Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Commun Biol ; 3(1): 678, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33188283

ABSTRACT

There is a life-long relationship between rhinovirus (RV) infection and the development and clinical manifestations of asthma. In this study we demonstrate that cultured primary bronchial epithelial cells from adults with asthma (n = 9) show different transcriptional and chromatin responses to RV infection compared to those without asthma (n = 9). Both the number and magnitude of transcriptional and chromatin responses to RV were muted in cells from asthma cases compared to controls. Pathway analysis of the transcriptionally responsive genes revealed enrichments of apoptotic pathways in controls but inflammatory pathways in asthma cases. Using promoter capture Hi-C we tethered regions of RV-responsive chromatin to RV-responsive genes and showed enrichment of these regions and genes at asthma GWAS loci. Taken together, our studies indicate a delayed or prolonged inflammatory state in cells from asthma cases and highlight genes that may contribute to genetic risk for asthma.


Subject(s)
Asthma/metabolism , Chromatin/metabolism , Epithelial Cells/physiology , Respiratory Mucosa/cytology , Rhinovirus/physiology , Adult , Asthma/genetics , Cells, Cultured , Humans , Transcription, Genetic
2.
Nat Commun ; 11(1): 2607, 2020 05 25.
Article in English | MEDLINE | ID: mdl-32451375

ABSTRACT

Quantification of pathogen and host biomarkers is essential for the diagnosis, monitoring, and treatment of infectious diseases. Here, we demonstrate sensitive and rapid quantification of bacterial load and cytokines from human biological samples to generate actionable hypotheses. Our digital assay measures IL-6 and TNF-α proteins, gram-negative (GN) and gram-positive (GP) bacterial DNA, and the antibiotic-resistance gene blaTEM with femtomolar sensitivity. We use our method to characterize bronchoalveolar lavage fluid from patients with asthma, and find elevated GN bacteria and IL-6 levels compared to healthy subjects. We then analyze plasma from patients with septic shock and find that increasing levels of IL-6 and blaTEM are associated with mortality, while decreasing IL-6 levels are associated with recovery. Surprisingly, lower GN bacteria levels are associated with higher probability of death. Applying decision-tree analysis to our measurements, we are able to predict mortality and rate of recovery from septic shock with over 90% accuracy.


Subject(s)
Cytokines/blood , DNA, Bacterial/blood , Shock, Septic/immunology , Shock, Septic/microbiology , Asthma/immunology , Asthma/microbiology , Bacterial Load , Biomarkers/analysis , Biomarkers/blood , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Case-Control Studies , Cytokines/analysis , DNA, Bacterial/genetics , Decision Trees , Genes, Bacterial , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/isolation & purification , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/isolation & purification , Host Microbial Interactions/immunology , Humans , Interleukin-6/analysis , Interleukin-6/blood , Multiplex Polymerase Chain Reaction/methods , Multiplex Polymerase Chain Reaction/statistics & numerical data , Prognosis , Sensitivity and Specificity , Shock, Septic/mortality , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/blood , beta-Lactam Resistance/genetics
3.
J Allergy Clin Immunol ; 144(5): 1214-1227.e7, 2019 11.
Article in English | MEDLINE | ID: mdl-31279011

ABSTRACT

BACKGROUND: The relationship between asthma, atopy, and underlying type 2 (T2) airway inflammation is complex. Although the bacterial airway microbiota is known to differ in asthmatic patients, the fungal and bacterial markers that discriminate T2-high (eosinophilic) and T2-low (neutrophilic/mixed-inflammation) asthma and atopy are still incompletely identified. OBJECTIVES: The aim of this study was to demonstrate the fungal microbiota structure of airways in asthmatic patients associated with T2 inflammation, atopy, and key clinical parameters. METHODS: We collected endobronchial brush (EB) and bronchoalveolar lavage (BAL) samples from 39 asthmatic patients and 19 healthy subjects followed by 16S gene and internal transcribed spacer-based microbiota sequencing. The microbial sequences were classified into exact sequence variants. The T2 phenotype was defined by using a blood eosinophil count with a threshold of 300 cells/µL. RESULTS: Fungal diversity was significantly lower in EB samples from patients with T2-high compared with T2-low inflammation; key fungal genera enriched in patients with T2-high inflammation included Trichoderma species, whereas Penicillium species was enriched in patients with atopy. In BAL fluid samples the dominant genera were Cladosporium, Fusarium, Aspergillus, and Alternaria. Using generalized linear models, we identified significant associations between specific fungal exact sequence variants and FEV1, fraction of exhaled nitric oxide values, BAL fluid cell counts, and corticosteroid use. Investigation of interkingdom (bacterial-fungal) co-occurrence patterns revealed different topologies between asthmatic patients and healthy control subjects. Random forest models with fungal classifiers predicted asthma status with 75% accuracy for BAL fluid samples and 80% accuracy for EB samples. CONCLUSIONS: We demonstrate clear differences in bacterial and fungal microbiota in asthma-associated phenotypes. Our study provides additional support for considering microbial signatures in delineating asthma phenotypes.


Subject(s)
Asthma/microbiology , Eosinophils/immunology , Fungi/genetics , Hypersensitivity, Immediate/microbiology , Microbiota/immunology , Neutrophils/immunology , Respiratory System/microbiology , Th2 Cells/immunology , Adult , Asthma/immunology , Cytokines/metabolism , Female , Fungi/immunology , Humans , Hypersensitivity, Immediate/immunology , Male , Microbiota/genetics , Middle Aged , Phenotype , RNA, Ribosomal, 16S/analysis
5.
PLoS One ; 13(3): e0193334, 2018.
Article in English | MEDLINE | ID: mdl-29534074

ABSTRACT

Basal airway epithelial cells (AEC) constitute stem/progenitor cells within the central airways and respond to mucosal injury in an ordered sequence of spreading, migration, proliferation, and differentiation to needed cell types. However, dynamic gene transcription in the early events after mucosal injury has not been studied in AEC. We examined gene expression using microarrays following mechanical injury (MI) in primary human AEC grown in submersion culture to generate basal cells and in the air-liquid interface to generate differentiated AEC (dAEC) that include goblet and ciliated cells. A select group of ~150 genes was in differential expression (DE) within 2-24 hr after MI, and enrichment analysis of these genes showed over-representation of functional categories related to inflammatory cytokines and chemokines. Network-based gene prioritization and network reconstruction using the PINTA heat kernel diffusion algorithm demonstrated highly connected networks that were richer in differentiated AEC compared to basal cells. Similar experiments done in basal AEC collected from asthmatic donor lungs demonstrated substantial changes in DE genes and functional categories related to inflammation compared to basal AEC from normal donors. In dAEC, similar but more modest differences were observed. We demonstrate that the AEC transcription signature after MI identifies genes and pathways that are important to the initiation and perpetuation of airway mucosal inflammation. Gene expression occurs quickly after injury and is more profound in differentiated AEC, and is altered in AEC from asthmatic airways. Our data suggest that the early response to injury is substantially different in asthmatic airways, particularly in basal airway epithelial cells.


Subject(s)
Bronchi/cytology , Bronchi/injuries , Chemokines/genetics , Epithelial Cells/metabolism , Gene Expression Profiling , Trachea/cytology , Trachea/injuries , Asthma/pathology , Bronchi/pathology , Epithelial Cells/pathology , Humans , Mechanical Phenomena , Signal Transduction , Time Factors , Trachea/pathology
6.
Microbiome ; 5(1): 67, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28666467

ABSTRACT

BACKGROUND: Progressive, chronic bacterial infection of the airways is a leading cause of death in cystic fibrosis (CF). Culture-independent methods based on sequencing of the bacterial 16S rRNA gene describe a distinct microbial community that decreases in richness and diversity with disease progression. Understanding the functional characteristics of the microbial community may aid in identifying potential therapies and may assist in management, but current methods are cumbersome. Here, we demonstrate the use of an oxidative metabolic assay as a complement to sequencing methods to describe the microbiome in the airways of patients with CF. METHODS: Expectorated sputum was collected from 16 CF subjects and 8 control subjects. The Biolog Gen III Microplate was used in a community-level physiological profiling (CLPP)-based assay to examine oxidative metabolic activity. 16S rRNA V4 amplicon sequencing was used to characterize the taxonomy and diversity of the samples. Correlations were then identified among the oxidative activity and taxonomy data. In an additional paired analysis, sputum from seven CF subjects were collected at two separate clinic visits and compared for oxidative activity, taxonomy, and diversity. RESULTS: Significant differences in oxidative metabolic activity, microbial taxonomy, and diversity were found between the CF and control sputum samples. Oxidative activity correlated positively with total genera but not with other measures of diversity or taxonomy, demonstrating that the metabolic assay complements the structural aspects of the microbiome. As expected, Pseudomonas was significantly enriched in CF samples, while Streptococcus and Prevotella were similarly abundant in both CF and control samples. Paired analysis of CF samples at separate clinic visits revealed comparable oxidative activity that correlated with similar stability in taxonomy and diversity. CONCLUSIONS: The CLPP assay used in this study complements existing sequencing methods to delineate the oxidative metabolic footprint of the CF airway bacterial community. This method may be useful to study the CF microbial community over time and with changes in disease state.


Subject(s)
Bacteria/metabolism , Cystic Fibrosis/microbiology , Microbiota/physiology , Respiratory System/microbiology , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Cystic Fibrosis/metabolism , DNA, Bacterial/analysis , Female , Humans , Male , Metabolic Networks and Pathways , Metabolome , Microbiota/genetics , Middle Aged , RNA, Ribosomal, 16S , Sputum/microbiology
8.
J Allergy Clin Immunol ; 131(6): 1496-503, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23534973

ABSTRACT

BACKGROUND: We previously reported an interaction between maternal asthma and the child's HLA-G genotype on the child's subsequent risk for asthma. The implicated single nucleotide polymorphism at +3142 disrupted a target site for the microRNA (miR)-152 family. We hypothesized that the interaction effect might be mediated by these miRs. OBJECTIVE: The objective of this study was to test this hypothesis in adults with asthma who are a subset of the same subjects who participated in our earlier family-based studies. METHODS: We measured soluble HLA-G (sHLA-G) concentrations in bronchoalveolar lavage fluid (n = 36) and plasma (n = 57) from adult asthmatic subjects with and without a mother with asthma, and HLA-G and miR-152 family (miR-148a, miR-148b, and miR-152) transcript levels in airway epithelial cells from the same subjects. RESULTS: miR-148b levels were significantly increased in airway epithelial cells from asthmatic subjects with an asthmatic mother compared with those seen in asthmatic subjects without an asthmatic mother, and +3142 genotypes were associated with sHLA-G concentrations in bronchoalveolar lavage fluid among asthmatic subjects with an asthmatic mother but not among those with a nonasthmatic mother. Neither effect was observed in the plasma (sHLA-G) or white blood cells (miRNA). CONCLUSION: These combined results are consistent with +3142 allele-specific targeting of HLA-G by the miR-152 family and support our hypothesis that miRNA regulation of sHLA-G in the airway is influenced by both the asthma status of the subject's mother and the subject's genotype. Moreover, we demonstrate that the effects of maternal asthma on the gene regulatory landscape in the airways of the mother's children persist into adulthood.


Subject(s)
Asthma/genetics , Asthma/immunology , HLA-G Antigens/immunology , MicroRNAs/genetics , Adult , Black or African American , Asthma/metabolism , Female , Genotype , HLA-G Antigens/blood , HLA-G Antigens/genetics , Humans , Lung/immunology , Lung/metabolism , Male , Maternal Exposure , Middle Aged , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , White People , Young Adult
9.
Respir Res ; 14: 4, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23327606

ABSTRACT

BACKGROUND: Human leukocyte antigen (HLA)-G is a nonclassical class I antigen with immunomodulatory roles including up-regulation of suppressor T regulatory lymphocytes. HLA-G was recently identified as an asthma susceptibility gene, and expression of a soluble isoform, HLA-G5, has been demonstrated in human airway epithelium. Increased presence of HLA-G5 has been demonstrated in bronchoalveolar lavage fluid recovered from patients with mild asthma; this suggests a role for this isoform in modulating airway inflammation though the mechanisms by which this occurs is unclear. Airway inflammation associated with Th2 cytokines such as IL-4 and IL-13 is a principal feature of asthma, but whether these cytokines elicit expression of HLA-G is not known. METHODS: We examined gene and protein expression of both soluble (G5) and membrane-bound (G1) HLA-G isoforms in primary differentiated human airway epithelial cells collected from normal lungs and grown in air-liquid interface culture. Cells were treated with up to 10 ng/ml of either IL-4, IL-5, or IL-13, or 100 ng/ml of the immunomodulatory cytokine IL-10, or 10,000 U/ml of the Th1-associated cytokine interferon-beta, for 24 hr, after which RNA was isolated for evaluation by quantitative PCR and protein was collected for Western blot analysis. RESULTS: HLA-G5 but not G1 was present in dAEC as demonstrated by quantitative PCR, western blot and confocal microscopy. Neither G5 nor G1 expression was increased by the Th2-associated cytokines IL-4, IL-5 or IL-13 over 24 hr, nor after treatment with IL-10, but was increased 4.5 ± 1.4 fold after treatment with 10,000 U/ml interferon-beta. CONCLUSIONS: These data demonstrate the constitutive expression of a T lymphocyte regulatory molecule in differentiated human airway epithelial cells that is not modulated by Th2-associated cytokines.


Subject(s)
Cytokines/immunology , Epithelial Cells/cytology , Epithelial Cells/immunology , HLA-G Antigens/immunology , Immunomodulation/immunology , Respiratory Mucosa/immunology , Th2 Cells/immunology , Cell Differentiation , Cells, Cultured , Humans , Respiratory Mucosa/cytology
10.
Urol Oncol ; 31(5): 566-71, 2013 Jul.
Article in English | MEDLINE | ID: mdl-21600800

ABSTRACT

OBJECTIVES: We sought to develop a clinical algorithm combining serum PSA with detection of TMPRSS2:ERG fusion and PCA3 in urine collected after digital rectal exam (post-DRE urine) to predict prostate cancer on subsequent biopsy. MATERIALS AND METHODS: Post-DRE urine was collected in 48 consecutive patients before prostate biopsy at 2 centers; quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used to detect PCA3 and TMPRSS2:ERG fusion transcript expression. Serum PSA was measured by clinical assay. The performance of TMPRSS2:ERG fusion, PCA3, and serum PSA as biomarkers predicting prostate cancer at biopsy was measured; a clinically practical algorithm combining serum PSA with TMPRSS2:ERG and PCA3 in post-DRE urine to predict prostate cancer was developed. RESULTS: Post-DRE urine sediment provided informative RNA in 45 patients; prostate cancer was present on subsequent biopsy in 15. TMPRSS2:ERG in post-DRE urine was associated with prostate cancer (OR = 12.02; P < 0.001). PCA3 had the highest sensitivity in predicting prostate cancer diagnosis (93%), whereas TMPRSS2:ERG had the highest specificity (87%). TMPRSS2:ERG had the greatest discriminatory value in predicting prostate cancer (AUC = 0.77 compared with 0.65 for PCA3 and 0.72 for serum PSA alone). Combining serum PSA, PCA3, and TMPRSS2:ERG in a multivariable algorithm optimized for clinical utility improved cancer prediction (AUC = 0.88; specificity = 90% at 80% sensitivity). CONCLUSIONS: A clinical algorithm specifying biopsy for all patients with PSA ≥ 10 ng/ml, while restricting biopsy among those with PSA <10 ng/ml to only those with detectable PCA3 or TMPRSS2:ERG in post-DRE urine, performed better than the individual biomarkers alone in predicting prostate cancer.


Subject(s)
Antigens, Neoplasm/urine , Oncogene Proteins, Fusion/urine , Prostate-Specific Antigen/blood , Prostatic Neoplasms/diagnosis , Aged , Antigens, Neoplasm/genetics , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Biomarkers, Tumor/urine , Biopsy , Humans , Logistic Models , Male , Middle Aged , Multivariate Analysis , Oncogene Proteins, Fusion/genetics , Predictive Value of Tests , Prostate/pathology , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
11.
Nat Biotechnol ; 29(8): 742-9, 2011 Jul 31.
Article in English | MEDLINE | ID: mdl-21804560

ABSTRACT

Noncoding RNAs (ncRNAs) are emerging as key molecules in human cancer, with the potential to serve as novel markers of disease and to reveal uncharacterized aspects of tumor biology. Here we discover 121 unannotated prostate cancer-associated ncRNA transcripts (PCATs) by ab initio assembly of high-throughput sequencing of polyA(+) RNA (RNA-Seq) from a cohort of 102 prostate tissues and cells lines. We characterized one ncRNA, PCAT-1, as a prostate-specific regulator of cell proliferation and show that it is a target of the Polycomb Repressive Complex 2 (PRC2). We further found that patterns of PCAT-1 and PRC2 expression stratified patient tissues into molecular subtypes distinguished by expression signatures of PCAT-1-repressed target genes. Taken together, our findings suggest that PCAT-1 is a transcriptional repressor implicated in a subset of prostate cancer patients. These findings establish the utility of RNA-Seq to identify disease-associated ncRNAs that may improve the stratification of cancer subtypes.


Subject(s)
Biomarkers, Tumor/genetics , Prostatic Neoplasms/genetics , RNA, Untranslated/genetics , Base Sequence , Biomarkers, Tumor/metabolism , Cell Growth Processes/physiology , Cluster Analysis , Cohort Studies , Computational Biology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , Enhancer of Zeste Homolog 2 Protein , Humans , Male , Molecular Sequence Data , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Untranslated/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reproducibility of Results , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
12.
Sci Transl Med ; 3(72): 72ra17, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21368222

ABSTRACT

Gene fusions involving ETS (erythroblastosis virus E26 transformation-specific) family transcription factors are found in ~50% of prostate cancers and as such can be used as a basis for the molecular subclassification of prostate cancer. Previously, we showed that marked overexpression of SPINK1 (serine peptidase inhibitor, Kazal type 1), which encodes a secreted serine protease inhibitor, defines an aggressive molecular subtype of ETS fusion-negative prostate cancers (SPINK1+/ETS⁻, ~10% of all prostate cancers). Here, we examined the potential of SPINK1 as an extracellular therapeutic target in prostate cancer. Recombinant SPINK1 protein (rSPINK1) stimulated cell proliferation in benign RWPE as well as cancerous prostate cells. Indeed, RWPE cells treated with either rSPINK1 or conditioned medium from 22RV1 prostate cancer cells (SPINK1+/ETS⁻) significantly increased cell invasion and intravasation when compared with untreated cells. In contrast, knockdown of SPINK1 in 22RV1 cells inhibited cell proliferation, cell invasion, and tumor growth in xenograft assays. 22RV1 cell proliferation, invasion, and intravasation were attenuated by a monoclonal antibody (mAb) to SPINK1 as well. We also demonstrated that SPINK1 partially mediated its neoplastic effects through interaction with the epidermal growth factor receptor (EGFR). Administration of antibodies to SPINK1 or EGFR (cetuximab) in mice bearing 22RV1 xenografts attenuated tumor growth by more than 60 and 40%, respectively, or ~75% when combined, without affecting PC3 xenograft (SPINK1⁻/ETS⁻) growth. Thus, this study suggests that SPINK1 may be a therapeutic target in a subset of patients with SPINK1+/ETS⁻ prostate cancer. Our results provide a rationale for both the development of humanized mAbs to SPINK1 and evaluation of EGFR inhibition in SPINK1+/ETS⁻ prostate cancers.


Subject(s)
Carrier Proteins/genetics , Gene Targeting , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Autocrine Communication , Carrier Proteins/metabolism , Cell Line , Cell Proliferation , ErbB Receptors/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transplantation, Heterologous , Trypsin Inhibitor, Kazal Pancreatic
13.
Am J Physiol Lung Cell Mol Physiol ; 299(5): L681-93, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20729386

ABSTRACT

IL-4 and IL-13 elicit several important responses in airway epithelium including chemokine secretion and mucous secretion that may contribute to airway inflammation, cell migration, and differentiation. These cytokines have overlapping but not identical effector profiles likely due to shared subunits in their receptor complexes. These receptors are variably described in epithelial cells, and the relative expression, localization, and function of these receptors in differentiated and repairing epithelial cells are not clear. We examined IL-4/IL-13 receptor expression and localization in primary airway epithelial cells collected from normal human lungs and grown under conditions yielding both undifferentiated and differentiated cells inclusive of basal, goblet, and ciliated cell phenotypes. Gene expression of the IL-4Rα, IL-2Rγc, IL-13Rα1, and IL-13Rα2 receptor subunits increased with differentiation, but different patterns of localization and protein abundance were seen for each subunit based on both differentiation and the cell subtypes present. Increased expression of receptor subunits observed in more differentiated cells was associated with more substantial functional responses to IL-4 stimulation including increased eotaxin-3 expression and accelerated migration after injury. We demonstrate substantial differences in IL-4/IL-13 receptor subunit expression and responsiveness to IL-4 based on the extent of airway epithelial cell differentiation and suggest that these differences may have functional consequences in airway inflammation.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/physiology , Receptors, Interleukin-13/metabolism , Receptors, Interleukin-4/metabolism , Respiratory Mucosa/cytology , Animals , Cell Movement , Cells, Cultured , Chemokine CCL26 , Chemokines, CC/genetics , Chemokines, CC/metabolism , Epithelial Cells/cytology , Humans , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Interleukin-13/genetics , Receptors, Interleukin-4/genetics , Respiratory Mucosa/physiology , Stress, Mechanical
15.
Cancer Cell ; 17(5): 443-54, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20478527

ABSTRACT

Chromosomal rearrangements fusing the androgen-regulated gene TMPRSS2 to the oncogenic ETS transcription factor ERG occur in approximately 50% of prostate cancers, but how the fusion products regulate prostate cancer remains unclear. Using chromatin immunoprecipitation coupled with massively parallel sequencing, we found that ERG disrupts androgen receptor (AR) signaling by inhibiting AR expression, binding to and inhibiting AR activity at gene-specific loci, and inducing repressive epigenetic programs via direct activation of the H3K27 methyltransferase EZH2, a Polycomb group protein. These findings provide a working model in which TMPRSS2-ERG plays a critical role in cancer progression by disrupting lineage-specific differentiation of the prostate and potentiating the EZH2-mediated dedifferentiation program.


Subject(s)
DNA-Binding Proteins/genetics , Gene Fusion , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Transcription Factors/genetics , Chromatin Immunoprecipitation , Disease Progression , Enhancer of Zeste Homolog 2 Protein , Humans , Male , Polycomb Repressive Complex 2 , Prostatic Neoplasms/genetics , Signal Transduction
16.
Proc Natl Acad Sci U S A ; 106(25): 10284-9, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-19487683

ABSTRACT

Breast cancer patients have benefited from the use of targeted therapies directed at specific molecular alterations. To identify additional opportunities for targeted therapy, we searched for genes with marked overexpression in subsets of tumors across a panel of breast cancer profiling studies comprising 3,200 microarray experiments. In addition to prioritizing ERBB2, we found AGTR1, the angiotensin II receptor type I, to be markedly overexpressed in 10-20% of breast cancer cases across multiple independent patient cohorts. Validation experiments confirmed that AGTR1 is highly overexpressed, in several cases more than 100-fold. AGTR1 overexpression was restricted to estrogen receptor-positive tumors and was mutually exclusive with ERBB2 overexpression across all samples. Ectopic overexpression of AGTR1 in primary mammary epithelial cells, combined with angiotensin II stimulation, led to a highly invasive phenotype that was attenuated by the AGTR1 antagonist losartan. Similarly, losartan reduced tumor growth by 30% in AGTR1-positive breast cancer xenografts. Taken together, these observations indicate that marked AGTR1 overexpression defines a subpopulation of ER-positive, ERBB2-negative breast cancer that may benefit from targeted therapy with AGTR1 antagonists, such as losartan.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Losartan/pharmacology , Receptor, Angiotensin, Type 1/biosynthesis , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Receptor, ErbB-2/genetics , Xenograft Model Antitumor Assays
17.
Nature ; 457(7231): 910-4, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19212411

ABSTRACT

Multiple, complex molecular events characterize cancer development and progression. Deciphering the molecular networks that distinguish organ-confined disease from metastatic disease may lead to the identification of critical biomarkers for cancer invasion and disease aggressiveness. Although gene and protein expression have been extensively profiled in human tumours, little is known about the global metabolomic alterations that characterize neoplastic progression. Using a combination of high-throughput liquid-and-gas-chromatography-based mass spectrometry, we profiled more than 1,126 metabolites across 262 clinical samples related to prostate cancer (42 tissues and 110 each of urine and plasma). These unbiased metabolomic profiles were able to distinguish benign prostate, clinically localized prostate cancer and metastatic disease. Sarcosine, an N-methyl derivative of the amino acid glycine, was identified as a differential metabolite that was highly increased during prostate cancer progression to metastasis and can be detected non-invasively in urine. Sarcosine levels were also increased in invasive prostate cancer cell lines relative to benign prostate epithelial cells. Knockdown of glycine-N-methyl transferase, the enzyme that generates sarcosine from glycine, attenuated prostate cancer invasion. Addition of exogenous sarcosine or knockdown of the enzyme that leads to sarcosine degradation, sarcosine dehydrogenase, induced an invasive phenotype in benign prostate epithelial cells. Androgen receptor and the ERG gene fusion product coordinately regulate components of the sarcosine pathway. Here, by profiling the metabolomic alterations of prostate cancer progression, we reveal sarcosine as a potentially important metabolic intermediary of cancer cell invasion and aggressivity.


Subject(s)
Disease Progression , Metabolomics , Prostatic Neoplasms/metabolism , Sarcosine/metabolism , Androgens/physiology , Cell Line , Cell Line, Tumor , Gene Knockdown Techniques , Glycine N-Methyltransferase/genetics , Glycine N-Methyltransferase/metabolism , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Sarcosine/analysis , Sarcosine/urine , Sarcosine Dehydrogenase/metabolism , Signal Transduction
18.
Science ; 322(5908): 1695-9, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-19008416

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is a mammalian histone methyltransferase that contributes to the epigenetic silencing of target genes and regulates the survival and metastasis of cancer cells. EZH2 is overexpressed in aggressive solid tumors by mechanisms that remain unclear. Here we show that the expression and function of EZH2 in cancer cell lines are inhibited by microRNA-101 (miR-101). Analysis of human prostate tumors revealed that miR-101 expression decreases during cancer progression, paralleling an increase in EZH2 expression. One or both of the two genomic loci encoding miR-101 were somatically lost in 37.5% of clinically localized prostate cancer cells (6 of 16) and 66.7% of metastatic disease cells (22 of 33). We propose that the genomic loss of miR-101 in cancer leads to overexpression of EZH2 and concomitant dysregulation of epigenetic pathways, resulting in cancer progression.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Histones/metabolism , MicroRNAs/genetics , Neoplasms/genetics , Prostatic Neoplasms/genetics , Transcription Factors/genetics , 3' Untranslated Regions , Algorithms , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Disease Progression , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Female , Genome, Human , Humans , Lysine/metabolism , Male , Methylation , MicroRNAs/metabolism , Neoplasm Metastasis , Neoplasms/metabolism , Polycomb Repressive Complex 2 , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Transcription Factors/metabolism
19.
Neoplasia ; 10(11): 1285-94, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18953438

ABSTRACT

Prostate cancer is the most common type of tumor found in American men and is the second leading cause of cancer death in males. To identify biomarkers that distinguish prostate cancer from normal, we compared multiple gene expression profiling studies. Through meta-analysis of expression array data from multiple prostate cancer studies, we identified GOLM1 (Golgi membrane protein 1, Golm 1) as consistently up-regulated in clinically localized prostate cancer. This observation was confirmed by reverse transcription-polymerase chain reaction (RT-PCR) and validated at the protein level by immunoblot assay and immunohistochemistry. Prostate epithelial cells were identified as the cellular source of GOLM1 expression using laser capture microdissection. Immunohistochemical staining localized the GOLM1 signal to the subapical cytoplasmic region, typical of a Golgi distribution. Surprisingly, GOLM1 immunoreactivity was detected in the supernatants of prostate cell lines and in the urine of patients with prostate cancer. The mechanism by which intact GOLM1 might be released from cells has not yet been elucidated. GOLM1 transcript levels were measured in urine sediments using quantitative PCR on a cohort of patients presenting for biopsy or radical prostatectomy. We found that urinary GOLM1 mRNA levels were a significant predictor of prostate cancer. Further, GOLM1 outperformed serum prostate-specific antigen (PSA) in detecting prostate cancer. The area under the receiver-operating characteristic curve was 0.622 for GOLM1 (P = .0009) versus 0.495 for serum PSA (P = .902). Our data indicating the up-regulation of GOLM1 expression and its appearance in patients' urine suggest GOLM1 as a potential novel biomarker for clinically localized prostate cancer.


Subject(s)
Membrane Proteins/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/urine , Cell Line , Cohort Studies , Epithelial Cells/metabolism , Gene Expression Profiling , Golgi Apparatus/metabolism , Humans , Immunohistochemistry , Male , Membrane Proteins/genetics , Membrane Proteins/urine , Prostate/cytology , Prostate/metabolism , Prostate-Specific Antigen/blood , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/urine , ROC Curve , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
20.
Cancer Res ; 68(18): 7629-37, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794152

ABSTRACT

Recurrent gene fusions involving E26 transformation-specific (ETS) transcription factors ERG, ETV1, ETV4, or ETV5 have been identified in 40% to 70% of prostate cancers. Here, we used a comprehensive fluorescence in situ hybridization (FISH) split probe strategy interrogating all 27 ETS family members and their five known 5' fusion partners in a cohort of 110 clinically localized prostate cancer patients. Gene rearrangements were only identified in ETS genes that were previously implicated in prostate cancer gene fusions including ERG, ETV1, and ETV4 (43%, 5%, and 5%, respectively), suggesting that a substantial fraction of prostate cancers (estimated at 30-60%) cannot be attributed to an ETS gene fusion. Among the known 5' gene fusion partners, TMPRSS2 was rearranged in 47% of cases followed by SLC45A3, HNRPA2B1, and C15ORF21 in 2%, 1%, and 1% of cases, respectively. Based on this comprehensive FISH screen, we have made four noteworthy observations. First, by screening the entire ETS transcription factor family for rearrangements, we found that a large fraction of prostate cancers (44%) cannot be ascribed to an ETS gene fusion, an observation which will stimulate research into identifying recurrent non-ETS aberrations in prostate cancers. Second, we identified SLC45A3 as a novel 5' fusion partner of ERG; previously, TMPRSS2 was the only described 5' partner of ERG. Third, we identified two prostate-specific, androgen-induced genes, FLJ35294 and CANT1, as 5' partners to ETV1 and ETV4. Fourth, we identified a ubiquitously expressed, androgen-insensitive gene, DDX5, fused in frame with ETV4, leading to the expression of a DDX5-ETV4 fusion protein.


Subject(s)
Adenovirus E1A Proteins/genetics , Cell Transformation, Neoplastic/genetics , DEAD-box RNA Helicases/genetics , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Base Sequence , Cell Line, Tumor , Gene Rearrangement , Humans , In Situ Hybridization, Fluorescence , Male , Molecular Sequence Data , Proto-Oncogene Proteins c-ets , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...