Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Cell ; 178(1): 160-175.e27, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31155233

ABSTRACT

Single-cell technologies have described heterogeneity across tissues, but the spatial distribution and forces that drive single-cell phenotypes have not been well defined. Combining single-cell RNA and protein analytics in studying the role of stromal cancer-associated fibroblasts (CAFs) in modulating heterogeneity in pancreatic cancer (pancreatic ductal adenocarcinoma [PDAC]) model systems, we have identified significant single-cell population shifts toward invasive epithelial-to-mesenchymal transition (EMT) and proliferative (PRO) phenotypes linked with mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription 3 (STAT3) signaling. Using high-content digital imaging of RNA in situ hybridization in 195 PDAC tumors, we quantified these EMT and PRO subpopulations in 319,626 individual cancer cells that can be classified within the context of distinct tumor gland "units." Tumor gland typing provided an additional layer of intratumoral heterogeneity that was associated with differences in stromal abundance and clinical outcomes. This demonstrates the impact of the stroma in shaping tumor architecture by altering inherent patterns of tumor glands in human PDAC.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Animals , Cell Proliferation , Coculture Techniques , Epithelial-Mesenchymal Transition , Female , HEK293 Cells , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinases/metabolism , RNA-Seq , STAT3 Transcription Factor/metabolism , Stromal Cells/metabolism , Transfection
2.
Nature ; 537(7618): 102-106, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27556950

ABSTRACT

Circulating tumour cells in women with advanced oestrogen-receptor (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer acquire a HER2-positive subpopulation after multiple courses of therapy. In contrast to HER2-amplified primary breast cancer, which is highly sensitive to HER2-targeted therapy, the clinical significance of acquired HER2 heterogeneity during the evolution of metastatic breast cancer is unknown. Here we analyse circulating tumour cells from 19 women with ER+/HER2- primary tumours, 84% of whom had acquired circulating tumour cells expressing HER2. Cultured circulating tumour cells maintain discrete HER2+ and HER2- subpopulations: HER2+ circulating tumour cells are more proliferative but not addicted to HER2, consistent with activation of multiple signalling pathways; HER2- circulating tumour cells show activation of Notch and DNA damage pathways, exhibiting resistance to cytotoxic chemotherapy, but sensitivity to Notch inhibition. HER2+ and HER2- circulating tumour cells interconvert spontaneously, with cells of one phenotype producing daughters of the opposite within four cell doublings. Although HER2+ and HER2- circulating tumour cells have comparable tumour initiating potential, differential proliferation favours the HER2+ state, while oxidative stress or cytotoxic chemotherapy enhances transition to the HER2- phenotype. Simultaneous treatment with paclitaxel and Notch inhibitors achieves sustained suppression of tumorigenesis in orthotopic circulating tumour cell-derived tumour models. Together, these results point to distinct yet interconverting phenotypes within patient-derived circulating tumour cells, contributing to progression of breast cancer and acquisition of drug resistance.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Receptor, ErbB-2/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Female , Humans , Neoplastic Cells, Circulating/drug effects , Phenotype , Receptor, ErbB-2/deficiency , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Signal Transduction
3.
Nat Commun ; 8: 14344, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181495

ABSTRACT

Metastasis-competent circulating tumour cells (CTCs) experience oxidative stress in the bloodstream, but their survival mechanisms are not well defined. Here, comparing single-cell RNA-Seq profiles of CTCs from breast, prostate and lung cancers, we observe consistent induction of ß-globin (HBB), but not its partner α-globin (HBA). The tumour-specific origin of HBB is confirmed by sequence polymorphisms within human xenograft-derived CTCs in mouse models. Increased intracellular reactive oxygen species (ROS) in cultured breast CTCs triggers HBB induction, mediated through the transcriptional regulator KLF4. Depletion of HBB in CTC-derived cultures has minimal effects on primary tumour growth, but it greatly increases apoptosis following ROS exposure, and dramatically reduces CTC-derived lung metastases. These effects are reversed by the anti-oxidant N-Acetyl Cysteine. Conversely, overexpression of HBB is sufficient to suppress intracellular ROS within CTCs. Altogether, these observations suggest that ß-globin is selectively deregulated in cancer cells, mediating a cytoprotective effect during blood-borne metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/blood , Neoplasms/genetics , beta-Globins/genetics , Animals , Antioxidants/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/genetics , Cytoprotection/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Reactive Oxygen Species/metabolism , Stress, Physiological , Up-Regulation/genetics , beta-Globins/metabolism
4.
Sci Rep ; 5: 11870, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26149121

ABSTRACT

Ergothioneine is a histidine thio-derivative isolated in 1909. In ergothioneine biosynthesis, the combination of a mononuclear non-heme iron enzyme catalyzed oxidative C-S bond formation reaction and a PLP-mediated C-S lyase (EgtE) reaction results in a net sulfur transfer from cysteine to histidine side-chain. This demonstrates a new sulfur transfer strategy in the biosynthesis of sulfur-containing natural products. Due to difficulties associated with the overexpression of Mycobacterium smegmatis EgtE protein, the proposed EgtE functionality remained to be verified biochemically. In this study, we have successfully overexpressed and purified M. smegmatis EgtE enzyme and evaluated its activities under different in vitro conditions: C-S lyase reaction using either thioether or sulfoxide as a substrate in the presence or absence of reductants. Results from our biochemical characterizations support the assignment of sulfoxide 4 as the native EgtE substrate and the involvement of a sulfenic acid intermediate in the ergothioneine C-S lyase reaction.


Subject(s)
Ergothioneine/biosynthesis , Lyases/metabolism , Sulfenic Acids/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ergothioneine/chemistry , Escherichia coli/metabolism , Lyases/genetics , Magnetic Resonance Spectroscopy , Mycobacterium smegmatis/enzymology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sulfenic Acids/chemistry
5.
Mol Cancer Res ; 13(5): 934-43, 2015 May.
Article in English | MEDLINE | ID: mdl-25678598

ABSTRACT

UNLABELLED: Epithelial-to-mesenchymal transition (EMT) has been implicated in models of tumor cell migration, invasion, and metastasis. In a search for candidate therapeutic targets to reverse this process, nontumorigenic MCF10A breast epithelial cells were infected with an arrayed lentiviral kinome shRNA library and screened for either suppression or enhancement of a 26-gene EMT RNA signature. No individual kinase gene knockdown was sufficient to induce EMT. In contrast, grouped epithelial markers were induced by knockdown of multiple kinases, including mitogen activated protein kinase 7 (MAPK7). In breast cancer cells, suppression of MAPK7 increased E-cadherin (CDH1) expression and inhibited cell migration. In an orthotopic mouse model, MAPK7 suppression reduced the generation of circulating tumor cells and the appearance of lung metastases. Together, these observations raise the possibility that targeting kinases that maintain mesenchymal cell properties in cancer cells, such as MAPK7, may lessen tumor invasiveness. IMPLICATIONS: Suppression of MAPK7 induces epithelial markers, reduces generation of circulating tumor cells and appearance of lung metastases.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Mitogen-Activated Protein Kinase 7/metabolism , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Animals , Antigens, CD , Breast Neoplasms/blood , Cadherins/biosynthesis , Cadherins/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mitogen-Activated Protein Kinase 7/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcriptome
6.
Science ; 349(6254): 1351-6, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26383955

ABSTRACT

Prostate cancer is initially responsive to androgen deprivation, but the effectiveness of androgen receptor (AR) inhibitors in recurrent disease is variable. Biopsy of bone metastases is challenging; hence, sampling circulating tumor cells (CTCs) may reveal drug-resistance mechanisms. We established single-cell RNA-sequencing (RNA-Seq) profiles of 77 intact CTCs isolated from 13 patients (mean six CTCs per patient), by using microfluidic enrichment. Single CTCs from each individual display considerable heterogeneity, including expression of AR gene mutations and splicing variants. Retrospective analysis of CTCs from patients progressing under treatment with an AR inhibitor, compared with untreated cases, indicates activation of noncanonical Wnt signaling (P = 0.0064). Ectopic expression of Wnt5a in prostate cancer cells attenuates the antiproliferative effect of AR inhibition, whereas its suppression in drug-resistant cells restores partial sensitivity, a correlation also evident in an established mouse model. Thus, single-cell analysis of prostate CTCs reveals heterogeneity in signaling pathways that could contribute to treatment failure.


Subject(s)
Androgen Antagonists/therapeutic use , Drug Resistance, Neoplasm/genetics , Neoplastic Cells, Circulating/metabolism , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Wnt Proteins/metabolism , Androgen Antagonists/pharmacology , Animals , Benzamides , Cell Line, Tumor , Humans , Male , Mice , Neoplastic Cells, Circulating/drug effects , Nitriles , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Splicing , Sequence Analysis, RNA/methods , Signal Transduction , Single-Cell Analysis/methods , Transcriptome , Wnt Proteins/genetics , Wnt-5a Protein , Xenograft Model Antitumor Assays
7.
Cancer Res ; 74(24): 7229-38, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25339351

ABSTRACT

Modeling the hematogenous spread of cancer cells to distant organs poses one of the greatest challenges in the study of human metastasis. Both tumor cell-intrinsic properties as well as interactions with reactive stromal cells contribute to this process, but identification of relevant stromal signals has been hampered by the lack of models allowing characterization of the metastatic niche. Here, we describe an implantable bioengineered scaffold, amenable to in vivo imaging, ex vivo manipulation, and serial transplantation for the continuous study of human metastasis in mice. Orthotopic or systemic inoculation of tagged human cancer cells into the mouse leads to the release of circulating tumor cells into the vasculature, which seed the scaffold, initiating a metastatic tumor focus. Mouse stromal cells can be readily recovered and profiled, revealing differential expression of cytokines, such as IL1ß, from tumor-bearing versus unseeded scaffolds. Finally, this platform can be used to test the effect of drugs on suppressing initiation of metastatic lesions. This generalizable model to study cancer metastasis may thus identify key stromal-derived factors with important implications for basic and translational cancer research.


Subject(s)
Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/drug therapy , Stromal Cells/metabolism , Animals , Biomedical Engineering/methods , Humans , Interleukin-1beta/biosynthesis , Mice , Neoplasms/pathology , Stromal Cells/pathology , Tissue Scaffolds
8.
Science ; 345(6193): 216-20, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-25013076

ABSTRACT

Circulating tumor cells (CTCs) are present at low concentrations in the peripheral blood of patients with solid tumors. It has been proposed that the isolation, ex vivo culture, and characterization of CTCs may provide an opportunity to noninvasively monitor the changing patterns of drug susceptibility in individual patients as their tumors acquire new mutations. In a proof-of-concept study, we established CTC cultures from six patients with estrogen receptor-positive breast cancer. Three of five CTC lines tested were tumorigenic in mice. Genome sequencing of the CTC lines revealed preexisting mutations in the PIK3CA gene and newly acquired mutations in the estrogen receptor gene (ESR1), PIK3CA gene, and fibroblast growth factor receptor gene (FGFR2), among others. Drug sensitivity testing of CTC lines with multiple mutations revealed potential new therapeutic targets. With optimization of CTC culture conditions, this strategy may help identify the best therapies for individual cancer patients over the course of their disease.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Molecular Targeted Therapy , Neoplastic Cells, Circulating/drug effects , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Cell Culture Techniques , Cell Separation , Class I Phosphatidylinositol 3-Kinases , Culture , Drug Screening Assays, Antitumor/methods , Estrogen Receptor alpha/genetics , Female , Gene Frequency , Humans , Mice , Microfluidics/methods , Mutation , Neoplastic Cells, Circulating/metabolism , Phosphatidylinositol 3-Kinases/genetics , Sequence Analysis, DNA , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cell Rep ; 5(6): 1679-89, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24360956

ABSTRACT

Epithelial-mesenchymal transition (EMT) is thought to contribute to cancer metastasis, but its underlying mechanisms are not well understood. To define early steps in this cellular transformation, we analyzed human mammary epithelial cells with tightly regulated expression of Snail-1, a master regulator of EMT. After Snail-1 induction, epithelial markers were repressed within 6 hr, and mesenchymal genes were induced at 24 hr. Snail-1 binding to its target promoters was transient (6-48 hr) despite continued protein expression, and it was followed by both transient and long-lasting chromatin changes. Pharmacological inhibition of selected histone acetylation and demethylation pathways suppressed the induction as well as the maintenance of Snail-1-mediated EMT. Thus, EMT involves an epigenetic switch that may be prevented or reversed with the use of small-molecule inhibitors of chromatin modifiers.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Epithelial-Mesenchymal Transition , Protein Processing, Post-Translational , Transcription Factors/metabolism , Acetylation , Carcinogenesis/metabolism , Epigenesis, Genetic , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/physiology , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , MCF-7 Cells , Methylation , Promoter Regions, Genetic , Snail Family Transcription Factors , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL