Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
NPJ Precis Oncol ; 2: 10, 2018.
Article in English | MEDLINE | ID: mdl-29872719

ABSTRACT

In addition to canonical oncoproteins, truncated isoforms and proteolysis products are implicated in both drug resistance and disease progression. In HER2-positive breast tumors, expression of truncated HER2 isoforms resulting from alternative translation and/or carboxy-terminal fragments (CTFs) resulting from proteolysis (collectively, t-erbB2) have been associated with shortened progression-free survival of patients. Thus, to advance clinical pathology and inform treatment decisions, we developed a high-selectivity cytopathology assay capable of distinguishing t-erbB2 from full-length HER2 expression without the need for isoform-specific antibodies. Our microfluidic, single-cell western blot, employs electrophoretic separations to resolve full-length HER2 from the smaller t-erbB2 in each ~28 pL single-cell lysate. Subsequently, a pan-HER2 antibody detects all resolved HER2 protein forms via immunoprobing. In analysis of eight breast tumor biopsies, we identified two tumors comprised of 15% and 40% t-erbB2-expressing cells. By single-cell western blotting of the t-erbB2-expressing cells, we observed statistically different ratios of t-erbB2 proteins to full-length HER2 expression. Further, target multiplexing and clustering analyses scrutinized signaling, including ribosomal S6, within the t-erbB2-expressing cell subpopulation. Taken together, cytometric assays that report both protein isoform profiles and signaling state offer cancer classification taxonomies with unique relevance to precisely describing drug resistance mechanisms in which oncoprotein isoforms/fragments are implicated.

2.
Adv Mater ; 28(2): 327-334, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26567472

ABSTRACT

Pore-gradient microgel arrays enable thousands of parallel high-resolution single-cell protein electrophoresis separations for targets accross a wide molecular mass (25-289 kDa), yet within 1 mm separation distances. Dual crosslinked hydrogels facilitate gel-pore expansion after electrophoresis for efficient and uniform immunoprobing. The photopatterned, light-activated, and acid-expandable hydrogel underpins single-cell protein analysis, here for oncoprotein-related signaling in human breast biopsy.


Subject(s)
Blotting, Western/instrumentation , Hydrogels , Single-Cell Analysis/instrumentation , Blotting, Western/methods , Breast Neoplasms/metabolism , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel/instrumentation , Electrophoresis, Polyacrylamide Gel/methods , Extracellular Signal-Regulated MAP Kinases/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hydrogels/chemistry , Hydrogels/radiation effects , Light , Nucleocytoplasmic Transport Proteins/metabolism , Porosity , Receptor, ErbB-2/metabolism , Single-Cell Analysis/methods , TOR Serine-Threonine Kinases/metabolism
3.
Breast Cancer Res Treat ; 148(1): 41-59, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25270120

ABSTRACT

The mechanisms by which breast cancer (BrC) can successfully metastasize are complex and not yet fully understood. Our goal was to identify tumor-induced stromal changes that influence metastatic cell behavior, and may serve as better targets for therapy. To identify stromal changes in cancer-bearing tissue, dual-species gene expression analysis was performed for three different metastatic BrC xenograft models. Results were confirmed by immunohistochemistry, flow cytometry, and protein knockdown. These results were validated in human clinical samples at the mRNA and protein level by retrospective analysis of cohorts of human BrC specimens. In pre-clinical models of BrC, systemic recruitment of S100A8+ myeloid cells-including myeloid-derived suppressor cells (MDSCs)-was promoted by tumor-derived factors. Recruitment of S100A8+ myeloid cells was diminished by inhibition of tumor-derived factors or depletion of MDSCs, resulting in fewer metastases and smaller primary tumors. Importantly, these MDSCs retain their ability to suppress T cell proliferation upon co-culture. Secretion of macrophage inhibitory factor (MIF) activated the recruitment of S100A8+ myeloid cells systemically. Inhibition of MIF, or depletion of MDSCs resulted in delayed tumor growth and lower metastatic burden. In human BrC specimens, increased mRNA and protein levels of S100A8+ infiltrating cells are highly associated with poor overall survival and shorter metastasis free survival of BrC patients, respectively. Furthermore, analysis of nine different human gene expression datasets confirms the association of increased levels of S100A8 transcripts with an increased risk of death. Recruitment of S100A8+ myeloid cells to primary tumors and secondary sites in xenograft models of BrC enhances cancer progression independent of their suppressive activity on T cells. In clinical samples, infiltrating S100A8+ cells are associated with poor overall survival. Targeting these molecules or associated pathways in cells of the tumor microenvironment may translate into novel therapeutic interventions and benefit patient outcome.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Myeloid Cells/pathology , Neoplasm Invasiveness/pathology , Tumor Microenvironment , Animals , Calgranulin A/biosynthesis , Cell Line, Tumor , Female , Flow Cytometry , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Tissue Array Analysis , Transcriptome
4.
PLoS One ; 7(10): e47995, 2012.
Article in English | MEDLINE | ID: mdl-23118918

ABSTRACT

Breast cancer is the most common cancer in women, and this prevalence has a major impact on health worldwide. Localized breast cancer has an excellent prognosis, with a 5-year relative survival rate of 85%. However, the survival rate drops to only 23% for women with distant metastases. To date, the study of breast cancer metastasis has been hampered by a lack of reliable metastatic models. Here we describe a novel in vivo model using human breast cancer xenografts in NOD scid gamma (NSG) mice; in this model human breast cancer cells reliably metastasize to distant organs from primary tumors grown within the mammary fat pad. This model enables the study of the entire metastatic process from the proper anatomical site, providing an important new approach to examine the mechanisms underlying breast cancer metastasis. We used this model to identify gene expression changes that occur at metastatic sites relative to the primary mammary fat pad tumor. By comparing multiple metastatic sites and independent cell lines, we have identified several gene expression changes that may be important for tumor growth at distant sites.


Subject(s)
Liver Neoplasms/secondary , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Animals , Cell Line, Tumor , Contraindications , Disease Models, Animal , Female , Gene Expression , Humans , Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , Lymphatic Metastasis , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Transcriptome
5.
Breast Cancer Res Treat ; 136(3): 683-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23089982

ABSTRACT

Therapies targeting the ERBB2 receptor, including the kinase inhibitor lapatinib (Tykerb, GlaxoSmithKline), have improved clinical outcome for women with ERBB2-amplified breast cancer. However, acquired resistance to lapatinib remains a significant clinical problem, and the mechanisms governing resistance remain poorly understood. We sought to define molecular alterations that confer an acquired lapatinib resistance phenotype in ER-/ERBB2+ human breast cancer cells. ERBB2-amplified SKBR3 breast cancer cells were rendered resistant to lapatinib via culture in increasing concentrations of the drug, and molecular changes associated with a resistant phenotype were interrogated using a collaborative enzyme-enhanced immunoassay platform and immunoblotting techniques for detection of phosphorylated signaling cascade proteins. Interestingly, despite apparent inactivation of the PI3K/AKT signaling pathway, resistant cells exhibited constitutive activation of mammalian target of rapamycin complex 1 (mTORC1) and were highly sensitive to mTOR inhibition with rapamycin and the dual PI3K/mTOR inhibitor NVP-BEZ235. These data demonstrate a role for downstream activation of mTORC1 in the absence of molecular alterations leading to PI3K/AKT hyperactivation as a potential mechanism of lapatinib resistance in this model of ERBB2+ breast cancer and support the rationale of combination or sequential therapy using ERBB2 and mTOR-targeting molecules to prevent or target resistance to lapatinib. Moreover, our data suggest that assessment of mTOR substrate phosphorylation (i.e., S6) may serve as a more robust biomarker to predict sensitivity to mTOR inhibitors in the context of lapatinib resistance than PI3K mutations, loss of PTEN and p-AKT levels.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Phosphatidylinositol 3-Kinases/metabolism , Proteins/metabolism , Quinazolines/pharmacology , Receptor, ErbB-2/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Lapatinib , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Mutation , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Receptor, ErbB-2/genetics , TOR Serine-Threonine Kinases
6.
Breast Cancer Res Treat ; 135(1): 79-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22535017

ABSTRACT

Cancer is caused by mutations in oncogenes and tumor suppressor genes, resulting in the deregulation of processes fundamental to the normal behavior of cells. The identification and characterization of oncogenes and tumor suppressors has led to new treatment strategies that have significantly improved cancer outcome. The advent of next generation sequencing has allowed the elucidation of the fine structure of cancer genomes, however, the identification of pathogenic changes is complicated by the inherent genomic instability of cancer cells. Therefore, functional approaches for the identification of novel genes involved in the initiation and development of tumors are critical. Here we report the first whole human genome in vivo RNA interference screen to identify functionally important tumor suppressor genes. Using our novel approach, we identify previously validated tumor suppressor genes including TP53 and MNT, as well as several novel candidate tumor suppressor genes including leukemia inhibitory factor receptor (LIFR). We show that LIFR is a key novel tumor suppressor, whose deregulation may drive the transformation of a significant proportion of human breast cancers. These results demonstrate the power of genome wide in vivo RNAi screens as a method for identifying novel genes regulating tumorigenesis.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Breast Neoplasms/genetics , Genes, Tumor Suppressor , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Genes, p53 , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , RNA Interference , RNA, Small Interfering
7.
PLoS One ; 7(12): e52106, 2012.
Article in English | MEDLINE | ID: mdl-23300597

ABSTRACT

MicroRNAs (miRs) are small, endogenous, non-coding RNAs that regulate the stability and/or translation of complementary mRNA targets. MiRs have emerged not only as critical modulators of normal physiologic processes, but their deregulation may significantly impact prostate and other cancers. The expression of miR-23b and miR-27b, which are encoded by the same miR cluster (miR-23b/-27b), are downregulated in metastatic, castration-resistant tumors compared to primary prostate cancer and benign tissue; however, their possible role in prostate cancer progression is unknown. We found that ectopic expression of miR-23b/-27b in two independent castration-resistant prostate cancer cell lines resulted in suppression of invasion and migration, as well as reduced survival in soft agar (a measure of anoikis). However, there was no effect of miR-23b/-27b on cell proliferation suggesting that these miRs function as metastasis (but not growth) suppressors in prostate cancer. Conversely, inhibition of miR-23b/-27b in the less aggressive androgen-dependent LNCaP prostate cancer cell line resulted in enhanced invasion and migration also without affecting proliferation. Mechanistically, we found that introduction of miR-23b/-27b in metastatic, castration-resistant prostate cancer cell lines resulted in a significant attenuation of Rac1 activity without affecting total Rac1 levels and caused increased levels of the tumor suppressor E-cadherin. Inhibition of these miRs had the opposite effect in androgen-dependent LNCaP cells. These results suggest that miR-23b/-27b are metastasis suppressors that might serve as novel biomarkers and therapeutic agents for castration-resistant disease.


Subject(s)
Cadherins/metabolism , Cell Movement , MicroRNAs/genetics , Neoplasms, Hormone-Dependent/genetics , Orchiectomy , Prostatic Neoplasms/genetics , rac1 GTP-Binding Protein/metabolism , Apoptosis , Blotting, Western , Cadherins/genetics , Cell Adhesion , Cell Proliferation , Flow Cytometry , Humans , Male , Neoplasms, Hormone-Dependent/secondary , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , rac1 GTP-Binding Protein/genetics
8.
EMBO J ; 31(4): 1028-40, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22157816

ABSTRACT

Tropomyosins are widespread actin-binding proteins that influence numerous cellular functions including actin dynamics, cell migration, tumour suppression, and Drosophila oocyte development. Synaptopodin is another actin-binding protein with a more restricted expression pattern in highly dynamic cell compartments such as kidney podocyte foot processes, where it promotes RhoA signalling by blocking the Smurf1-mediated ubiquitination of RhoA. Here, we show that synaptopodin has a shorter half-life but shares functional properties with the highly stable tropomyosin. Transgenic expression of synaptopodin restores oskar mRNA localization in Drosophila oocytes mutant for TmII, thereby rescuing germline differentiation and fertility. Synaptopodin restores stress fibres in tropomyosin-deficient human MDA-MB 231 breast cancer cells and TPMα-depleted fibroblasts. Gene silencing of TPMα but not TPMß causes loss of stress fibres by promoting Smurf1-mediated ubiquitination and proteasomal degradation of RhoA. Functionally, overexpression of synaptopodin or RhoA(K6,7R) significantly reduces MDA-MB 231 cell migration. Our findings elucidate RhoA stabilization by structurally unrelated actin-binding proteins as a conserved mechanism for regulation of stress fibre dynamics and cell motility in a cell type-specific fashion.


Subject(s)
Microfilament Proteins/physiology , Neoplasms/genetics , Tropomyosin/genetics , Tropomyosin/physiology , rhoA GTP-Binding Protein/physiology , Animals , Animals, Genetically Modified , Cell Line, Tumor , Humans , Mice , NIH 3T3 Cells , Neoplasms/pathology
9.
Virus Res ; 142(1-2): 78-84, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19200445

ABSTRACT

Neurotropic herpesviruses express viral deoxyuridine triphosphate nucleotidohydrolase (dUTPase) and uracil DNA glycosylase (UDG) enzymes which may reduce uracil misincorporation into viral DNA, particularly in neurons of infected ganglia. The simian varicella virus (SVV) dUTPase (ORF 8) and UDG (ORF 59) share 37.7% and 53.9% amino acid identity, respectively, with varicella-zoster virus (VZV) homologs. Infectious SVV mutants defective in either dUTPase (SVV-dUTPase(-)) or UDG (SVV-UDG(-)) activity or both (SVV-dUTPase(-)/UDG(-)) were constructed using recA assisted restriction endonuclease cleavage (RARE) and a cosmid recombination system. Loss of viral dUTPase and UDG enzymatic activity was confirmed in CV-1 cells infected with the SVV mutants. The SVV-dUTPase(-), SVV-UDG(-), and SVV-dUTPase(-)/UDG(-) mutants replicated as efficiently as wild-type SVV in cell culture. SVV dUTPase and UDG expression was detected in tissues derived from acutely infected animals, but not in tissues derived from latently infected animals. Further studies will evaluate the pathogenesis of SVV dUTPase and UDG mutants and their potential as varicella vaccines.


Subject(s)
Herpesviridae Infections/virology , Pyrophosphatases/metabolism , Uracil-DNA Glycosidase/metabolism , Varicellovirus/enzymology , Viral Proteins/metabolism , Virus Replication , Amino Acid Sequence , Animals , Base Sequence , Chickenpox/virology , Chlorocebus aethiops , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Herpesvirus 3, Human/physiology , Humans , Molecular Sequence Data , Pyrophosphatases/chemistry , Pyrophosphatases/genetics , Sequence Alignment , Uracil-DNA Glycosidase/chemistry , Uracil-DNA Glycosidase/genetics , Varicellovirus/chemistry , Varicellovirus/genetics , Varicellovirus/physiology , Vero Cells , Viral Proteins/genetics
10.
J Gen Virol ; 89(Pt 3): 741-750, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18272766

ABSTRACT

Recombinant simian varicella viruses (rSVVs) were engineered to express respiratory syncytial virus (RSV) antigens. The RSV surface glycoprotein G and second matrix protein M2 (22k) genes were cloned into the SVV genome, and recombinant viruses were characterized in vitro and in vivo. rSVVs were also engineered to express the membrane-anchored or secreted forms of the RSV-G protein as well as an RSV G lacking its chemokine mimicry motif (CX3C), which may have different effects on priming the host immune response. The RSV genes were efficiently expressed in rSVV/RSV-infected Vero cells as RSV-G and -M2 transcripts were detected by RT-PCR, and RSV antigens were detected by immunofluorescence and immunoblot assays. The rSVVs replicated efficiently in Vero cell culture. Rhesus macaques immunized with rSVV/RSV-G and rSVV/RSV-M2 vaccines produced antibody responses to SVV and RSV antigens. The results demonstrate that recombinant varicella viruses are suitable vectors for the expression of RSV antigens and may represent a novel vaccine strategy for immunization against both pathogens.


Subject(s)
Antibodies, Viral/blood , Herpesvirus 3, Human/immunology , Recombination, Genetic , Respiratory Syncytial Viruses/immunology , Viral Envelope Proteins/immunology , Viral Matrix Proteins/immunology , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , Chickenpox/immunology , Chickenpox/prevention & control , Chickenpox Vaccine/administration & dosage , Chickenpox Vaccine/genetics , Chickenpox Vaccine/immunology , Chlorocebus aethiops , Genetic Vectors , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/metabolism , Macaca mulatta , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/genetics , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...