Microfluidics for single cell analysis Huabing Yin1 and Damian Marshall2 Substantial evidence shows that the heterogeneity of individual cells within a genetically identical population can be critical to their chance of survival. Methods that use average responses from a population often mask the difference from individual cells. To fully understand cell-to-cell variability, a complete analysis of an individual cell, from its live state to cell lysates, is essential. Highly sensitive detection of multiple components and high throughput analysis of a large number of individual cells remain the key challenges to realise this aim. In this context, microfluidics and lab-on-a-chip technology have emerged as the most promising avenue to address these challenges.In this review, we will focus on the recent development in microfluidics that are aimed at total single cell analysis on chip, that is, from an individual live cell to its gene and proteins. We also discuss the opportunities that microfluidic based single cell analysis can bring into the drug discovery process. Addresses 1 Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK 2 LGC Ltd, Teddington, UK Corresponding author: Yin, Huabing (huabing.yin@glasgow.ac.uk) Current Opinion in Biotechnology 2012, 23:110–119 This review comes from a themed issue on Analytical biotechnology Edited by Wei E. Huang and Jizhong Zhou Available online 29th November 2011 0958-1669/$ – see front matter # 2011 Elsevier Ltd. All rights reserved. DOI 10.1016/j.copbio.2011.11.002 Introduction Cellular analysis underpins many fields including life science, diagnostics, the pharmaceutical industry and renewable energy [1,2]. Conventional cell-based assays measure the average response from a population of cells, assuming that an average response is representative of a typical cell within a population. However, this simplification can result in a misleading interpretation. For example, an average of 50% protein expression in a cell population can represent either a 100% response in half the cells or a 50% response in all. A plethora of evidence has shown cellular heterogeneity commonly exists within an isogenic or clonal population [3]. This heterogeneity can arise through genetic drift, differences in cell development or cell cycle status, differences in cell age due to proliferation and passaging as well as non-genetic heterogeneity arising from the inherent stochasticity of cellular processes. Whether in isolation or caused through a combination of the above events, cellular heterogeneity can dramatically influence cellular decision making and cell fate [4], however, this can be masked by the average response from a population. One approach to solve this dilemma is to analyse a population at individual cell level. However, multiple individual cells are required to obtain statistically meaningful data, and therefore high throughput analysis is critical. A number of single cell analysis methods have been established. Some conventional examples are listed in Table 1. Microscopic imaging of a cell is the most obvious approach and has been well established for wide range of applications, such as physiological studies, measurements of gene and protein expression. However, assays on single cells are difficult to perform. The patch-clamp technique enables highly sensitive measurement of changes in ion channels, however it requires high skills to perform and has limitations when detecting complex changes. Furthermore, both of these methods are low throughput. Conventional high throughput tools for single cell analysis include well established methods such as Flow Cytometry (and Imaging Flow Cytometry, Fluorescence Activated Cell Sorting) that can detect, sort and collect cells with desired properties. However, as data are only collected at a single time point, these techniques still do not permit dynamic monitoring of cell response. Microfluidics has emerged as a powerful enabling technology for investigating the inherent complexity of cellular systems [5]. Typical microfluidic channels have dimension of tens to hundreds of microns that are comparable to the size of a single cell (10 mm in size and roughly 1 pL in volume) [5]. At this length scale, the physical behaviour of fluids is fundamentally different from that seen in large channels: Laminar flow forms when two fluid streams come together and mixing of molecules across their interface only occurs through diffusion. This phenomenon permits the accurate realisation of complex molecular trajectories and has been effectively exploited in many circumstances, such as in gradient formation [6,7] and in localised stimuli delivery to subcellular compartments [8]. In addition to the conventional methods already described, new microtechnology based tools for single cell analysis have also emerged in the last decade; for example, optical/magnetic tweezing and the use of Available online at www.sciencedirect.com Current Opinion in Biotechnology 2012, 23:110–119 www.sciencedirect.com patterned substrates (Table 1). However, these can only address challenges in one or a few aspects of single cell analysis. However, from an individual live cell to its gene and proteins. By contrast, microfluidics has been rapidly developed into a powerful approach capable of integrating multiple functions for micrototal analysis of biological systems (mTAS) and single cell analysis [2,9 ]. A rapid increasing body of new discoveries have been enabled by microfluidics that would not be possible otherwise. As discussed in this review, whole-genome molecular haplotyping of single human cells has recently been achieved using microfluidic technology [10 ]. It is arguable that microfluidics will be a central part of the next generation sequencing focused towards personal genomics. Capabilities in other single cell ‘omic’ studies, including metabolomics, transcriptomics and proteomics [11 ] have also been significantly accelerated partly owing to the rapid development of microfluidics. This advancement will enable information on the subtle variability that exists in biological systems to be revealed, and will have significant implications for understanding the progression of a disease as well as potential of transforming system biology. As an enabling technology, microfluidics owes its power to handing small volumes of liquid (nanoliter to picoliter), streamlining multiple procedures on a single chip, with scope for parallelization. Recently developed droplet-microfluidics has also emerged as a new forerunner for single cell encapsulation and analysis with massive parallelization. High throughput screening of rare cells to a drug library has been achieved, providing addition information on cell heterogeneity response [12 ]. The use of microdroplet confinement has enabled new insights into the nature of quorum sensing, suggesting it is a ‘‘cell-autonomous mechanism for diffusion or efficiency sensing’’ [9 ]. In this review, we will discuss various microfluidic applications for single cell analysis. We focus on recent developments aimed at total single cell analysis on chip, that is, from an individual live cell to its gene and proteins. We recognise the rapidly expanding number of applications for single cell analysis and do not attempt to cover all these topics. Instead, we will focus on the opportunities that microfluidic based single cell analysis can bring into the drug discovery process. Single cell analysis on chip: overview Most cellular processes are not isolated single events, but are interconnected and hierarchically organised from molecules to a whole cell. As such, multiparameter analysis, for example, coupling epigenetic to gene expression or protein expression to physiological measurement at the level of single cells would enable a better understanding of the whole process. To achieve this, a complete analysis of an individual cell, from its live state to cell lyses, is essential. We define such a process as ‘‘total analysis of single cells’’ and list it as one of the three strategies that could be employed for cell analysis on chip (Figure 1). The three strategies are roughly categorised by the subject of interest, that is, whole cells or cell lysates or both, and therefore the associated on chip operations vary. In the last decade numerous technologies have been developed for each operation making it impracticable to list them all [1,2,13,14]. However, a few major approaches are illustrated for each operation. Although it appears that ‘‘total analysis of single cells’’ is just a sequential combination of the other two strategies (i.e. using whole cell and cell lysates respectively), it is far from trivial. The total amount of analyte in a single cell is Microfluidics technology Yin and Marshall 111 Table 1 Comparison of exampled approaches to single cell analysis Approaches Main applications Key advantage Key disadvantage Microscopic imaging  Morphological studies  Gene and protein expressions  Intracellular communications Generic and well established Difficult to perform assays on single cells Patch clamp  Ion channel studies Very sensitive Limited applications Flow cytometry  Gene and protein expression  Population studies Very high throughput Requires labeled cells in suspension Tweezing (e.g. optical, magnetic)  Manipulation  Single cell mechanics Low force range (pN) Requires complex optical systems Patterned substrates  Cell–cell communication studies  Controlled cell proliferation and differentiation, guidance Simple and versatile Requires fabrication capability Microfluidics  All of the above, that is, a wide range of applications from cell manipulation to total single cell analysis Enabling technology for integrated total single cell analysis Has not yet gained popular acceptance www.sciencedirect.com Current Opinion in Biotechnology 2012, 23:110–119 very low (N.B. a single somatic cell weighs 500 pg) [15]. Furthermore, the majority of the targets of interest are of low copy number and among abundant interferences. These complexities and limitations make the detection of an analytes in a single living cell highly challenging. Whether the combination of these operations is even capable of appropriately sensitive detection is perhaps the first thing to consider. For example, mass spectrometry is the major tool for proteomics. However, integration of mass spectrometry based proteomics and on chip single cell analysis has yet to be achieved. The challenges for single cell analysis on chip are substantial but also provide great opportunities to advance technologies in the field of microfluidics and its associated applications (see below). Single cell analysis on chip: challenges and prospects A detailed understanding of the complex heterogeneity in cell populations and its impact on cell behaviour and biological responses can only be achieved using highly sensitive methods with resolution at the single cell and ideally subcellular level (e.g. down to the level of single molecules). In addition, since the processes occur with varying time scales (e.g. from seconds to hours), dynamic control of conditions is essential. However, to achieve total single cell analysis, the development of high throughput systems capable of handling and analysing individual cells is essential. In this section, we highlight the recent advances towards this goal. Spatiotemporal single cell manipulation Several single cell immobilisation methods have been developed, including microwells and traps. Traps can function via a range of means including physical geometry, hydrodynamics, magnetic force, dielectrophoresis and optical or acoustic tweezers [16,17 ,18] (Figure 2). In the early stages of trap development, enhancement of trapping efficiency has been a major focus – single cell trapping efficiency has now reached 97% with optimised trap architectures [18]. Recent development has seen a tendency towards cell–cell interaction and long term measurement of cell activities [18,20,21,22 ,23]. Of special interest is recent work that tracks the lineages of hundreds of single cells in parallel for detailed study of the time scales of heterogeneity in a population [22 ] (Figure 2). Via a similar method, the mechanisms governing hematopoietic stem cell fate decision (e.g. selfrenewal and differentiation processes) were revealed [23]. Recently, hydrodynamic cell trapping systems have shown great promise for high throughput handing and manipulation of single cells that would otherwise be impractical [17 ,19 ,24 ] (Figure 2). Various designs of microwell, weir and microjail arrays have been used to passively trap hundreds of single cells. Controlled pairing and fusing of different cell types for fusionmediated reprogramming has also been achieved [19 ] (Figure 2). Successful long term observation of the response of normal and disordered single cells to drugs 112 Analytical biotechnology Figure 1 Strategies (a) Whole cell (Biological analysis) - On chip culture - Immobilisation - Trapping - Chemical - Biological (e.g cell-cell) - Physical - Label-based (e.g. fluorescence luminescence) - Label free (e.g. impedance, Raman, AFM) - μwell - μFACS - DEP - Optical tweezers - Dynamic flow B4. DetectionB3. Separation - CE - Affinity binding - qPCR, RT-PCR - Electrochemical - ELISA - ESI-MS, MS B. Cell lysates (B2-4) (b) Cell lysates (Biochemical analysis) A. Whole cell (A1-4) Total single cell analysis Cells (clonal/wild) - On chip culture - Immobilisation - Trapping - Chemical - Electrical - Physical - Optical B1. Live cell handling B2. Lysis A1. Live cell handling A2. Stimulation A3. inspection (non-invasive) A4. Selection Operations Current Opinion in Biotechnology Overview of microfluidic approaches for single cell analysis via three different strategies. The total single cell analysis requires the sequential combination of strategies A and B. The approaches listed for each operation are not exhaustive. More detail can be found in several excellent reviews [1,2,13,14]. Abbreviations: CE, Capillary Electrophoresis; ESIMS, Electrospray Ionization Mass Spectrometry; MS, Mass Spectrometry; DEP, Dielectrophoresis; ELISA, Enzyme linked Immunosorbent Assays; qPCR, quantitative Polymerase Chain Reaction; RT-PCR, Reverse Transcription Polymerase Chain Reaction; mFACS, Micro Fluorescence Activated Cell Sorting. Current Opinion in Biotechnology 2012, 23:110–119 www.sciencedirect.com demonstrates the potential of microfluidics in personalised diagnostics [24 ]. Single living cell detection Although cell response can be measured quantitatively in many ways, non-invasive methods (and treatments) are required for living cell detection. At present fluorescence techniques remain the most common methods owing to their established nature and abundance of commercially available probes. Since cell-to-cell variability is temporally and spatially dependent [4], high content quantification of all components involved would be ideal. A single time pointfluorescencemeasurement,such as flowcytometry,is well suited for high throughput single component quantification. Spatiotemporal measurement of protein translocation, interactions and modification rely on imaging technologies such as Fluorescence Resonance Energy Transfer (FRET) and Quantitative Time-Lapse Fluorescence Microscopy [13,25]. However, the large data rich files generated by these techniques compromise through- put. Recently, an intriguing approach has been developed for both high content and high throughput detection [26]. It used a photomultiplier to combine one-dimensional imaging with microfluidic flowcytometry and demonstrated its usefulnessinhighcontentscreeningwitha speedofseveral thousands cells per second. This work, together with the work from Chung et al. [27] illustrates that integration between microfluidics, automation and conventional microscopy could expand the ability of a biological lab for high resolution and large-scale quantitative exper- iments. Development of label free techniques continues. A powerful method capable of measuring growth rate at the single cell level was developed using a Suspended Microchannel Resonator technique coupled with microfluidic control [28 ]. It is envisaged that this method will contribute to our understanding of many cellular processes that affect cell growth. Cell lysate analysis Substantial developments have been made to integrate major analytical methods currently used for genomics and proteomic for single cell analysis [11 ]. A few notable examples that could contribute to future developments are discussed below. Gene Gene expression analysis of single cells has been demonstrated by various groups [29,30 ,31]. For example, Toriello et al. have developed integrated microfluidic devices that couple single cell selection and capture, enzymatic reaction and quantitative detection all on a single platform [30 ] (Figure 3). By quantifying mRNA from two distinct populations at the single cell level, they illustrated that stochastic variations in gene expression and silencing within single cells is masked by bulk measurements [30 ]. Quake and colleagues have pioneered large-scale gene expression analysis from single cells. The technology they developed has been exploited in a wide range of applications, such as a recent work on the whole-genome molecular haplotyping of single human metaphase cell [10 ] (Figure 3). Indeed, developments in this field advance rapidly, and many technologies have started to enter commercial markets. Examples of companies offering the technology include Fluidigm Corporation, Oxford Nanopore Technology, and Genechip Affymetrix. These technologies are massively expanding the throughput for gene expression measurements as well our understanding of the differential gene expression profiles that underlie cell behaviours. For example, Petriv et al. recently used the Fluidigm dynamic array system to perform 80,000 RTqPCR assays to map the expression of micro RNA’s in the hematopoietic hierarchy and showed using single cells the major reprogramming events that occurs upon cell differentiation [32]. Epigenetics Changes in gene expression within a cell population can be caused by mechanisms other than the underlying DNA sequence. Processes such as DNA methylation or histone deacetylation can activate or silence genes without genomic alteration. These epigenetic processes are a major factor in non-Mendelian disease such as Alzheimer’s and Parkinson’s disease [33] and can influence the way cells behave in response to drugs [34]. Unlike gene expression studies the application of single cell approaches to epigenetic analysis has so far been limited. However, a recent study by Kantlehner et al. [35] demonstrated an approach for DNA methylation profiling within the regulatory CpG islands of single cells for genes which are aberrantly methylated in several types of cancer. Their single cell approach was aided by the fact they could reduce sample volumes down from the standard 0.2–1.5 mL PCR volumes to 5 mL which increased their experimental success rate. This type of analysis would therefore lend itself to microfluidic approaches where the reactions volumes could be reduced even further and it is probably only a matter of time before these studies are reported in the literature. Protein In comparison to gene expression analysis, measuring proteins from a single cell represents another level of challenge [11 ,14]. This is due to the extremely low concentrations of proteins (sometimes less than 1000 copies per cell) and that, unlike DNA or RNA, proteins cannot be directly amplified. Currently, single cell proteomics is still in its infancy. Two main development focuses are prominent, namely, sensitivity enhancement and improved integration and automation. Microfluidics technology Yin and Marshall 113 www.sciencedirect.com Current Opinion in Biotechnology 2012, 23:110–119 114 Analytical biotechnology Figure 2 1 2 1 Load green cells ‘up’ 2 Transfer cells ‘down’ 3 Load red cells ‘down’ (a) (b) (c) t=0 500 Taken from D. Di Carlo et. al, Analytical Chemistry 750 1000 1200 min off 30 µm off on21 Pho84 Hsp12 Rps8b Taken from A. M. Skelly et. al, Nature Methods (d) Scale bar: 5µm All scale bars: 10µm Taken from A.C. Rowat et. al, PNAS 1E1D1C1B Gd V~ 1A Cells Live Dead 488nm 555nm 1 Dyes 50µm2 2A 2B 2D Laser Slit 2C 2E Taken from E. Brouzes et. al, PNAS Scale bar: 50 µm Current Opinion in Biotechnology Current Opinion in Biotechnology 2012, 23:110–119 www.sciencedirect.com Innovative combinations of microfluidic devices and optical microscopy have proven to be an efficient way to cope with the detection of low number of proteins [36 ,37]. The whole process is essential to this, namely single cell manipulation, separation and detection all in one device. This not only minimises loss of protein and reduces contamination but also enables a correlation between protein expression and the phenotypic stage of a singe cell at a specific time point (i.e. when it is lysed). A good example is shown in the work from Huang et al. where individual b2 adrenergic receptors from a single cell were successfully counted in a microfluidic channel using cylindrical optics [36 ]. With the aim towards single cell proteomics, Salehi-Reyhani et al. have developed an integrated microfluidic antibody capture chip and demonstrated detection of the human tumour suppressor protein p53 from a single cell [37]. Despite many advances in fluorescence based technologies, problems in spectra overlapping and uncertainties from labeling procedures ultimately restrict the total number of simultaneous measurements. A promising alternative is to combine single cell analysis with mass spectrometry. Currently, full integration has been challenged by a lack of an effective interface between the mass spectrometer and miniaturised sample preparation Microfluidics technology Yin and Marshall 115 (Figure 2 Legend) Single cell manipulation on chip. (A) Cell trapping by hydrodynamic focusing principles. (1) A schematic and (2) A phase contrast image of an array of high quantity single-cell isolates. Reproduced by permission of the American Chemical Society [17 ]. (B) Single cell manipulation enabling precise control of cell-to-cell contact for cell fusion. (1) to (3) show schematics and corresponding red/green micrographs of a three-step cellloading protocol. Reproduced by permission of Nature [19 ]. (C) Restrained and monitored growth of individual cells allowing interrogation of the lineage of single cells. (1) Bright field image showing single progenitor cells constrained to grow in a line. (2) Variations of three different protein expressions in lineages of cells were revealed. Reproduced by permission of the National Academy of Sciences [22 ]. (D) Droplet microfluidic for high throughput single cell manipulation and analysis. A series of schematics (1) and corresponding micrographs (2) showing 5 optimised modules employed for analysis of cell viability. Reproduced by permission of the National Academy of Sciences [12 ]. Figure 3 cell-sorting region (a) control channels amplification region (c) (b) 2 Taken from H.C. Fan et. al, Nature biotechnology 3 Sample inlet 3 valve pump RTDs Reactor Heater Gold cell capture pad Hold chamber Affinity capture chamber Anode CE separation channel 1 cm Taken from N.M. Toriello et. al, PNASTaken from J.S. Mellors et. al, Analytical chemistry 50 heme α 23+ α 24+ α 22+ α 21+ α 20+ α 19+ α 18+β 22+ β 21+ β 20+ β 19+ β 18+ β 17+ β 16+ 40 30 20 10 0 50 40 30 20 IoncountIoncount 10 0 600 650 700 750 800 m/z 850 900 950 1000 600 650 700 750 800 m/z 850 900 950 1000 80 μm Chromosomes released by protease digestion Partitioning of chromosome suspension into 48 chambers Capture of a metaphase cell 1 5. Multiple strand displacement amplification of isolated chromosomes 6. Retrieval of amplified DNA for downstream analyses 100 μm 4.3.2. 1. Single chromosome C SC B B Current Opinion in Biotechnology Individual cell analysis from a live cell to gene or protein expressions. (A) An integrated microfluidic approach for whole-genome molecular haplotyping of single cells. (1) An overview image of the device. (2–6) The stages that a cell progresses through during analysis. Reproduced by permission of Nature [10 ]. (B) integration of microfluidic device with ESI-MS for on line single cell analysis. (1) A diagram showing the basic operation. (2) Mass spectra generated by a normal single cell lysis and (3) by a cell with higher haemoglobin content. Reproduced by permission of the American Chemical Society [38 ]. (C) An integrated device for gene expression analysis from a single cell. The processes of single cell capture, mRNA transcription and amplification were all integrated on chip. Reproduced by permission of the National Academy of Sciences [30 ]. www.sciencedirect.com Current Opinion in Biotechnology 2012, 23:110–119 platform [38 ,39] (Figure 3). However, this is starting to be addressed. For example, Mao et al. [40], have recently reported the development of a microfluidic system which allows high throughput nanoelectrospray into the mass spectrometer providing sensitivity comparable to the larger volume standard capillary emitters. While this is not a fully integrated system it does open the possibility for handling the volumes obtained from single cells and presenting them to the mass spectrometer. Achieving this complete integration between the two platforms will be an important step forward towards single cell proteomics. Metabolites The cellular metabolome can provide a highly sensitive measure of a cellular phenotype with quantification of intracellular metabolite concentrations forming an integral part of systems biology and pre-clinical drug toxicity analysis [41]. Similar to protein analysis, single cell metabolomics is challenging due to the small quantities of analytes which are present in single cells, with a typical 1 pL cell volume containing metabolite concentrations in the low femtomole range [15]. In common with single cell protein analysis, direct amplification of the analytes cannot be performed. To date, most metabolite profiling studies on single cells have used approaches based on molecular sensors such as FRET [42] which limits the analysis to specific metabolites and prevents ‘omics’ level detection. More recently however, mass spectrometry based approaches have reached the limits of detection required for single cell metabolomics making the technology suitable for use in combination with microfluidic systems [38 ,43]. For example, Zenobi’s group developed a microfluidic device which can position single cells onto a specialisedslideformetabolome analysis ofADP,ATP, GTP,and UDP-Glucose by matrix assisted laser desorption ionization (MALDI) mass spectrometry. Meanwhile, Ramsey’s group coupled a microfluidic system which incorporated elements for single cell lysis, a solution electrophoresis channel, where cellular constituents can be separated, and an electrosmotic pump to direct the eluted cell components to the mass spectrometer. These microfluidic based mass spectrometry systems, while still at the experimental stage, open up the possibility for multiple simultaneous quantitative detection of multiple metabolites at the single cell level and could uncover the subtle metabolite concentration differences that are currently hidden due to stochastic variability. Cells in droplets In the last few years droplet microfluidics has emerged as a promising avenue for single cell encapsulation, dynamic living cell assay, and single cell immunoanalysis [12 ,44,45] (Figure 2). In these cases, microfluidic devices enable high throughput generation of femtoliter sized and picoliter sized aqueous droplets in an immiscible carrier, such as oil. These droplets are effectively nanolabs that accommodate single cells and host all subsequent reactions. High throughput cytotoxicity screening of drugs [12 ] (Figure 2), in-droplet cell lysis and intracellular content analysis have all been illustrated [46]. An advantage of in-droplet analysis is that the droplet confines the lysate, preventing its dilution through diffusion. Currently, most of these analyses are based on fluorescence and therefore suffer from the limitations described above. Recently, online Mass spectrometry of individual microdroplets has been demonstrated [47 ]; this could greatly enhance the sensitivity of droplet-based single cell analysis. Microfluidic single cell analysis in drug discovery Cell based assays form fundamental practices at various stages of the drug discovery process. To reduce the cost of discovery, high throughput screening using robotics and multi-well plates is the principal tool for pharmaceutical industry. In general, the average response from a population in a well (tens or hundreds of cells) is used as the readout. However, the increasing evidence of heterogeneous responses from individual cells invites the introduction of new strategies capable of revealing information at both the individual and population level. We envisage that the development of microfluidic single cell analysis is oneof the most attractive approaches towards this goal, and illustrate its potential with a few recent examples below. Concentration profiling The concentration and time course of a drug (and multiple drugs) interacting with a cell are essential in evaluating its efficacy [48]. Laminar flow within microfluidic devices provides a unique advantage to create purpose designed concentration profiles (e.g. gradients) for various applications, such as the study of chemotaxis [7]. Dependent on the cell type studied, the drug concentration can be delivered by either flowing over an adhered cell layer on a substrate or by encapsulation together with cells in a microdroplet [12 ]. In the case of adhered cells, different gradient generators have been reported for creating concentration profiles for the pharmacological screening of voltage-gated human hERG K+ channels [49] and toxicity tests [50]. Such systems enable the rapid acquisition of a large amount of high content data that is essential to draw statistically meaningful conclusions. It should also be noted that shear stress can induce similar response to the chemical stimuli for adherent cells [51] and the high flow rate used for gradient generation might damage shear sensitive cells [50]. The droplet-based cytotoxicity screen is of specific interest for large scale single cell based screening [12 ] as it offers high throughput, precise delivery and powerful manipulation permitting a multitude of possibilities. Miniaturisation for future discoveries In recognition of the challenges that current target-driven drug discovery faces, there is an ongoing paradigm shift 116 Analytical biotechnology Current Opinion in Biotechnology 2012, 23:110–119 www.sciencedirect.com towards pathway-driven drug discovery [52]. Multiparameter phenotype profiling of a compound is essential to understand the biological pathway but faces many challenges [52]. In this context, microfluidic technology will be one of the enabling technologies to address these challenges, as exemplified in the many new discoveries that would not be possible by conventional methods [53,54]. For example, recently, Bao et al. has developed a microfluidic system that enables precise timing control with integration of multiple simultaneous experiments. With this system, tracking responses of individual cells across multiple stimulations was achieved, leading to the new finding that the variability in G-protein-Coupled Signalling is associated with long-lived cell state difference rather than from stochasticity [54]. Conclusions The impact of the rapid expansion of high throughput single cell analysis is evident in its great potential for numerous applications, including drug discovery, diagnostics, cancer research, regenerative medicine, system and synthetic biology, and many others. The implementation of microfluidic technologies in single cell analysis is one of the most promising approaches that not only offers infor- mationrich,highthroughputscreeningbutalsoenablesthe creation of innovative conditions that are impractical or impossible by conventional means. The possibilities for distinguishing the difference between individual cells and the benefits from miniaturisation (e.g. confinement) have led to many discoveries in both traditional biopharmaceutical communities and in emerging fields such as synthetic biology [9 ,52]. Recent research has only just started to show the full potential of microfluidics in this field. Major challenges still remain. Full integration of microfluidic (and the miniaturised platforms) for single cell manipulation with established analytic methods, such as mass spectrometry and NMR, traditionally used for high sensitivity single molecule detection is still some way off. Developing effective strategies for extracting system level information the very large data sets generated by total single cell analysis of population is essential. New breakthroughs in these areas will enable microfluidics approaches to transition from an interesting research topic to routinely used tool for drug screening and diagnostics. Acknowledgements HY acknowledge support from the Royal Academy of Engineering, also the Engineering and Physical Science Research Council (EPSRC). DM acknowledges support from the UK National Measurement System. References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest 1. El-Ali J, Sorger PK, Jensen KF: Cells on chips. Nature 2006, 442:403-411. 2. Dittrich PS, Tachikawa K, Manz A: Micro total analysis systems. Latest advancements and trends. Anal Chem 2006, 78:3887-3907. 3. Walling MA, Shepard JRE: Cellular heterogeneity and live cell arrays. Chem Soc Rev 2011, 40:4049-4076. 4. Perkins TJ, Swain PS: Strategies for cellular decision-making. Mol Syst Biol 2009, 5:12. 5. Whitesides GM: The origins and the future of microfluidics. Nature 2006, 442:368-373. 6. Ji BZ, Cusack M, Freer A, Dobson PS, Gadegaard N, Yin HB: Control of crystal polymorph in microfluidics using molluscan 28 kDa Ca(2+)-binding protein. Integr Biol 2010, 2:528-535. 7. Jeon NL, Baskaran H, Dertinger SKW, Whitesides GM, Van de Water L, Toner M: Neutrophil chemotaxis in linear and complex gradients of interleukin-8 formed in a microfabricated device. Nat Biotechnol 2002, 20:826-830. 8. Takayama S, Ostuni E, LeDuc P, Naruse K, Ingber DE, Whitesides GM: Selective chemical treatment of cellular microdomains using multiple laminar streams. Chem Biol 2003, 10:123-130. 9.  Vincent ME, Liu WS, Haney EB, Ismagilov RF: Microfluidic stochastic confinement enhances analysis of rare cells by isolating cells and creating high density environments for control of diffusible signals. Chem Soc Rev 2010, 39:974-984. An insightful review on studying single cells in confined microdroplet environments. The confinement limits the diffusion of secreted signalling molecules and therefore enables an individual cell to rapidly condition its microenvironment. Unique advantages of this approach were summarised, including observations of new function and behaviours of bacteria (e.g. quorum sensing as cell-autonomous mechanism for diffusion), increase in detection speed, and enhancement in cultivation of rare cells. 10.  Fan HC, Wang JB, Potanina A, Quake SR: Whole-genome molecular haplotyping of single cells. Nat Biotechnol 2011, 29: 51-+. A good demonstration of exploiting microfluidic technology for wholegenome molecular haplotyping of single human cells. The authors claimed this is the first demonstration of a molecular-based, wholegenome haplotyping technique amenable for personal genomics. 11.  Wang DJ, Bodovitz S: Single cell analysis: the new frontier in ‘omics’. Trends Biotechnol 2010, 28:281-290. 12.  Brouzes E, Medkova M, Savenelli N, Marran D, Twardowski M, Hutchison JB, Rothberg JM, Link DR, Perrimon N, Samuels ML: Droplet microfluidic technology for single-cell highthroughput screening. Proc Natl Acad Sci USA 2009, 106:14195-14200. An intriguing illustration of droplet-based microfluidic technology for high throughput screening of single mammalian cells. This integrated microfluidic system allowed quantitative cytotoxicity screening of a library of drugs with encapsulated human monocytic U937 cells. An interesting optically coded droplet library for droplet identification was devised. 13. Muzzey D, van Oudenaarden A: Quantitative time-lapse fluorescence microscopy in single cells. Annu Rev Cell Dev Biol 2009, 25:301-327. 14. Rubakhin SS, Romanova EV, Nemes P, Sweedler JV: Profiling metabolites and peptides in single cells. Nat Methods 2011, 8(4 Suppl.):S20-S29. 15. Schmid A, Kortmann H, Dittrich PS, Blank LM: Chemical and biological single cell analysis. Curr Opin Biotechnol 2010, 21:12-20. 16. Voldman J: Electrical forces for microscale cell manipulation. Annu Rev Biomed Eng 2006, 8:425-454. 17.  Di Carlo D, Aghdam N, Lee LP: Single-cell enzyme concentrations, kinetics, and inhibition analysis using highdensity hydrodynamic cell isolation arrays. Anal Chem 2006, 78:4925-4930. The authors have pioneered the development of hydrodynamic trapping of cells using microstructures and demonstrated a robust, fast way of forming high density single cell arrays. The ordered single-cell arrays were applied for measuring single-cell enzyme kinetics, illustrating its high throughput and robust nature. Microfluidics technology Yin and Marshall 117 www.sciencedirect.com Current Opinion in Biotechnology 2012, 23:110–119 18. Kobel S, Valero A, Latt J, Renaud P, Lutolf M: Optimization of microfluidic single cell trapping for long-term on-chip culture. Lab Chip 2010, 10:857-863. 19.  Skelley AM, Kirak O, Suh H, Jaenisch R, Voldman J: Microfluidic control of cell pairing and fusion. Nat Methods 2009, 6:147-152. Weir structured microfluidic traps were developed to capture and pair thousand of cells with control of cell types. High efficiencies of up to 89% were reported with electrical fusion of paired cells. The reprogramming in hybrids between mouse embryonic stem cells and mouse embryonic fibroblasts were observed. 20. Kirschbaum M, Jaeger MS, Duschl C: Correlating short-term Ca(2+) responses with long-term protein expression after activation of single T cells. Lab Chip 2009, 9:3517-3525. 21. Frimat JP, Becker M, Chiang YY, Marggraf U, Janasek D, Hengstler JG, Franzke J, West J: A microfluidic array with cellular valving for single cell co-culture. Lab Chip 2011, 11:231-237. 22.  Rowat AC, Bird JC, Agresti JJ, Rando OJ, Weitz DA: Tracking lineages of single cells in lines using a microfluidic device. Proc Natl Acad Sci USA 2009, 106:18149-18154. An innovative use of microfluidic technology to track multiple lineages of single cells by trapping and confining the growth of individual cells in line for up to 8 generations. The work illustrated that the time scale of phenotypic persistence is protein-specific, and the possibility of studying temporal fluctuations in protein levels. 23. Lecault V, VanInsberghe M, Sekulovic S, Knapp D, Wohrer S, Bowden W, Viel F, McLaughlin T, Jarandehei A, Miller M et al.: High-throughput analysis of single hematopoietic stem cell proliferation in microfluidic cell culture arrays. Nat Methods 2011, 8:581-593. 24.  Faley SL, Copland M, Wlodkowic D, Kolch W, Seale KT, Wikswo JP, Cooper JM: Microfluidic single cell arrays to interrogate signalling dynamics of individual, patient-derived hematopoietic stem cells. Lab Chip 2009, 9:2659-2664. Aligned with reference [14], this work used hydrodynamic traps to immobilise non-adherent individual stem cells for investigating chemoresistance. Dynamic, on chip cell assays successfully revealed the difference in the response of normal and diseased cells. The work also provided new insights into cellular behaviours which were not apparent when using conventional methods. 25. Spiller DG, Wood CD, Rand DA, White MRH: Measurement of single-cell dynamics. Nature 2010, 465:736-745. 26. McKenna BK, Evans JG, Cheung MC, Ehrlich DJ: A parallel microfluidic flow cytometer for high-content screening. Nat Methods 2011, 8:401-441. 27. Chung KH, Crane MM, Lu H: Automated on-chip rapid microscopy, phenotyping and sorting of C. elegans. Nat Methods 2008, 5:637-643. 28.  Godin M, Delgado FF, Son SM, Grover WH, Bryan AK, Tzur A, Jorgensen P, Payer K, Grossman AD, Kirschner MW, Manalis SR: Using buoyant mass to measure the growth of single cells. Nat Methods 2010, 7: 387–U370. A highly sensitive mass measurement method that can detect ‘instantaneous’ growth rate of individual cells. It is a good complementary method to optical approaches for single cell study. 29. Kalisky T, Quake SR: Single-cell genomics. Nat Methods 2011, 8:311-314. 30.  Toriello NM, Douglas ES, Thaitrong N, Hsiao SC, Francis MB, Bertozzi CR, Mathies RA: Integrated microfluidic bioprocessor for single-cell gene expression analysis. Proc Natl Acad Sci USA 2008, 105:20173-20178. A complete process from single cell capture to its gene expression was achieved on an integrated microfluidic device. The measurement of variations of mRNA knockdown in individual Jurkat cells revealed the stochasticity in gene expression and silencing within single cells. 31. Marcy Y, Ouverney C, Bik EM, Losekann T, Ivanova N, Martin HG, Szeto E, Platt D, Hugenholtz P, Relman DA, Quake SR: Dissecting biological ‘‘dark matter’’ with single-cell genetic analysis of rare and uncultivated TM7 microbes from the human mouth. Proc Natl Acad Sci USA 2007, 104:11889-11894. 32. Petriv OI, Kuchenbauer F, Delaney AD, Lecault V, White A, Kent D, Marmolejo L, Heuser M, Berg T, Copley M et al.: Comprehensive microRNA expression profiling of the hematopoietic hierarchy. Proc Natl Acad Sci USA 2010, 107:15443-15448. 33. Schumacher A, Petronis A: Epigenetics of complex diseases: from general theory to laboratory experiments. Curr Top Microbiol Immunol 2006, 310:81-115. 34. Wilkinson GR: Drug therapy – drug metabolism and variability among patients in drug response. N Engl J Med 2005, 352:2211-2221. 35. Kantlehner M, Kirchner R, Hartmann P, Ellwart JW, AlunniFabbroni M, Schumacher A: A high-throughput DNA methylation analysis of a single cell. Nucleic Acids Res 2011, 39:E44-E68. 36.  Huang B, Wu HK, Bhaya D, Grossman A, Granier S, Kobilka BK, Zare RN: Counting low-copy number proteins in a single cell. Science 2007, 315:81-84. Illustrated an innovative combination of micro-sized channel with cylindrical optics to enable single molecule counting. The microdevice was developed for cell manipulation, lysis, labeling, separation and protein quantification, all on a single chip. 37. Salehi-Reyhani A, Kaplinsky J, Burgin E, Novakova M, Demello AJ, Templer RH, Parker P, Neil MAA, Ces O, French P et al.: A first step towards practical single cell proteomics: a microfluidic antibody capture chip with TIRF detection. Lab Chip 2011, 11:1256-1261. 38.  Mellors JS, Jorabchi K, Smith LM, Ramsey JM: Integrated microfluidic device for automated single cell analysis using electrophoretic separation and electrospray ionization mass spectrometry. Anal Chem 2010, 82:967-973. Continuous on chip lysis of erythrocytes with real time electrophoretic separation and electrospray ionization mass spectrometry was reported. This work is perhaps the most integrated demonstration to date for combining single cell study with mass spectrometry. 39. Rubakhin SS, Churchill JD, Greenough WT, Sweedler JV: Profiling signaling peptides in single mammalian cells using mass spectrometry. Anal Chem 2006, 78:7267-7272. 40. Mao P, Wang H, Yang P, Wang DJ: Multinozzle emitter arrays for nanoelectrospray mass spectrometry. Anal Chem 2011, 83:6082-6089. 41. Robertson DG, Watkins PB, Reily MD: Metabolomics in toxicology: preclinical and clinical applications. Toxicol Sci 2011, 120:S146-S170. 42. Okumoto S: Imaging approach for monitoring cellular metabolites and ions using genetically encoded biosensors. Curr Opin Biotechnol 2010, 21:45-54. 43. Amantonico A, Urban PL, Oh JY, Zenobi R: Interfacing microfluidics and laser desorption/ionization mass spectrometry by continuous deposition for application in single cell analysis. Chimia 2009, 63:185-188. 44. Huebner A, Srisa-Art M, Holt D, Abell C, Hollfelder F, Demello AJ, Edel JB: Quantitative detection of protein expression in single cells using droplet microfluidics. Chem Commun 2007, 12:1218-1220. 45. Martino C, Zagnoni M, Sandison ME, Chanasakulniyom M, Pitt AR, Cooper JM: Intracellular protein determination using dropletbased immunoassays. Anal Chem 2011, 83:5361-5368. 46. He MY, Edgar JS, Jeffries GDM, Lorenz RM, Shelby JP, Chiu DT: Selective encapsulation of single cells and subcellular organelles into picoliter- and femtoliter-volume droplets. Anal Chem 2005, 77:1539-1544. 47.  Fidalgo LM, Whyte G, Ruotolo BT, Benesch JLP, Stengel F, Abell C, Robinson CV, Huck WTS: Coupling microdroplet microreactors withmassspectrometry:readingthecontentsofsingledroplets online. Angew Chem Int Ed 2009, 48:3665-3668. On line ESI-MS analysis of components encapsulated in microdroplets was reported. A key to this was to separate the carrier fluid (oil) which interferes with the ESI process. 48. Yin HB, Pattrick N, Zhang XL, Klauke N, Cordingley HC, Haswell SJ, Cooper JM: Quantitative comparison between microfluidic and microtiter plate formats for cell-based assays. Anal Chem 2008, 80:179-185. 118 Analytical biotechnology Current Opinion in Biotechnology 2012, 23:110–119 www.sciencedirect.com 49. Pihl J, Sinclair J, Sahlin E, Karlsson M, Petterson F, Olofsson J, Orwar O: Microfluidic gradient-generating device for pharmacological profiling. Anal Chem 2005, 77:3897-3903. 50. Tirella A, Marano M, Vozzi F, Ahluwalia A: A microfluidic gradient maker for toxicity testing of bupivacaine and lidocaine. Toxicol In Vitro 2008, 22:1957-1964. 51. Yin HB, Zhang XL, Pattrick N, Klauke N, Cordingley HC, Haswell SJ, Cooper JM: Influence of hydrodynamic conditions on quantitative cellular assays in microfluidic systems. Anal Chem 2007, 79:7139-7144. 52. Feng Y, Mitchison TJ, Bender A, Young DW, Tallarico JA: Multiparameter phenotypic profiling: using cellular effects to characterize small-molecule compounds. Nat Rev Drug Discov 2009, 8:567-578. 53. Sung JH, Kam C, Shuler ML: A microfluidic device for a pharmacokinetic-pharmacodynamic (PK-PD) model on a chip. Lab Chip 2010, 10:446-455. 54. Bao XR, Fraser IDC, Wall EA, Quake SR, Simon MI: Variability in G-protein-coupled signaling studied with microfluidic devices. Biophys J 2010, 99:2414-2422. Microfluidics technology Yin and Marshall 119 www.sciencedirect.com Current Opinion in Biotechnology 2012, 23:110–119