome that orchestrates the transcriptional reprogramming of other cell types in the microenvironment. Downregulation of miR-320 and upregulation of one of its direct targets, ETS2 (v-ets erythroblastosis virus E26 oncogene homolog 2) are critical events in Pten-deleted stromal fibroblasts responsible for inducing this oncogenic secretome, which in turn promotes tumour angiogenesis and tumour-cell invasion. Expression of the Pten-miR-320-Ets2-regulated secretome distinguished human normal breast stroma from tumour stroma and robustly correlated with recurrence in breast cancer patients. This work reveals miR-320 as a critical component of the Pten tumour-suppressor axis that acts in stromal fibroblasts to reprogramme the tumour microenvironment and curtail tumour progression.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Tumor Microenvironment/genetics , Animals , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Female , Fibroblasts/metabolism , Humans , Kaplan-Meier Estimate , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism
9.
Cytokine Growth Factor Rev ; 21(2-3): 119-26, 2010.
Article in English | MEDLINE | ID: mdl-20226717

ABSTRACT

The systemic administration of oncolytic virus (OV) is often inefficient due to clearance of the virus by host defense mechanism and spurious targeting of non-cancer tissues through the bloodstream. Cell mediated OV delivery could hide the virus from host defenses and direct them toward tumors: Mesenchymal and neural stem cells have been described to possess tumor-homing ability as well as the capacity to deliver OVs. In this review, we will focus on approaches where OV and carrier cells are utilized for cancer therapy. Effective cellular internalization and replication of OVs need to occur both in cancer and carrier cells. We thus will discuss the current challenges faced by the use of OV delivery via carrier cells.


Subject(s)
Genetic Vectors , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Stem Cells/virology , Adenoviridae/genetics , Adenoviridae/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/virology , Neurons/cytology , Neurons/virology , Oncolytic Viruses/genetics , Stem Cells/cytology , Transduction, Genetic , Virus Replication
10.
Cell Death Differ ; 17(2): 221-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19521422

ABSTRACT

Recent data draw close parallels between cancer, including glial brain tumors, and the biology of stem and progenitor cells. At the same time, it has become clear that one of the major roles that microRNAs play is in the regulation of stem cell biology, differentiation, and cell 'identity'. For example, microRNAs have been increasingly implicated in the regulation of neural differentiation. Interestingly, initial studies in the incurable brain tumor glioblastoma multiforme strongly suggest that microRNAs involved in neural development play a role in this disease. This encourages the idea that certain miRs allow continued tumor growth through the suppression of differentiation and the maintenance of the stem cell-like properties of tumor cells. These concepts will be explored in this article.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , MicroRNAs/genetics , Stem Cells/pathology , Stem Cells/physiology , Brain Neoplasms/pathology , Cell Differentiation/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans
11.
Oncogene ; 27(30): 4249-54, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18345032

ABSTRACT

Oncolytic herpes simplex viruses (HSVs), in clinical trials for the treatment of malignant gliomas, are assumed to be selective for tumor cells because their replication is strongly attenuated in quiescent cells, but not in cycling cells. Oncolytic selectivity is thought to occur because mutations in viral ICP6 (encoding a viral ribonucleotide reductase function) and/or gamma34.5 function are respectively complemented by mammalian ribonucleotide reductase and GADD34, whose genes are expressed in cycling cells. However, it is estimated that only 5-15% of malignant glioma cells are in mitosis at any one time. Therefore, effective replication of HSV oncolytic viruses might be limited to a subpopulation of tumor cells, since at any one time the majority of tumor cells would not be cycling. However, we report that an HSV with defective ICP6 function replicates in quiescent cultured murine embryonic fibroblasts obtained from mice with homozygous p16 deletions. Furthermore, intracranial inoculation of this virus into the brains of p16-/- mice provides evidence of viral replication that does not occur when the virus is injected into the brains of wild-type mice. These approaches provide in vitro and in vivo evidence that ICP6-negative HSVs are 'molecularly targeted,' because they replicate in quiescent tumor cells carrying specific oncogene deletions, independent of cell cycle status.


Subject(s)
Cell Cycle/physiology , Genes, p16 , Oncolytic Viruses/genetics , Simplexvirus/genetics , Simplexvirus/physiology , Viral Proteins/genetics , Virus Replication/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cell Cycle/genetics , Cells, Cultured , Glioma/genetics , Glioma/therapy , Homozygote , Humans , Mice , Mice, Knockout , Mutation/physiology , Oncolytic Virotherapy
12.
Gene Ther ; 14(22): 1555-63, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17984995

ABSTRACT

This is the second part of a review summarizing progress and prospects in gene therapy clinical research. Twenty key diseases/strategies are succinctly described and commented on by leaders in the field. This part includes clinical trials for skin diseases, neurological disorders, HIV/AIDS, ornithine transcarbamylase deficiency, alpha(1)-antitrypsin deficiency, haemophilia and cancer.


Subject(s)
Genetic Therapy/trends , Clinical Trials as Topic , Gene Transfer Techniques/adverse effects , Gene Transfer Techniques/trends , Genetic Therapy/methods , Genetic Vectors , Humans , Neoplasms/therapy , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/trends
13.
Gene Ther ; 13(24): 1731-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16871231

ABSTRACT

An increasing number of oncolytic viruses have been developed and studied for cancer therapy. In response to needs for non-invasive monitoring and imaging of oncolytic virotherapy, several different approaches, including a positron emission tomography-based method, a method using secreted marker peptides, and optical imaging-based methods, have been reported. Among these modalities, we utilized the luciferase-based bioluminescent assay/imaging systems to determine the kinetics and dynamics of a productive viral infection. The replication cycle of herpes simplex virus type 1 (HSV-1) is punctuated by a temporal cascade of three classes of viral genes: immediate-early (IE), early (E) and late (L) genes. U(L)39- and gamma(1)34.5-deleted, replication-conditional HSV-1 mutants that express firefly luciferase under the control of the IE4/5 or strict-late gC promoters were generated. These oncolytic viruses were examined in cultured cells and a mouse tumor model. IE promoter- and strict-late promoter-mediated luciferase expression was confirmed to indicate viral infection and replication, respectively. Incorporation of a strict-late promoter-driven luciferase cassette into oncolytic HSV-1 vectors would be useful for assessing tumor oncolysis in preclinical tumor treatment studies.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Herpesvirus 1, Human/genetics , Neoplasms, Experimental/therapy , Oncolytic Virotherapy/methods , Promoter Regions, Genetic , Animals , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Gene Expression/drug effects , Genetic Engineering , Genetic Vectors/administration & dosage , Herpes Simplex/genetics , Humans , Luciferases/genetics , Luminescent Proteins/genetics , Mice , Mice, Nude , Models, Animal , Neoplasms, Experimental/virology , Neuroglia/enzymology , Phosphonoacetic Acid/pharmacology , Purines/pharmacology , Roscovitine , Vero Cells , Virus Replication
14.
Gene Ther ; 13(8): 705-14, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16421599

ABSTRACT

Replication-conditional herpes simplex virus (HSV)-based vectors have great potential in the treatment of various types of cancers including brain tumors. HSV mutants lacking the U(L)39 gene and both copies of the gamma(1)34.5 gene (e.g. MGH1, G207) have been demonstrated to possess oncolytic effects as well as potent anticancer vaccination effects without compromising safety. Such mutants thus provide optimal templates to produce novel oncolytic HSV vectors for cancer gene therapy applications. In order to accomplish quick and efficient construction of oncolytic HSV vectors, a novel BAC-based method designated as 'HSVQuik system' was developed. This system sequentially utilizes two different site-specific recombination systems to introduce virtually any transgene cassettes of interest into the deleted U(L)39 locus (Flp-FRT in Escherichia coli) and to release the vector genome sequence from the procaryotic plasmid backbone (Cre-loxP in Vero cells). Taking advantage of the HSVQuik system, we constructed three oncolytic HSV vectors that express mouse IL4, CD40 ligand and 6CK, respectively. In vivo therapeutic experiments using two luciferase-labeled syngeneic mouse brain tumor models revealed that expression of these immunomodulators significantly enhanced antitumor efficacy of oncolytic HSV. The HSVQuik system, together with luciferase-labeled tumor models, should expedite the process of generating and evaluating oncolytic HSV vectors for cancer gene therapy applications.


Subject(s)
Cancer Vaccines/genetics , Genetic Engineering/methods , Neoplasms/therapy , Oncolytic Virotherapy , Simplexvirus/genetics , Animals , CD40 Ligand/genetics , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Chemokine CCL21 , Chemokines, CC/genetics , Chlorocebus aethiops , Female , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Interleukin-4/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Neoplasm Transplantation , Neoplasms/genetics , Oncolytic Viruses/genetics , Plasmids , Vero Cells , Virus Replication
15.
Cancer Gene Ther ; 13(3): 225-33, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16138122

ABSTRACT

Gene therapy is a potentially useful approach in the treatment of human brain tumors, which are notoriously refractory to conventional approaches. Most human clinical trials to date have been unsuccessful in terms of improving patient outcome. Recent improvements in viral vectors, the development of stem cell technology, and increased understanding of the mechanism of action of therapeutic transgenes provide hope that the next generation of gene therapeutics may show increased efficacy in treatment of this devastating disease.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy , Gene Transfer Techniques , Genetic Vectors , Humans , Interferons/therapeutic use , Interleukins/therapeutic use , Transgenes/physiology
16.
Neurology ; 64(11): 1838-45, 2005 Jun 14.
Article in English | MEDLINE | ID: mdl-15955931

ABSTRACT

The neurofibromatoses are a diverse group of genetic conditions that share a predisposition to the development of tumors of the nerve sheath. Schwannomatosis is a recently recognized third major form of neurofibromatosis (NF) that causes multiple schwannomas without vestibular tumors diagnostic of NF2. Patients with schwannomatosis represent 2.4 to 5% of all patients requiring schwannoma resection and approximately one third of patients with schwannomatosis have anatomically localized disease with tumors limited to a single limb or segment of spine. Epidemiologic studies suggest that schwannomatosis is as common as NF2, but that familial occurrence is inexplicably rare. Patients with schwannomatosis overwhelmingly present with pain, and pain remains the primary clinical problem and indication for surgery. Diagnostic criteria for schwannomatosis are needed for both clinicians and researchers, but final diagnostic certainly will await the identification of the schwannomatosis locus itself.


Subject(s)
Neurilemmoma/diagnosis , Neurofibromatosis 2/diagnosis , Diagnosis, Differential , Humans , Neurilemmoma/pathology , Neurilemmoma/physiopathology , Neuroma, Acoustic/diagnosis
17.
Gene Ther ; 11(15): 1195-204, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15164098

ABSTRACT

The expression of genes from genomic loci can be relatively complex, utilizing exonic, intronic and flanking sequences to regulate tissue and developmental specificity. Infectious bacterial artificial chromosomes (iBACs) have been shown to deliver and express large genomic loci (up to 135 kb) into primary cells for functional analyses. The delivery of large genomic DNA inserts allows the expression of complex loci and of multiple splice variants. Herein, we demonstrate for the first time that an iBAC will deliver and correctly express in human glioma cells the entire CDKN2A/CDKN2B genomic region, which encodes for at least three important cell-cycle regulatory proteins (p16(INK4a), p14(ARF) and p15(INK4b)). Two of these proteins are expressed from overlapping genes, utilizing alternative splicing and promoter usage. The delivered locus expresses each gene at physiological levels and cellular responses (apoptosis versus growth arrest) occur dependent on cellular p53 status, as expected. The work further demonstrates the potential of the iBAC system for the delivery of genomic loci whose expression is mediated by complex splicing and promoter usage both for gene therapy applications and functional genomics studies.


Subject(s)
Chromosomes, Artificial, Bacterial , DNA/administration & dosage , Genes, p16 , Genetic Therapy/methods , Glioma/therapy , Apoptosis , Cell Division , Cell Line, Tumor , Gene Expression , Gene Expression Profiling , Genomics , Glioma/virology , Herpes Simplex/genetics , Herpesvirus 1, Human , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction
18.
Gene Ther ; 10(15): 1225-33, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12858187

ABSTRACT

The owl monkey (Aotus trivirgatus) has served as the standard non-human primate model of herpes simplex virus-1 (HSV-1) infection because it is highly susceptible to HSV-1 encephalitis. Owl monkeys, however, are expensive, difficult to obtain, and difficult to maintain in captivity, thus greatly hampering the efficiency of preclinical gene therapy trials for brain tumors using HSV-1-based vectors. We have therefore compared the susceptibility of the common marmoset (Callithrix jacchus) with the owl monkey in a model of intracerebral inoculation of wildtype HSV-1 F-strain at increasing titers. The common marmosets consistently succumbed earlier to viral encephalitis than the owl monkeys. The histological evaluation of the common marmoset revealed extensive HSV-1 infection with a concomitant yet less marked inflammatory response compared to the owl monkeys. PCR for HSV-1 demonstrated a similar extra-CNS shedding route in both experimental models. Our findings show that the common marmoset is at least as susceptible to intracerebral HSV-infection as the owl monkey and that it can therefore serve as a valid and reliable experimental model for the important preclinical safety tests of HSV-based therapeutic viral vector constructs in the brain.


Subject(s)
Callithrix/virology , Disease Models, Animal , Encephalitis, Herpes Simplex/pathology , Genetic Vectors/toxicity , Herpesvirus 1, Human/pathogenicity , Animals , Brain/virology , Disease Susceptibility , Encephalitis, Herpes Simplex/etiology , Female , Herpesvirus 1, Human/genetics , Kidney/virology , Liver/virology , Male , Polymerase Chain Reaction/methods , Survival Rate
20.
Gene Ther ; 10(11): 983-90, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12756419

ABSTRACT

Several clinical trials have or are being performed testing the safety and efficacy of different strains of oncolytic viruses (OV) for malignant cancers. OVs represent either naturally occurring or genetically engineered strains of viruses that exhibit relatively selective replication in tumor cells. Several types of OV have been derived from herpes simplex virus 1 (HSV1). Tumor oncolysis depends on the processes of initial OV infection of tumor, followed by subsequent propagation of OV within the tumor itself. The role of the immune responses in these processes has not been extensively studied. On the contrary, effects of the immune response on the processes of wild-type HSV1 infection and propagation in the central nervous system have been studied and described in detail. The first line of defense against a wild-type HSV1 infection in both naive and immunized individuals is provided by innate humoral (complement, cytokines, chemokines) and cellular (macrophages, neutrophils, NK cells, gammadelta T cells, and interferon-producing cells) responses. These orchestrate the lysis of virions and virus-infected cells as well as provide a link to effective adaptive immunity. The role of innate defenses in curtailing the oncolytic effect of genetically engineered HSV has only recently been studied, but several of the same host responses appear to be operative in limiting anticancer effects by the replicating virus. The importance of this knowledge lies in finding avenues to modulate such initial innate responses, in order to allow for increased oncolysis of tumors while minimizing host toxicity.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Neoplasms/therapy , Viruses/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/virology , Central Nervous System/immunology , Central Nervous System/virology , Complement Inactivator Proteins/therapeutic use , Complement System Proteins/immunology , Cyclophosphamide/therapeutic use , Herpes Simplex/complications , Humans , Immunosuppressive Agents/therapeutic use , Neoplasms/immunology , Neoplasms/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...