151
|
Rosàs-Canyelles E, Dai T, Li S, Herr AE. Mouse-to-mouse variation in maturation heterogeneity of smooth muscle cells. LAB ON A CHIP 2018; 18:1875-1883. [PMID: 29796562 PMCID: PMC6019577 DOI: 10.1039/c8lc00216a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Smooth muscle cell (SMC) heterogeneity plays an important role in vascular remodeling, a life-threatening hallmark of many vascular diseases. However, the characterization of SMCs at the single-cell level is stymied by drawbacks of contemporary single-cell protein measurements, including antibody probe cross-reactivity, chemical fixation artifacts, limited isoform-specific probes, low multiplexing and difficulty sampling cells with irregular morphologies. To scrutinize healthy vessels for subpopulations of SMCs with proliferative-like phenotypes, we developed a high-specificity, multiplexed single-cell immunoblotting cytometry tool for unfixed, uncultured primary cells. We applied our assay to demonstrate maturation stage profiling of aortic SMCs freshly isolated from individual mice. After ensuring unbiased sampling of SMCs (80-120 μm in length), we performed single-SMC electrophoretic protein separations, which resolve protein signal from off-target antibody binding, and immunoblotted for differentiation markers α-SMA, CNN-1 and SMMHC (targets ranging from 34 kDa to 227 kDa). We identified a subpopulation of immature-like SMCs, supporting the recently-established mechanism that only a subset of SMCs is responsible for vascular remodeling. Furthermore, the low sample requirements of our assay enable single-mouse resolution studies, which minimizes animal sacrifice and experimental costs while reporting animal-to-animal phenotypic variation, essential for achieving reproducibility and surmounting the drawbacks of pooling primary cells from different animals.
Collapse
|
152
|
Das TK, Esernio J, Cagan RL. Restraining Network Response to Targeted Cancer Therapies Improves Efficacy and Reduces Cellular Resistance. Cancer Res 2018; 78:4344-4359. [DOI: 10.1158/0008-5472.can-17-2001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/04/2017] [Accepted: 05/21/2018] [Indexed: 11/16/2022]
|
153
|
Ding C, Zhang C, Yin X, Cao X, Cai M, Xian Y. Near-Infrared Fluorescent Ag 2S Nanodot-Based Signal Amplification for Efficient Detection of Circulating Tumor Cells. Anal Chem 2018; 90:6702-6709. [PMID: 29722265 DOI: 10.1021/acs.analchem.8b00514] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The level of circulating tumor cells (CTCs) plays a critical role in tumor metastasis and personalized therapy, but it is challenging for highly efficient capture and detection of CTCs because of the extremely low concentration in peripheral blood. Herein, we report near-infrared fluorescent Ag2S nanodot-based signal amplification combing with immune-magnetic spheres (IMNs) for highly efficient magnetic capture and ultrasensitive fluorescence labeling of CTCs. The near-infrared fluorescent Ag2S nanoprobe has been successfully constructed through hybridization chain reactions using aptamer-modified Ag2S nanodots, which can extremely improve the imaging sensitivity and reduce background signal of blood samples. Moreover, the antiepithelial-cell-adhesion-molecule (EpCAM) antibody-labeled magnetic nanospheres have been used for highly capture rare tumor cells in whole blood. The near-infrared nanoprobe with signal amplification and IMNs platform exhibits excellent performance in efficient capture and detection of CTCs, which shows great potential in cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Caiping Ding
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Cuiling Zhang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Xueyang Yin
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Xuanyu Cao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Meifang Cai
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| | - Yuezhong Xian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , China
| |
Collapse
|
154
|
Chen S, El-Heliebi A, Schmid J, Kashofer K, Czyż ZT, Polzer BM, Pantel K, Kroneis T, Sedlmayr P. Target Cell Pre-enrichment and Whole Genome Amplification for Single Cell Downstream Characterization. J Vis Exp 2018:56394. [PMID: 29863657 PMCID: PMC6101176 DOI: 10.3791/56394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Rare target cells can be isolated from a high background of non-target cells using antibodies specific for surface proteins of target cells. A recently developed method uses a medical wire functionalized with anti-epithelial cell adhesion molecule (EpCAM) antibodies for in vivo isolation of circulating tumor cells (CTCs)1. A patient-matched cohort in non-metastatic prostate cancer showed that the in vivo isolation technique resulted in a higher percentage of patients positive for CTCs as well as higher CTC counts as compared to the current gold standard in CTC enumeration. As cells cannot be recovered from current medical devices, a new functionalized wire (referred to as Device) was manufactured allowing capture and subsequent detachment of cells by enzymatic treatment. Cells are allowed to attach to the Device, visualized on a microscope and detached using enzymatic treatment. Recovered cells are cytocentrifuged onto membrane-coated slides and harvested individually by means of laser microdissection or micromanipulation. Single-cell samples are then subjected to single-cell whole genome amplification allowing multiple downstream analysis including screening and target-specific approaches. The procedure of isolation and recovery yields high quality DNA from single cells and does not impair subsequent whole genome amplification (WGA). A single cell's amplified DNA can be forwarded to screening and/or targeted analysis such as array comparative genome hybridization (array-CGH) or sequencing. The device allows ex vivo isolation from artificial rare cell samples (i.e. 500 target cells spiked into 5 mL of peripheral blood). Whereas detachment rates of cells are acceptable (50 - 90%), the recovery rate of detached cells onto slides spans a wide range dependent on the cell line used (<10 - >50%) and needs some further attention. This device is not cleared for the use in patients.
Collapse
Affiliation(s)
- Shukun Chen
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz
| | - Amin El-Heliebi
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz
| | - Julia Schmid
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz
| | | | - Zbigniew T Czyż
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM
| | | | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf
| | - Thomas Kroneis
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz; Sahlgrenska Cancer Center, University of Gothenburg;
| | - Peter Sedlmayr
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz
| |
Collapse
|
155
|
Walz JA, Mace CR. Correlation of Cell Surface Biomarker Expression Levels with Adhesion Contact Angle Measured by Lateral Microscopy. Anal Chem 2018; 90:6572-6579. [DOI: 10.1021/acs.analchem.8b00268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jenna A. Walz
- Department of Chemistry, Tufts University, 62 Talbot Avenue, Medford, Massachusetts 02155, United States
| | - Charles R. Mace
- Department of Chemistry, Tufts University, 62 Talbot Avenue, Medford, Massachusetts 02155, United States
| |
Collapse
|
156
|
Aneuploid CTC and CEC. Diagnostics (Basel) 2018; 8:diagnostics8020026. [PMID: 29670052 PMCID: PMC6023477 DOI: 10.3390/diagnostics8020026] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 12/11/2022] Open
Abstract
Conventional circulating tumor cell (CTC) detection technologies are restricted to large tumor cells (> white blood cells (WBCs)), or those unique carcinoma cells with double positive expression of surface epithelial cell adhesion molecule (EpCAM) for isolation, and intracellular structural protein cytokeratins (CKs) for identification. With respect to detecting the full spectrum of highly heterogeneous circulating rare cells (CRCs), including CTCs and circulating endothelial cells (CECs), it is imperative to develop a strategy systematically coordinating all tri-elements of nucleic acids, biomarker proteins, and cellular morphology, to effectively enrich and comprehensively identify CRCs. Accordingly, a novel strategy integrating subtraction enrichment and immunostaining-fluorescence in situ hybridization (SE-iFISH), independent of cell size variation and free of hypotonic damage as well as anti-EpCAM perturbing, has been demonstrated to enable in situ phenotyping multi-protein expression, karyotyping chromosome aneuploidy, and detecting cytogenetic rearrangements of the ALK gene in non-hematologic CRCs. Symbolic non-synonymous single nucleotide variants (SNVs) of both the TP53 gene (P33R) in each single aneuploid CTCs, and the cyclin-dependent kinase inhibitor 2A (CDKN2A) tumor suppressor gene in each examined aneuploid CECs, were identified for the first time across patients with diverse carcinomas. Comprehensive co-detecting observable aneuploid CTCs and CECs by SE-iFISH, along with applicable genomic and/or proteomic single cell molecular profiling, are anticipated to facilitate elucidating how those disparate categories of aneuploid CTCs and CECs cross-talk and functionally interplay with tumor angiogenesis, therapeutic drug resistance, tumor progression, and cancer metastasis.
Collapse
|
157
|
Kang CC, Ward TM, Bockhorn J, Schiffman C, Huang H, Pegram MD, Herr AE. Electrophoretic cytopathology resolves ERBB2 forms with single-cell resolution. NPJ Precis Oncol 2018; 2:10. [PMID: 29872719 PMCID: PMC5871910 DOI: 10.1038/s41698-018-0052-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 02/10/2018] [Accepted: 02/20/2018] [Indexed: 12/20/2022] Open
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.
Collapse
Affiliation(s)
- Chi-Chih Kang
- 1Department of Bioengineering, University of California Berkeley, Berkeley, CA 94720 USA
| | - Toby M Ward
- 2Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Jessica Bockhorn
- 2Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Courtney Schiffman
- 3Division of Biostatistics, School of Public Health, University of California Berkeley, Berkeley, CA 94720 USA
| | - Haiyan Huang
- 4Department of Statistics, University of California Berkeley, Berkeley, CA 94720 USA
| | - Mark D Pegram
- 2Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Amy E Herr
- 1Department of Bioengineering, University of California Berkeley, Berkeley, CA 94720 USA
| |
Collapse
|
158
|
Mong J, Tan MH. Size-Based Enrichment Technologies for Non-cancerous Tumor-Derived Cells in Blood. Trends Biotechnol 2018; 36:511-522. [PMID: 29559166 DOI: 10.1016/j.tibtech.2018.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 02/19/2018] [Accepted: 02/21/2018] [Indexed: 01/09/2023]
Abstract
Enumeration of circulating tumor cells (CTCs) in the bloodstream can predict prognosis and survival in cancer patients. However, CTC rarity and heterogeneity pose challenges in using them as biomarkers. Recent publications have reported new classes of circulating, non-cancerous tumor-derived cells present in cancer patients but not in healthy controls; these include cancer-associated macrophages, tumor-endothelial clusters (TECs), and cancer-associated fibroblasts (CAFs). Well-established marker-dependent CTC enrichment technologies will miss this group of circulating cells. To maximize our chance of finding useful circulating biomarkers in cancer patients, we propose the use of size-based enrichment technologies to isolate both cancerous and non-cancerous cells in circulation. We review their biological properties and discuss device features to consider in their enrichment.
Collapse
Affiliation(s)
- Jamie Mong
- Biodevices and Diagnostics, Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore
| | - Min-Han Tan
- Biodevices and Diagnostics, Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore; National Cancer Centre Singapore, Singapore 169610, Singapore; Sengkang General Hospital, Singapore 544886, Singapore; Concord Cancer Hospital, Singapore 289891, Singapore.
| |
Collapse
|
159
|
Citi V, Del Re M, Martelli A, Calderone V, Breschi MC, Danesi R. Phosphorylation of AKT and ERK1/2 and mutations of PIK3CA and PTEN are predictive of breast cancer cell sensitivity to everolimus in vitro. Cancer Chemother Pharmacol 2018; 81:745-754. [PMID: 29476223 DOI: 10.1007/s00280-018-3543-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/13/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Everolimus is the hydroxyethyl derivative of sirolimus and a strong inhibitor of mammalian target of rapamycin (mTOR). This drug has immunosuppressive and anticancer activities and the present in vitro study was aimed at identifying the cellular and molecular profiles of breast cancer cells predictive of sensitivity to everolimus. MATERIALS AND METHODS MCF-7, T-47D, ZR-75-1, CAMA-1, HCC-1500 and MCF-10A cells were used and viability was assessed using WST-1 dye. Sensitivity to everolimus was correlated with phosphorylation of AKT (Ser473/Thr308), mTOR (Ser2448), and ERK1/2 (Thr202/Tyr204) and mutational profile of KRAS, NRAS, BRAF, PIK3CA, PTEN, TSC1, TSC2 and FRAP genes. Protein phosphorylation was evaluated by AlphaScreen SureFire, while the mutational status was examined by digital droplet PCR and Sanger sequencing. RESULTS Everolimus showed a transient growth inhibition in non-tumorigenic cells, while in tumorigenic lines the drug suppressed the proliferation in a concentration-dependent manner but with different potency (IC50) and efficacy (Emax), being ZR-75-1 the most sensitive and T47D the least sensitive. MCF-7, T47D and HCC1500 had activating mutations in PIK3CA gene, while loss-of-activity PTEN mutations were detected in sensitive cell lines, including ZR-75-1, which showed no changes or minimal increase in the amount of p-AKT(Ser473/Thr308) and p-ERK1/2(Thr202/Tyr204) induced by everolimus compared to the resistant cell line T47D in which phosphorylation of AKT and ERK was increased. CONCLUSIONS Cellular levels of p-AKT(Ser473/Thr308) and p-ERK1/2(Thr202/Tyr204), activating mutations of PIK3CA and inactivating mutations of PTEN may predict response to everolimus in breast cancer cells; these findings have potential applications for treatment personalization of everolimus in breast cancer patients.
Collapse
Affiliation(s)
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, 55, Via Roma, 56126, Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | | | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, 55, Via Roma, 56126, Pisa, Italy.
| |
Collapse
|
160
|
Poudineh M, Sargent EH, Pantel K, Kelley SO. Profiling circulating tumour cells and other biomarkers of invasive cancers. Nat Biomed Eng 2018; 2:72-84. [DOI: 10.1038/s41551-018-0190-5] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 01/09/2018] [Indexed: 02/07/2023]
|
161
|
Lee MCG, Sun B. Quantitation of nonspecific protein adsorption at solid–liquid interfaces for single-cell proteomics. CAN J CHEM 2018. [DOI: 10.1139/cjc-2017-0304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Protein nonspecific adsorption that occurred at the solid–liquid interface has been subjected to intense physical and chemical characterizations due to its crucial role in a wide range of applications, including food and pharmaceutical industries, medical implants, biosensing, and so on. Protein-adsorption caused sample loss has largely hindered the studies of single-cell proteomics; the prevention of such loss requires the understanding of protein–surface adsorption at the proteome level, in which the competitive adsorption of thousands and millions of proteins with vast dynamic range occurs. To this end, we feel the necessity to review current methodologies on their potentials to characterize — more specifically to quantify — the proteome-wide adsorption. We hope this effort can help advancing single-cell proteomics and trace proteomics.
Collapse
Affiliation(s)
| | - Bingyun Sun
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
162
|
Lim LC, Lim YM. Proteome Heterogeneity in Colorectal Cancer. Proteomics 2018; 18. [PMID: 29316255 DOI: 10.1002/pmic.201700169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/17/2017] [Indexed: 01/26/2023]
Abstract
Tumor heterogeneity is an important feature of colorectal cancer (CRC) manifested by dynamic changes in gene expression, protein expression, and availability of different tumor subtypes. Recent publications in the past 10 years have revealed proteome heterogeneity between different colorectal tumors and within the same tumor site. This paper reviews recent research works on the proteome heterogeneity in CRC, which includes the heterogeneity within a single tumor (intratumor heterogeneity), between different anatomical sites at the same organ, and between primary and metastatic sites (intertumor heterogeneity). The potential use of proteome heterogeneity in precision medicine and its implications in biomarker discovery and therapeutic outcomes will be discussed. Identification of the unique proteome landscape between and within individual tumors is imperative for understanding cancer biology and the management of CRC patients.
Collapse
Affiliation(s)
- Lay Cheng Lim
- Centre for Cancer Research, Faculty of Medicine and Health Sciences, University of Tunku Abdul Rahman, Selangor, Malaysia
| | - Yang Mooi Lim
- Centre for Cancer Research, Faculty of Medicine and Health Sciences, University of Tunku Abdul Rahman, Selangor, Malaysia
| |
Collapse
|
163
|
Mondal M, Liao R, Guo J. Highly Multiplexed Single-Cell Protein Analysis. Chemistry 2018; 24:7083-7091. [PMID: 29194810 DOI: 10.1002/chem.201705014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Indexed: 12/17/2022]
Abstract
Single-cell proteomic analysis is crucial to advance our understanding of normal physiology and disease pathogenesis. The comprehensive protein profiling in individual cells of a heterogeneous sample can provide new insights into many important biological issues, such as the regulation of inter- and intracellular signaling pathways or the varied cellular compositions of normal and diseased tissues. With highly multiplexed molecular imaging of many different protein biomarkers in patient biopsies, diseases can be accurately diagnosed to guide the selection of the ideal treatment. In this Minireview, we will describe the recent technological advances of single-cell proteomic assays, discuss their advantages and limitations, highlight their applications in biology and precision medicine, and present the current challenges and potential solutions.
Collapse
Affiliation(s)
- Manas Mondal
- Biodesign Institute & School of Molecular Sciences, Arizona State University, Tempe, Arizona, 85287, USA
| | - Renjie Liao
- Biodesign Institute & School of Molecular Sciences, Arizona State University, Tempe, Arizona, 85287, USA
| | - Jia Guo
- Biodesign Institute & School of Molecular Sciences, Arizona State University, Tempe, Arizona, 85287, USA
| |
Collapse
|
164
|
Lin JMG, Kang CC, Zhou Y, Huang H, Herr AE, Kumar S. Linking invasive motility to protein expression in single tumor cells. LAB ON A CHIP 2018; 18:371-384. [PMID: 29299576 PMCID: PMC5771853 DOI: 10.1039/c7lc01008g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The invasion of malignant cells into tissue is a critical step in the progression of cancer. While it is increasingly appreciated that cells within a tumor differ in their invasive potential, it remains nearly unknown how these differences relate to cell-to-cell variations in protein expression. Here, we introduce a microfluidic platform that integrates measurements of invasive motility and protein expression for single cells, which we use to scrutinize human glioblastoma tumor-initiating cells (TICs). Our live-cell imaging microdevice is comprised of polyacrylamide microchannels that exhibit tissue-like stiffness and present chemokine gradients along each channel. Due to intrinsic differences in motility, cell subpopulations separate along the channel axis. The separated cells are then lysed in situ and each single-cell lysate is subjected to western blotting in the surrounding polyacrylamide matrix. We observe correlations between motility and Nestin and EphA2 expression. We identify protein-protein correlations within single TICs, which would be obscured with population-based assays. The integration of motility traits with single-cell protein analysis - on the same cell - offers a new means to identify druggable targets of invasive capacity.
Collapse
Affiliation(s)
- Jung-Ming G Lin
- UC-Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, CA 94720, USA.
| | | | | | | | | | | |
Collapse
|
165
|
Abstract
Cancer immunotherapy fights against cancer by modulating the immune response and is delivering encouraging results in clinical treatments. However, it is challenging to achieve durable response in all cancer patients during treatment due to the diversity and dynamic nature of immune system as well as inter- and intratumor heterogeneity. A comprehensive assessment of system immunity and tumor microenvironment is crucial for effective and safe cancer therapy, which can potentially be resolved by single-cell proteomic analysis. Single-cell proteomic technologies enable system-wide profiling of protein levels in a number of single cells within the immune system and tumor microenvironment, and thereby provide direct assessment of the functional state of the immune cells and tumor-immune interaction that could be used to evaluate efficacy of immunotherapy and to improve clinical outcome. In this chapter, we summarized current single-cell proteomic technologies and their applications in cancer immunotherapy.
Collapse
|
166
|
Neoh KH, Hassan AA, Chen A, Sun Y, Liu P, Xu KF, Wong AS, Han RP. Rethinking liquid biopsy: Microfluidic assays for mobile tumor cells in human body fluids. Biomaterials 2018; 150:112-124. [DOI: 10.1016/j.biomaterials.2017.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/21/2017] [Accepted: 10/02/2017] [Indexed: 12/27/2022]
|
167
|
Peng L, Cantor DI, Huang C, Wang K, Baker MS, Nice EC. Tissue and plasma proteomics for early stage cancer detection. Mol Omics 2018; 14:405-423. [PMID: 30251724 DOI: 10.1039/c8mo00126j] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The pursuit of novel and effective biomarkers is essential in the struggle against cancer, which is a leading cause of mortality worldwide. Here we discuss the relative advantages and disadvantages of the most frequently used proteomics techniques, concentrating on the latest advances and application of tissue and plasma proteomics for novel cancer biomarker discovery.
Collapse
Affiliation(s)
- Liyuan Peng
- Dept of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy
- Chengdu
- P. R. China
| | - David I. Cantor
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Macquarie University
- New South Wales
- Australia
| | - Canhua Huang
- Dept of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy
- Chengdu
- P. R. China
| | - Kui Wang
- Dept of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy
- Chengdu
- P. R. China
| | - Mark S. Baker
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University
- Australia
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University
- Clayton
- Australia
| |
Collapse
|
168
|
Abstract
Cell-matrix and cell-cell interactions influence intracellular signalling and play an important role in physiologic and pathologic processes. Detachment of cells from the surrounding microenvironment alters intracellular signalling. Here, we demonstrate and characterise an integrated microfluidic device to culture single and clustered cells in tuneable microenvironments and then directly analyse the lysate of each cell in situ, thereby eliminating the need to detach cells prior to analysis. First, we utilise microcontact printing to pattern cells in confined geometries. We then utilise a microscale isoelectric focusing (IEF) module to separate, detect, and analyse lamin A/C from substrate-adhered cells seeded and cultured at varying (500, 2000, and 9000 cells per cm2) densities. We report separation performance (minimum resolvable pI difference of 0.11) that is on par with capillary IEF and independent of cell density. Moreover, we map lamin A/C and β-tubulin protein expression to morphometric information (cell area, circumference, eccentricity, form factor, and cell area factor) of single cells and observe poor correlation with each of these parameters. By eliminating the need for cell detachment from substrates, we enhance detection of cell receptor proteins (CD44 and β-integrin) and dynamic phosphorylation events (pMLCS19) that are rendered undetectable or disrupted by enzymatic treatments. Finally, we optimise protein solubilisation and separation performance by tuning lysis and electrofocusing (EF) durations. We observe enhanced separation performance (decreased peak width) with longer EF durations by 25.1% and improved protein solubilisation with longer lysis durations. Overall, the combination of morphometric analyses of substrate-adhered cells, with minimised handling, will yield important insights into our understanding of adhesion-mediated signalling processes.
Collapse
Affiliation(s)
- Elaine J Su
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA.
| | | |
Collapse
|
169
|
Abstract
This review by Micalizzi et al. discusses advances in analysis and characterization of circulating tumor cells (CTCs) from patients with cancer. It focuses on the current knowledge of CTC biology and the potential clinical implications. Advances in the enrichment and analysis of rare cells from the bloodstream have allowed for detection and characterization of circulating tumor cells (CTCs) from patients with cancer. The analysis of CTCs has provided significant insight into the metastatic process. Studies on the biology of CTCs have begun to elucidate the molecular mechanisms of CTC generation, intravasation, survival, interactions with components of the blood, extravasation, and colonization of distant organs. Additionally, the study of CTCs has exposed dramatic intrapatient and interpatient heterogeneity and their evolution over time. In this review, we focus on the current knowledge of CTC biology and the potential clinical implications.
Collapse
Affiliation(s)
- Douglas S Micalizzi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, 02129, USA.,Department of Medicine, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, 02129, USA.,Department of Surgery, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, 02129, USA.,Department of Medicine, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
170
|
Vlassakis J, Herr AE. Joule Heating-Induced Dispersion in Open Microfluidic Electrophoretic Cytometry. Anal Chem 2017; 89:12787-12796. [PMID: 29110464 DOI: 10.1021/acs.analchem.7b03096] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
While protein electrophoresis conducted in capillaries and microchannels offers high-resolution separations, such formats can be cumbersome to parallelize for single-cell analysis. One approach for realizing large numbers of concurrent separations is open microfluidics (i.e., no microchannels). In an open microfluidic device adapted for single-cell electrophoresis, we perform 100s to 1000s of simultaneous separations of endogenous proteins. The microscope slide-sized device contains cells isolated in microwells located in a ∼40 μm polyacrylamide gel. The gel supports protein electrophoresis after concurrent in situ chemical lysis of each isolated cell. During electrophoresis, Joule (or resistive) heating degrades separation performance. Joule heating effects are expected to be acute in open microfluidic devices, where a single, high-conductivity buffer expedites the transition from cell lysis to protein electrophoresis. Here, we test three key assertions. First, Joule heating substantially impacts analytical sensitivity due to diffusive losses of protein out of the open microfluidic electrophoretic (EP) cytometry device. Second, increased analyte diffusivity due to autothermal runaway Joule heating is a dominant mechanism that reduces separation resolution in EP cytometry. Finally, buffer exchange reduces diffusive losses and band broadening, even when handling single-cell lysate protein concentrations in an open device. We develop numerical simulations of Joule heating-enhanced diffusion during electrophoresis and observe ∼50% protein loss out of the gel, which is reduced using the buffer exchange. Informed by analytical model predictions of separation resolution (with Joule heating), we empirically demonstrate nearly fully resolved separations of proteins with molecular mass differences of just 4 kDa or 12% (GAPDH, 36 kDa; PS6, 32 kDa) in each of 129 single cells. The attained separation performance with buffer exchange is relevant to detection of currently unmeasurable protein isoforms responsible for cancer progression.
Collapse
Affiliation(s)
- Julea Vlassakis
- Department of Bioengineering and ‡The UC Berkeley/UCSF Graduate Program in Bioengineering, University of California Berkeley , Berkeley, California 94720, United States
| | - Amy E Herr
- Department of Bioengineering and ‡The UC Berkeley/UCSF Graduate Program in Bioengineering, University of California Berkeley , Berkeley, California 94720, United States
| |
Collapse
|
171
|
Markou A, Lazaridou M, Paraskevopoulos P, Chen S, Świerczewska M, Budna J, Kuske A, Gorges TM, Joosse SA, Kroneis T, Zabel M, Sedlmayr P, Alix-Panabières C, Pantel K, Lianidou ES. Multiplex Gene Expression Profiling of In Vivo Isolated Circulating Tumor Cells in High-Risk Prostate Cancer Patients. Clin Chem 2017; 64:297-306. [PMID: 29122836 DOI: 10.1373/clinchem.2017.275503] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/11/2017] [Indexed: 11/06/2022]
Abstract
BACKGROUND Molecular characterization of circulating tumor cells (CTCs) is important for selecting patients for targeted treatments. We present, for the first time, results on gene expression profiling of CTCs isolated in vivo from high-risk prostate cancer (PCa) patients compared with CTC detected by 3 protein-based assays-CellSearch®, PSA-EPISPOT, and immunofluorescence of CellCollector® in vivo-captured CTCs-using the same blood draw. METHODS EpCAM-positive CTCs were isolated in vivo using the CellCollector from 108 high-risk PCa patients and 36 healthy volunteers. For 27 patients, samples were available before and after treatment. We developed highly sensitive multiplex RT-qPCR assays for 14 genes (KRT19, EpCAM, CDH1, HMBS, PSCA, ALDH1A1, PROM1, HPRT1, TWIST1, VIM, CDH2, B2M, PLS3, and PSA), including epithelial markers, stem cell markers, and epithelial-to-mesenchymal-transition (EMT) markers. RESULTS We observed high heterogeneity in gene expression in the captured CTCs for each patient. At least 1 marker was detected in 74 of 105 patients (70.5%), 2 markers in 45 of 105 (40.9%), and 3 markers in 16 of 105 (15.2%). Epithelial markers were detected in 31 of 105 (29.5%) patients, EMT markers in 46 of 105 (43.8%), and stem cell markers in 15 of 105 (14.3%) patients. EMT-marker positivity was very low before therapy (2 of 27, 7.4%), but it increased after therapy (17 of 27, 63.0%), whereas epithelial markers tended to decrease after therapy (2 of 27, 7.4%) compared with before therapy (13 of 27, 48.1%). At least 2 markers were expressed in 40.9% of patients, whereas the positivity was 19.6% for CellSearch, 38.1% for EPISPOT, and 43.8% for CellCollector-based IF-staining. CONCLUSIONS The combination of in vivo CTC isolation with downstream RNA analysis is highly promising as a high-throughput, specific, and ultrasensitive approach for multiplex liquid biopsy-based molecular diagnostics.
Collapse
Affiliation(s)
- Athina Markou
- Analysis of Circulating Tumor Cells Lab, Department of Chemistry, University of Athens, Athens, Greece
| | - Marifili Lazaridou
- Analysis of Circulating Tumor Cells Lab, Department of Chemistry, University of Athens, Athens, Greece
| | | | - Shukun Chen
- Institute for Cell Biology, Histology and Embryology, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Monika Świerczewska
- Department of Histology and Embryology, Poznań University of Medical Sciences, Poznań, Poland
| | - Joanna Budna
- Department of Histology and Embryology, Poznań University of Medical Sciences, Poznań, Poland
| | - Andra Kuske
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias M Gorges
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Kroneis
- Institute for Cell Biology, Histology and Embryology, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Maciej Zabel
- Department of Histology and Embryology, Poznań University of Medical Sciences, Poznań, Poland
| | - Peter Sedlmayr
- Institute for Cell Biology, Histology and Embryology, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Catherine Alix-Panabières
- University Institute for Clinical Research (IURC), Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier Saint-Eloi Hospital, EA2415, Montpellier, France
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Evi S Lianidou
- Analysis of Circulating Tumor Cells Lab, Department of Chemistry, University of Athens, Athens, Greece;
| |
Collapse
|
172
|
Phillips TM. Recent advances in CE and microchip-CE in clinical applications: 2014 to mid-2017. Electrophoresis 2017; 39:126-135. [PMID: 28853177 DOI: 10.1002/elps.201700283] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 11/11/2022]
Abstract
CE and microchip CE (ME) are powerful tools for the analysis of a number of different analytes and have been applied to a variety of clinical fields and human samples. This review will present an overview of the most recent applications of these techniques to different areas of clinical medicine during the period of 2014 to mid-2017. CE and ME have been applied to clinical chemistry, drug detection and monitoring, hematology, infectious diseases, oncology, endocrinology, neonatology, nephrology, and genetic screening. Samples examined range from serum, plasma, and urine to lest utilized materials such as tears, cerebral spinal fluid, sweat, saliva, condensed breath, single cells, and biopsy tissue. Examples of clinical applications will be given along with the various detection systems employed.
Collapse
Affiliation(s)
- Terry M Phillips
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
173
|
Renier C, Pao E, Che J, Liu HE, Lemaire CA, Matsumoto M, Triboulet M, Srivinas S, Jeffrey SS, Rettig M, Kulkarni RP, Di Carlo D, Sollier-Christen E. Label-free isolation of prostate circulating tumor cells using Vortex microfluidic technology. NPJ Precis Oncol 2017; 1:15. [PMID: 29872702 PMCID: PMC5859469 DOI: 10.1038/s41698-017-0015-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/28/2017] [Accepted: 03/05/2017] [Indexed: 01/21/2023] Open
Abstract
There has been increased interest in utilizing non-invasive "liquid biopsies" to identify biomarkers for cancer prognosis and monitoring, and to isolate genetic material that can predict response to targeted therapies. Circulating tumor cells (CTCs) have emerged as such a biomarker providing both genetic and phenotypic information about tumor evolution, potentially from both primary and metastatic sites. Currently, available CTC isolation approaches, including immunoaffinity and size-based filtration, have focused on high capture efficiency but with lower purity and often long and manual sample preparation, which limits the use of captured CTCs for downstream analyses. Here, we describe the use of the microfluidic Vortex Chip for size-based isolation of CTCs from 22 patients with advanced prostate cancer and, from an enumeration study on 18 of these patients, find that we can capture CTCs with high purity (from 1.74 to 37.59%) and efficiency (from 1.88 to 93.75 CTCs/7.5 mL) in less than 1 h. Interestingly, more atypical large circulating cells were identified in five age-matched healthy donors (46-77 years old; 1.25-2.50 CTCs/7.5 mL) than in five healthy donors <30 years old (21-27 years old; 0.00 CTC/7.5 mL). Using a threshold calculated from the five age-matched healthy donors (3.37 CTCs/mL), we identified CTCs in 80% of the prostate cancer patients. We also found that a fraction of the cells collected (11.5%) did not express epithelial prostate markers (cytokeratin and/or prostate-specific antigen) and that some instead expressed markers of epithelial-mesenchymal transition, i.e., vimentin and N-cadherin. We also show that the purity and DNA yield of isolated cells is amenable to targeted amplification and next-generation sequencing, without whole genome amplification, identifying unique mutations in 10 of 15 samples and 0 of 4 healthy samples.
Collapse
Affiliation(s)
- Corinne Renier
- Vortex Biosciences Inc., 1490 O’Brien Drive, Suite E, Menlo Park, CA 94025 USA
| | - Edward Pao
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, PO Box 951600, Los Angeles, CA 90095 USA
| | - James Che
- Vortex Biosciences Inc., 1490 O’Brien Drive, Suite E, Menlo Park, CA 94025 USA
| | - Haiyan E. Liu
- Vortex Biosciences Inc., 1490 O’Brien Drive, Suite E, Menlo Park, CA 94025 USA
| | | | - Melissa Matsumoto
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, PO Box 951600, Los Angeles, CA 90095 USA
| | - Melanie Triboulet
- Department of Surgery, Stanford University School of Medicine, MSLS Bldg, 1201 Welch Road, Stanford, CA 94305 USA
| | - Sandy Srivinas
- Department of Medicine, Stanford University School of Medicine, 875 Blake Wilbur Drive, Stanford, CA 94305 USA
| | - Stefanie S. Jeffrey
- Department of Surgery, Stanford University School of Medicine, MSLS Bldg, 1201 Welch Road, Stanford, CA 94305 USA
| | - Matthew Rettig
- Departments of Medicine Urology, UCLA Medical Center, Los Angeles, CA 90095 USA
- Department of Medicine, VA Greater Los Angeles Healthcare System-West Los Angeles, Los Angeles, CA 90073 USA
- Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095 USA
| | - Rajan P. Kulkarni
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, PO Box 951600, Los Angeles, CA 90095 USA
- Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095 USA
- California NanoSystems Institute, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095 USA
- Division of Dermatology, UCLA Medical Center, 52-121 CHS, Los Angeles, CA 90095 USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, PO Box 951600, Los Angeles, CA 90095 USA
- Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095 USA
- California NanoSystems Institute, 570 Westwood Plaza, Building 114, Los Angeles, CA 90095 USA
| | | |
Collapse
|