Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Crit Rev Oncol Hematol ; 112: 72-79, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28325267

ABSTRACT

Enzastaurin is an orally administered drug that was intended for the treatment of solid and haematological cancers. It was initially developed as an isozyme specific inhibitor of protein kinase Cß (PKCß), which is involved in both the AKT and MAPK signalling pathways that are active in many cancers. Enzastaurin had shown encouraging preclinical results for the prevention of angiogenesis, inhibition of proliferation and induction of apoptosis as well as showing limited cytotoxicity within phase I clinical trials. However, during its assessment in phase II and III clinical trials the efficacy of enzastaurin was poor both in combination with other drugs and as a single agent. In this review, we will discuss the development of enzastaurin from drug design to clinical testing, exploring target identification, validation and preclinical assessment. Finally, we will consider the clinical evaluation of enzastaurin as an example of the challenges associated with drug development. In particular, we discuss the poor translation of drug efficacy from preclinical animal models, inappropriate end point analysis, limited standards in phase I clinical trials, insufficient use of biomarker analysis and also patient stratification, all of which contributed to the failure to achieve approval of enzastaurin as an anticancer therapeutic.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Indoles/pharmacology , Animals , Humans , Neoplasms/drug therapy , Protein Kinase C beta/antagonists & inhibitors
2.
Cancer Gene Ther ; 20(2): 125-32, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23306614

ABSTRACT

We have recently found a novel oncogene, named cancer upregulated gene 2 (CUG2), which activates Ras and mitogen-activated protein kinases (MAPKs), including ERK, JNK and p38 MAPK. Because activation of these signaling pathways has previously been shown to enhance cancer cell susceptibility to oncolysis by certain viruses, we examined whether vesicular stomatitis virus (VSV) could function as a potential therapeutic agent by efficiently inducing cytolysis in cells transformed by CUG2. Unexpectedly, NIH3T3 cells stably expressing CUG2 (NIH-CUG2) were resistant to VSV because of the activation of signal transducers and activators of transcription 1 (STAT1). The result was supported by evidence showing that suppression of STAT1 with short interference RNA (siRNA) renders cells susceptible to VSV. Furthermore, 2'-5' oligoadenylate synthetase-like (OASL) 2 was the most affected by STAT1 expression level among anti-viral proteins and furthermore suppression of OASL2 mRNA level caused NIH-CUG2 cells to succumb to VSV as seen in NIH-CUG2 cells treated with STAT1 siRNA. In addition, Colon26L5 carcinoma cells stably expressing CUG2 (Colon26L5-CUG2) exhibited resistance to VSV, whereas Colon26L5 stably expressing a control vector yielded to VSV infection. Moreover, Colon26L5-CUG2 cells stably suppressing STAT1 succumbed to VSV infection, resulting in apoptosis. Taken together, we propose that VSV treatment combined with the selective regulation of genes such as STAT1 and OASL2 will improve therapeutic outcomes for CUG2-overexpressing tumors.


Subject(s)
2',5'-Oligoadenylate Synthetase/genetics , Colonic Neoplasms/genetics , Nuclear Proteins/genetics , Oncolytic Viruses/genetics , STAT1 Transcription Factor/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Cell Line, Tumor , Chromosomal Proteins, Non-Histone , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Mice , NIH 3T3 Cells , Nuclear Proteins/metabolism , Oncolytic Virotherapy , Oncolytic Viruses/pathogenicity , STAT1 Transcription Factor/metabolism , Signal Transduction , Vesicular Stomatitis/genetics , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/genetics
3.
Br J Cancer ; 104(2): 290-9, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21179029

ABSTRACT

BACKGROUND: Although the naturally occurring reovirus causes only mild symptoms in humans, it shows considerable potential as an oncolytic agent because of its innate ability to target cancer cells. In immunocompromised hosts, however, wild-type reovirus can target healthy tissues, including heart, liver, pancreas and neural structures. METHODS: We characterized an attenuated form of reovirus (AV) derived from a persistently infected cell line through sequence analysis, as well as western blot and in vitro transcription and translation techniques. To examine its pathogenesis and oncolytic potential, AV reovirus was tested on healthy embryonic stem cells, various non-transformed and transformed cell lines, and in severe combined immunodeficiency (SCID) mice with tumour xenografts. RESULTS: Sequence analysis of AV reovirus revealed a premature STOP codon in its sigma 1 attachment protein. Western blot and in vitro translation confirmed the presence of a truncated σ1. In comparison to wild-type reovirus, AV reovirus did not kill healthy stem cells or induce black tail formation in SCID mice. However, it did retain its ability to target cancer cells and reduce tumour size. CONCLUSION: Despite containing a truncated attachment protein, AV reovirus still preferentially targets cancer cells, and compared with wild-type reovirus it shows reduced toxicity when administered to immunodeficient hosts, suggesting the potential use of AV reovirus in combination cancer therapy.


Subject(s)
Oncolytic Virotherapy , Reoviridae/pathogenicity , Animals , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Humans , Immunohistochemistry , Mice , Mice, SCID , Microscopy, Electron , Protein Biosynthesis , Reoviridae/genetics , Transcription, Genetic , Transplantation, Heterologous , Virulence
4.
Oncogene ; 29(27): 3990-6, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20473328

ABSTRACT

Replication-competent oncolytic viruses hold great potential for the clinical treatment of many cancers. Importantly, many oncolytic virus candidates, such as reovirus and myxoma virus, preferentially infect cancer cells bearing abnormal cellular signaling pathways. Reovirus and myxoma virus are highly responsive to activated Ras and Akt signaling pathways, respectively, for their specificity for viral oncolysis. However, considering the complexity of cancer cell populations, it is possible that other tumor-specific signaling pathways may also contribute to viral discrimination between normal versus cancer cells. Because carcinogenesis is a multistep process involving the accumulation of both oncogene activations and the inactivation of tumor suppressor genes, we speculated that not only oncogenes but also tumor suppressor genes may have an important role in determining the tropism of these viruses for cancer cells. It has been previously shown that many cellular tumor suppressor genes, such as p53, ATM and Rb, are important for maintaining genomic stability; dysfunction of these tumor suppressors may disrupt intact cellular antiviral activity due to the accumulation of genomic instability or due to interference with apoptotic signaling. Therefore, we speculated that cells with dysfunctional tumor suppressors may display enhanced susceptibility to challenge with these oncolytic viruses, as previously seen with adenovirus. We report here that both reovirus and myxoma virus preferentially infect cancer cells bearing dysfunctional or deleted p53, ATM and Rb tumor suppressor genes compared to cells retaining normal counterparts of these genes. Thus, oncolysis by these viruses may be influenced by both oncogenic activation and tumor suppressor status.


Subject(s)
Genes, Tumor Suppressor , Myxoma virus/physiology , Neoplasms/genetics , Neoplasms/virology , Oncolytic Viruses/physiology , Reoviridae/physiology , Viral Tropism , Animals , Apoptosis/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Deletion , Gene Knockdown Techniques , Humans , Mice , Neoplasms/pathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
5.
Cancer Gene Ther ; 17(5): 307-14, 2010 May.
Article in English | MEDLINE | ID: mdl-20075984

ABSTRACT

As we have recently found a novel oncogene, the cancer-upregulated gene 2 (CUG2), which was elevated in a variety of tumor tissues such as the ovary, liver, lung and pancreas, we examined whether reovirus could efficiently induce cytolysis in cancer cells expressing CUG2 and thus be used as a potential cancer therapeutic agent. In this study, we describe experiments in which we use reovirus to treat NIH3T3 cells stably expressing either CUG2 (NIH-CUG2) or vector only (NIH-Vec). NIH-CUG2 cells readily support reoviral proliferation and undergo apoptosis, whereas NIH-Vec cells are highly resistant to reoviral infection and virus-induced apoptosis. This notable result may be explained by the observation that CUG2 expression inhibits PKR activation, leading to reoviral proliferation in nonpermissive NIH3T3 cells. Furthermore, reovirus infection results in almost complete regression of tumorgenic NIH-CUG2 cells in transplanted nude mice. As we found that CUG2 enhances activation of MAPK (ERK, JNK and p38), Src kinase and Ras, we examined whether CUG2 confers reoviral replication independent of the Ras or p38 MAPK signaling pathway. From these experiments we found that either inhibition of p38 MAPK or Ras blocks reoviral proliferation even in the presence of CUG2 but inhibition of ERK, JNK and Src kinase does not, indicating that activation of p38 MAPK and Ras has critical roles in reoviral replication in CUG2-expressing tumor cells. Accordingly, we propose that reovirus can be useful in the treatment of transformed cells expressing CUG2, which is commonly detected in various tumor tissues.


Subject(s)
Apoptosis/physiology , Nuclear Proteins/metabolism , Reoviridae/physiology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Cell Line , Chromosomal Proteins, Non-Histone , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , NIH 3T3 Cells , Nuclear Proteins/genetics , Proliferating Cell Nuclear Antigen/metabolism , Reoviridae/growth & development , p38 Mitogen-Activated Protein Kinases/genetics , ras Proteins/genetics
6.
Cancer Gene Ther ; 17(5): 365-72, 2010 May.
Article in English | MEDLINE | ID: mdl-20075985

ABSTRACT

Many oncolytic viruses are currently being tested as potential cancer therapeutic agents. To be effective, these viruses must replicate and propagate efficiently through the tumor mass. However, it is possible that the hypoxia that characterizes many tumors may be an obstacle to viral therapy because of its inhibition of viral replication and propagation. We, therefore, decided to test how oncolytic reovirus and its target cells respond to hypoxia. We found that reovirus infection suppresses hypoxia inducible factor (HIF)-1alpha protein levels (but not transcript abundance) in colon cancer HCT116 cells under CoCl(2) or hypoxia. Reovirus infection was able to reduce HIF-1alpha levels in both von Hippel Lindau (VHL)-/- renal carcinoma A498 and p53-/- HCT116 cells, indicating that the decrease of HIF-1alpha mediated by reovirus requires neither VHL nor p53 proteins. However, treatment with the inhibitor MG132 restored HIF-1alpha levels, suggesting that reovirus-induced HIF-1alpha decrease needs proteosomal activity. A498 VHL-/- cells with constitutive expression of HIF-1alpha were relatively resistant to reovirus-induced apoptosis when compared with A498 VHL+/+ cells. However, we found that the use of YC-1 to target HIF-1alpha promoted reovirus-induced apoptosis in A498 VHL-/- cells. Accordingly, we propose that reovirus may be used together with YC-1 as a potential therapeutic agent against chemoresistant or radioresistant tumors that are hypoxic and show increased levels of HIF-1alpha.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oncolytic Viruses/physiology , Tumor Suppressor Protein p53/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Down-Regulation , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Oncolytic Viruses/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Cancer Gene Ther ; 16(5): 453-61, 2009 May.
Article in English | MEDLINE | ID: mdl-19096445

ABSTRACT

Although reovirus has been used in tests as a potential cancer therapeutic agent against a variety of cancer cells, its application to hepatocellular carcinoma cells, in which the hepatitis B virus (HBV) X (HBX) protein of HBV plays a primary role, has not yet been explored. Here, we describe experiments in which we use reovirus to treat Chang liver carcinoma cells expressing either a vector only (Chang-vec) or a vector encoding HBX protein (Chang-HBX). Although Chang-vec cells readily support reoviral proliferation and undergo apoptosis, Chang-HBX cells are highly resistant to reoviral infection and virus-induced apoptosis, even though HBX protein induces activation of Ras and inactivation of PKR, which are normally thought to enhance reoviral oncolysis. The resistance of Chang-HBX cells to reovirus may instead be explained by HBX-induced downregulation of death receptor 5 and activation of Stat1. Phosphorylated Stat1 activates interferon (IFN)-stimulated regulatory element (ISRE)- and IFN-gamma-activated sequence (GAS)-mediated transcription, leading to the production of IFN-beta, whereas the reduced expression of Stat1 with its siRNA results in a decrease in IFN-beta production, by which Chang-HBX cells eventually succumb to reovirus infection. This result further indicates that HBX induces the establishment of an antiviral state through Stat1 activation. Thus, it appears that active Ras does not override the antiviral effect mediated by the activation of Stat1. Accordingly, we report that HBX, an oncoprotein of HBV, can prevent reoviral oncolysis of hepatocellular carcinoma. This suggests there may be limits to the practical application of reovirus in the treatment of human cancers already expressing other oncoviral proteins.


Subject(s)
Carcinoma, Hepatocellular/therapy , Gene Expression Regulation , Liver Neoplasms/therapy , Reoviridae/metabolism , Trans-Activators/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Humans , Interferon-alpha/immunology , Interferon-alpha/metabolism , Liver Neoplasms/pathology , Mice , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Reoviridae/genetics , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins , eIF-2 Kinase/metabolism , ras Proteins/metabolism
8.
Cancer Gene Ther ; 14(10): 867-72, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17627293

ABSTRACT

Reovirus type 3 Dearing has demonstrated oncolytic efficacy in vitro and in vivo against a variety of cancer cell lines, tumor xenografts and syngeneic cancer models. In this study, we investigated the effectiveness of reovirus against aberrant crypt foci (ACF) and colon cancer induced by the carcinogen azoxymethane (AOM) in an immunocompetent rat model. Sprague-Dawley rats received 15 mg/kg AOM intraperitoneally once per week for 4 weeks and reovirus was administered rectally once a week for 5 weeks starting 20 weeks after the last dose of AOM. Two weeks after completion of reovirus therapy, animals were examined for tumor burden in the colon and other tissues. Reovirus-treated animals showed a decrease in total ACF numbers (P=0.014), in large ACFs (P=0.0069) and in tumor number (P=0.03) compared to vehicle-treated animals. Fewer obstructing tumors in the colon (P=0.07) and duodenum (P=0.03) and reduced hepatic metastases were also noted. In addition, a tumor cell line derived from hepatic metastases was found to be susceptible to reovirus in vitro. Our results show that repeated rectal reovirus administration had some efficacy in the treatment and prevention of AOM-induced ACFs, colon cancers and metastases.


Subject(s)
Adenocarcinoma/prevention & control , Azoxymethane/toxicity , Carcinogens/toxicity , Colonic Neoplasms/prevention & control , Orthoreovirus/physiology , Precancerous Conditions/prevention & control , Adenocarcinoma/chemically induced , Adenocarcinoma/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/immunology , Female , Lymphocytes/immunology , Male , Precancerous Conditions/chemically induced , Precancerous Conditions/immunology , Rats , Rats, Sprague-Dawley
9.
Oncogene ; 26(28): 4124-34, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17213803

ABSTRACT

Reovirus shows considerable potential as an oncolytic agent for Ras-activated tumors and is currently in clinical trials. Here we ask whether such tumor cell lines can acquire resistance to reoviral oncolysis. We challenged human HT1080 fibrosarcoma cells that carry a Ras mutation by prolonged exposure to reovirus, thereby yielding highly virus-resistant HTR1 cells. These cells are persistently infected with reovirus, exhibit high Ras activity and retain the original Ras gene mutation, showing that resistance to reovirus can be displayed in cells with active Ras. The HTR1 cells also exhibit reduced cellular cathepsin B activity, which normally contributes to viral entry and activation. Persistently infected HTR1 cells were not tumorigenic in vivo, whereas immunologically cured virus-free HTR1 cells were highly tumorigenic. Thus, acquisition of resistance to reovirus may constrain therapeutic strategies. To determine whether reoviral resistance is associated with a general reduction in apoptotic potential, we challenged the HTR1 cells with apoptotic inducers and E1B-defective adenovirus, resulting in significant apoptosis and cell death following both approaches. Therefore, even if resistance to reoviral oncolysis should arise in tumor cells in vivo, other therapeutic strategies may nevertheless remain effective.


Subject(s)
Fibrosarcoma/pathology , Oncogene Protein p21(ras)/physiology , Reoviridae/physiology , Base Sequence , Cathepsin B/metabolism , Cell Line , Cell Line, Tumor , DNA Primers , Fibrosarcoma/virology , Humans , Mutation , Oncogene Protein p21(ras)/genetics
10.
Br J Cancer ; 95(8): 1020-7, 2006 Oct 23.
Article in English | MEDLINE | ID: mdl-17047650

ABSTRACT

The use of oncolytic viruses has received considerable attention in recent years and many viruses have proved to be effective against a variety of cancer models and a few are currently being used in clinical trials. However, the possible emergence and outcome of virus-resistant tumour cells has not been addressed. We previously reported the effective use of reovirus against lymphoid malignancies, including the Burkitt's lymphoma cell line Raji. Here we isolated in vitro persistently infected (PI) Raji cells, and cells 'cured' of persistent reovirus infection ('cured' cells). Both PI and cured Raji cells resisted reovirus infection and cell killing in vitro. In vivo, the PI cells were non-tumorigenic in SCID mice, but cured cells regained the parental cells' ability to form tumours. Tumour xenografts from the cured cells, however, were highly susceptible to reovirus oncolysis in vivo. This susceptibility was due to the proteolytic environment within tumours that facilitates reovirus infection and cell killing. Our results show that persistent infection by reovirus impedes tumour development and that although PI cells cleared of reovirus are tumorigenic, they are killed upon rechallenge with reovirus. Both the PI and cured states are therefore not likely to be significant barriers to reovirus oncolytic therapy.


Subject(s)
Burkitt Lymphoma/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Xenograft Model Antitumor Assays , Animals , Burkitt Lymphoma/pathology , Burkitt Lymphoma/virology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cysteine Proteinase Inhibitors/pharmacology , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Mammalian orthoreovirus 3/drug effects , Mammalian orthoreovirus 3/physiology , Mice , Mice, SCID , Oncolytic Viruses/drug effects , Time Factors , Virion/drug effects , Virion/physiology
11.
Int J Oral Maxillofac Surg ; 35(8): 746-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16777381

ABSTRACT

Expression of the oncofoetal glycoprotein, carcinoembryonic antigen (CEA), has been observed in a number of malignancies and is also being pursued as a target for anti-cancer therapy. This study explored the status of this biochemical entity in the oral squamous cell carcinoma (SCC) in South India caused by extensive chewing habits. Squamous cell carcinoma in the study belonged to grade I and grade II. Tumour staging of the patients recruited in the study ranged from T2N1M0 to T4N3M0. Of the grade II cases studied, 88% (7 out of 8) showed expression of CEA. The 2 cases of grade I SCC of buccal mucosa also showed positive anti-CEA staining. If the results from this pilot study can be validated with a larger sample size, a role can be attributed to this tumour marker in oral neoplasia, thereby opening up avenues for using CEA as an additional diagnostic marker in oral SCC in this population and as a possible target for anti-cancer therapy.


Subject(s)
Carcinoembryonic Antigen/analysis , Carcinoma, Squamous Cell/chemistry , Mouth Neoplasms/chemistry , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/etiology , Female , Humans , India , Male , Middle Aged , Mouth Neoplasms/etiology , Pilot Projects , Tobacco, Smokeless/adverse effects
12.
J Biol Chem ; 276(50): 47013-20, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11600495

ABSTRACT

The candidate tumor suppressor gene, ING1, encodes several protein isoforms as a result of alternative splicing that may possess agonistic and antagonistic roles in the control of cell proliferation and apoptosis. Recently a related gene, ING2, was isolated in human whose expression is increased in adenocarcinomas. Little is known about the cellular function and regulation of these ING family members, but the fact that ING proteins contain a plant homeodomain finger suggests that these proteins may modulate transcription factor-mediated pathways. To elucidate how ING may interact in different tissues to modulate function, we used amphibian metamorphosis as a model system in which a single stimulus, thyroid hormone (TH), initiates tissue-specific proliferation, differentiation, and apoptosis. We have isolated the first Xenopus laevis ING2 and demonstrate that transcript levels increase in response to TH treatment. We provide evidence for the existence of splice variants that are differentially expressed in tissues with different TH-induced fates. Western blots using an antibody directed against the highly conserved C-terminal end of ING proteins reveal a tissue-specific pattern of ING isoform expression in adult Xenopus tissues. Analyses of premetamorphic tadpole tissues show a TH-induced accumulation of ING proteins in tail, whereas the levels in the leg are not affected. This TH-induced accumulation is also observed in serum-free tail organ cultures and is prevented by inhibitors of tail apoptosis. Therefore, this work presents the first link between ING expression and a hormonally regulated nuclear transcription factor-mediated apoptotic response opening the possibility that ING family members may be involved in transducing the signal initiated by TH that determines cell fate.


Subject(s)
Homeodomain Proteins/biosynthesis , Receptors, Cytoplasmic and Nuclear , Thyroid Hormones/metabolism , Tumor Suppressor Proteins , Xenopus Proteins , Alternative Splicing , Amino Acid Sequence , Animals , Apoptosis , Base Sequence , Blotting, Northern , Blotting, Western , Cell Differentiation , Cell Division , Cell Lineage , Cell Nucleus/metabolism , Cloning, Molecular , Culture Media, Serum-Free/pharmacology , DNA, Complementary/metabolism , Female , Gene Library , Genes, Tumor Suppressor , Homeodomain Proteins/genetics , Humans , Male , Metamorphosis, Biological , Mice , Molecular Sequence Data , Organ Culture Techniques , Protein Isoforms , Protein Structure, Tertiary , RNA, Messenger/metabolism , Receptors, Thyroid Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction , Thyroid Hormones/pharmacology , Tissue Distribution , Triiodothyronine/pharmacology , Xenopus , Xenopus laevis
13.
Br J Cancer ; 85(1): 55-63, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11437402

ABSTRACT

Studies have suggested that an imbalance of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) may contribute to the malignant phenotype of gliomas. In this study, we have undertaken a detailed analysis of expression of the TIMP family in normal human brain and malignant gliomas at both the mRNA and protein level. Reverse transcription-PCR (RT-PCR) analyses of total RNA from surgical tumour specimens revealed unique expression patterns for the 4 members of the TIMP family, with TIMP-1 and -4 showing positive and negative correlations, respectively, with glioma malignancy. By RT-PCR, TIMP-2 and TIMP-3 expression did not change with tumour grade. In situ hybridization localized TIMP-1 to glial tumour cells and also to the surrounding tumour vasculature. TIMP-4 transcripts were predominantly localized to tumour cells, though minor expression was found in vessels. Recombinant TIMP-4 reduced invasion of U251 glioma cells through Matrigel, and U87 clones overexpressing TIMP-4 showed reduced invasive capacity in vitro. TIMP-4, but not TIMP-1, blocked Membrane Type-1-MMP-mediated progelatinase-A (MMP-2) activation in human umbilical vein endothelial cells. The differential expression and localization of individual TIMPs may contribute to the pathophysiology of human malignant gliomas, particularly with regard to tumour vascularization.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinases/biosynthesis , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Enzyme Activation/drug effects , Enzyme Precursors/metabolism , Gelatinases/metabolism , Gene Expression Regulation, Neoplastic , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Humans , In Situ Hybridization , Metalloendopeptidases/metabolism , Neoplasm Invasiveness , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/pharmacology , Tumor Cells, Cultured , Tissue Inhibitor of Metalloproteinase-4
14.
J Natl Cancer Inst ; 93(12): 903-12, 2001 Jun 20.
Article in English | MEDLINE | ID: mdl-11416111

ABSTRACT

BACKGROUND: Reovirus is a naturally occurring oncolytic virus that usurps activated Ras-signaling pathways of tumor cells for its replication. Ras pathways are activated in most malignant gliomas via upstream signaling by receptor tyrosine kinases. The purpose of this study was to determine the effectiveness of reovirus as an experimental treatment for malignant gliomas. METHODS: We investigated whether reovirus would infect and lyse human glioma cell lines in vitro. We also tested the effect of injecting live reovirus in vivo on human gliomas grown subcutaneously or orthotopically (i.e., intracerebrally) in mice. Finally, reovirus was tested ex vivo against low-passage cell lines derived from human glioma specimens. All P values were two-sided. RESULTS: Reovirus killed 20 (83%) of 24 established malignant glioma cell lines tested. It caused a dramatic and often complete tumor regression in vivo in two subcutaneous (P =.0002 for both U251N and U87) and in two intracerebral (P =.0004 for U251N and P =.0009 for U87) human malignant glioma mouse models. As expected, serious toxic effects were found in these severely immunocompromised hosts. In a less immunocompromised mouse model, a single intratumoral inoculation of live reovirus led to a dramatic prolongation of survival (compared with control mice treated with dead virus; log-rank test, P<.0001 for both U251N and U87 cell lines). The animals treated with live virus also appeared to be healthier and gained body weight (P =.0001). We then tested the ability of reovirus to infect and kill primary cultures of brain tumors removed from patients and found that it killed nine (100%) of nine glioma specimens but none of the cultured meningiomas. CONCLUSIONS: Reovirus has potent activity against human malignant gliomas in vitro, in vivo, and ex vivo. Oncolysis with reovirus may be a potentially useful treatment for a broad range of human cancers.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Mammalian orthoreovirus 3/physiology , Animals , Brain Neoplasms/pathology , Brain Neoplasms/virology , Female , Glioma/pathology , Glioma/virology , Humans , Male , Mammalian orthoreovirus 3/isolation & purification , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Survival Rate , Transplantation, Heterologous , Tumor Cells, Cultured
15.
Nucleic Acids Res ; 29(10): 2052-8, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11353074

ABSTRACT

The ING1 candidate tumor suppressor is downregulated in a variety of primary tumors and established cancer cell lines. Blocking its expression experimentally promotes unregulated growth in vitro and in vivo, using cell and animal models. Alternative splicing products encode proteins that localize to the nucleus, inhibit cell cycle progression and affect apoptosis in different model systems. Here we show that ING1 proteins translocate to the nucleolus 12-48 h after UV-induced DNA damage. When a small 50 amino acid portion of ING1 was fused to green fluorescent protein, the fusion protein was efficiently targeted to the nucleolus, indicating that ING1 possesses an intrinsic nucleolar targeting sequence (NTS). We mapped this activity to two distinct 4 amino acid regions, which individually direct fused heterologous proteins to the nucleolus. Overexpression of ING1 induced apoptosis of primary fibroblasts in the presence and absence of UV exposure. In contrast, NTS mutants of ING1 that were not targeted to the nucleolus did not efficiently induce apoptosis when overexpressed and instead protected cells from UV-induced apoptosis. Taken together, these results indicate that UV induces ING1 to translocate to the nucleolus and that this translocation may facilitate apoptosis.


Subject(s)
Cell Nucleolus/metabolism , Cell Nucleolus/radiation effects , Protein Sorting Signals/physiology , Proteins/chemistry , Proteins/metabolism , Amino Acid Sequence , Apoptosis/radiation effects , Cell Cycle Proteins , Cells, Cultured , Consensus Sequence , DNA Damage/radiation effects , DNA-Binding Proteins , Fibroblasts , Fluorescent Antibody Technique, Indirect , Gene Products, tat/genetics , Gene Products, tat/metabolism , Genes, Tumor Suppressor/genetics , Humans , Inhibitor of Growth Protein 1 , Intracellular Signaling Peptides and Proteins , Kinetics , Molecular Sequence Data , Mutation/genetics , Nuclear Localization Signals/genetics , Nuclear Localization Signals/physiology , Nuclear Proteins , Protein Binding , Protein Sorting Signals/genetics , Protein Transport/radiation effects , Proteins/genetics , RNA Polymerase I/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription, Genetic/radiation effects , Transfection , Tumor Suppressor Proteins , Ultraviolet Rays
16.
Oncogene ; 19(29): 3256-65, 2000 Jul 06.
Article in English | MEDLINE | ID: mdl-10918582

ABSTRACT

Recent data indicating that overexpression of caveolin-1 as well as c-myc are relatively common features of advanced prostate cancer prompted us to test for potential cooperative interactions between caveolin-1 and c-myc that would be consistent with malignant progression. We used the well-characterized Rat1AmycERT cells to show that the caveolin-1 gene is down-regulated at the level of transcription by c-myc. By maintaining relatively high levels of caveolin-1 with an adenoviral vector or in stably transfected clones we show that caveolin-1 can suppress c-myc-induced apoptosis. Further we established human prostate cancer cell lines with the mycER construct and show that clones with increased caveolin-1 are more resistant to myc-induced apoptosis and have increased capacity for growth in soft agar when c-myc is activated.


Subject(s)
Apoptosis , Caveolins , Down-Regulation , Membrane Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Caveolin 1 , Cell Line , Humans , Male , Membrane Proteins/metabolism , Prostatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-myc/genetics , Rats
17.
J Cell Biochem ; 78(1): 151-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10797574

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) exists in large multimeric complexes, but its function and binding partners in these complexes are unclear. We explored these issues by chromatographic and immunochemical analyses of Cdk5 and p25(nck5a) (a neuronal Cdk5 activator) and their associated proteins from bovine brain. Mono-S column enzyme eluates were divided into three fractions and analyzed by gel filtration. The majority of p25(nck5a) from Mono-S fractions I, II, and III eluted from the gel filtration column at approximately 60, 200, and 400 kDa, respectively, and Cdk5 was abundant in fractions >400 kDa. We characterized these macromolecular structures by immunoprecipitating p25(nck5a), followed by a second immunoprecipitation of remaining unbound proteins using a Cdk5 antibody. The p25(nck5a) immunoprecipitates showed association with Cdk5. Amphiphysin was detected in the 400-kDa complex and synapsin I in the >400 kDa structure. The Cdk5 immunoprecipitates, however, revealed abundant retained Cdk5 but no remaining p25(nck5a), indicating that Cdk5 in macromolecular structures is mostly unassociated with p25(nck5a). Thus, we demonstrate: an amphiphysin-associated 400-kDa Cdk5/p25(nck5a) complex, a synapsin I-associated >400-kDa Cdk5/p25(nck5a) complex, and nck5a-free Cdk5 complexes (200 to >400 kDa). Amphiphysin acts as a Cdk5/p25(nck5a) substrate in the 400-kDa complex and we speculate that Cdk5/p25(nck5a) participates in amphiphysin-mediated endocytosis.


Subject(s)
Brain/metabolism , Cyclin-Dependent Kinases/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Animals , Cattle , Chromatography, Gel , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/isolation & purification , Nerve Tissue Proteins/isolation & purification , Phosphorylation
18.
Neuro Oncol ; 2(3): 145-50, 2000 07.
Article in English | MEDLINE | ID: mdl-11302334

ABSTRACT

Malignant gliomas maintain a poor prognosis and survival rate due to their marked local invasive growth and neovascularization. Matrix metalloproteinases (MMPs) have been implicated in glioma invasion and angiogenesis, but it is unknown whether they are produced by the tumor cells or surrounding stroma. Using in situ hybridization and immunohistochemistry, we found expression of mRNA for both gelatinase-A (MMP2) and gelatinase-B (MMP9) localized to tumor cells and vascular structures in glioma sections. Gelatinase-A protein expression was detected most prominently in tumor cells, with very little signal seen in vasculature. Gelatinase-B protein expression was prominent in vascular structures but was also expressed in tumor cells. Our data show that these proteases are produced by glioma cells and vascular structures and suggest that synthetic MMP inhibitors might be useful in this disease.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
19.
J Bacteriol ; 181(21): 6747-55, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10542177

ABSTRACT

Current bacterial taxonomy is mostly based on phenotypic criteria, which may yield misleading interpretations in classification and identification. As a result, bacteria not closely related may be grouped together as a genus or species. For pathogenic bacteria, incorrect classification or misidentification could be disastrous. There is therefore an urgent need for appropriate methodologies to classify bacteria according to phylogeny and corresponding new approaches that permit their rapid and accurate identification. For this purpose, we have devised a strategy enabling us to resolve phylogenetic clusters of bacteria by comparing their genome structures. These structures were revealed by cleaving genomic DNA with the endonuclease I-CeuI, which cuts within the 23S ribosomal DNA (rDNA) sequences, and by mapping the resulting large DNA fragments with pulsed-field gel electrophoresis. We tested this experimental system on two representative bacterial genera: Salmonella and Pasteurella. Among Salmonella spp., I-CeuI mapping revealed virtually indistinguishable genome structures, demonstrating a high degree of structural conservation. Consistent with this, 16S rDNA sequences are also highly conserved among the Salmonella spp. In marked contrast, the Pasteurella strains have very different genome structures among and even within individual species. The divergence of Pasteurella was also reflected in 16S rDNA sequences and far exceeded that seen between Escherichia and Salmonella. Based on this diversity, the Pasteurella haemolytica strains we analyzed could be divided into 14 phylogenetic groups and the Pasteurella multocida strains could be divided into 9 groups. If criteria for defining bacterial species or genera similar to those used for Salmonella and Escherichia coli were applied, the striking phylogenetic diversity would allow bacteria in the currently recognized species of P. multocida and P. haemolytica to be divided into different species, genera, or even higher ranks. On the other hand, strains of Pasteurella ureae and Pasteurella pneumotropica are very similar to those of P. multocida in both genome structure and 16S rDNA sequence and should be regarded as strains within this species. We conclude that large-scale genome structure can be a sensitive indicator of phylogenetic relationships and that, therefore, I-CeuI-based genomic mapping is an efficient tool for probing the phylogenetic status of bacteria.


Subject(s)
Genome, Bacterial , Pasteurella/genetics , Phylogeny , Salmonella/genetics , Base Sequence , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Electrophoresis, Gel, Pulsed-Field , Endodeoxyribonucleases/metabolism , Molecular Sequence Data , Pasteurella/classification , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 23S/genetics , Salmonella/classification
20.
Neurosci Res ; 34(1): 21-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10413323

ABSTRACT

Neurofibrillary tangles (NFT) in Alzheimer's disease (AD) consist largely of hyperphosphorylated tau protein. Many of the phosphorylation sites on tau are serine/threonine-proline sequences, several of which are phosphorylated in vitro by neuronal Cdc2-like kinase (Nclk), a kinase composed of Cdk5 and its activator(s). Thus, tau hyperphosphorylation in AD may result in part from deregulation of Nclk. To test this hypothesis, we examined Nclk activity in prefrontal and cerebellar cortex from 15 postmortem AD and 16 age-matched control subjects, and corrected either for Cdk5 level or for neuronal loss. The ratio of Nclk activity in prefrontal versus cerebellar cortex was then compared. When corrections were made for neuronal loss, the ratios of kinase activity in prefrontal versus cerebellar cortex were significantly higher in AD (6.45+/-0.86) than the controls (3.13+/-0.46; P = 0.003). This finding is consistent with a role for Nclk in the pathogenesis of NFT in AD.


Subject(s)
Alzheimer Disease/enzymology , Cerebellar Cortex/enzymology , Cyclin-Dependent Kinases/metabolism , Neurofibrillary Tangles/enzymology , Prefrontal Cortex/enzymology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Case-Control Studies , Cerebellar Cortex/pathology , Cyclin-Dependent Kinase 5 , Female , Humans , Male , Middle Aged , Neurofibrillary Tangles/pathology , Prefrontal Cortex/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...