High-throughput physical phenotyping of cell differentiation

In this report, we present multiparameter deformability cytometry (m-DC), in which we explore a large set of parameters describing the physical phenotypes of pluripotent cells and their derivatives. m-DC utilizes microfluidic inertial focusing and hydrodynamic stretching of single cells in conjunction with high-speed video recording to realize high-throughput characterization of over 20 different cell motion and morphology-derived parameters. Parameters extracted from videos include size, deformability, deformation kinetics, and morphology. We train support vector machines that provide evidence that these additional physical measurements improve classification of induced pluripotent stem cells, mesenchymal stem cells, neural stem cells, and their derivatives compared to size and deformability alone. In addition, we utilize visual interactive stochastic neighbor embedding to visually map the high-dimensional physical phenotypic spaces occupied by these stem cells and their progeny and the pathways traversed during differentiation. This report demonstrates the potential of m-DC for improving understanding of physical differences that arise as cells differentiate and identifying cell subpopulations in a label-free manner. Ultimately, such approaches could broaden our understanding of subtle changes in cell phenotypes and their roles in human biology.

[1]  M. Radmacher,et al.  From molecules to cells: imaging soft samples with the atomic force microscope. , 1992, Science.

[2]  Gideon Rechavi,et al.  Directed differentiation of human embryonic stem cells into functional retinal pigment epithelium cells. , 2009, Cell stem cell.

[3]  Sean C. Bendall,et al.  viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia , 2013, Nature Biotechnology.

[4]  David R. Myers,et al.  Cellular softening mediates leukocyte demargination and trafficking, thereby increasing clinical blood counts , 2016, Proceedings of the National Academy of Sciences.

[5]  Jason P Beech,et al.  Sorting cells by size, shape and deformability. , 2012, Lab on a chip.

[6]  P. Janmey,et al.  Cell mechanics: integrating cell responses to mechanical stimuli. , 2007, Annual review of biomedical engineering.

[7]  Todd Sulchek,et al.  Mechanical stiffness as an improved single-cell indicator of osteoblastic human mesenchymal stem cell differentiation. , 2014, Journal of biomechanics.

[8]  David M Gamm,et al.  Induced Pluripotent Stem Cell-Derived Retinal Pigmented Epithelium: A Comparative Study Between Cell Lines and Differentiation Methods. , 2016, Journal of ocular pharmacology and therapeutics : the official journal of the Association for Ocular Pharmacology and Therapeutics.

[9]  H. Blau,et al.  Nuclear reprogramming to a pluripotent state by three approaches , 2010, Nature.

[10]  Mehmet Toner,et al.  Deformability-based cell selection with downstream immunofluorescence analysis. , 2016, Integrative biology : quantitative biosciences from nano to macro.

[11]  Philip Poronnik,et al.  PKC-alpha-mediated remodeling of the actin cytoskeleton is involved in constitutive albumin uptake by proximal tubule cells. , 2005, American journal of physiology. Renal physiology.

[12]  Farshid Guilak,et al.  Viscoelastic properties of human mesenchymally-derived stem cells and primary osteoblasts, chondrocytes, and adipocytes. , 2008, Journal of biomechanics.

[13]  Dino Di Carlo,et al.  High-Throughput Assessment of Cellular Mechanical Properties. , 2015, Annual review of biomedical engineering.

[14]  Junichi Kiryu,et al.  Characterization of Human Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium Cell Sheets Aiming for Clinical Application , 2014, Stem cell reports.

[15]  Todd Sulchek,et al.  Microfluidic cellular enrichment and separation through differences in viscoelastic deformation. , 2015, Lab on a chip.

[16]  Hongshen Ma,et al.  Microfluidic deformability analysis of the red cell storage lesion. , 2015, Journal of biomechanics.

[17]  Tadashi Yokoi,et al.  Generation of retinal ganglion cells with functional axons from human induced pluripotent stem cells , 2015, Scientific Reports.

[18]  Amy C. Rowat,et al.  Screening cell mechanotype by parallel microfiltration , 2015, Scientific Reports.

[19]  B. Stanger,et al.  Adult cell plasticity in vivo: de-differentiation and transdifferentiation are back in style , 2016, Nature Reviews Molecular Cell Biology.

[20]  Yo Sup Moon,et al.  Quantitative Diagnosis of Malignant Pleural Effusions by Single-Cell Mechanophenotyping , 2013, Science Translational Medicine.

[21]  Elizabeth G Loboa,et al.  Cytoskeletal and focal adhesion influences on mesenchymal stem cell shape, mechanical properties, and differentiation down osteogenic, adipogenic, and chondrogenic pathways. , 2012, Tissue engineering. Part B, Reviews.

[22]  Enrico Bertini,et al.  The cytoskeletal arrangements necessary to neurogenesis , 2016, Oncotarget.

[23]  Yi Zheng,et al.  Recent advances in microfluidic techniques for single-cell biophysical characterization. , 2013, Lab on a chip.

[24]  Clive N Svendsen,et al.  Human iPSC‐Derived Neural Progenitors Preserve Vision in an AMD‐Like Model , 2015, Stem cells.

[25]  Caroline Brandl,et al.  In-Depth Characterisation of Retinal Pigment Epithelium (RPE) Cells Derived from Human Induced Pluripotent Stem Cells (hiPSC) , 2014, NeuroMolecular Medicine.

[26]  S. Suresh,et al.  Cell and molecular mechanics of biological materials , 2003, Nature materials.

[27]  Carlos G Dotti,et al.  Neuronal polarity: demarcation, growth and commitment. , 2012, Current opinion in cell biology.

[28]  U. Lendahl,et al.  Nestin mRNA expression correlates with the central nervous system progenitor cell state in many, but not all, regions of developing central nervous system. , 1995, Brain research. Developmental brain research.

[29]  Frank Bradke,et al.  Control of neuronal polarity and plasticity--a renaissance for microtubules? , 2009, Trends in cell biology.

[30]  Mauricio González,et al.  Cytoskeletal organization of human mesenchymal stem cells (MSC) changes during their osteogenic differentiation , 2004, Journal of cellular biochemistry.

[31]  Kyoko Ohno-Matsui,et al.  Differentiation/Purification Protocol for Retinal Pigment Epithelium from Mouse Induced Pluripotent Stem Cells as a Research Tool , 2016, PloS one.

[32]  Hongshen Ma,et al.  Microfluidic cell-phoresis enabling high-throughput analysis of red blood cell deformability and biophysical screening of antimalarial drugs. , 2015, Lab on a chip.

[33]  Julia B. Guidry,et al.  Cytoskeletal Expression and Remodeling in Pluripotent Stem Cells , 2016, PloS one.

[34]  Marie-Pier Champagne,et al.  Pax6 is required for delta-catenin/neurojugin expression during retinal, cerebellar and cortical development in mice. , 2006, Developmental biology.

[35]  U. Keyser,et al.  Real-time deformability cytometry: on-the-fly cell mechanical phenotyping , 2015, Nature Methods.

[36]  Naoki Nishishita,et al.  Pigment Epithelium-Derived Factor Secreted from Retinal Pigment Epithelium Facilitates Apoptotic Cell Death of iPSC , 2013, Scientific Reports.

[37]  A. Bennaceur-Griscelli,et al.  Morphological Analysis of Human Induced Pluripotent Stem Cells During Induced Differentiation and Reverse Programming , 2014, BioResearch open access.

[38]  Saravanan Karumbayaram,et al.  From Skin Biopsy to Neurons Through a Pluripotent Intermediate Under Good Manufacturing Practice Protocols , 2012, Stem cells translational medicine.

[39]  Yi Zhang,et al.  Embryonic stem cell and induced pluripotent stem cell: an epigenetic perspective , 2012, Cell Research.

[40]  Jane Goodall,et al.  Mitf regulation of Dia1 controls melanoma proliferation and invasiveness. , 2006, Genes & development.

[41]  Jeremy J Mao,et al.  Cytoskeletal Changes of Mesenchymal Stem Cells During Differentiation , 2007, ASAIO journal.

[42]  David W. M. Marr,et al.  Viscoelasticity as a biomarker for high-throughput flow cytometry. , 2013, Biophysical journal.

[43]  Richard Superfine,et al.  The Rho GEFs LARG and GEF-H1 regulate the mechanical response to force on integrins , 2011, Nature Cell Biology.

[44]  Vera C. Fonseca,et al.  Cellular mechanical properties reflect the differentiation potential of adipose-derived mesenchymal stem cells , 2012, Proceedings of the National Academy of Sciences.

[45]  Dino Di Carlo,et al.  Hydrodynamic stretching of single cells for large population mechanical phenotyping , 2012, Proceedings of the National Academy of Sciences.

[46]  R. Hochmuth,et al.  Micropipette aspiration of living cells. , 2000, Journal of biomechanics.

[47]  M. Ramalho-Santos,et al.  Open chromatin in pluripotency and reprogramming , 2010, Nature Reviews Molecular Cell Biology.

[48]  Jin Woong Kim,et al.  The physical origins of transit time measurements for rapid, single cell mechanotyping. , 2016, Lab on a chip.

[49]  Enrico Bertini,et al.  Cytoskeletal dynamics during in vitro neurogenesis of induced pluripotent stem cells (iPSCs) , 2016, Molecular and Cellular Neuroscience.

[50]  A. Pollock,et al.  Calbindin Expression in Renal Tubular Epithelial Cells. ALTERED SODIUM PHOSPHATE CO-TRANSPORT IN ASSOCIATION WITH CYTOSKELETAL REARRANGEMENT (*) , 1995, The Journal of Biological Chemistry.