Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 11(2): 124, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054850

ABSTRACT

Despite the introduction of novel targeted therapies, chemotherapy still remains the primary treatment for metastatic melanoma in poorly funded healthcare environments or in case of disease relapse, with no reliable molecular markers for progression-free survival (PFS) available. As chemotherapy primarily eliminates cancer cells by apoptosis, we here evaluated if the expression of key apoptosis regulators (Bax, Bak, Bcl-2, Bcl-xL, Smac, Procaspase-9, Apaf-1, Procaspase-3 and XIAP) allows prognosticating PFS in stage III/IV melanoma patients. Following antibody validation, marker expression was determined by automated and manual scoring of immunohistochemically stained tissue microarrays (TMAs) constructed from treatment-naive metastatic melanoma biopsies. Interestingly and counter-intuitively, low expression of the pro-apoptotic proteins Bax, Bak and Smac indicated better prognosis (log-rank p < 0.0001, p = 0.0301 and p = 0.0227 for automated and p = 0.0422, p = 0.0410 and p = 0.0073 for manual scoring). These findings were independently validated in the cancer genome atlas (TCGA) metastatic melanoma cohort (TCGA-SKCM) at transcript level (log-rank p = 0.0004, p = 0.0104 and p = 0.0377). Taking expression heterogeneity between the markers in individual tumour samples into account allowed defining combinatorial Bax, Bak, Smac signatures that were associated with significantly increased PFS (p = 0.0002 and p = 0.0028 at protein and transcript level, respectively). Furthermore, combined low expression of Bax, Bak and Smac allowed predicting prolonged PFS (> 12 months) on a case-by-case basis (area under the receiver operating characteristic curve (ROC AUC) = 0.79). Taken together, our results therefore suggest that Bax, Bak and Smac jointly define a signature with potential clinical utility in chemotherapy-treated metastatic melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/analysis , Biomarkers, Tumor/analysis , Melanoma/drug therapy , Mitochondrial Proteins/analysis , Skin Neoplasms/drug therapy , bcl-2 Homologous Antagonist-Killer Protein/analysis , bcl-2-Associated X Protein/analysis , Aged , Apoptosis Regulatory Proteins/genetics , Biomarkers, Tumor/genetics , Down-Regulation , Female , Gene Expression Profiling , Humans , Image Interpretation, Computer-Assisted , Immunohistochemistry , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma/secondary , Middle Aged , Mitochondrial Proteins/genetics , Pattern Recognition, Automated , Predictive Value of Tests , Progression-Free Survival , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Time Factors , Tissue Array Analysis , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
2.
Nat Med ; 21(8): 955-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26168295

ABSTRACT

Colon cancer prevention currently relies on colonoscopy using white light to detect and remove polyps, but small and flat polyps are difficult to detect and frequently missed when using this technique. Fluorescence colonoscopy combined with a fluorescent probe specific for a polyp biomarker may improve polyp detection. Here we describe GE-137, a water-soluble probe consisting of a 26-amino acid cyclic peptide that binds the human tyrosine kinase c-Met conjugated to a fluorescent cyanine dye. Intravenous administration of GE-137 leads to its accumulation specifically in c-Met-expressing tumors in mice, and it is safe and well tolerated in humans. Fluorescence colonoscopy in patients receiving intravenous GE-137 enabled visualization of all neoplastic polyps that were visible with white light (38), as well as an additional nine polyps that were not visible with white light. This first-in-human pilot study shows that molecular imaging using an intravenous fluorescent agent specific for c-Met is feasible and safe, and that it may enable the detection of polyps missed by other techniques.


Subject(s)
Colorectal Neoplasms/diagnosis , Intestinal Polyps/diagnosis , Peptides, Cyclic/drug effects , Proto-Oncogene Proteins c-met/analysis , Amino Acid Sequence , Animals , Cell Line, Tumor , Female , Fluorescence , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data
3.
PLoS One ; 7(12): e51835, 2012.
Article in English | MEDLINE | ID: mdl-23284784

ABSTRACT

GNAS/Gnas encodes G(s)α that is mainly biallelically expressed but shows imprinted expression in some tissues. In Albright Hereditary Osteodystrophy (AHO) heterozygous loss of function mutations of GNAS can result in ectopic ossification that tends to be superficial and attributable to haploinsufficiency of biallelically expressed G(s)α. Oed-Sml is a point missense mutation in exon 6 of the orthologous mouse locus Gnas. We report here both the late onset ossification and occurrence of benign cutaneous fibroepithelial polyps in Oed-Sml. These phenotypes are seen on both maternal and paternal inheritance of the mutant allele and are therefore due to an effect on biallelically expressed G(s)α. The ossification is confined to subcutaneous tissues and so resembles the ossification observed with AHO. Our mouse model is the first with both subcutaneous ossification and fibroepithelial polyps related to G(s)α deficiency. It is also the first mouse model described with a clinically relevant phenotype associated with a point mutation in G(s)α and may be useful in investigations of the mechanisms of heterotopic bone formation. Together with earlier results, our findings indicate that G(s)α signalling pathways play a vital role in repressing ectopic bone formation.


Subject(s)
Disease Models, Animal , GTP-Binding Protein alpha Subunits, Gs/physiology , Mutation/genetics , Ossification, Heterotopic/etiology , Skin Diseases/etiology , Subcutaneous Tissue/pathology , Animals , Chromogranins , Female , Male , Mice , Mice, Knockout , Ossification, Heterotopic/pathology , Phenotype , Skin Diseases/pathology
4.
Biomark Med ; 5(2): 227-48, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21473728

ABSTRACT

There has been increased interest in the analysis of protein biomarkers in clinical tumor tissues in recent years. Tissue-based biomarker assays can add value and aid decision-making at all stages of drug development, as well as being developed for use as predictive biomarkers and for patient stratification and prognostication in the clinic. However, there must be an awareness of the legal and ethical issues related to the sourcing of human tissue samples. This article also discusses the limits of scope and critical aspects on the successful use of the following tissue-based methods: immunohistochemistry, tissue microarrays and automated image analysis. Future advances in standardization of tissue biobanking methods, immunohistochemistry and quantitative image analysis techniques are also discussed.


Subject(s)
Biomarkers, Tumor/analysis , Neoplasm Proteins/analysis , Tissue and Organ Harvesting/methods , Automation , Clinical Trials as Topic , Diagnostic Imaging , European Union , Humans , Immunohistochemistry , Organ Preservation , Tissue Array Analysis , Tissue and Organ Harvesting/ethics , Tissue and Organ Harvesting/legislation & jurisprudence , United States
5.
Leuk Res ; 34(2): 210-20, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19709744

ABSTRACT

Mutations in the BLM gene cause human Bloom syndrome (BS), an autosomal recessive disorder of growth retardation, immunodeficiency and cancer predisposition. Homozygous null Blm(m3/m3) mice are cancer prone with a 5-fold increased risk of cancer compared with Blm(m3/+) and Blm(+/+) mice. Irradiation of Blm(m3/m3) mice increased the risk to 28-fold. Tumors occurred mainly in the hematopoietic system and were similar to those in BS based on detailed histologic and immunohistochemical analyses. Irradiated Blm-deficient mice thus provide a novel model for understanding accelerated malignancies in BS and a new platform for investigating the molecular basis for a wide range of hematopoietic neoplasms.


Subject(s)
Disease Models, Animal , Hematologic Neoplasms/pathology , RecQ Helicases/deficiency , Animals , Bloom Syndrome/complications , Hematologic Neoplasms/genetics , Mice , Mice, Knockout
6.
Clin Cancer Res ; 15(12): 4138-46, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19509160

ABSTRACT

PURPOSE: To determine the efficacy of AZD0530, an orally active small molecule Src inhibitor, in human pancreatic cancer xenografts and to seek biomarkers predictive of activity. EXPERIMENTAL DESIGN: Sixteen patient-derived pancreatic cancer xenografts from the PancXenoBank collection at Johns Hopkins were treated with AZD0530 (50 mg/kg/day, p.o.) for 28 days. Baseline gene expression profiles of differently expressed genes in 16 tumors by Affymetrix U133 Plus 2.0 gene array were used to predict AZD0530 sensitivity in an independent group of eight tumors using the K-Top Scoring Pairs (K-TSP) method. RESULTS: Three patient tumors of 16 were found to be sensitive to AZD0530, defined as tumor growth <50% compared with control tumors (100%). Western blot and/or immunohistochemistry results showed that AZD0530 administration resulted in the down-regulation of Src, FAK, p-FAK, p-paxillin, p-STAT-3, and XIAP in sensitive tumor xenografts compared with control tumors. The K-TSP classifier identified one gene pair (LRRC19 and IGFBP2) from the 16 training cases based on a decision rule. The classifier achieved 100% and 83.3% of sensitivity and specificity in an independent test set that consists of eight xenograft cases. CONCLUSIONS: AZD0530 treatment significantly inhibits the tumor growth in a subset of human pancreatic tumor xenografts. One gene pair (LRRC19 and IGFBP2) identified by the K-TSP classifier has high predictive power for AZD0530 sensitivity, suggesting the potential for this gene pair as biomarker for pancreatic tumor sensitivity to AZD0530.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodioxoles/pharmacology , Biomarkers, Tumor/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Quinazolines/pharmacology , Animals , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Gene Expression Profiling , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Paxillin/antagonists & inhibitors , Paxillin/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
8.
Proc Natl Acad Sci U S A ; 105(50): 19904-9, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19064922

ABSTRACT

Large-scale cancer genome projects will soon be able to sequence many cancer genomes to comprehensively identify genetic changes in human cancer. Genome-wide association studies have also identified putative cancer associated loci. Functional validation of these genetic mutations in vivo is becoming a challenge. We describe here a DNA transposon-based platform that permits us to explore the oncogenic potential of genetic mutations in the mouse. Briefly, promoter-less human cancer gene cDNAs were first cloned into Sleeping Beauty (SB) transposons. DNA transposition in the mouse that carried both the transposons and the SB transposase made it possible for the cDNAs to be expressed from an appropriate endogenous genomic locus and in the relevant cell types for tumor development. Consequently, these mice developed a broad spectrum of tumors at very early postnatal stages. This technology thus complements the large-scale cancer genome projects.


Subject(s)
DNA Mutational Analysis/methods , DNA Transposable Elements/genetics , DNA, Neoplasm/genetics , Neoplasms/genetics , Oncogenes , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Genetic Complementation Test , Genome, Human , Humans , Mice , Mice, Transgenic , Mutation , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Transposases/genetics
9.
Science ; 316(5824): 608-11, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17463290

ABSTRACT

MicroRNAs are a class of small RNAs that are increasingly being recognized as important regulators of gene expression. Although hundreds of microRNAs are present in the mammalian genome, genetic studies addressing their physiological roles are at an early stage. We have shown that mice deficient for bic/microRNA-155 are immunodeficient and display increased lung airway remodeling. We demonstrate a requirement of bic/microRNA-155 for the function of B and T lymphocytes and dendritic cells. Transcriptome analysis of bic/microRNA-155-deficient CD4+ T cells identified a wide spectrum of microRNA-155-regulated genes, including cytokines, chemokines, and transcription factors. Our work suggests that bic/microRNA-155 plays a key role in the homeostasis and function of the immune system.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Immune System/physiology , MicroRNAs/physiology , T-Lymphocytes/immunology , 3' Untranslated Regions , Animals , Cytokines/biosynthesis , Gene Expression Regulation , Gene Targeting , Homeostasis , Immunoglobulin G/biosynthesis , Lung/pathology , Lung Diseases/immunology , Lung Diseases/pathology , Lymphocyte Activation , Mice , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/physiology , Salmonella Infections, Animal/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Vaccination
10.
Toxicol Pathol ; 35(3): 366-75, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17455084

ABSTRACT

Immunohistochemistry is an indispensable tool in human pathology enabling immunophenotypic characterization of tumor cells. Immunohistochemical analyses of mouse models of human hematopoietic neoplasias have become an important aspect for comparison of murine entities with their human counterparts. The aim of this study was to establish a diagnostic antibody panel for analysis of murine lymphomas/leukemias, useful in formalin-fixed/paraffin-embedded tissue. Overall, 48 antibodies (4 rabbit monoclonal, 12 rabbit polyclonal, 2 goat polyclonal, 11 rat, and 19 mouse monoclonal), which were either mouse-specific (14) or cross-reactive with murine tissue (34) were tested for staining quality and diagnostic value in 468 murine hematopoietic neoplasms. Specific staining was achieved with 29 antibodies, of which 18 were human antibodies cross-reactive with murine tissue. Only 23 (B220, BCL-2, BCL-6, CD117, CD138 (2x), CD3 (2x), CD43, CD45, CD5, CD79 alpha cy, cyclin D1, Ki-67 (2x), Mac-3, Mac-2, lysozyme, mast cell tryptase, MPO, Pax-5, TdT, and TER-119) were regarded as valuable for diagnostic evaluation. Immunohistochemistry was also established in an automated immunostainer for high throughput analysis. The antibody panel developed is useful for the classification of murine lymphomas and leukemias analyzed, and a valuable tool for human and veterinary pathologists involved in the diagnostic interpretation of murine models of hematopoietic neoplasias.


Subject(s)
Aging/immunology , Antibodies/immunology , Cross Reactions , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Immunohistochemistry/methods , Animals , Antibodies/genetics , Female , Fixatives/chemistry , Formaldehyde/chemistry , Hematologic Neoplasms/classification , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Paraffin Embedding
11.
Proc Natl Acad Sci U S A ; 104(11): 4501-5, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360553

ABSTRACT

Genetic mosaics produced by FLP/FRT induced mitotic recombination have been widely used in Drosophila to study gene function in development. Recently, the Cre/loxP system has been applied to induce mitotic recombination in mouse embryonic stem cells and in many adult mouse tissues. We have used this strategy to generate a previously undescribed p53 mouse model in which expression of a ubiquitously expressed recombinase in a heterozygous p53 knockout animal produces mitotic recombinant clones homozygous for the p53 mutation. The induction of loss of heterozygosity in a few cells in an otherwise normal tissue mimics genetic aspects of tumorigenesis more closely than existing models and has revealed the possible cell autonomous nature of Wnt3. Our results suggest that inducible mitotic recombination can be used for clonal analysis of mutants in the mouse.


Subject(s)
Genes, p53 , Mitosis , Recombination, Genetic , Alleles , Animals , DNA Nucleotidyltransferases/genetics , Drosophila , Genotype , Li-Fraumeni Syndrome/genetics , Loss of Heterozygosity , Mice , Models, Genetic , Neoplasms/genetics
12.
Genesis ; 45(1): 51-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17216607

ABSTRACT

SALL4 is a homologue of the Drosophila homeotic gene spalt, a zinc finger transcription factor, required for inner cell mass proliferation in early embryonic development. It also interacts with other transcription factors to control the development of the anorectal region, kidney, heart, limbs, and brain. Truncating mutations in SALL4 cause Okihiro syndrome, manifest as Duane anomaly, radial ray defects and sensorineural and conductive deafness. We report the characterization of a novel murine Sall4 null allele created by bacterial recombineering in ES cells. Homozygous mutant mice exhibit early embryonic lethality. Heterozygous mutant mice recapitulate phenotypic features of Okihiro syndrome including deafness, lower anogenital tract abnormalities, renal hypoplasia, anencephaly, Hirschprung's disease, and skeletal defects. This phenotype shows important differences in cardiac and ear manifestations to previously characterized Sall4 mutant alleles and should prove useful for the investigation of the influence of modifier alleles and protein interactions on the transcriptional regulatory function of Sall4.


Subject(s)
DNA-Binding Proteins/physiology , Organogenesis , Transcription Factors/physiology , Animals , DNA-Binding Proteins/genetics , Duane Retraction Syndrome/embryology , Duane Retraction Syndrome/genetics , Embryonic Development , Mice , Microscopy, Electron, Scanning , Mutation , Transcription Factors/genetics
13.
Hum Mol Genet ; 12(20): 2645-56, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-12928478

ABSTRACT

Heterozygous carriers of mutations in the BRCA2 gene have a high risk of developing breast and other cancers. In these individuals, BRCA2 appears to act as a tumour suppressor gene, in that loss of the wild type allele is frequently observed within tumours, leading to loss of BRCA2 function. Because BRCA2 functions in DNA repair via homologous recombination, this leads to genomic instability. However, it is unclear whether loss of the wild type allele is stochastic or if heterozygosity for BRCA2 mutation carries a phenotype that contributes to tumorigenic progression. Here we demonstrate that, in a specific vertebrate cell type, the chicken B cell line DT40, heterozygosity for a BRCA2 mutation has a distinct phenotype. This is characterized by a reduced growth rate, increased cell death, heightened sensitivity to specific DNA damaging agents and reduced RAD51 focus formation after irradiation. Thus in certain cell types, genome instability might be driven directly by heterozygosity for BRCA2 mutation.


Subject(s)
BRCA2 Protein/genetics , Heterozygote , Mutation , Alleles , Animals , Avian Proteins , B-Lymphocytes/metabolism , Cell Death , Cell Division , Cell Survival , Chickens , DNA Damage , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Genome , Humans , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Models, Genetic , Phenotype , Plasmids/metabolism , Rad51 Recombinase , Recombination, Genetic , Transfection
14.
Hum Mol Genet ; 11(7): 841-51, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11929857

ABSTRACT

Carriers of mutations in the BRCA2 gene have a high risk of developing breast and other cancers. The BRCA2 gene, which is located on human chromosome 13, encodes a very large protein of only poorly understood function. To define regions of sequence conservation and highlight potentially functionally important domains, we have cloned and characterized the chicken BRCA2 gene, the first non-mammalian BRCA2 gene to be described. The gene is organized similarly to the human BRCA2 gene, but is more compact and is localized to the subtelomeric region of chicken chromosome 1q, within a region that contains other genes from human chromosome 13. The chicken BRCA2 gene encodes a protein of 3399 amino acids, which is poorly conserved with mammalian BRCA2 proteins, having only 37% amino acid identity overall with human BRCA2. However, certain domains are much more highly conserved, indicating functional significance. We describe genes with some of these conserved domains in organisms as diverse as intracellular parasites, mosquitoes and plants. The evolutionarily divergent chicken BRCA2 sequence may also be useful in assigning the large number of sequence variants that have been described in the human BRCA2 gene which are of unknown significance in disease causation.


Subject(s)
BRCA2 Protein/genetics , Mutation , Protein Structure, Tertiary , Sequence Analysis, DNA , Amino Acid Sequence , Animals , Avian Proteins , Chickens , Chromosome Mapping , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Phylogeny , Sequence Alignment , Sequence Analysis, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...