Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Vet Pharmacol Ther ; 46(6): 401-412, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37198956

ABSTRACT

The ATP-binding cassette transporter P-glycoprotein (P-gp) limits the oral bioavailability of many drugs. Although P-gp has been well studied in humans and mice, little is known about the substrate specificities of many of its species orthologs. To address this, we performed in vitro analysis of P-gp transporter function using HEK293 cells stably expressing human, ovine, porcine, canine, and feline P-gp. We also employed a human physiologically based pharmacokinetic (PBPK) model to assess variations in digoxin exposure resulting from altered P-gp function. Compared to human P-gp, sheep P-gp had significantly less digoxin efflux (2.3-fold ±0.04 vs. 1.8-fold ±0.03, p < .0001) and all species orthologs had significantly less quinidine efflux compared with human P-gp (p < .05). Human P-gp also had significantly greater efflux of talinolol compared to sheep and dog P-gp (1.9-fold ±0.04 vs. 1.6-fold ±0.06, p = .003 and 1.6-fold ±0.05, p = .0002, respectively). P-gp expression protected all lines against paclitaxel-induced toxicity, with sheep P-gp being significantly less protective. The inhibitor verapamil demonstrated dose-dependent inhibition of all P-gp orthologs. Finally, a PBPK model showed digoxin exposure was sensitive to altered P-gp activity. Overall, our study found that species differences in this major drug transporter exist and that the appropriate species ortholog of P-gp should be evaluated during veterinary drug development.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Digoxin , Humans , Animals , Dogs , Cats , Sheep , Mice , Swine , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , HEK293 Cells , ATP Binding Cassette Transporter, Subfamily B/metabolism , Digoxin/metabolism , Verapamil
2.
PLoS Genet ; 9(4): e1003464, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23637631

ABSTRACT

Gene fusions, like BCR/ABL1 in chronic myelogenous leukemia, have long been recognized in hematologic and mesenchymal malignancies. The recent finding of gene fusions in prostate and lung cancers has motivated the search for pathogenic gene fusions in other malignancies. Here, we developed a "breakpoint analysis" pipeline to discover candidate gene fusions by tell-tale transcript level or genomic DNA copy number transitions occurring within genes. Mining data from 974 diverse cancer samples, we identified 198 candidate fusions involving annotated cancer genes. From these, we validated and further characterized novel gene fusions involving ROS1 tyrosine kinase in angiosarcoma (CEP85L/ROS1), SLC1A2 glutamate transporter in colon cancer (APIP/SLC1A2), RAF1 kinase in pancreatic cancer (ATG7/RAF1) and anaplastic astrocytoma (BCL6/RAF1), EWSR1 in melanoma (EWSR1/CREM), CDK6 kinase in T-cell acute lymphoblastic leukemia (FAM133B/CDK6), and CLTC in breast cancer (CLTC/VMP1). Notably, while these fusions involved known cancer genes, all occurred with novel fusion partners and in previously unreported cancer types. Moreover, several constituted druggable targets (including kinases), with therapeutic implications for their respective malignancies. Lastly, breakpoint analysis identified new cell line models for known rearrangements, including EGFRvIII and FIP1L1/PDGFRA. Taken together, we provide a robust approach for gene fusion discovery, and our results highlight a more widespread role of fusion genes in cancer pathogenesis.


Subject(s)
Gene Fusion , Protein-Tyrosine Kinases , Genomics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics
3.
Proc Natl Acad Sci U S A ; 109(5): E252-9, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22233809

ABSTRACT

Defining the molecular genetic alterations underlying pancreatic cancer may provide unique therapeutic insight for this deadly disease. Toward this goal, we report here an integrative DNA microarray and sequencing-based analysis of pancreatic cancer genomes. Notable among the alterations newly identified, genomic deletions, mutations, and rearrangements recurrently targeted genes encoding components of the SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeling complex, including all three putative DNA binding subunits (ARID1A, ARID1B, and PBRM1) and both enzymatic subunits (SMARCA2 and SMARCA4). Whereas alterations of each individual SWI/SNF subunit occurred at modest-frequency, as mutational "hills" in the genomic landscape, together they affected at least one-third of all pancreatic cancers, defining SWI/SNF as a major mutational "mountain." Consistent with a tumor-suppressive role, re-expression of SMARCA4 in SMARCA4-deficient pancreatic cancer cell lines reduced cell growth and promoted senescence, whereas its overexpression in a SWI/SNF-intact line had no such effect. In addition, expression profiling analyses revealed that SWI/SNF likely antagonizes Polycomb repressive complex 2, implicating this as one possible mechanism of tumor suppression. Our findings reveal SWI/SNF to be a central tumor suppressive complex in pancreatic cancer.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/physiology , Genes, Tumor Suppressor , Pancreatic Neoplasms/metabolism , Transcription Factors/physiology , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Profiling , Humans , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transcription Factors/genetics , Transcriptome
4.
Proc Natl Acad Sci U S A ; 109(46): E3196-205, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23112155

ABSTRACT

The mutational activation of oncogenes drives cancer development and progression. Classic oncogenes, such as MYC and RAS, are active across many different cancer types. In contrast, "lineage-survival" oncogenes represent a distinct and emerging class typically comprising transcriptional regulators of a specific cell lineage that, when deregulated, support the proliferation and survival of cancers derived from that lineage. Here, in a large collection of colorectal cancer cell lines and tumors, we identify recurrent amplification of chromosome 13, an alteration highly restricted to colorectal-derived cancers. A minimal region of amplification on 13q12.2 pinpoints caudal type homeobox transcription factor 2 (CDX2), a regulator of normal intestinal lineage development and differentiation, as a target of the amplification. In contrast to its described role as a colorectal tumor suppressor, CDX2 when amplified is required for the proliferation and survival of colorectal cancer cells. Further, transcriptional profiling, binding-site analysis, and functional studies link CDX2 to Wnt/ß-catenin signaling, itself a key oncogenic pathway in colorectal cancer. These data characterize CDX2 as a lineage-survival oncogene deregulated in colorectal cancer. Our findings challenge a prevailing view that CDX2 is a tumor suppressor in colorectal cancer and uncover an additional piece in the multistep model of colorectal tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Gene Amplification , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , CDX2 Transcription Factor , Cell Line, Tumor , Cell Survival , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 13/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Homeodomain Proteins/genetics , Humans , Mice , NIH 3T3 Cells , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Wnt Signaling Pathway/genetics
5.
BMC Genomics ; 14: 624, 2013 Sep 16.
Article in English | MEDLINE | ID: mdl-24041470

ABSTRACT

BACKGROUND: Pancreatic cancer is a deadly disease with a five-year survival of less than 5%. A better understanding of the underlying biology may suggest novel therapeutic targets. Recent surveys of the pancreatic cancer genome have uncovered numerous new alterations; yet systematic functional characterization of candidate cancer genes has lagged behind. To address this challenge, here we have devised a highly-parallel RNA interference-based functional screen to evaluate many genomically-nominated candidate pancreatic cancer genes simultaneously. RESULTS: For 185 candidate pancreatic cancer genes, selected from recurrently altered genomic loci, we performed a pooled shRNA library screen of cell growth/viability across 10 different cell lines. Knockdown-associated effects on cell growth were assessed by enrichment or depletion of shRNA hairpins, by hybridization to barcode microarrays. A novel analytical approach (COrrelated Phenotypes for On-Target Effects; COPOTE) was used to discern probable on-target knockdown, based on identifying different shRNAs targeting the same gene and displaying concordant phenotypes across cell lines. Knockdown data were integrated with genomic architecture and gene-expression profiles, and selected findings validated using individual shRNAs and/or independent siRNAs. The pooled shRNA library design delivered reproducible data. In all, COPOTE analysis identified 52 probable on-target gene-knockdowns. Knockdown of known oncogenes (KRAS, MYC, SMURF1 and CCNE1) and a tumor suppressor (CDKN2A) showed the expected contrasting effects on cell growth. In addition, the screen corroborated purported roles of PLEKHG2 and MED29 as 19q13 amplicon drivers. Most notably, the analysis also revealed novel possible oncogenic functions of nucleoporin NUP153 (ostensibly by modulating TGFß signaling) and Kruppel-like transcription factor KLF5 in pancreatic cancer. CONCLUSIONS: By integrating physical and functional genomic data, we were able to simultaneously evaluate many candidate pancreatic cancer genes. Our findings uncover new facets of pancreatic cancer biology, with possible therapeutic implications. More broadly, our study provides a general strategy for the efficient characterization of candidate genes emerging from cancer genome studies.


Subject(s)
DNA, Neoplasm/genetics , Genes, Neoplasm , Pancreatic Neoplasms/genetics , Cell Line, Tumor , Comparative Genomic Hybridization , Gene Knockdown Techniques , Gene Library , Genomics , High-Throughput Nucleotide Sequencing , Humans , Kruppel-Like Transcription Factors/genetics , RNA Interference , RNA, Small Interfering/genetics , Sequence Analysis, DNA , Transcriptome
6.
PLoS Genet ; 4(5): e1000081, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18535672

ABSTRACT

Pancreatobiliary cancers have among the highest mortality rates of any cancer type. Discovering the full spectrum of molecular genetic alterations may suggest new avenues for therapy. To catalogue genomic alterations, we carried out array-based genomic profiling of 31 exocrine pancreatic cancers and 6 distal bile duct cancers, expanded as xenografts to enrich the tumor cell fraction. We identified numerous focal DNA amplifications and deletions, including in 19% of pancreatobiliary cases gain at cytoband 18q11.2, a locus uncommonly amplified in other tumor types. The smallest shared amplification at 18q11.2 included GATA6, a transcriptional regulator previously linked to normal pancreas development. When amplified, GATA6 was overexpressed at both the mRNA and protein levels, and strong immunostaining was observed in 25 of 54 (46%) primary pancreatic cancers compared to 0 of 33 normal pancreas specimens surveyed. GATA6 expression in xenografts was associated with specific microarray gene-expression patterns, enriched for GATA binding sites and mitochondrial oxidative phosphorylation activity. siRNA mediated knockdown of GATA6 in pancreatic cancer cell lines with amplification led to reduced cell proliferation, cell cycle progression, and colony formation. Our findings indicate that GATA6 amplification and overexpression contribute to the oncogenic phenotypes of pancreatic cancer cells, and identify GATA6 as a candidate lineage-specific oncogene in pancreatobiliary cancer, with implications for novel treatment strategies.


Subject(s)
Biliary Tract Neoplasms/genetics , GATA6 Transcription Factor/genetics , Gene Amplification , Pancreatic Neoplasms/genetics , Animals , Biliary Tract Neoplasms/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human, Pair 18/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Mice , Pancreatic Neoplasms/metabolism , Transplantation, Heterologous/pathology , Transplantation, Heterologous/veterinary
7.
Cancer Res ; 67(18): 8504-10, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875689

ABSTRACT

Prostate cancer is clinically heterogeneous, ranging from indolent to lethal disease. Expression profiling previously defined three subtypes of prostate cancer, one (subtype-1) linked to clinically favorable behavior, and the others (subtypes-2 and -3) linked with a more aggressive form of the disease. To explore disease heterogeneity at the genomic level, we carried out array-based comparative genomic hybridization (array CGH) on 64 prostate tumor specimens, including 55 primary tumors and 9 pelvic lymph node metastases. Unsupervised cluster analysis of DNA copy number alterations (CNA) identified recurrent aberrations, including a 6q15-deletion group associated with subtype-1 gene expression patterns and decreased tumor recurrence. Supervised analysis further disclosed distinct patterns of CNA among gene-expression subtypes, where subtype-1 tumors exhibited characteristic deletions at 5q21 and 6q15, and subtype-2 cases harbored deletions at 8p21 (NKX3-1) and 21q22 (resulting in TMPRSS2-ERG fusion). Lymph node metastases, predominantly subtype-3, displayed overall higher frequencies of CNA, and in particular gains at 8q24 (MYC) and 16p13, and loss at 10q23 (PTEN) and 16q23. Our findings reveal that prostate cancers develop via a limited number of alternative preferred genetic pathways. The resultant molecular genetic subtypes provide a new framework for investigating prostate cancer biology and explain in part the clinical heterogeneity of the disease.


Subject(s)
Prostatic Neoplasms/genetics , Chromosome Aberrations , Cluster Analysis , DNA, Neoplasm/genetics , Gene Dosage , Gene Expression Profiling , Humans , Male , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/classification , Prostatic Neoplasms/pathology
8.
Am J Surg Pathol ; 32(2): 205-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18223322

ABSTRACT

Growing evidence suggests that only a fraction of prostate cancers detected clinically are potentially lethal. An important clinical issue is identifying men with indolent cancer who might be spared aggressive therapies with associated morbidities. Previously, using microarray analysis we defined 3 molecular subtypes of prostate cancer with different gene-expression patterns. One, subtype-1, displayed features consistent with more indolent behavior, where an immunohistochemical marker (AZGP1) for subtype-1 predicted favorable outcome after radical prostatectomy. Here we characterize a second candidate tissue biomarker, hCAP-D3, expressed in subtype-1 prostate tumors. hCAP-D3 expression, assayed by RNA in situ hybridization on a tissue microarray comprising 225 cases, was associated with decreased tumor recurrence after radical prostatectomy (P=0.004), independent of pathologic tumor stage, Gleason grade, and preoperative prostate-specific antigen levels. Simultaneous assessment of hCAP-D3 and AZGP1 expression in this tumor set improved outcome prediction. We have previously demonstrated that hCAP-D3 is induced by androgen in prostate cells. Extending this finding, Gene Set Enrichment Analysis revealed enrichment of androgen-responsive genes in subtype-1 tumors (P=0.019). Our findings identify hCAP-D3 as a new biomarker for subtype-1 tumors that improves prognostication, and reveal androgen signaling as an important biologic feature of this potentially clinically favorable molecular subtype.


Subject(s)
Adenosine Triphosphatases/genetics , Androgens/metabolism , Biomarkers, Tumor/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/physiology , Multiprotein Complexes/genetics , Prostatic Neoplasms/genetics , Adenosine Triphosphatases/metabolism , Adipokines , Androgens/genetics , Biomarkers, Tumor/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , In Situ Hybridization , Male , Multiprotein Complexes/metabolism , Neoplasm Recurrence, Local , Prognosis , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , RNA, Neoplasm/analysis , Signal Transduction , Survival Rate , Tissue Array Analysis
9.
Cancer Res ; 65(20): 9200-5, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230380

ABSTRACT

Genetic instability plays a central role in the development and progression of human cancer. Two major classes of genetic instability, microsatellite instability (MSI) and chromosome instability (microsatellite stable; MSS), are best understood in the context of colon cancer, where MSI tumors represent approximately 15% of cases, and compared with MSS tumors, more often arise in the proximal colon and display favorable clinical outcome. To further explore molecular differences, we profiled gene expression in a set of 18 colon cancer cell lines using cDNA microarrays representing approximately 21,000 different genes. Supervised analysis identified a robust expression signature distinguishing MSI and MSS samples. As few as eight genes predicted with high accuracy the underlying genetic instability in the original and in three independent sample sets, comprising 13 colon cancer cell lines, 61 colorectal tumors, and 87 gastric tumors. Notably, the MSI signature was retained despite genetically correcting the underlying instability, suggesting the signature reflects a legacy of the tumor having arisen from MSI, rather than sensing the ongoing state of MSI. Our findings support a model in which MSI and MSS preferentially target different genes and pathways in cancer. Further, among the MSI signature genes, our findings implicate a role of elevated metallothionein expression in the clinical behavior of MSI cancers.


Subject(s)
Colonic Neoplasms/genetics , Genomic Instability , Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Cell Line, Tumor , Chromosomal Instability , Colonic Neoplasms/classification , Colonic Neoplasms/metabolism , Gene Expression Profiling , HCT116 Cells , Humans , Microsatellite Repeats/genetics , MutL Protein Homolog 1 , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
10.
Semin Ultrasound CT MR ; 34(3): 236-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23768890

ABSTRACT

Head and neck malignancies, including squamous cell carcinoma, lymphoma, and thyroid cancer, are a major cause of morbidity and mortality worldwide and frequently present with cervical lymphadenopathy. Distinguishing normal from malignant lymph nodes is critical for accurate staging, prognosis, and determination of optimal therapeutic options. Gray-scale, power, and color Doppler ultrasonography offers an inexpensive yet effective method in identifying abnormal cervical lymph nodes. Sonographic nodal features that should be assessed include size, shape, echotexture (including microcalcifications and cystic changes), presence of an echogenic hilus, and vascularity. Although no single sonographic feature can accurately distinguish malignant from normal nodes, a combination of these characteristics can help to make this determination.


Subject(s)
Carcinoma/diagnostic imaging , Carcinoma/secondary , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/secondary , Image Enhancement/methods , Lymph Nodes/diagnostic imaging , Ultrasonography/methods , Lymphatic Metastasis , Neck
11.
Cancer Res ; 72(10): 2657-71, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22419663

ABSTRACT

EGFRvIII is a tumor-specific variant of the epidermal growth factor receptor (EGFR). Although EGFRvIII is most commonly found in glioblastoma, its expression in other tumor types remains controversial. In this study, we investigated EGFRvIII expression and amplification in primary breast carcinoma. Our analyses confirmed the presence of EGFRvIII, but in the absence of amplification or rearrangement of the EGFR locus. Nested reverse transcriptase PCR and flow cytometry were used to detect a higher percentage of positive cases. EGFRvIII-positive cells showed increased expression of genes associated with self-renewal and epithelial-mesenchymal transition along with a higher percentage of stem-like cells. EGFRvIII also increased in vitro sphere formation and in vivo tumor formation. Mechanistically, EGFRvIII mediated its effects through the Wnt/ß-catenin pathway, leading to increased ß-catenin target gene expression. Inhibition of this pathway reversed the observed effects on cancer stem cell (CSC) phenotypes. Together, our findings show that EGFRvIII is expressed in primary breast tumors and contributes to CSC phenotypes in breast cancer cell lines through the Wnt pathway. These data suggest a novel function for EGFRvIII in breast tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , ErbB Receptors/genetics , Neoplastic Stem Cells/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Genes, erbB-1 , Humans , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Phenotype , Wnt Signaling Pathway
12.
Genome Biol ; 13(8): R75, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22929540

ABSTRACT

BACKGROUND: Molecular characterization of tumors has been critical for identifying important genes in cancer biology and for improving tumor classification and diagnosis. Long non-coding RNAs, as a new, relatively unstudied class of transcripts, provide a rich opportunity to identify both functional drivers and cancer-type-specific biomarkers. However, despite the potential importance of long non-coding RNAs to the cancer field, no comprehensive survey of long non-coding RNA expression across various cancers has been reported. RESULTS: We performed a sequencing-based transcriptional survey of both known long non-coding RNAs and novel intergenic transcripts across a panel of 64 archival tumor samples comprising 17 diagnostic subtypes of adenocarcinomas, squamous cell carcinomas and sarcomas. We identified hundreds of transcripts from among the known 1,065 long non-coding RNAs surveyed that showed variability in transcript levels between the tumor types and are therefore potential biomarker candidates. We discovered 1,071 novel intergenic transcribed regions and demonstrate that these show similar patterns of variability between tumor types. We found that many of these differentially expressed cancer transcripts are also expressed in normal tissues. One such novel transcript specifically expressed in breast tissue was further evaluated using RNA in situ hybridization on a panel of breast tumors. It was shown to correlate with low tumor grade and estrogen receptor expression, thereby representing a potentially important new breast cancer biomarker. CONCLUSIONS: This study provides the first large survey of long non-coding RNA expression within a panel of solid cancers and also identifies a number of novel transcribed regions differentially expressed across distinct cancer types that represent candidate biomarkers for future research.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling , RNA, Long Noncoding/genetics , Carcinoma/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans
SELECTION OF CITATIONS
SEARCH DETAIL