Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Int J Oncol ; 52(1): 47-54, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115565

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most common primary malignant brain tumors. Unraveling the molecular and genetic complexity that determines GBM's pronounced migratory property could provide new options for therapeutic targeting that may significantly complement current surgical and chemoradiation therapy and alter the current poor outcome. In this study, we establish stable AJAP1 overexpressing glioma cells in order to examine in vivo tumor growth. We examine AJAP1 localization by confocal microscopy and AJAP1's functional effect on migration and invasion across surfaces coated with laminin. Finally, analysis of AJAP1 expression in murine xenografts and GBM primary tumors revealed its association with tumor growth and survival. Stable overexpression of AJAP1 promotes adherence, decreases invasion of glioma cells through an extracellular-like matrix, and slows migration in the presence of laminin. These observations are reversed by gene knockdown using multiple siRNAs. Additionally, overexpression of AJAP1 decreases colony formation in glioma cells, and leads to smaller tumor growth with increased survival in glioma xenograft mice. Loss of AJAP1 protein expression predicts worse survival in GBM patients. AJAP1 overexpression decreases cell motility in the presence of laminin and decreases tumor growth in xenografts. Its loss of expression predicts worse survival in patients. This study extends our prior observations and implicates AJAP1 as a potential prognostic marker and a viable target for therapeutic intervention in GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion Molecules/biosynthesis , Cell Movement/physiology , Glioblastoma/metabolism , Glioblastoma/pathology , Animals , Apoptosis/physiology , Brain Neoplasms/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Growth Processes/physiology , Cell Membrane/metabolism , Gene Knockdown Techniques , Glioblastoma/genetics , Heterografts , Humans , Mice , Neoplasm Invasiveness , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
2.
Int J Oncol ; 47(5): 1703-10, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26397460

ABSTRACT

The homeobox transcription factor orthodenticle homeobox 2 (OTX2) plays a critical role in very early neurogenesis, but can become oncogenic when aberrantly expressed later in life. We previously discovered its novel oncogenic role in the malignant childhood brain tumor medulloblastoma and hypothesize an oncogenic role in retinoblastoma. Primary retinoblastoma tumors and cell lines were analyzed by quantitative-PCR, immunoblotting and immunohistochemistry for OTX2. The effect of modulating OTX2 expression on tumorigenesis was tested pharmacologically and by siRNA. A lentiviral shRNA-engineered vector was used for conditional knockdown studies on tumor growth in vivo. A luciferase reporter assay was used to analyze ATRA's effect on OTX2's promoter. In this study on retinoblastoma, OTX2 was frequently amplified and/or overexpressed in primary tumors and cell lines. Knockdown of OTX2 expression by siRNA or pharmacologic inhibition by all-trans retinoic acid (ATRA) repressed OTX2 expression and cell proliferation and significantly decreased tumor growth in vivo. Loss of OTX2 expression also resulted in decreased expression of C-MYC and CRX, genes previously implicated in retinoblastoma tumorigenesis. Loss of OTX2 expression increased the phosphorylation of RB, a potential mechanism of modulating cell proliferation. Aberrant expression of OTX2 may contribute to the development of retinoblastoma. OTX2 may serve as a common transcription factor that interlinks multiple tumor-driving pathways. These results also show that OTX2 can be genetically and pharmacologically targeted, providing an exciting new therapeutic option that may be less toxic and more efficacious than current treatments.


Subject(s)
Homeodomain Proteins/genetics , Otx Transcription Factors/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Retinoblastoma/genetics , Retinoblastoma/therapy , Signal Transduction/genetics , Trans-Activators/genetics , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Homeodomain Proteins/biosynthesis , Humans , Otx Transcription Factors/genetics , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/biosynthesis , Retinoblastoma/pathology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Trans-Activators/biosynthesis , Tretinoin/administration & dosage
3.
CNS Neurosci Ther ; 20(5): 429-37, 2014 May.
Article in English | MEDLINE | ID: mdl-24483339

ABSTRACT

AIMS: Down-regulation of AJAP1 in glioblastoma multiforme (GBM) has been reported. However, the expression profiles of AJAP1 in gliomas and the underlying mechanisms of AJAP1 function on invasion are still poorly understood. METHODS: The gene profiles of AJAP1 in glioma patients were studied among four independent cohorts. Confocal imaging was used to analyze the AJAP1 localization. After AJAP1 overexpression in GBM cell lines, cellular polarity, cytoskeleton distribution, and antitumor effect were investigated in vitro and in vivo. RESULTS: AJAP1 expression was significantly decreased in gliomas compared with normal brain in REMBRANDT and CGCA cohorts. Additionally, low AJAP1 expression was associated with worse survival in GBMs in REMBRANDT and TCGA U133A cohorts and was significantly associated with classical and mesenchymal subtypes of GBMs among four cohorts. Confocal imaging indicated AJAP1 localized in cell membranes in low-grade gliomas and AJAP1-overexpressing GBM cells, but difficult to assess in high-grade gliomas due to its absence. AJAP1 overexpression altered the cytoskeleton and cellular polarity in vitro and inhibited the tumor growth in vivo. CONCLUSIONS: AJAP1 is dysregulated at an early stage of gliomagenesis and may suppress glioma cell invasion and proliferation, which suggests that AJAP1 may be a potential diagnostic and prognostic marker for gliomas.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Cell Adhesion Molecules/metabolism , Cytoskeleton/metabolism , Glioma/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cohort Studies , Cytoskeleton/ultrastructure , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Humans , Kaplan-Meier Estimate , Mice, Nude , Microscopy, Confocal , Neoplasm Staging , Neoplasm Transplantation
4.
Int J Oncol ; 44(4): 1243-51, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24481586

ABSTRACT

Previous studies identified the frequent loss of adherens junction-associated protein 1 (AJAP1) expression in glioblastoma (GBM) and its correlation with worse survival. AJAP1 may suppress glioma cell migration, which plays an important role in tumor progression in malignant gliomas such as GBM. However, the role of AJAP1 in cell cycle arrest or apoptosis and resistance to chemotherapy remains unclear. Based on microarray screening results, quantitative PCR and luciferase plasmid reporter constructs were used to evaluate the possible regulatory role of AJAP1 on MAGEA2 expression and function. Cell death assays, TUNEL and other markers of apoptosis were utilized to detect cell apoptosis. Restoration of AJAP1 expression in glioma cells was analyzed after temozolomide exposure. AJAP1 suppressed the expression of MAGEA2 and inhibited the transcriptional activity of MAGEA2 in glioma cells. As AJAP1 expression decreased MAGEA2 protein expression apoptosis increased moderately. Consistent with increased cell death, the induced loss of MAGEA2 expression correlated with increased caspase 3/7 activity, BCL2/BAX ratio and TUNEL signal. AJAP1 expression enhanced cell death in the presence of temozolomide. This study suggests AJAP1 may also function as a pro-apoptotic factor and potentiate cell death by temozolomide in glioma cells. This effect may be partially explained by AJAP1-mediated gene regulation of MAGEA2.


Subject(s)
Apoptosis/genetics , Brain Neoplasms/pathology , Cell Adhesion Molecules/genetics , Glioblastoma/pathology , Melanoma-Specific Antigens/biosynthesis , Neoplasm Proteins/biosynthesis , Adherens Junctions , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Movement , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , HEK293 Cells , Histone Deacetylases/metabolism , Humans , In Situ Nick-End Labeling , Temozolomide , Transcription, Genetic , Tumor Suppressor Protein p53/biosynthesis , bcl-2-Associated X Protein/biosynthesis
5.
Neurosurg Focus ; 32(4): E8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22463118

ABSTRACT

Intracerebral hemorrhage (ICH) is a subtype of stoke that may cause significant morbidity and mortality. Brain injury due to ICH initially occurs within the first few hours as a result of mass effect due to hematoma formation. However, there is increasing interest in the mechanisms of secondary brain injury as many patients continue to deteriorate clinically despite no signs of rehemorrhage or hematoma expansion. This continued insult after primary hemorrhage is believed to be mediated by the cytotoxic, excitotoxic, oxidative, and inflammatory effects of intraparenchymal blood. The main factors responsible for this injury are thrombin and erythrocyte contents such as hemoglobin. Therapies including thrombin inhibitors, N-methyl-D-aspartate antagonists, chelators to bind free iron, and antiinflammatory drugs are currently under investigation for reducing this secondary brain injury. This review will discuss the molecular mechanisms of brain injury as a result of intraparenchymal blood, potential targets for therapeutic intervention, and treatment strategies currently in development.


Subject(s)
Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Hemin/physiology , Stroke/metabolism , Stroke/pathology , Thrombin/physiology , Antithrombins/therapeutic use , Cerebral Hemorrhage/complications , Hemin/antagonists & inhibitors , Hemin/metabolism , Humans , Iron Chelating Agents/therapeutic use , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/physiopathology , Signal Transduction/drug effects , Signal Transduction/physiology , Stroke/etiology , Thrombin/antagonists & inhibitors , Thrombin/metabolism
6.
Mol Cancer Res ; 10(2): 208-17, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22241217

ABSTRACT

Glioblastoma is universally fatal because of its propensity for rapid recurrence due to highly migratory tumor cells. Unraveling the genomic complexity that underlies this migratory characteristic could provide therapeutic targets that would greatly complement current surgical therapy. Using multiple high-resolution genomic screening methods, we identified a single locus, adherens junctional associated protein 1 (AJAP1) on chromosome 1p36 that is lost or epigenetically silenced in many glioblastomas. We found AJAP1 expression absent or reduced in 86% and 100% of primary glioblastoma tumors and cell lines, respectively, and the loss of expression correlates with AJAP1 methylation. Restoration of AJAP1 gene expression by transfection or demethylation agents results in decreased tumor cell migration in glioblastoma cell lines. This work shows the significant loss of expression of AJAP1 in glioblastoma and provides evidence of its role in the highly migratory characteristic of these tumors.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Adhesion Molecules/genetics , Cell Movement/genetics , Epigenesis, Genetic , Glioblastoma/genetics , Glioblastoma/pathology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Proliferation , DNA Methylation/drug effects , Decitabine , Gene Expression Regulation, Neoplastic , Humans , RNA, Small Interfering
7.
Anticancer Agents Med Chem ; 10(7): 543-55, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20950259

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most common and most aggressive types of primary brain tumors in humans. Even with aggressive surgical resections using state of the art preoperative and intraoperative neuroim-aging, along with the most recent techniques in radiotherapy and chemotherapy, the prognosis for GBM patients remains dismal. Survival after diagnosis is about 12-14 months. The tumor cells which already have migrated into normal brain tissue beyond the surgical resection margin account for the inability to effectively treat this tumor. Understanding how to control the migration of GBM cells is paramount to future therapies. In this review, we will focus on the emerging targets and agents which are being exploited to inhibit the migration of glioma cells in GBM.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain/pathology , Glioblastoma/drug therapy , Glioblastoma/pathology , Molecular Targeted Therapy , Antineoplastic Agents/pharmacology , Brain Neoplasms/therapy , Cell Movement , Combined Modality Therapy , Cytokines/metabolism , Female , Glioblastoma/therapy , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Male , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology , Prognosis , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction
8.
Neuro Oncol ; 12(9): 956-66, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20472715

ABSTRACT

Glioblastoma multiforme (GBM) is one of the deadliest tumors afflicting humans, and the mechanisms of its onset and progression remain largely undefined. Our attempts to elucidate its molecular pathogenesis through DNA copy-number analysis by genome-wide digital karyotyping and single nucleotide polymorphism arrays identified a dramatic focal amplification on chromosome 1q32 in 4 of 57 GBM tumors. Quantitative real-time PCR measurements revealed that HDMX is the most commonly amplified and overexpressed gene in the 1q32 locus. Further genetic screening of 284 low- and high-grade gliomas revealed that HDMX amplifications occur solely in pediatric and adult GBMs and that they are mutually exclusive of TP53 mutations and MDM2 amplifications. Here, we demonstrate that HDMX regulates p53 to promote GBM growth and attenuates tumor response to chemotherapy. In GBM cells, HDMX overexpression inhibits p53-mediated transcriptional activation of p21, releases cells from G0 to G1 phase, and enhances cellular proliferation. HDMX overexpression does not affect the expression of PUMA and BAX proapoptotic genes. While in GBM cells treated with the chemotherapeutic agent 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), HDMX appears to stabilize p53 and promote phosphorylation of the DNA double-stranded break repair protein H2AX, up-regulate the DNA repair gene VPX, stimulate DNA repair, and confer resistance to BCNU. In summary, HDMX exhibits bona fide oncogenic properties and offers a promising molecular target for GBM therapeutic intervention.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation/genetics , Glioblastoma/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Carmustine/pharmacology , Cell Cycle Proteins , Child , Drug Resistance, Neoplasm/genetics , Gene Amplification , Gene Expression , Genome-Wide Association Study , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Immunoblotting , In Situ Hybridization, Fluorescence , Mice , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins/genetics , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
9.
Cancer Res ; 70(1): 181-91, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028867

ABSTRACT

OTX2 is a developmentally regulated transcription factor involved in early morphogenesis of the central nervous system. This gene is amplified and overexpressed in medulloblastoma cell lines, but the nature and extent of its genetic alterations in primary tumors have not been evaluated. Analysis of a large cohort of primary medulloblastomas revealed frequent focal copy number gain of a region minimally containing OTX2 as a single gene. OTX2 copy number gain was restricted to tumor subtypes that did not express a molecular signature of Wnt or Shh pathway activation. FISH analysis revealed copy number gain in a subset of cells within medulloblastoma samples, suggesting a late event in tumor progression. Gain of OTX2 copy number was associated with the presence of anaplastic histologic features and shorter survival in medulloblastoma patients. In support of a functional role, ectopic OTX2 expression enhanced proliferation and tumorigenicity of immortalized primary cells, whereas OTX2 knockdown in medulloblastoma cells prolonged the survival of animals bearing xenograft tumors. Mechanistic investigations revealed upregulation of MYC as a potential mechanism whereby OTX2 promotes tumor progression. Our findings define OTX2 as an important oncogenic driver in medulloblastoma.


Subject(s)
Cerebellar Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Medulloblastoma/genetics , Otx Transcription Factors/genetics , Animals , Blotting, Western , Cerebellar Neoplasms/metabolism , Disease Progression , Gene Dosage , Genes, myc/genetics , Hedgehog Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Medulloblastoma/metabolism , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Polymorphism, Single Nucleotide
10.
Pharmgenomics Pers Med ; 3: 111-27, 2010.
Article in English | MEDLINE | ID: mdl-23226047

ABSTRACT

Glioblastoma multiforme (GBM) remains one of the most malignant primary central nervous system tumors. Personalized therapeutic approaches have not become standard of care for GBM, but science is fast approaching this goal. GBM's heterogeneous genomic landscape and resistance to radiotherapy and chemotherapy make this tumor one of the most challenging to treat. Recent advances in genome-wide studies and genetic profiling show that there is unlikely to be a single genetic or cellular event that can be effectively targeted in all patients. Instead, future therapies will likely require personalization for each patient's tumor genotype or proteomic profile. Over the past year, many investigations specifically focused simultaneously on strategies to target oncogenic pathways, angiogenesis, tumor immunology, epigenomic events, glioma stem cells (GSCs), and the highly migratory glioma cell population. Combination therapy targeting multiple pathways is becoming a fast growing area of research, and many studies put special attention on small molecule inhibitors. Because GBM is a highly vascular tumor, therapy that directs monoclonal antibodies or small molecule tyrosine kinase inhibitors toward angiogenic factors is also an area of focus for the development of new therapies. Passive, active, and adoptive immunotherapies have been explored by many studies recently, and epigenetic regulation of gene expression with microRNAs is also becoming an important area of study. GSCs can be useful targets to stop tumor recurrence and proliferation, and recent research has found key molecules that regulate GBM cell migration that can be targeted by therapy. Current standard of care for GBM remains nonspecific; however, pharmacogenomic studies are underway to pave the way for patient-specific therapies that are based on the unique aberrant pathways in individual patients. In conclusion, recent studies in GBM have found many diverse molecular targets possible for therapy. The next obstacle in treating this fatal tumor is ascertaining which molecules in each patient should be targeted and how best to target them, so that we can move our current nonspecific therapies toward the realm of personalized medicine.

11.
Cancer Res ; 69(23): 9105-11, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19920201

ABSTRACT

Glioblastoma multiforme is the most prevalent type of adult brain tumor and one of the deadliest tumors known to mankind. The genetic understanding of glioblastoma multiforme is, however, limited, and the molecular mechanisms that facilitate glioblastoma multiforme cell survival and growth within the tumor microenvironment are largely unknown. We applied digital karyotyping and single nucleotide polymorphism arrays to screen for copy-number changes in glioblastoma multiforme samples and found that the most frequently amplified region is at chromosome 7p11.2. The high resolution of digital karyotyping and single nucleotide polymorphism arrays permits the precise delineation of amplicon boundaries and has enabled identification of the minimal region of amplification at chromosome 7p11.2, which contains two genes, EGFR and SEC61gamma. SEC61gamma encodes a subunit of a heterotrimeric protein channel located in the endoplasmic reticulum (ER). In addition to its high frequency of gene amplification in glioblastoma multiforme, SEC61gamma is also remarkably overexpressed in 77% of glioblastoma multiforme but not in lower-grade gliomas. The small interfering RNA-mediated knockdown of SEC61gamma expression in tumor cells led to growth suppression and apoptosis. Furthermore, we showed that pharmacologic ER stress agents induce SEC61gamma expression in glioblastoma multiforme cells. Together, these results indicate that aberrant expression of SEC61gamma serves significant roles in glioblastoma multiforme cell survival likely via a mechanism that is involved in the cytoprotective ER stress-adaptive response to the tumor microenvironment.


Subject(s)
Brain Neoplasms/genetics , Endoplasmic Reticulum/genetics , Glioblastoma/genetics , Membrane Proteins/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Growth Processes/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gene Amplification , Genes, erbB-1 , Glioblastoma/metabolism , Glioblastoma/pathology , HeLa Cells , Humans , Membrane Proteins/biosynthesis , Proto-Oncogene Mas , RNA, Small Interfering/genetics , SEC Translocation Channels , Tunicamycin/pharmacology
12.
Clin Med Oncol ; 3: 39-52, 2009 Apr 08.
Article in English | MEDLINE | ID: mdl-19777070

ABSTRACT

In the adult population, glioblastoma multiforme is one of the most common primary brain tumors encountered. Unfortunately, this highly malignant tumor represents over 50% of all types of primary central nervous system gliomas. The vast majority of GBMs develops quite rapidly without clinical, radiological, or morphologic evidence of a less malignant precursor lesion (primary or de novo GBMs), as compared to secondary GBMs that develop slowly by progression from diffuse low-grade astrocytomas. These GBM subtypes must be kept in mind because they may constitute distinct disease entities. Even though they look histologically quite similar, they likely involve different genetic alterations and signaling pathways. Decades of surgical therapy, radiotherapy, and chemotherapy have failed to drastically change survival. Clearly, we do not fully understand this tumor; however, the exciting genetic revolution in glioma research over the past decade is providing a promising outlook for exploring this tumor at the genetic level. Science has begun to elucidate the numerous genetic alterations and critical signaling pathways, and it has opened new exciting areas of research such as glioma stem cell biology and neoangiogenesis. This work has already begun to improve our understanding of GBM cell proliferation, migration, and invasion. Indeed, exciting novel targeted therapies are making their way to clinical trials based on this increased knowledge. This review provides the current understanding of GBM oncogenomics, signaling pathways, and glioma stem cell biology and discusses the potential new therapeutic targets on the horizon.

13.
Expert Opin Investig Drugs ; 18(8): 1061-83, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19555299

ABSTRACT

Malignant gliomas such as glioblastoma multiforme (GBM) present some of the greatest challenges in the management of cancer patients worldwide, despite notable recent achievements in oncology. Even with aggressive surgical resections using state-of-the-art preoperative and intraoperative neuroimaging, along with recent advances in radiotherapy and chemotherapy, the prognosis for GBM patients remains dismal: median survival after diagnosis is about 14 months. Established good prognostic factors are limited, but include young age, high Karnofsky Performance Status (KPS), high mini-mental status examination score, O6-methylguanine methyltransferase promoter methylation, and resection of > 98% of the tumor. Standard treatment includes resection, followed by concurrent chemotherapy and radiotherapy. GBM research is being conducted worldwide at a remarkable pace, with some of the more recent promising studies focused on identification of aberrant genetic events and signaling pathways, tumor stem cell identification and characterization, modulation of tumor immunological responses, combination therapies, and understanding of the rare long-term survivors. Past treatment strategies have failed for various reasons; however, newer strategies in trials today and on the horizon encourage optimism. To help illustrate 'where we have been' with this fatal disease and 'where we are going' with contemporary studies, we include in this review a detailed history of Phase III clinical trials for GBM, with a final emphasis on exciting new treatment strategies that offer hope for future GBM therapy.


Subject(s)
Central Nervous System Neoplasms/therapy , Glioblastoma/therapy , Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/epidemiology , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Clinical Trials, Phase III as Topic , Combined Modality Therapy , Glioblastoma/epidemiology , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Neurosurgical Procedures , Radiotherapy , Treatment Outcome
14.
BMC Med Genomics ; 2: 22, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19426505

ABSTRACT

BACKGROUND: Microorganisms have been associated with many types of human diseases; however, a significant number of clinically important microbial pathogens remain to be discovered. METHODS: We have developed a genome-wide approach, called Digital Karyotyping Microbe Identification (DK-MICROBE), to identify genomic DNA of bacteria and viruses in human disease tissues. This method involves the generation of an experimental DNA tag library through Digital Karyotyping (DK) followed by analysis of the tag sequences for the presence of microbial DNA content using a compiled microbial DNA virtual tag library. RESULTS: To validate this technology and to identify pathogens that may be associated with human cancer pathogenesis, we used DK-MICROBE to determine the presence of microbial DNA in 58 human tumor samples, including brain, ovarian, and colorectal cancers. We detected DNA from Human herpesvirus 6 (HHV-6) in a DK library of a colorectal cancer liver metastasis and in normal tissue from the same patient. CONCLUSION: DK-MICROBE can identify previously unknown infectious agents in human tumors, and is now available for further applications for the identification of pathogen DNA in human cancer and other diseases.

15.
Expert Opin Ther Targets ; 13(6): 701-18, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19409033

ABSTRACT

Glioblastoma is the commonest primary brain tumor, as well as the deadliest. Malignant gliomas such as glioblastoma multiforme (GBM) present some of the greatest challenges in the management of cancer patients worldwide, despite notable recent achievements in oncology. Even with aggressive surgical resections using state-of-the-art preoperative and intraoperative neuroimaging, along with recent advances in radiotherapy and chemotherapy, the prognosis for GBM patients remains dismal: survival after diagnosis is about 1 year. Established prognostic factors are limited, but include age, Karnofsky performance status, mini-mental status examination score, O6-methylguanine methyltransferase promoter methylation and extent of surgery. Standard treatment includes resection of > 95% of the tumor, followed by concurrent chemotherapy and radiotherapy. Nevertheless, GBM research is being conducted worldwide at a remarkable pace, in the laboratory and at the bedside, with some of the more recent promising studies focused on identification of aberrant genetic events and signaling pathways to develop molecular-based targeted therapies, tumor stem cell identification and characterization, modulation of tumor immunological responses and understanding of the rare long-term survivors. With this universally fatal disease, any small breakthrough will have a significant impact on survival and provide hope to the thousands of patients who receive this diagnosis annually. This review describes the epidemiology, clinical presentation, pathology and tumor immunology, with a focus on understanding the molecular biology that underlies the current targeted therapeutics being tested.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Combined Modality Therapy , Glioblastoma/diagnosis , Glioblastoma/genetics , Humans , Prognosis , Risk Factors
16.
Mol Ther ; 16(8): 1366-71, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18560415

ABSTRACT

Glycogen storage disease type II (Pompe disease; MIM 232300) stems from the deficiency of acid alpha-glucosidase (GAA; acid maltase; EC 3.2.1.20), which primarily involves cardiac and skeletal muscles. An adeno-associated virus 2/8 (AAV2/8) vector containing the muscle creatine kinase (MCK) (CK1) reduced glycogen content by approximately 50% in the heart and quadriceps in GAA-knockout (GAA-KO) mice; furthermore, an AAV2/8 vector containing the hybrid alpha-myosin heavy chain enhancer-/MCK enhancer-promoter (MHCK7) cassette reduced glycogen content by >95% in heart and >75% in the diaphragm and quadriceps. Transduction with an AAV2/8 vector was higher in the quadriceps than in the gastrocnemius. An AAV2/9 vector containing the MHCK7 cassette corrected GAA deficiency in the distal hindlimb, and glycogen accumulations were substantially cleared by human GAA (hGAA) expression therein; however, the analogous AAV2/7 vector achieved much lower efficacy. Administration of the MHCK7-containing vectors significantly increased striated muscle function as assessed by increased Rotarod times at 18 weeks after injection, whereas the CK1-containing vector did not increase Rotarod performance. Importantly, type IIb myofibers in the extensor digitalis longus (EDL) were transduced, thereby correcting a myofiber type that is unresponsive to enzyme replacement therapy. In summary, AAV8 and AAV9-pseudotyped vectors containing the MHCK7 regulatory cassette achieved enhanced efficacy in Pompe disease mice.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Glycogen Storage Disease Type II/therapy , Muscle, Striated/metabolism , Animals , Creatine Kinase, MM Form/genetics , Creatine Kinase, MM Form/metabolism , Enhancer Elements, Genetic/genetics , Female , Genetic Vectors/genetics , Glycogen/metabolism , Glycogen Storage Disease Type II/genetics , Hindlimb/metabolism , Hindlimb/pathology , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Striated/enzymology , Muscle, Striated/pathology , Myocardium/metabolism , Myosin Heavy Chains/genetics , Promoter Regions, Genetic/genetics , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , Transduction, Genetic , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
17.
Blood ; 108(8): 2726-35, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16763212

ABSTRACT

SALL4, a human homolog to Drosophila spalt, is a novel zinc finger transcriptional factor essential for development. We cloned SALL4 and its isoforms (SALL4A and SALL4B). Through immunohistochemistry and real-time reverse-transcription-polymerase chain reaction (RT-PCR), we demonstrated that SALL4 was constitutively expressed in human primary acute myeloid leukemia (AML, n = 81), and directly tested the leukemogenic potential of constitutive expression of SALL4 in a murine model. SALL4B transgenic mice developed myelodysplastic syndrome (MDS)-like features and subsequently AML that was transplantable. Increased apoptosis associated with dysmyelopoiesis was evident in transgenic mouse marrow and colony-formation (CFU) assays. Both isoforms could bind to beta-catenin and synergistically enhanced the Wnt/beta-catenin signaling pathway. Our data suggest that the constitutive expression of SALL4 causes MDS/AML, most likely through the Wnt/beta-catenin pathway. Our murine model provides a useful platform to study human MDS/AML transformation, as well as the Wnt/beta-catenin pathway's role in the pathogenesis of leukemia stem cells.


Subject(s)
DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Oncogenes , Transcription Factors/genetics , Alternative Splicing , Animals , Apoptosis , Base Sequence , Cloning, Molecular , Colony-Forming Units Assay , DNA, Complementary/genetics , DNA, Neoplasm/genetics , DNA-Binding Proteins/metabolism , Gene Expression , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Neoplasm Transplantation , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Signal Transduction , Transcription Factors/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
18.
J Biol Chem ; 281(28): 18918-26, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16670092

ABSTRACT

SALL1 is a member of the SAL gene family that encodes a group of putative developmental transcription factors. SALL1 plays a critical role during kidney development as mutations of the human SALL1 gene cause Townes-Brocks syndrome, which is associated with kidney malformation. Deletion of the mouse Sall1 gene results in renal agenesis or severe dysgenesis. To date, little is known about the molecular mechanisms controlling the regulation of SALL1 expression. This report describes the cloning and characterization of the human SALL1 gene promoter. Consensus binding sites were identified for several transcription factors, with multiple sites for WT1 and SIX1. In transient transfection assays, SALL1 promoter activity was higher in HEK-293 human kidney cells and COS-7 monkey kidney cells than in NIH-3T3 fibroblasts, consistent with its role in kidney development. Transcription from the SALL1 promoter was strikingly activated by the SIX1 protein. Utilizing a luciferase reporter gene assay, endogenous or exogenously added SIX1 activated the SALL1 promoter. Overexpression of SIX1 induced a significant increase in the endogenous SIX1 protein. In addition, co-expression of SIX1 and Eya1 resulted in a significant increase in the SALL1 promoter activity when compared with either SIX1 or Eya1 alone. Finally, we demonstrate that SIX1 was able to bind to the SALL1 promoter by retardation assays and that deletion of the putative element of SIX1 significantly diminishes the SALL1 promoter activity response to SIX1 stimulation. Our findings, when taken together, indicate that SALL1 is a likely target gene for SIX1 during kidney development.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Kidney/embryology , Transcription Factors/biosynthesis , Transcriptional Activation , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Homeodomain Proteins/genetics , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Structure, Tertiary , Sequence Homology, Nucleic Acid , Transcription Factors/genetics
19.
Life Sci ; 78(6): 614-21, 2006 Jan 02.
Article in English | MEDLINE | ID: mdl-16137713

ABSTRACT

Chemokine-like factor 1 (CKLF1) exhibits chemotactic effects on leukocytes. Its amino acid sequence shares similarity with those of TARC/CCL17 and MDC/CCL22, the cognate ligands for CCR4. The chemotactic effects of CKLF1 for CCR4-transfected cells could be desensitized by TARC/CCL17 and markedly inhibited by PTX. CKLF1 induced a calcium flux in CCR4-transfected cells and fully desensitized a subsequent response to TARC/CCL17, and TARC/CCL17 could partly desensitize the response to CKLF1. CKLF1 caused significant receptor internalization in pCCR4-EGFP transfected cells. Taken together, CKLF1 is a novel functional ligand for CCR4.


Subject(s)
Chemokines/pharmacology , Receptors, Chemokine/metabolism , Calcium/metabolism , Cells, Cultured , Chemokine CCL17 , Chemokines, CC/pharmacology , Chemotaxis , Humans , Ligands , MARVEL Domain-Containing Proteins , Receptors, CCR4 , Recombinant Proteins/pharmacology , Transfection
20.
Cancer Res ; 65(3): 919-24, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15705891

ABSTRACT

Through digital karyotyping of permanent medulloblastoma cell lines, we found that the homeobox gene OTX2 was amplified more than 10-fold in three cell lines. Gene expression analyses showed that OTX2 transcripts were present at high levels in 14 of 15 (93%) medulloblastomas with anaplastic histopathologic features. Knockdown of OTX2 expression by siRNAs inhibited medulloblastoma cell growth in vitro, whereas pharmacologic doses of all-trans retinoic acid repressed OTX2 expression and induced apoptosis only in medulloblastoma cell lines that expressed OTX2. These observations suggest that OTX2 is essential for the pathogenesis of anaplastic medulloblastomas and that these tumors may be amenable to therapy with all-trans-retinoic acid.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Homeodomain Proteins/genetics , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Nerve Tissue Proteins/genetics , Trans-Activators/genetics , Tretinoin/pharmacology , Brain Neoplasms/metabolism , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Gene Amplification , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/biosynthesis , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Oncogenes/drug effects , Oncogenes/genetics , Otx Transcription Factors , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Trans-Activators/antagonists & inhibitors , Trans-Activators/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL