Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
J Transl Genet Genom ; 5(4): 423-442, 2021.
Article in English | MEDLINE | ID: mdl-35342877

ABSTRACT

Aim: To molecularly characterize the tumor microenvironment and evaluate immunologic parameters in canine glioma patients before and after treatment with oncolytic human IL-12-expressing herpes simplex virus (M032) and in treatment naïve canine gliomas. Methods: We assessed pet dogs with sporadically occurring gliomas enrolled in Stage 1 of a veterinary clinical trial that was designed to establish the safety of intratumoral oncoviral therapy with M032, a genetically modified oncolytic herpes simplex virus. Specimens from dogs in the trial and dogs not enrolled in the trial were evaluated with immunohistochemistry, NanoString, Luminex cytokine profiling, and multi-parameter flow cytometry. Results: Treatment-naive canine glioma microenvironment had enrichment of Iba1 positive macrophages and minimal numbers of T and B cells, consistent with previous studies identifying these tumors as immunologically "cold". NanoString mRNA profiling revealed enrichment for tumor intrinsic pathways consistent with suppression of tumor-specific immunity and support of tumor progression. Oncolytic viral treatment induced an intratumoral mRNA transcription signature of tumor-specific immune responses in 83% (5/6) of canine glioma patients. Changes included mRNA signatures corresponding with interferon signaling, lymphoid and myeloid cell activation, recruitment, and T and B cell immunity. Multiplexed protein analysis identified a subset of oligodendroglioma subjects with increased concentrations of IL-2, IL-7, IL-6, IL-10, IL-15, TNFα, GM-CSF between 14 and 28 days after treatment, with evidence of CD4+ T cell activation and modulation of IL-4 and IFNγ production in CD4+ and CD8+ T cells isolated from peripheral blood. Conclusion: These findings indicate that M032 modulates the tumor-immune microenvironment in the canine glioma model.

2.
Oncogene ; 36(25): 3562-3575, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28166199

ABSTRACT

Dysregulation of the NF-κB transcription factor occurs in many cancer types. Krüppel-like family of transcription factors (KLFs) regulate the expression of genes involved in cell proliferation, differentiation and survival. Here, we report a new mechanism of NF-κB activation in glioblastoma through depletion of the KLF6 tumor suppressor. We show that KLF6 transactivates multiple genes negatively controlling the NF-κB pathway and consequently reduces NF-κB nuclear localization and downregulates NF-κB targets. Reconstitution of KLF6 attenuates their malignant phenotype and induces neural-like differentiation and senescence, consistent with NF-κB pathway inhibition. KLF6 is heterozygously deleted in 74.5% of the analyzed glioblastomas and predicts unfavorable patient prognosis suggesting that haploinsufficiency is a clinically relevant means of evading KLF6-dependent regulation of NF-κB. Together, our study identifies a new mechanism by which KLF6 regulates NF-κB signaling, and how this mechanism is circumvented in glioblastoma through KLF6 loss.


Subject(s)
Gene Deletion , Glioblastoma/genetics , Glioblastoma/metabolism , Haploinsufficiency , Kruppel-Like Transcription Factors/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/metabolism , Male , NF-kappa B/genetics , Proto-Oncogene Proteins/metabolism , Transcriptional Activation
4.
Gene Ther ; 22(4): 348-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25427614

ABSTRACT

Pathophysiological hypoxia, which fosters the glioma stem-like cell (GSC) phenotype, is present in high-grade gliomas and has been linked to tumor development, invasiveness and resistance to chemotherapy and radiation. Oncolytic virotherapy with engineered herpes simplex virus-1 (HSV-1) is a promising therapy for glioblastoma; however, the efficacy of γ(1)34.5-deleted HSVs, which have been used in clinical trials, was diminished in hypoxia. We investigated the ability of a chimeric human cytolomegalovirus (HCMV)/HSV-1 virus, which expresses the human CMV protein kinase R evasion gene IRS1 and is in preparation for clinical trials, to infect and kill adult and pediatric patient-derived glioblastoma xenografts in hypoxia and normoxia. Infectivity, cytotoxicity and viral recovery were significantly greater with the chimeric virus compared with the γ(1)34.5-deleted virus, regardless of oxygen tension. The chimeric virus infected and killed CD133+ GSCs similarly to wild-type HSV-1. Increased activation of mitogen-activated protein kinase p38 and its substrate heat-shock protein 27 (Hsp27) was seen after viral infection in normoxia compared with hypoxia. Hsp27 knockdown or p38 inhibition reduced virus recovery, indicating that the p38 pathway has a role in the reduced efficacy of the γ(1)34.5-deleted virus in hypoxia. Taken together, these findings demonstrate that chimeric HCMV/HSV-1 efficiently targets both CD133+ GSCs and glioma cells in hypoxia.


Subject(s)
Cytomegalovirus/metabolism , Glioblastoma/therapy , Herpesvirus 1, Human/genetics , Oncolytic Virotherapy , Protein Kinases/metabolism , Viral Proteins/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Cytomegalovirus/genetics , Glioblastoma/metabolism , HSP27 Heat-Shock Proteins/metabolism , Humans , Mice, Nude , Organisms, Genetically Modified , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Gene Ther ; 21(11): 984-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25119379

ABSTRACT

Limited expression and distribution of nectin-1, the major herpes simplex virus (HSV) type-1 entry-receptor, within tumors has been proposed as an impediment to oncolytic HSV (oHSV) therapy. To determine whether resistance to oHSVs in malignant peripheral nerve sheath tumors (MPNSTs) was explained by this hypothesis, nectin-1 expression and oHSV viral yields were assessed in a panel of MPNST cell lines using γ134.5-attenuated (Δγ134.5) oHSVs and a γ134.5 wild-type (wt) virus for comparison. Although there was a correlation between nectin-1 levels and viral yields with the wt virus (R=0.75, P =0.03), there was no correlation for Δγ134.5 viruses (G207, R7020 or C101) and a modest trend for the second-generation oHSV C134 (R=0.62, P=0.10). Nectin-1 overexpression in resistant MPNST cell lines did not improve Δγ134.5 oHSV output. While multistep replication assays showed that nectin-1 overexpression improved Δγ134.5 oHSV cell-to-cell spread, it did not confer a sensitive phenotype to resistant cells. Finally, oHSV yields were not improved with increased nectin-1 in vivo. We conclude that nectin-1 expression is not the primary obstacle of productive infection for Δγ134.5 oHSVs in MPNST cell lines. In contrast, viruses that are competent in their ability to counter the antiviral response may derive benefit with higher nectin-1 expression.


Subject(s)
Cell Adhesion Molecules/metabolism , Nerve Sheath Neoplasms/metabolism , Oncolytic Viruses/physiology , Receptors, Virus/metabolism , Simplexvirus/physiology , Animals , Cell Line, Tumor , Chlorocebus aethiops , Cricetulus , Humans , Mice , Nectins , Nerve Sheath Neoplasms/virology , Oncolytic Virotherapy , Oncolytic Viruses/metabolism
6.
Gene Ther ; 18(11): 1098-102, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21544094

ABSTRACT

Oncolytic viruses have been combined with standard cancer therapies to increase therapeutic efficacy. Given the sequential activation of herpes viral genes (herpes simplex virus-1, HSV-1) and the temporal cellular changes induced by ionizing radiation, we hypothesized an optimal temporal sequence existed in combining oncolytic HSV-1 with ionizing radiation. Murine U-87 glioma xenografts were injected with luciferase encoding HSV-1, and ionizing radiation (IR) was given at times before or after viral injection. HSV-1 replication and tumor-volume response were followed. Radiation given 6-9 h after HSV-1 injection resulted in maximal viral luciferase expression and infectious viral production in tumor xenografts. The greatest xenograft regression was also seen with radiation given 6 h after viral injection. We then tested if HSV-1 replication had a dose response to ionizing radiation. HSV-1 luciferase expression exhibited a dose response as xenografts were irradiated from 0 to 5 Gy. There was no difference in viral luciferase expression as IR dose increased from 5 Gy up to 20 Gy. These results suggest that the interaction of IR with the HSV-1 lytic cycle can be manipulated for therapeutic gain by delivering IR at a specific time within viral replicative cycle.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Herpesvirus 1, Human/growth & development , Oncolytic Virotherapy/methods , Radiation, Ionizing , Virus Replication/radiation effects , Animals , Combined Modality Therapy , Dose-Response Relationship, Radiation , Herpesvirus 1, Human/radiation effects , Mice , Mice, Nude , Oncolytic Viruses/growth & development , Oncolytic Viruses/radiation effects , Virus Replication/genetics
7.
Cancer Gene Ther ; 16(10): 794-805, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19363468

ABSTRACT

Malignant forms of glioma, the most common primary brain tumors, remain poorly responsive to multimodality therapeutic interventions, including chemotherapy. Suppressed apoptosis and extraordinary invasiveness are important distinctive features that contribute to the malignant phenotype of glioma. We have developed the vascular endothelial growth factor receptor 1 (VEGFR-1/flt-1) conditional replicating adenoviral vector (CRAdRGDflt-IL24) encoding the interleukin-24 (IL-24) gene. We investigated whether a combination of CRAdRGDflt-IL24-mediated oncolytic virotherapy and chemotherapy using temozolomide (TMZ) produces increased cytotoxicity against human glioma cells in comparison with these agents alone. Combination of CRAdRGDflt-IL24 and TMZ significantly enhanced cytotoxicity in vitro, inhibited D54MG tumor growth and prolonged survival of mice harboring intracranial human glioma xenografts in comparison with CRAdRGDflt-IL24 or TMZ alone. These data indicate that combined treatment with CRAdRGDflt-IL24-mediated oncolytic virotherapy and TMZ chemotherapy provides a promising approach for glioma therapy.


Subject(s)
Brain Neoplasms/therapy , Dacarbazine/analogs & derivatives , Glioma/therapy , Interleukins/genetics , Oncolytic Virotherapy/methods , Vascular Endothelial Growth Factor Receptor-1/genetics , Adenoviridae/genetics , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/virology , Cell Growth Processes/genetics , Cell Line, Tumor , Combined Modality Therapy , Dacarbazine/pharmacology , Female , Genetic Therapy/methods , Genetic Vectors/genetics , Glioma/drug therapy , Glioma/genetics , Glioma/virology , Humans , Mice , Mice, Nude , Promoter Regions, Genetic , Recombinant Proteins/pharmacology , Temozolomide , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Xenograft Model Antitumor Assays
8.
Magn Reson Med ; 61(3): 615-25, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19235262

ABSTRACT

The poor prognosis associated with malignant glioma is largely attributable to its invasiveness and robust angiogenesis. Angiogenesis involves host-tumor interaction and requires in vivo evaluation. Despite their versatility, few studies have used mouse glioma models with perfusion MRI approaches, and generally lack longitudinal study design. Using a micro-MRI system (8.5 Tesla), a novel dual bolus-tracking perfusion MRI strategy was implemented. Using the small molecule contrast agent Magnevist, dynamic contrast enhanced MRI was implemented in the intracranial 4C8 mouse glioma model to determine K(trans) and v(e), indices of tumor vascular permeability and cellularity, respectively. Dynamic susceptibility contrast MRI was subsequently implemented to assess both cerebral blood flow and volume, using the macromolecular superparamagnetic iron oxide, Feridex, which circumvented tumor bolus susceptibility curve distortions from first-pass extravasation. The high-resolution parametric maps obtained over 4 weeks, indicated a progression of tumor vascularization, permeability, and decreased cellularity with tumor growth. In conclusion, a comprehensive array of key parameters were reliably quantified in a longitudinal mouse glioma study. The syngeneic 4C8 intracerebral mouse tumor model has excellent characteristics for studies of glioma angiogenesis. This approach provides a useful platform for noninvasive and highly diagnostic longitudinal investigations of anti-angiogenesis strategies in a relevant orthotopic animal model.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/physiopathology , Ferric Compounds , Glioma/blood supply , Glioma/physiopathology , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Neovascularization, Pathologic/pathology , Animals , Blood Flow Velocity , Cell Line, Tumor , Contrast Media , Female , Mice , Mice, Inbred C57BL , Molecular Weight , Permeability
9.
Gene Ther ; 14(14): 1111-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17495948

ABSTRACT

Combined treatment using adenoviral (Ad)-directed enzyme/prodrug therapy and radiation therapy has the potential to become a powerful method of cancer therapy. We have developed an Ad vector encoding a mutant bacterial cytosine deaminase (bCD) gene (AdbCD-D314A), which has a higher affinity for cytosine than wild-type bCD (bCDwt). The purpose of this study was to evaluate cytotoxicity in vitro and therapeutic efficacy in vivo of the combination of AdbCD-D314A with the prodrug 5-fluorocytosine (5-FC) and ionizing radiation against human glioma. The present study demonstrates that AdbCD-D314A infection resulted in increased 5-FC-mediated cell killing, compared with AdbCDwt. Furthermore, a significant increase in cytotoxicity following AdbCD-D314A and radiation treatment of glioma cells in vitro was demonstrated as compared to AdbCDwt. Animal studies showed significant inhibition of subcutaneous or intracranial tumor growth of D54MG glioma xenografts by the combination of AdbCD-D314A/5-FC with ionizing radiation as compared with either agent alone, and with AdbCDwt/5-FC plus radiation. The results suggest that the combination of AdbCD-D314A/5-FC with radiation produces markedly increased cytotoxic effects in cancer cells in vitro and in vivo. These data indicate that combined treatment with this novel mutant enzyme/prodrug therapy and radiotherapy provides a promising approach for cancer therapy.


Subject(s)
Adenoviridae/genetics , Brain Neoplasms/therapy , Cytosine Deaminase/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glioma/therapy , Animals , Antimetabolites/therapeutic use , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Cell Line, Tumor , Combined Modality Therapy , Cytosine/metabolism , Cytosine Deaminase/metabolism , Escherichia coli/enzymology , Flucytosine/therapeutic use , Genes, Transgenic, Suicide , Genetic Vectors/genetics , Glioma/diagnostic imaging , Glioma/metabolism , Humans , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Prodrugs/therapeutic use , Radiography , Transplantation, Heterologous
10.
Gene Ther ; 14(13): 1045-54, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17429445

ABSTRACT

Oncolytic herpes simplex virus (HSV)-1 gamma(1)34.5-deletion mutants (Deltagamma(1)34.5 HSV) are promising agents for tumor therapy. The attenuating mutation renders the virus aneurovirulent but also limits late viral protein synthesis and efficient replication in many tumors. We tested whether one function of gamma(1)34.5 gene, which mediates late viral protein synthesis through host protein kinase R (PKR) antiviral response evasion, could be restored, without restoring the neurovirulence. We have previously reported the construction of two chimeric Deltagamma(1)34.5 HSV vectors (chimeric HSV), C130 and C134, which express the human cytomegalovirus (HCMV) PKR-evasion genes TRS1 and IRS1, respectively. We now demonstrate the following. The HCMV/HSV-1 chimeric viruses (i) maintain late viral protein synthesis in the human malignant glioma cells tested (D54-MG, U87-MG and U251-MG); (ii) replicate to higher titers than Deltagamma(1)34.5 HSV in malignant glioma cells in vitro and in vivo; (iii) are aneurovirulent; and (iv) are superior to other Deltagamma(1)34.5 HSV with both improved reduction of tumor volumes in vivo, and improved survival in two experimental murine brain tumor models. These findings demonstrate that transfer of HCMV IRS1 or TRS1 gene into Deltagamma(1)34.5 HSV significantly improves replication in malignant gliomas without restoring wild-type neurovirulence, resulting in enhanced tumor reduction and prolonged survival.


Subject(s)
Brain Neoplasms/therapy , Cytomegalovirus/genetics , Genetic Therapy/methods , Glioblastoma/therapy , Herpesvirus 1, Human/genetics , Oncolytic Virotherapy/methods , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Chimera , Genetic Engineering , Glioblastoma/pathology , Glioma , Mice , Mice, SCID , Neoplasm Transplantation , Neuroblastoma , Neurons/pathology , Neurons/virology , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Transplantation, Heterologous , Virus Replication
11.
Cancer Gene Ther ; 14(1): 45-56, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16990846

ABSTRACT

Lack of effective therapy of primary brain tumors has promoted the development of novel experimental approaches utilizing oncolytic viruses combined with gene therapy. Towards this end, we have assessed a conditionally replication-competent, gamma(1)34.5-deleted herpes simplex virus type 1 (HSV-1) expressing cytosine deaminase (CD) for treatment of malignant brain tumors. Our results are summarized as follows: (i) a recombinant HSV (M012) was constructed in which both copies of the gamma(1)34.5 gene were replaced with the bacterial CD gene, under the control of the cellular promoter Egr-1; (ii) M012-infected cells in vitro efficiently convert 5-fluorocytosine (5-FC) to 5-fluorouracil, thereby enhancing cytotoxicity of neighboring, uninfected cells; (iii) both direct and bystander cytotoxicity of murine neuroblastoma and human glioma cell lines after infection with M012 were demonstrated; (iv) direct intracerebral inoculation of A/J mice demonstrated lack of neurotoxicity at doses similar to G207, a gamma(1)34.5-deleted HSV with demonstrated safety in human patient trials and (v) intratumoral injection of M012 into Neuro-2a flank tumors in combination with 5-FC administration significantly reduced tumor growth versus tumors treated with R3659 combined with 5-FC, or treated with M012 alone. Thus, M012 is a promising new oncolytic HSV vector with an enhanced prodrug-mediated, antineoplastic effect that is safe for intracranial administration.


Subject(s)
Bacteria/enzymology , Brain Neoplasms/therapy , Cytosine Deaminase/genetics , Simplexvirus/genetics , Animals , Brain Neoplasms/drug therapy , Chlorocebus aethiops , Female , Fluorouracil/therapeutic use , Genetic Engineering , Humans , Mice , Vero Cells
12.
Gene Ther ; 14(6): 523-32, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17167498

ABSTRACT

Adenovirus serotype 5 (Ad5) is widely used in the development of gene therapy protocols. However, current gene therapy strategies involving brain are mostly based on intra-cranial injection. A major obstacle for systemically administered vectors to infect brain tissue is the blood-brain barrier (BBB). One strategy to cross the BBB is transcytosis, a transcellular transport process that shuttles a molecule from one side of the cell to the other side. Recently, melanotransferrin (MTf)/P97 was found to be able to cross the BBB and accumulate in brain. We thus hypothesize that re-directing Ad5 vectors to the MTf transcytosis pathway may facilitate Ad5 vectors to cross the BBB. To test this hypothesis, we constructed a bi-specific adaptor protein containing the extracellular domain of the coxsackie-adenovirus receptor (CAR) and the full-length melanotransferrin (sCAR-MTf), and investigated its ability to re-direct Ad5 vectors to the MTf transcytosis pathway. We found this adaptor protein could re-direct Ad5 to the MTf transcytosis pathway in an in vitro BBB model, and the transcytosed Ad5 viral particles retained their native infectivity. The sCAR-MTf-mediated Ad5 transcytosis was temperature- and dose dependent. In addition, we examined the directionality of sCAR-MTf-mediated Ad5 transcytosis, and found the efficiency of apical-to-basal transcytosis was much higher than that of basal-to-apical direction, supporting a role of this strategy in transporting Ad5 vectors towards the brain. Taken together, our study demonstrated that re-directing Ad5 to the MTf transcytosis pathway could facilitate gene delivery across the BBB.


Subject(s)
Adenoviridae Infections/metabolism , Adenoviridae/genetics , Blood-Brain Barrier/virology , Genetic Therapy/methods , Neoplasm Proteins/genetics , Antigens, Neoplasm , Biological Transport , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Gene Expression , Genetic Engineering , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HeLa Cells , Humans , Luciferases/genetics , Melanoma-Specific Antigens , Neoplasm Proteins/metabolism , Receptors, Virus/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Transduction, Genetic/methods
13.
Gene Ther ; 9(1): 75-80, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11850725

ABSTRACT

The herpes simplex virus (HSV) recombinant virus R7020 is an attenuated virus designed as a candidate for immunization against both HSV-1 and HSV-2 infections. It was extensively tested in an experimental animal system and in a healthy human adult population without significant untoward effects. We report on the use of R7020 with ionizing radiation as an oncolytic agent for hepatomas. Two hepatoma cell lines were studied, Hep3B and Huh7. R7020 replicated to higher titers in Hep3B cells than in Huh7 cells. Tissue culture studies correlated with hepatoma xenograft responses to R7020. R7020 was more effective in mediating Hep3B tumor xenograft regression compared with Huh7. Ionizing radiation combined with R7020 also showed differential results in antitumor efficacy between the two cell lines in tumor xenografts. Ionizing radiation enhanced the replication of R7020 in Hep3B xenografts. Moreover, the combination of ionizing radiation and virus caused a greater regression of xenograft volume than either R7020 or radiation alone. Ionizing radiation had no effect on the replication of R7020 virus in Huh7 xenografts. These results indicate that a regimen involving infection with an appropriate herpesvirus such as R7020 in combination with ionizing radiation can be highly effective in eradicating certain tumor xenografts.


Subject(s)
Genetic Therapy/methods , Herpes Simplex Virus Vaccines/administration & dosage , Herpesvirus 1, Human , Herpesvirus 2, Human , Liver Neoplasms, Experimental/therapy , Animals , Combined Modality Therapy , Humans , Liver Neoplasms, Experimental/radiotherapy , Mice , Mice, Nude , Neoplasm Transplantation , Tumor Cells, Cultured , Virus Replication/radiation effects
14.
Cancer Res ; 61(23): 8470-9, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11731430

ABSTRACT

The failure and/or toxicity of conventional therapies for many types of human cancers underscore the need for development of safe and effective alternative treatments. Toward this goal, we describe the direct oncolytic activity of RNA-based vectors derived from poliovirus, termed replicons, which are genetically incapable of producing infectious virus. These replicons are cytopathic in vitro for human tumor cells originating from brain, breast, lung, ovary, and skin (melanoma). The cytopathic effects in a malignant glioma cell line were associated with nuclear DNA condensation, indicative of cells undergoing apoptosis. Injection of replicons into established xenograft flank tumors in scid mice resulted in oncolytic activity and extended survival. Inoculation of replicons into established intracranial xenograft tumors in scid mice resulted in tumor infection within 8 h and extended survival. Histological analysis revealed that replicons had infected tumor cells at the site of inoculation and, most importantly, diffused to infect tumor cells that had metastasized from the initial site of implantation. The wide spectrum of cytopathic activity for human tumors combined with effective distribution after in vivo inoculation establishes the therapeutic potential of poliovirus replicons for a variety of cancers.


Subject(s)
Neoplasms/therapy , Poliovirus/physiology , Replicon/physiology , Animals , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Brain Neoplasms/virology , Cytopathogenic Effect, Viral , Glioma/pathology , Glioma/therapy , Glioma/virology , HeLa Cells , Humans , Mice , Mice, SCID , Neoplasms/pathology , Neoplasms/virology , Poliovirus/genetics , RNA, Viral/administration & dosage , RNA, Viral/genetics , Xenograft Model Antitumor Assays
15.
Arch Biochem Biophys ; 394(2): 167-72, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11594730

ABSTRACT

Based on previous findings of increased nitric oxide synthase (NOS) expression in human gliomas (4), we hypothesized that peroxynitrite, a highly reactive metabolite of nitric oxide (NO) and superoxide (O(*-)(2)), might be increased in these tumors in vivo. Here we demonstrate that nitrotyrosine (a footprint of peroxynitrite protein modification) is present in human malignant gliomas. Furthermore, we show that p53, a key tumor suppressor protein, has evidence of peroxynitrite-mediated modifications in gliomas in vivo. Experiments in vitro demonstrate that peroxynitrite treatment of recombinant wild-type p53 at physiological concentrations results in formation of higher molecular weight aggregates, tyrosine nitration, and loss of specific DNA binding. Peroxynitrite treatment of human glioma cell lysates similarly resulted in selective tyrosine nitration of p53 and was also associated with loss of p53 DNA binding ability. These data indicate that tyrosine nitration of proteins occurs in human gliomas in vivo, that p53 may be a target of peroxynitrite in these tumors, and that physiological concentrations of peroxynitrite can result in a loss of p53 DNA binding ability in vitro. These findings raise the possibility that peroxynitrite may contribute to loss of wild-type p53 functional activity in gliomas by posttranslational protein modifications.


Subject(s)
Glioblastoma/metabolism , Glioma/metabolism , Peroxynitrous Acid/metabolism , Tumor Suppressor Protein p53/metabolism , Tyrosine/analogs & derivatives , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Glioblastoma/pathology , Glioma/pathology , Humans , Immunohistochemistry , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Peroxynitrous Acid/chemistry , Peroxynitrous Acid/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/drug effects , Tyrosine/analysis , Tyrosine/chemistry , Tyrosine/metabolism
16.
J Biol Chem ; 276(42): 38755-61, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11514582

ABSTRACT

Members of the degenerin/epithelial Na(+) channel superfamily of ion channels subserve many functions, ranging from whole body sodium handling to mechanoelectrical transduction. We studied brain Na(+) channel 2 (BNaC-2) in planar lipid bilayers to examine its single channel properties and regulation by Ca(2+). Upon incorporation of vesicles made from membranes of oocytes expressing either wild-type (WT) BNaC-2 or BNaC-2 with a gain-of-function (GF) point mutation (G433F), functional channels with different properties were obtained. WT BNaC-2 resided in a closed state with short openings, whereas GF BNaC-2 was constitutively activated; a decrease in the pH in the trans compartment of the bilayer activated WT BNaC-2 and decreased its permeability for Na(+) over K(+). Moreover, these maneuvers made the WT channel more resistant to amiloride. In contrast, GF BNaC-2 did not respond to a decrease in pH, and its amiloride sensitivity and selectivity for Na(+) over K(+) were unaffected by this pH change. Buffering the bathing solutions with EGTA to reduce the free [Ca(2+)] to <10 nm increased WT single channel open probability 10-fold, but not that of GF BNaC-2. Ca(2+) blocked both WT and GF BNaC-2 in a dose- and voltage-dependent fashion; single channel conductances were unchanged. A drop in pH reduced the ability of Ca(2+) to inhibit these channels. These results show that BNaC-2 is an amiloride-sensitive sodium channel and suggest that pH activation of these channels could be, in part, a consequence of H(+) "interference" with channel regulation by Ca(2+).


Subject(s)
Brain/metabolism , Calcium/metabolism , Hydrogen-Ion Concentration , Ion Channels/chemistry , Lipid Bilayers/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Sodium Channels/chemistry , Sodium Channels/genetics , Acid Sensing Ion Channels , Animals , Chelating Agents/pharmacology , Cloning, Molecular , Degenerin Sodium Channels , Egtazic Acid/pharmacology , Epithelial Sodium Channels , Kinetics , Membrane Proteins , Nerve Tissue Proteins/metabolism , Oocytes/metabolism , Point Mutation , Protein Binding , Sodium Channels/metabolism , Xenopus
17.
Cancer Res ; 61(7): 3084-91, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306491

ABSTRACT

Fas transduces not only apoptotic signals through various pathways but also angiogenic and proinflammatory responses in vivo. Human glioma cells express Fas although sensitivity to Fas-mediated cell death is variable, suggesting that Fas may have functions other than apoptosis in these cells. In this study, we addressed alternative functions of Fas expressed on human gliomas by Fas ligation in three human glioma cell lines, CRT-MG, U373-MG, and U87-MG, and the in vivo expression of Fas and chemokines in human glioblastoma multiforme (GBM). Herein, we demonstrate that: (a) stimulation with agonistic anti-Fas monoclonal antibody CH-11 and human recombinant soluble Fas ligand induces expression of the CC chemokine MCP-1 and the CXC chemokine interleukin-8 by human glioma cell lines at the mRNA and protein levels in a dose- and time-dependent manner; (b) selective pharmacological inhibitors of MEK1 (U0126 and PD98059) and p38 mitogen-activated protein kinase (MAPK) (SB202190) suppress Fas-mediated chemokine expression in a dose-dependent manner; (c) Fas ligation on human glioma cells leads to activation of both extracellular signal-regulated kinases ERK1/ERK2 and p38 MAPK; and (d) GBM samples express higher levels of Fas compared with normal control brain, which correlates with increased interleukin 8 expression. These findings indicate that Fas ligation on human glioma cells leads to the selective induction of chemokine expression, which involves the ERK1/ERK2 and p38 MAPK signaling pathways. Therefore, the Fas-Fas ligand system in human brain tumors may be involved not only in apoptotic processes but also in the provocation of angiogenic and proinflammatory responses.


Subject(s)
Chemokines/biosynthesis , Glioma/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinases/physiology , fas Receptor/physiology , Adult , Apoptosis/physiology , Glioblastoma/enzymology , Glioblastoma/immunology , Glioblastoma/metabolism , Glioma/enzymology , Glioma/immunology , Humans , Interleukin-8/biosynthesis , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Tumor Cells, Cultured , fas Receptor/biosynthesis , p38 Mitogen-Activated Protein Kinases
18.
Cancer Res ; 61(5): 2154-61, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280780

ABSTRACT

Tumors of the central nervous system (CNS) often have sustained expression of labile genes, including angiogenic growth factors and immunosuppressive cytokines, which promote tumor progression. Stabilization of the RNA transcripts for these genes, such as vascular endothelial growth factor (VEGF), is an important molecular pathway for this up-regulation. HuR, a member of the Elav family of RNA-binding proteins, has been implicated in this pathway through its binding to adenine and uridine (AU)-rich stability elements (ARE) located in the 3' untranslated regions (3'-UTRs) of the mRNA. Whereas three of the Elav family members (Hel-N1, HuC, and HuD) are restricted to young and mature neurons, HuR is more broadly expressed, including proliferating cells of the developing CNS. Because RNA stabilization of labile genes may promote tumor growth, we analyzed and compared the expression pattern of HuR in 35 freshly resected and cultured CNS tumors to determine whether there was any correlation with tumor grade or histological type. We found that HuR mRNA was consistently expressed in all of the tumors, regardless of cell origin or degree of malignancy. Using a novel HuR-specific polyclonal antibody, we found that strong HuR protein expression was limited to high-grade malignancies (glioblastoma multiforme and medulloblastoma). Within the glioblastoma multiforme, prominent HuR expression was also detected in perinecrotic areas in which angiogenic growth factors are up-regulated. To further define its role as a potential RNA stabilizer, we analyzed whether HuR could bind to the stability motifs within the 3'-UTRs of cytokines and growth factors linked to brain tumor progression. We used a novel ELISA-based RNA binding assay and focused on the 3'-UTRs of angiogenic factors VEGF, COX-2, and (interleukin) IL-8 as well as the immunomodulating factors IL-6, transforming growth factor (TGF)-beta and tumor necrosis factor (TNF)-alpha as potential RNA ligands. Our results indicated overall a very high binding affinity to these RNA targets. A comparison of these ligands revealed a hierarchy of binding affinities with the angiogenic factors, and TGF-beta showing the highest (Kd of 1.8-3.4 nM), and TNF-alpha the lowest (Kd of 18.3 nM). The expression pattern of HuR, coupled with the RNA binding data, strongly suggests a role for this protein in the posttranscriptional regulation of these genes in CNS tumors.


Subject(s)
3' Untranslated Regions/metabolism , Angiogenesis Inducing Agents/genetics , Antigens, Surface , Brain Neoplasms/metabolism , Cytokines/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions/genetics , Adenine Nucleotides/metabolism , Amino Acid Sequence , Angiogenesis Inducing Agents/biosynthesis , Astrocytoma/genetics , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Division/physiology , Cytokines/biosynthesis , Disease Progression , ELAV Proteins , ELAV-Like Protein 1 , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Medulloblastoma/genetics , Medulloblastoma/metabolism , Medulloblastoma/pathology , Meningioma/genetics , Meningioma/metabolism , Meningioma/pathology , Molecular Sequence Data , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Uracil Nucleotides/metabolism
19.
Physiol Genomics ; 5(1): 21-33, 2001 Feb 07.
Article in English | MEDLINE | ID: mdl-11161003

ABSTRACT

Gene expression profiling of three human temporal lobe brain tissue samples (normal) and four primary glioblastoma multiforme (GBM) tumors using oligonucleotide microarrays was done. Moreover, confirmation of altered expression was performed by whole cell patch clamp, immunohistochemical staining, and RT-PCR. Our results identified several ion and solute transport-related genes, such as N-methyl-d-aspartate (NMDA) receptors, alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA)-2 receptors, GABA(A) receptor subunits alpha3, beta1, beta2, and beta3, the glutamate transporter, the glutamate/aspartate transporter II, the potassium channel K(V)2.1, hK(V)beta3, and the sodium/proton exchanger 1 (NHE-1), that are all downregulated in the tumors compared with the normal tissues. In contrast, aquaporin-1, possibly aquaporins-3 and -5, and GLUT-3 message appeared upregulated in the tumors. Our results also confirmed previous work showing that osteopontin, nicotinamide N-methyltransferase, murine double minute 2 (MDM2), and epithelin (granulin) are upregulated in GBMs. We also demonstrate for the first time that the cytokine and p53 binding protein, macrophage migration inhibitory factor (MIF), appears upregulated in GBMs. These results indicate that the modulation of ion and solute transport genes and heretofore unsuspected cytokines (i.e., MIF) may have profound implications for brain tumor cell biology and thus may identify potential useful therapeutic targets in GBMs.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Profiling , Aquaporin 1 , Aquaporins/analysis , Blood Group Antigens , Brain Neoplasms/metabolism , Brain Neoplasms/physiopathology , Humans , Immunohistochemistry , Macrophage Migration-Inhibitory Factors/analysis , Membrane Potentials/drug effects , N-Methylaspartate/analysis , N-Methylaspartate/pharmacology , Oligonucleotide Array Sequence Analysis , Patch-Clamp Techniques , Potassium Channels/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/physiology , Reverse Transcriptase Polymerase Chain Reaction , Temporal Lobe/cytology , Temporal Lobe/physiology
20.
Mol Med ; 7(9): 598-608, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11778649

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is a hypervascularized and locally infiltrating brain tumor of astroglial origin with a very poor prognosis. An X-linked c-fos oncogene-inducible mitogenic, morphogenic, and angiogenic factor, endothelial growth factor-D (VEGF-D), is the newest mammalian member of VEGF family. We analyzed VEGF-D in GBM because of its high angiogenic potential and its linkage to the X chromosome. MATERIALS AND METHODS: Nonmalignant brain and GBM tissue sections as well as GBM cell lines were analyzed by immunofluorescence for the expression of VEGF-D, factor VIII (endothelial cell marker), glial-fibrillary acidic protein (GFAP) (astrocytic cell lineage cytoplasmic marker), and several Fos family transcription factors, including c-Fos and Fra-1. The proteins were also detected by Western blots. The differences between genotypes of normal brain and GBM cells were examined by cDNA microarrays. RESULTS AND CONCLUSIONS: GBM expressed ubiquitously VEGF-D, which colocalized with GFAP. Contrary to our expectations, low levels of c-Fos were detected in GBM cells. However, we identified another Fos family member, Fra-1, together with its transcriptional activation partner, c-Jun, as being stably up-regulated in GBM cells. Furthermore, we demonstrated that a fra-1 transgene induced VEGF-D expression in cultured cells and GBM cell stimulation evoked a sustained increase in both Fra-1 and VEGF-D levels. This study reveals that an up-regulation of AP-1 factors may be a hallmark of GBM. Because VEGF-D activates VEGF receptor 2 and 3, receptors important for tumor angiogenesis, it may represent an X-linked/AP-1-regulated onco-angiogen in human GBM. The VEGF-D system and AP-1 activity appear to be very attractive targets for new molecular diagnostics and rational molecular anti-cancer therapies.


Subject(s)
Brain/metabolism , Endothelial Growth Factors/metabolism , Glioblastoma/metabolism , Neovascularization, Pathologic , X Chromosome/genetics , Animals , Brain/pathology , Endothelial Growth Factors/genetics , Genetic Linkage , Glioblastoma/blood supply , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunohistochemistry , Mice , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor D
SELECTION OF CITATIONS
SEARCH DETAIL
...