Open access peer-reviewed chapter

Time-Lapse Microscopy

Written By

John L. Collins, Bart van Knippenberg, Kai Ding and Alexander V. Kofman

Submitted: 08 August 2018 Reviewed: 29 August 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.81199

From the Edited Volume

Cell Culture

Edited by Radwa Ali Mehanna

Chapter metrics overview

1,780 Chapter Downloads

View Full Metrics

Abstract

Time-lapse microscopy is a powerful, versatile and constantly developing tool for real-time imaging of living cells. This review outlines the advances of time-lapse microscopy and refers to the most interesting reports, thus pointing at the fact that the modern biology and medicine are entering the thrilling and promising age of molecular cinematography.

Keywords

  • time-lapse
  • microscopy
  • real-time
  • imaging
  • cell

1. Introduction

Originally described as time-lapse cinemicrography (microphotography) [1], the modern time-lapse microscopy (TLM) emerged as a powerful and continuously improving tool for studying the cellular processes and cell-cell interactions with the applications ranging from fundamental aspects of molecular and cell biology to medical practice. The related time-lapse photography is more relevant to observing non-microscopic objects, such as plants and landscapes. TLM is the technique of capturing the sequence of microscopic images at regular intervals. TLM allows scientists to observe cellular dynamics and behavior of the population of living cells as well as of the single living cell within the population [2, 3]. Live cell imaging and the first non-sophisticated TLM techniques were pioneered at the very beginning of the twentieth century [4]. However, to be visible in the light microscope, the cells are to be subjected to fixation and staining, the processes that kill the cells. Introduction of phase-contrast microscopy in 1940s, development of fluorescent and multidimensional microscopy, flow cytometry and computational tools made live cell imaging a widespread approach and prompted scientists to consider TLM as an essential technique that carries an enormous promise for basic biological science and medicine. For this review, we focused on mammalian cell cultures, although TLM can also be efficiently employed to study prokaryotic cells and unicellular microorganisms. In the absence of up-to-date comprehensive review on TLM advances, our aim was to familiarize the readers with the current advances of TLM methodology and provide for the reference guide to the most interesting reports where TLM has been utilized both for biological research and clinical purposes.

Advertisement

2. Time-lapse microscopy: from making movies to bedside

2.1. Versatility of TLM

In this part, we will briefly review some selected publications, which highlight the rapid development of TLM as a versatile discovery tool within the broad scope of modern biology and medicine. Importance of TLM as a new method in biological research was highlighted by Burton [5]. The progress of tissue culture methods, phase-contrast microscopy (see below) and real-time imaging by TLM enabled scientists to overcome the major limitation of traditional microscopy; preparation of very thin transparent samples, which required tissue fixation and did not make it possible to investigate living cells, let alone, and biological processes over time in the same sample. Early reports demonstrated the feasibility of TLM for comparative studies of cultured cells [6, 7, 8] and for monitoring living blood and lymph cells [1], cell division [9, 10] and reaction of cells to varying contents of electrolytes in perfusion chambers [11]. TLM was helpful to decode the process of multinucleation in the developing skeletal muscles [12] and to describe the variable cytotoxic response toward allografts [13, 14].

TLM is a suitable tool to monitor cell motility and migration, including quantitative assessment of migration, such as the number of migrating cells and the distance [15, 16, 17, 18, 19, 20]. In multicellular organisms, the directed and coordinated cell migration (chemotaxis) occurs during embryonic development, tissue regeneration and inflammatory response [21], while cancer cells migrate into surrounding tissues and the vasculature. To monitor chemotaxis, TLM can be used together with the Dunn chamber [21, 22, 23]; Boyden chamber [24, 25]; Bridge chamber [26]; LOCOMOTIS, the motility tracking system [27] and other types of chambers for cell visualization and TLM applications [28, 29, 30]. TLM was employed to study embryonic stem cells [31]; hematopoietic progenitor cells [20, 32, 33, 34]; mesenchymal stem cells [35]; activated lymphocytes forming lymphocytes colonies [36]; primordial germ cells, a migratory cell population that will eventually give rise to the gametes [37, 38, 39]; the migration route of progenitor cells in cell cultures obtained from live chicken embryos [40, 41]; microglial cells [42, 43, 44]; olfactory cells from schizophrenia patients [45]; neurons [46]; chemokines that drive migration of megakaryocytes from the proliferative osteoblastic niche within the bone marrow to the capillary-rich vascular niche, which is an essential step for platelet production [47]; migration of osteoclasts toward bone surfaces [48]; motility of cultured endothelial cells to study remodeling of their intercellular junctions [49]; generation of a complete polarized epithelial monolayer by the epithelial cells of mammary gland [17]; movement of cancer cells that were cultured under hypoxic conditions [50] and treated with salinomycin [24]; individual cell motility in fibroblastoid L929 cells [51]; human osteosarcoma MG-63 cells [52]; B35 neuroblastoma cells transiently expressing GFP and C6 glioma cells after staining with Hoechst 33258 [16] and motility of L5222 leukemia cells within the mesentery and migration of induced pluripotent cells during their early reprograming [53]. Of note, most studies are devoted to neural stem cells [18, 19, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63] due to growing clinical importance.

TLM allows investigators to visualize and characterize cell-cell contacts [46, 52, 64, 65, 66, 67, 68, 69, 70, 71]. The most interesting reports are concerned with the contacts between the various types of stem/progenitor cells as well as the tumor-environment cell interactions: the importance of proper cell-cell contacts level for their correct positioning and cell polarity during organogenesis [39], glial-neuronal interactions [72, 73, 74], interactions between microglia and brain tumors [75], between astrocytes and neural progenitor cells [42], between mesenchymal stem cells and human myoblasts [76], dendritic cells [77], endothelial cells [78], cancer cells [79], extracellular matrix molecules [80], between erythroblastic islands in bone marrow [81], between neural progenitor cells [62, 82, 83, 84], between neural cells and hematopoietic stem cells that migrate to the central nervous system [85], hematopoietic stem cells and stromal cells [20], endothelial progenitor cells and cardiac myocytes [86], between induced pluripotent cells during the early reprogramming phase [53], vesicle traffic through intercellular bridges between prostate cancer cells [87] and synaptic contacts [88, 89, 90, 91, 92, 93, 94].

Cell division and cell death can be well investigated with TLM [50, 52, 95, 96, 97]. Division and growth of both labeled [96, 98, 99, 100] and non-labeled [101, 102] cells in culture [52, 95, 98, 103, 104, 105] and tissue slices [106], including monitoring of a single cell [95, 99, 107, 108, 109, 110, 111, 112], can be observed and assessed with TLM. The fluorescent ubiquitination-based cell cycle indicator (FUCCI) system can effectively label individual G1, S/G2/M and G1/S-transition phase nuclei as red, green and yellow, respectively, to visualize the real-time cell cycle transitions in living mammalian cells [113, 114, 115, 116]. Microinjection of complementary RNA to cyclin B1 was reported as a tool for TLM studying meiosis [117]. Real-time imaging was employed to monitor nuclear envelope breakdown, which is one of the major morphological changes during mitosis [118] and apoptosis [119]; nucleolar assembly after mitosis [120]; tracking of template DNA strands during mitosis [121, 122]; preferred mitotic orientation of daughter cells, which is important for their following self-organization and tissue formation [123, 124]; interkinetic nuclear migration toward the apical surface in epithelial cells [125, 126]; multinucleation of skeletal muscle cells [12]; asymmetric division of stem cells [127, 128, 129]; identification and characterization of cell division genes by combining RNA interference, time-lapse microscopy and computational image processing [130]; cytokinesis [131, 132]; cleavage furrow [133]; abscission by using TLM in combination with electron microscopy [134, 135] and mitotic synchronization in the cell population [136]. The observations related to cellular senescence and various forms of cell death include re-entry into the cell cycle [10, 124, 137, 138, 139] and variable frequency of divisions [140]; changes in mitotic and interphase duration [141, 142, 143, 144, 145, 146, 147]; short G1 phase, which is a distinctive feature of mouse embryonic stem cells [148]; delayed G2 phase [149]; neosis, the term used for karyokinesis via nuclear budding followed by asymmetric, intracellular cytokinesis [150]; secretion of exosomes with anti-apoptotic microRNAs [151]; apoptosis [119, 152, 153, 154, 155, 156]; phagocytosis of apoptotic cells [157]; necrosis [158]; autophagy [159]; mitotic catastrophe [52, 143] and phototoxicity [160].

TLM can also be used to study intracellular dynamics of subcellular organelles [161, 162], natural cellular proteins and reporters, introduced nanoparticles and even physiological effects of small inorganic molecules and gases. Time-lapse imaging was used to monitor and quantify movements and changes in mitochondria [163, 164, 165]; Golgi apparatus [166]; centrosomes and microtubules [167, 168, 169, 170]; centromeres [171]; cellular membrane [172]; dendritic spines [173]; dynamics of interkinetic nuclear migration [174, 175]; intercellular uptake and distribution of nano-sized (less than 100 mkm) ceramic particles [176]; intracellular translocation of p65 and IkappaB-alpha proteins [177]; intracellular distribution of integrin beta1 and F-actin [178]; fluctuations in Notch signaling to maintain neural progenitors [179]; re-localization of PP1gamma, which is implicated in multiple cell cycle-related processes including regulation of chromosome segregation and cytokinesis [180]; movement of the replication origin region of the chromosome during the cell cycle in Bacillus subtilis [181]; dynamics of 53BP1 protein in DNA-damage response [182]; measuring gene dynamics with luciferase as a reporter [183]; colocalization of MAP kinases in mitochondria [184]; clustering of acetylcholine receptor on myotubes [185]; multiple chromosomal populations of topoisomerase II [186]; focal points for chromosome condensation and decondensation [187]; intracellular calcium dynamics [188, 189] and single-cell time-lapse imaging of intracellular O2 [190].

Although TLM is mostly used with cultured mammalian cells and live cells in tissues, the significant number of reports indicates that TLM could be employed to observe and study prokaryotic cells and other unicellular and multicellular organisms as well as viruses. Here, we mention only few examples, such as time-lapse imaging of growth, cell-cell contacts and formation of spherical granules in E. coli [191, 192, 193, 194]; time-lapse visualization of bacterial colony morphologies in the special bacterial chamber MOCHA [195]; screening and assessing effects of antibiotics, such as antibiotics-bacteria interactions [196, 197, 198, 199] and studying yeasts [200, 201, 202] and viruses [203, 204, 205, 206, 207]. The smaller microorganisms, analogously to intracellular structures, usually require higher magnification and more sophisticated microscopic equipment.

2.2. TLM technical approaches

TLM monitoring of mammalian cells usually requires the inverted microscope, which is fully or partially enclosed by a cell incubator (environmental chamber), a partly sealed transparent box that maintains the temperature, humidity and even partial gas (carbon dioxide) pressure, protects cultured cells from the light and allows the investigator to manipulate with the microscope in order to choose the field of view and adjust other imaging parameters [208, 209, 210]. The TLM chambers and devices underwent significant improvements over the time, from the simple glass tissue chambers and manual capturing sequences of images to the automated high-resolution microscopes and sophisticated computerized equipment for long-term TLM observations [154, 162, 211, 212, 213, 214, 215, 216, 217, 218, 219]. The up-to-date portable live cell culture monitor (CytoSMART Technologies, Eindhoven, The Netherlands) works within the regular CO2 incubator. The culture flask (T-flask, Petri dish, wells or any other transparent vessel) is positioned onto the lens of the device; the field of view is chosen by the investigator, and the cell growth and migration can be monitored and analyzed in the real-time mode by accessing the cloud [52].

The phase-contrast method of imaging is based on the ability of materials with a different refractive index to delay the passage of the light through the sample by different amounts, so that they appear darker or brighter. This is the most common TLM technique that is used since 1950s [1, 6, 7, 11] for studying different types of cells and microorganisms both alone and in combination with electron microscopy [220, 221, 222]. The so-called differential interference contrast (DIC) microscopy (Nomarski microscopy) also produces high-contrast images of transparent non-stained biological objects, and it has been broadly used for TLM [223, 224, 225, 226]. Fluorescent TLM dating back in 1950s TLM [9, 227] can be used nowadays with fluorescent proteins-reporters [207, 228, 229, 230, 231], fluorescent nanoparticles [232, 233] and membrane dyes [160, 234, 235]. As the further proof of TLM flexibility, we present some reports where TLM is combined with other advanced microscopy techniques: multiplexed or multifield (recording of many fields simultaneously) TLM [236, 237], confocal TLM [156, 171, 207, 238, 239, 240, 241, 242], multi-photon TLM [58, 243, 244, 245], the so-called four-dimensional imaging (three-dimensional images over time) [242, 246], time-lapse bioluminescence analysis [247], Forster resonance energy transfer (FRET) microscopy [248], time-lapse optical coherence tomography [249, 250, 251], in toto imaging to image and track every single cell movement and division during the development of organs and tissues [241] and other innovative approaches [50, 252]. TLM can be used to monitor not only cultured cells (cell population and single cell [109] but also living cells in tissue slices up to a depth of 60 micrometers in brain slices, in regions where cell bodies remain largely uninjured by the tissue preparation and are visible in their natural environment [229, 253]. For real-time observation of corneal cells in a living mouse, a novel microscope system was designed, which consists of an upright fluorescence microscope for visualization of corneal cells, a mouse-holding unit for immobilization of the animal and the eye and a set of gimbals which permit observation of a wide area of corneal surface without refocusing [254].

TLM would not be possible without an automated image analysis, which is used to extract meaningful data from the bulk of images. Automated cell tracking faces problems associated with high cell density; cell mobility; cell division; multiple cell parameters such as object size, position or texture; cell lysis or overlap of cells [255]. A variety of algorithms, including segmentation (the process of partitioning a digital image into multiple sets of pixels or segments) algorithms, have been developed, and they are constantly improving. For most datasets, a preprocessing step is needed before information can be extracted. Irregular illumination and shading effects can be removed by using a background subtraction method. Other commonly used techniques include contrast enhancement and noise filtering [256]. In some cases, registration is needed to align subsequent image frames and compensate for unwanted movements. Global movements can be caused by movement of the specimen or imaging equipment, but local deformations in the specimen might also have to be corrected for. This is especially the case when considering TLM of living animals, which is heavily affected by breathing and heartbeat [257]. At higher magnifications, when studying intracellular dynamics, cell migration itself might also be considered an unwanted movement that has to be corrected [258]. Object detection is a set of techniques to separate objects of interest from the background. The objects of interest can be cells or intracellular particles [130, 259]. Basic segmentation techniques can be sufficient to detect individual cells, although more advanced techniques are still being developed to cope with increasingly complex data [260, 261]. Finally, several analysis techniques are available to quantify the different types of cell behavior over time, for example, trajectory analysis for assessing trajectory length and directional persistence [262]. By now, various algorithms are designed for quantifying and tracking cell migration [3] and single cell motility [261, 263]; cell proliferation [264]; cell cycle and cell lineage analysis [107]; changes in mitotic and interphase duration [141]; cell-cell contacts [52]; studying specific cells and tissues [265] and specific intracellular processes such as transcription [99] or morphogenesis [266]; colocalization of cells and intracellular markers [184]; tracking cellular organelles [258]; highlighting the certain cell type within tissues or mixed cell cultures [267]; clustered, overlapping or dying cells [268]; in toto imaging of developing organisms, tissues and organs [241] and assessing development and selection of embryos for in vitro fertilization [269, 270].

2.3. TLM for assisted reproductive technology and its promise for clinical medicine

TLM is emerging as a promising clinical technique for selecting embryos for transplantation, although the discussion is still under way whether TLM may become an alternative to preimplantation genetic screening [271, 272]. The so-called morphokinetic analysis [273] by TLM is aimed to assess the number, development and quality (viability) of embryos by monitoring cleavage anomalies, multinucleation [274] or specific cell cycle kinetics [274, 275] and cleavage divisions [276], aneuploidy [277, 278], which is considered as a key causal factor of delays in embryonic development toward a blastocyte [278], and even chromosomal abnormalities [279]. Although more clinical research is required to finally prove that TLM can identify the best embryo for transfer and has an advantage over the conventional incubation of embryos [280], TLM is under consideration for patenting as a method for selecting embryos for implantation [281, 282]. TLM can also be used for sperm motility analysis [283].

One of the potential medical applications of TLM is the assessment of ex vivo engineered cells for cell therapy of degenerative and inherited disorders and other human pathologies like cancer [284, 285, 286, 287, 288]. TLM can also be used for diagnostics, for example, for detecting abnormalities in cell behavior in human dystrophic muscle cultures [289] or estimating tumor malignancy [290] in drug discovery [291], for testing gene therapeutic agents [292] and for evaluating side effects of antibiotics [293] and efficacy of chemotherapeutics [294, 295]. TLM is a valuable tool for understanding the pathogenesis of certain disorders, such as dysplastic erythroblast formation of erythroblasts from the patient with congenital dyserythropoietic anemia [296], thrombus formation [224], IgE-mediated mast cell degranulation and recovery [297], imaging of disease progression in deep brain areas using fluorescence microendoscopy [298], reprogramming in induced pluripotent cells [110] and other applications.

Advertisement

3. Conclusion

TLM is a powerful and versatile tool in modern biological research, with the immense potential for future clinical applications. One of the probably underexplored features of TLM is its promise to further characterize heterogeneity of cells within tissues [144], in particular, stem/progenitor cells and differentiating cells [299] as well as cancer cells [300]. Some of the above-mentioned methods are associated with unavoidable costs (expensive equipment, such as lenses, filters and sensors, and their damage due to high humidity within the incubator), non-natural impacts on living cells by the high excitation energy of lasers and bleaching/degradation of the fluorochromes over time, which influences quantification of long-running processes. However, the growing number of reports about new improvements and advances in TLM techniques and TLM-related applications that provide valuable information, which is not imageable by other techniques, makes it possible to conclude that the era of microcinematography in biomedical research has just begun.

Advertisement

Acknowledgments

We thank Mr. Joffry Maltha (CytoSMART Technologies) for assistance with preparation of this manuscript. The work was partially supported by the National Cancer Institute, USA, Award Number R37CA229417.

Advertisement

Conflict of interest

None declared.

References

  1. 1. Klausewitz W. Cytodiagnostic studies on living blood and lymph cells of some Amphibia by means of micro-time lapse film and phase contrast microscopy. Zeitschrift für Zellforschung und Mikroskopische Anatomie. 1953;39(1):1-35
  2. 2. Baker M. Cellular imaging: Taking a long, hard look. Nature. 2010;466(7310):1137-1140
  3. 3. Svensson CM, Medyukhina A, Belyaev I, Al-Zaben N, Figge MT. Untangling cell tracks: Quantifying cell migration by time lapse image data analysis. Cytometry Part A: The Journal of the International Society for Analytical Cytology. 2018;93(3):357-370
  4. 4. Landecker H. Seeing things: From microcinematography to live cell imaging. Nature Methods. 2009;6(10):707-709
  5. 5. Burton AL. Time-lapse phase-contrast cinephotomicrography: A new method in biological research. Canadian Medical Association Journal. 1962;87:20-26
  6. 6. Borysko E, Sapranauskas P. A new technique for comparative phase-contrast and electron microscope studies of cells grown in tissue culture, with an evaluation of the technique by means of time-lapse cinemicrographs. Bulletin of the Johns Hopkins Hospital. 1954;95(2):68-79
  7. 7. Kramis NJ. Time-lapse, phase contrast cine photomicrography of tissue culture cells. Journal of the Biological Photographic Association. 1956;24(1):27-29
  8. 8. Rose GG. Time-lapse cinemicrography of cells in tissue culture. Bulletin of the Johns Hopkins Hospital. 1965;116:33-68
  9. 9. Montgomery PO, Bonner WA. Ultra-violet time lapse motion picture observations of mitosis in newt cells. Experimental Cell Research. 1959;17(3):378-384
  10. 10. Froese G. Division delay in HeLa cells in Chinese hamster cells. A time-lapse study. International Journal of Radiation Biology and Related Studies in Physics, Chemistry, and Medicine. 1966;10(4):353-367
  11. 11. Overman RR, Pomerat CM. Electrolytes and plasma expanders. I. Reaction of human cells in perfusion chambers with phase contrast, time-lapse cine records. Zeitschrift für Zellforschung und Mikroskopische Anatomie. 1956;45(1):2-17
  12. 12. Capers CR. Multinucleation of skeletal muscle in vitro. The Journal of Biophysical and Biochemical Cytology. 1960;7:559-566
  13. 13. Terasaki PI, Cannon JA, Longmire WP Jr, Chamberlain CC. Antibody response to homografts: V. Cytotoxic effects upon lymphocytes as measured by time-lapse cinematography. Annals of the New York Academy of Sciences. 1960;87:258-265
  14. 14. Sharp JA, Smiddy FG. Time-lapse cinemicrography of lymphoid tissue cultured in normal and in uraemic serum. Nature. 1962;193:191-192
  15. 15. Jain P, Worthylake RA, Alahari SK. Quantitative analysis of random migration of cells using time-lapse video microscopy. Journal of Visualized Experiments: JoVE. 2012;63:e3585
  16. 16. Dai L, Alt W, Schilling K, Retzlik J, Gieselmann V, Magin TM, et al. A fast and robust quantitative time-lapse assay for cell migration. Experimental Cell Research. 2005;311(2):272-280
  17. 17. Wick N, Thurner S, Paiha K, Sedivy R, Vietor I, Huber LA. Quantitative measurement of cell migration using time-lapse videomicroscopy and non-linear system analysis. Histochemistry and Cell Biology. 2003;119(1):15-20
  18. 18. Arocena M, Zhao M, Collinson JM, Song B. A time-lapse and quantitative modelling analysis of neural stem cell motion in the absence of directional cues and in electric fields. Journal of Neuroscience Research. 2010;88(15):3267-3274
  19. 19. Hossain WA, D’Sa C, Morest DK. Interactive roles of fibroblast growth factor 2 and neurotrophin 3 in the sequence of migration, process outgrowth, and axonal differentiation of mouse cochlear ganglion cells. Journal of Neuroscience Research. 2008;86(11):2376-2391
  20. 20. Wagner W, Saffrich R, Wirkner U, Eckstein V, Blake J, Ansorge A, et al. Hematopoietic progenitor cells and cellular microenvironment: Behavioral and molecular changes upon interaction. Stem Cells. 2005;23(8):1180-1191
  21. 21. Wells CM, Ridley AJ. Analysis of cell migration using the Dunn chemotaxis chamber and time-lapse microscopy. Methods in Molecular Biology. 2005;294:31-41
  22. 22. Muinonen-Martin AJ, Veltman DM, Kalna G, Insall RH. An improved chamber for direct visualisation of chemotaxis. PLoS One. 2010;5(12):e15309
  23. 23. Vasaturo A, Caserta S, Russo I, Preziosi V, Ciacci C, Guido S. A novel chemotaxis assay in 3-D collagen gels by time-lapse microscopy. PLoS One. 2012;7(12):e52251
  24. 24. Kopp F, Hermawan A, Oak PS, Herrmann A, Wagner E, Roidl A. Salinomycin treatment reduces metastatic tumor burden by hampering cancer cell migration. Molecular Cancer. 2014;13:16
  25. 25. Boyden S. The chemotactic effect of mixtures of antibody and antigen on polymorphonuclear leucocytes. The Journal of Experimental Medicine. 1962;115:453-466
  26. 26. Zigmond SH. Ability of polymorphonuclear leukocytes to orient in gradients of chemotactic factors. The Journal of Cell Biology. 1977;75(2 Pt 1):606-616
  27. 27. Lynch AE, Triajianto J, Routledge E. Low-cost motility tracking system (LOCOMOTIS) for time-lapse microscopy applications and cell visualisation. PLoS One. 2014;9(8):e103547
  28. 28. Buhler H, Adamietz R, Abeln T, Diaz-Carballo D, Nguemgo-Kouam P, Hero T, et al. Automated multichamber time-lapse videography for long-term in vivo observation of migrating cells. In Vivo. 2017;31(3):329-334
  29. 29. Mathieu E, Paul CD, Stahl R, Vanmeerbeeck G, Reumers V, Liu C, et al. Time-lapse lens-free imaging of cell migration in diverse physical microenvironments. Lab on a Chip. 2016;16(17):3304-3316
  30. 30. Funahashi J, Nakamura H. Time-lapse imaging system with shell-less culture chamber. Development, Growth & Differentiation. 2014;56(4):305-309
  31. 31. Coll JL, Ben-Ze’ev A, Ezzell RM, Rodriguez Fernandez JL, Baribault H, Oshima RG, et al. Targeted disruption of vinculin genes in F9 and embryonic stem cells changes cell morphology, adhesion, and locomotion. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(20):9161-9165
  32. 32. Denkers IA, Dragowska W, Jaggi B, Palcic B, Lansdorp PM. Time lapse video recordings of highly purified human hematopoietic progenitor cells in culture. Stem Cells. 1993;11(3):243-248
  33. 33. Wuchter P, Leinweber C, Saffrich R, Hanke M, Eckstein V, Ho AD, et al. Plerixafor induces the rapid and transient release of stromal cell-derived factor-1 alpha from human mesenchymal stromal cells and influences the migration behavior of human hematopoietic progenitor cells. Cell and Tissue Research. 2014;355(2):315-326
  34. 34. Fonseca AV, Freund D, Bornhauser M, Corbeil D. Polarization and migration of hematopoietic stem and progenitor cells rely on the RhoA/ROCK I pathway and an active reorganization of the microtubule network. Journal of Biological Chemistry. 2010;285(41):31661-31671
  35. 35. Lee DH, Park BJ, Lee MS, Lee JW, Kim JK, Yang HC, et al. Chemotactic migration of human mesenchymal stem cells and MC3T3-E1 osteoblast-like cells induced by COS-7 cell line expressing rhBMP-7. Tissue Engineering. 2006;12(6):1577-1586
  36. 36. Donnenberg AD, Cameron J. Cinemicroscopic studies of lymphocyte behavior in semi-solid medium: The polyclonal origin of lymphocyte colonies. Experimental Hematology. 1985;13(1):29-35
  37. 37. Dudley B, Molyneaux K. In vivo germ line stem cell migration: A mouse model. Methods in Molecular Biology. 2011;750:117-129
  38. 38. Srihawong T, Kuwana T, Siripattarapravat K, Tirawattanawanich C. Chicken primordial germ cell motility in response to stem cell factor sensing. The International Journal of Developmental Biology. 2015;59(10-12):453-460
  39. 39. Paksa A, Bandemer J, Hoeckendorf B, Razin N, Tarbashevich K, Minina S, et al. Repulsive cues combined with physical barriers and cell-cell adhesion determine progenitor cell positioning during organogenesis. Nature Communications. 2016;7:11288
  40. 40. Song J, Yue Q, Munsterberg A. Time-lapse imaging of chick cardiac precursor cells. Methods in Molecular Biology. 2011;769:359-372
  41. 41. Masyuk M, Morosan-Puopolo G, Brand-Saberi B, Theiss C. Combination of in ovo electroporation and time-lapse imaging to study migrational events in chicken embryos. Developmental Dynamics: An Official Publication of the American Association of the Anatomists. 2014;243(5):690-698
  42. 42. Kornyei Z, Szlavik V, Szabo B, Gocza E, Czirok A, Madarasz E. Humoral and contact interactions in astroglia/stem cell co-cultures in the course of glia-induced neurogenesis. Glia. 2005;49(3):430-444
  43. 43. Carbonell WS, Murase S, Horwitz AF, Mandell JW. Migration of perilesional microglia after focal brain injury and modulation by CC chemokine receptor 5: An in situ time-lapse confocal imaging study. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2005;25(30):7040-7047
  44. 44. Braun H, Buhnemann C, Neumann J, Reymann KG. Preparation of a tissue-like cortical primary culture from embryonic rats using Matrigel and serum free start V medium. Journal of Neuroscience Methods. 2006;157(1):32-38
  45. 45. Fan Y, Abrahamsen G, Mills R, Calderon CC, Tee JY, Leyton L, et al. Focal adhesion dynamics are altered in schizophrenia. Biological Psychiatry. 2013;74(6):418-426
  46. 46. Famulski JK, Trivedi N, Howell D, Yang Y, Tong Y, Gilbertson R, et al. Siah regulation of Pard3A controls neuronal cell adhesion during germinal zone exit. Science. 2010;330(6012):1834-1838
  47. 47. Mazharian A. Assessment of megakaryocyte migration and chemotaxis. Methods in Molecular Biology. 2012;788:275-288
  48. 48. Nevius E, Pinho F, Dhodapkar M, Jin H, Nadrah K, Horowitz MC, et al. Oxysterols and EBI2 promote osteoclast precursor migration to bone surfaces and regulate bone mass homeostasis. The Journal of Experimental Medicine. 2015;212(11):1931-1946
  49. 49. Guo R, Sakamoto H, Sugiura S, Ogawa M. Endothelial cell motility is compatible with junctional integrity. Journal of Cellular Physiology. 2007;211(2):327-335
  50. 50. Kamlund S, Strand D, Janicke B, Alm K, Oredsson S. Influence of salinomycin treatment on division and movement of individual cancer cells cultured in normoxia or hypoxia evaluated with time-lapse digital holographic microscopy. Cell Cycle. 2017;16(21):2128-2138
  51. 51. Hartmann-Petersen R, Walmod PS, Berezin A, Berezin V, Bock E. Individual cell motility studied by time-lapse video recording: Influence of experimental conditions. Cytometry. 2000;40(4):260-270
  52. 52. Dosch J, Hadley E, Wiese C, Soderberg M, Houwman T, Ding K, et al. Time-lapse microscopic observation of non-dividing cells in cultured human osteosarcoma MG-63 cell line. Cell Cycle. 2018;17(2):174-181
  53. 53. Megyola CM, Gao Y, Teixeira AM, Cheng J, Heydari K, Cheng EC, et al. Dynamic migration and cell-cell interactions of early reprogramming revealed by high-resolution time-lapse imaging. Stem Cells. 2013;31(5):895-905
  54. 54. Puche AC, Bovetti S. Studies of adult neural stem cell migration. Methods in Molecular Biology. 2011;750:227-240
  55. 55. Ito H, Morishita R, Tabata H, Nagata K. Visualizing septin and cell dynamics in mammalian brain slices. Methods in Cell Biology. 2016;136:295-309
  56. 56. Hughes EG, Kang SH, Fukaya M, Bergles DE. Oligodendrocyte progenitors balance growth with self-repulsion to achieve homeostasis in the adult brain. Nature Neuroscience. 2013;16(6):668-676
  57. 57. Zhang RL, LeTourneau Y, Gregg SR, Wang Y, Toh Y, Robin AM, et al. Neuroblast division during migration toward the ischemic striatum: A study of dynamic migratory and proliferative characteristics of neuroblasts from the subventricular zone. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2007;27(12):3157-3162
  58. 58. Zhao LR, Nam SC. Multiphoton microscope imaging: The behavior of neural progenitor cells in the rostral migratory stream. Neuroscience Letters. 2007;425(2):83-88
  59. 59. Comte I, Kim Y, Young CC, van, der Harg JM, Hockberger P, Bolam PJ, et al. Galectin-3 maintains cell motility from the subventricular zone to the olfactory bulb. Journal of Cell Science. 2011;124(Pt 14):2438-2447
  60. 60. Sonego M, Oberoi M, Stoddart J, Gajendra S, Hendricusdottir R, Oozeer F, et al. Drebrin regulates neuroblast migration in the postnatal mammalian brain. PLoS One. 2015;10(5):e0126478
  61. 61. Sonego M, Gajendra S, Parsons M, Ma Y, Hobbs C, Zentar MP, et al. Fascin regulates the migration of subventricular zone-derived neuroblasts in the postnatal brain. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2013;33(30):12171-12185
  62. 62. Kulesa P, Bronner-Fraser M, Fraser S. In ovo time-lapse analysis after dorsal neural tube ablation shows rerouting of chick hindbrain neural crest. Development. 2000;127(13):2843-2852
  63. 63. Noctor SC, Martinez-Cerdeno V, Ivic L, Kriegstein AR. Cortical neurons arise in symmetric and asymmetric division zones and migrate through specific phases. Nature Neuroscience. 2004;7(2):136-144
  64. 64. Chen S, Bremer AW, Scheideler OJ, Na YS, Todhunter ME, Hsiao S, et al. Interrogating cellular fate decisions with high-throughput arrays of multiplexed cellular communities. Nature Communications. 2016;7:10309
  65. 65. Merouane A, Rey-Villamizar N, Lu Y, Liadi I, Romain G, Lu J, et al. Automated profiling of individual cell-cell interactions from high-throughput time-lapse imaging microscopy in nanowell grids (TIMING). Bioinformatics. 2015;31(19):3189-3197
  66. 66. Hirata Y, Li HW, Takahashi K, Ishii H, Sykes M, Fujisaki J. MHC class I expression by donor hematopoietic stem cells is required to prevent NK cell attack in allogeneic, but not syngeneic recipient mice. PLoS One. 2015;10(11):e0141785
  67. 67. Ciccocioppo R, Cangemi GC, Kruzliak P, Gallia A, Betti E, Badulli C, et al. Ex vivo immunosuppressive effects of mesenchymal stem cells on Crohn’s disease mucosal T cells are largely dependent on indoleamine 2,3-dioxygenase activity and cell-cell contact. Stem Cell Research & Therapy. 2015;6:137
  68. 68. Tomura M, Mori YS, Watanabe R, Tanaka M, Miyawaki A, Kanagawa O. Time-lapse observation of cellular function with fluorescent probe reveals novel CTL-target cell interactions. International Immunology. 2009;21(10):1145-1150
  69. 69. Jensen AM, Raff MC. Continuous observation of multipotential retinal progenitor cells in clonal density culture. Developmental Biology. 1997;188(2):267-279
  70. 70. Shih YT, Wang MC, Peng HH, Chen TF, Chen L, Chang JY, et al. Modulation of chemotactic and pro-inflammatory activities of endothelial progenitor cells by hepatocellular carcinoma. Cellular Signalling. 2012;24(3):779-793
  71. 71. Yamazaki K, Roberts RA, Spooncer E, Dexter TM, Allen TD. Cellular interactions between 3T3 cells and interleukin-3-dependent multipotent haemopoietic cells: A model system for stromal-cell-mediated haemopoiesis. Journal of Cellular Physiology. 1989;139(2):301-312
  72. 72. Ioannidou K, Edgar JM, Barnett SC. Time-lapse imaging of glial-axonal interactions. Current Protocols in Neuroscience. 2015;72:2.23.1-2.23.14
  73. 73. Ioannidou K, Anderson KI, Strachan D, Edgar JM, Barnett SC. Time-lapse imaging of the dynamics of CNS glial-axonal interactions in vitro and ex vivo. PLoS One. 2012;7(1):e30775
  74. 74. Louhivuori LM, Jansson L, Turunen PM, Jantti MH, Nordstrom T, Louhivuori V, et al. Transient receptor potential channels and their role in modulating radial glial-neuronal interaction: A signaling pathway involving mGluR5. Stem Cells and Development. 2015;24(6):701-713
  75. 75. Bayerl SH, Niesner R, Cseresnyes Z, Radbruch H, Pohlan J, Brandenburg S, et al. Time lapse in vivo microscopy reveals distinct dynamics of microglia-tumor environment interactions-a new role for the tumor perivascular space as highway for trafficking microglia. Glia. 2016;64(7):1210-1226
  76. 76. Black AB, Dahlenburg H, Pepper K, Nacey C, Pontow S, Kuhn MA, et al. Human myoblast and mesenchymal stem cell interactions visualized by videomicroscopy. Human Gene Therapy Methods. 2015;26(6):193-196
  77. 77. Silva AM, Oliveira MI, Sette L, Almeida CR, Oliveira MJ, Barbosa MA, et al. Resveratrol as a natural anti-tumor necrosis factor-alpha molecule: Implications to dendritic cells and their crosstalk with mesenchymal stromal cells. PLoS One. 2014;9(3):e91406
  78. 78. Ern C, Krump-Konvalinkova V, Docheva D, Schindler S, Rossmann O, Bocker W, et al. Interactions of human endothelial and multipotent mesenchymal stem cells in cocultures. The Open Biomedical Engineering Journal. 2010;4:190-198
  79. 79. Al-toub M, Vishnubalaji R, Hamam R, Kassem M, Aldahmash A, Alajez NM. CDH1 and IL1-beta expression dictates FAK and MAPKK-dependent cross-talk between cancer cells and human mesenchymal stem cells. Stem Cell Research & Therapy. 2015;6:135
  80. 80. Su PJ, Tran QA, Fong JJ, Eliceiri KW, Ogle BM, Campagnola PJ. Mesenchymal stem cell interactions with 3D ECM modules fabricated via multiphoton excited photochemistry. Biomacromolecules. 2012;13(9):2917-2925
  81. 81. Allen TD, Testa NG. Cellular interactions in erythroblastic islands in long-term bone marrow cultures, as studied by time-lapse video. Blood Cells. 1991;17(1):29-38. Discussion 9-43
  82. 82. Baghbaderani BA, Behie LA, Mukhida K, Hong M, Mendez I. New bioengineering insights into human neural precursor cell expansion in culture. Biotechnology Progress. 2011;27(3):776-787
  83. 83. Kulesa PM, Fraser SE. In ovo time-lapse analysis of chick hindbrain neural crest cell migration shows cell interactions during migration to the branchial arches. Development. 2000;127(6):1161-1172
  84. 84. Fok-Seang J, Mathews GA, Ffrench-Constant C, Trotter J, Fawcett JW. Migration of oligodendrocyte precursors on astrocytes and meningeal cells. Developmental Biology. 1995;171(1):1-15
  85. 85. Gottschling S, Eckstein V, Saffrich R, Jonas A, Uhrig M, Krause U, et al. Primitive and committed human hematopoietic progenitor cells interact with primary murine neural cells and are induced to undergo self-renewing cell divisions. Experimental Hematology. 2007;35(12):1858-1871
  86. 86. Koyanagi M, Brandes RP, Haendeler J, Zeiher AM, Dimmeler S. Cell-to-cell connection of endothelial progenitor cells with cardiac myocytes by nanotubes: A novel mechanism for cell fate changes? Circulation Research. 2005;96(10):1039-1041
  87. 87. Vidulescu C, Clejan S, O’Connor KC. Vesicle traffic through intercellular bridges in DU 145 human prostate cancer cells. Journal of Cellular and Molecular Medicine. 2004;8(3):388-396
  88. 88. Nishiyama N, Colonna J, Shen E, Carrillo J, Nishiyama H. Long-term in vivo time-lapse imaging of synapse development and plasticity in the cerebellum. Journal of Neurophysiology. 2014;111(1):208-216
  89. 89. Kopel H, Schechtman E, Groysman M, Mizrahi A. Enhanced synaptic integration of adult-born neurons in the olfactory bulb of lactating mothers. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2012;32(22):7519-7527
  90. 90. Wang L, Kisaalita WS. Administration of BDNF/ginsenosides combination enhanced synaptic development in human neural stem cells. Journal of Neuroscience Methods. 2011;194(2):274-282
  91. 91. Li Q, Deng Z, Zhang Y, Zhou X, Nagerl UV, Wong ST. A global spatial similarity optimization scheme to track large numbers of dendritic spines in time-lapse confocal microscopy. IEEE Transactions on Medical Imaging. 2011;30(3):632-641
  92. 92. Li J, Erisir A, Cline H. In vivo time-lapse imaging and serial section electron microscopy reveal developmental synaptic rearrangements. Neuron. 2011;69(2):273-286
  93. 93. Yampolsky P, Pacifici PG, Lomb L, Giese G, Rudolf R, Roder IV, et al. Time lapse in vivo visualization of developmental stabilization of synaptic receptors at neuromuscular junctions. Journal of Biological Chemistry. 2010;285(45):34589-34596
  94. 94. Walsh MK, Lichtman JW. In vivo time-lapse imaging of synaptic takeover associated with naturally occurring synapse elimination. Neuron. 2003;37(1):67-73
  95. 95. Piltti KM, Cummings BJ, Carta K, Manughian-Peter A, Worne CL, Singh K, et al. Live-cell time-lapse imaging and single-cell tracking of in vitro cultured neural stem cells—Tools for analyzing dynamics of cell cycle, migration, and lineage selection. Methods. 2018;133:81-90
  96. 96. Yang R, Wang M, Wang J, Huang X, Yang R, Gao WQ. Cell division mode change mediates the regulation of cerebellar granule neurogenesis controlled by the sonic hedgehog Signaling. Stem Cell Reports. 2015;5(5):816-828
  97. 97. Ortega F, Berninger B, Costa MR. Primary culture and live imaging of adult neural stem cells and their progeny. Methods in Molecular Biology. 2013;1052:1-11
  98. 98. Daynac M, Morizur L, Kortulewski T, Gauthier LR, Ruat M, Mouthon MA, et al. Cell sorting of neural stem and progenitor cells from the adult mouse subventricular zone and live-imaging of their cell cycle dynamics. Journal of Visualized Experiments: JoVE. 2015;103:53247
  99. 99. Blanchoud S, Nicolas D, Zoller B, Tidin O, Naef F. CAST: An automated segmentation and tracking tool for the analysis of transcriptional kinetics from single-cell time-lapse recordings. Methods. 2015;85:3-11
  100. 100. Pacini G, Marino A, Migliarini S, Brilli E, Pelosi B, Maddaloni G, et al. A Tph2GFP reporter stem cell line to model in vitro and in vivo serotonergic neuron development and function. ACS Chemical Neuroscience. 2017;8:1043-1052
  101. 101. Suga M, Kii H, Niikura K, Kiyota Y, Furue MK. Development of a monitoring method for nonlabeled human pluripotent stem cell growth by time-lapse image analysis. Stem Cells Translational Medicine. 2015;4(7):720-730
  102. 102. Haetscher N, Feuermann Y, Wingert S, Rehage M, Thalheimer FB, Weiser C, et al. STAT5-regulated microRNA-193b controls haematopoietic stem and progenitor cell expansion by modulating cytokine receptor signalling. Nature Communications. 2015;6:8928
  103. 103. Azimi MS, Motherwell JM, Murfee WL. An ex vivo method for time-lapse imaging of cultured rat mesenteric microvascular networks. Journal of Visualized Experiments: JoVE. 2017;120. DOI: 10.3791/55183
  104. 104. Mackay DR, Ullman KS, Rodesch CK. Time-lapse imaging of mitosis after siRNA transfection. Journal of Visualized Experiments: JoVE. 2010;40:1878
  105. 105. Lin S, Fonteno S, Satish S, Bhanu B, Talbot P. Video bioinformatics analysis of human embryonic stem cell colony growth. Journal of Visualized Experiments: JoVE. 2010;39:1933
  106. 106. Pilaz LJ, Silver DL. Live imaging of mitosis in the developing mouse embryonic cortex. Journal of Visualized Experiments: JoVE. 2014;88. DOI: 10.3791/51298
  107. 107. Errington RJ, Chappell SC, Khan IA, Marquez N, Wiltshire M, Griesdoorn VD, et al. Time-lapse microscopy approaches to track cell cycle and lineage progression at the single-cell level. Current Protocols in Cytometry. 2013;64(1):12.4.1-12.4.13
  108. 108. Scherf N, Franke K, Glauche I, Kurth I, Bornhauser M, Werner C, et al. On the symmetry of siblings: Automated single-cell tracking to quantify the behavior of hematopoietic stem cells in a biomimetic setup. Experimental Hematology. 2012;40(2):119-30 e9
  109. 109. Ortega F, Costa MR, Simon-Ebert T, Schroeder T, Gotz M, Berninger B. Using an adherent cell culture of the mouse subependymal zone to study the behavior of adult neural stem cells on a single-cell level. Nature Protocols. 2011;6(12):1847-1859
  110. 110. Smith ZD, Nachman I, Regev A, Meissner A. Dynamic single-cell imaging of direct reprogramming reveals an early specifying event. Nature Biotechnology. 2010;28(5):521-526
  111. 111. Sillitoe K, Horton C, Spiller DG, White MR. Single-cell time-lapse imaging of the dynamic control of NF-kappaB signalling. Biochemical Society Transactions. 2007;35(Pt 2):263-266
  112. 112. Errington RJ, Marquez N, Chappell SC, Wiltshire M, Smith PJ. Time-lapse microscopy approaches to track cell cycle progression at the single-cell level. Current Protocols in Cytometry. 2005;31(1):12.4.1-12.4.11
  113. 113. Hashimoto H, Yuasa S, Tabata H, Tohyama S, Hayashiji N, Hattori F, et al. Time-lapse imaging of cell cycle dynamics during development in living cardiomyocyte. Journal of Molecular and Cellular Cardiology. 2014;72:241-249
  114. 114. Mort RL, Ford MJ, Sakaue-Sawano A, Lindstrom NO, Casadio A, Douglas AT, et al. Fucci2a: A bicistronic cell cycle reporter that allows Cre mediated tissue specific expression in mice. Cell Cycle. 2014;13(17):2681-2696
  115. 115. Miwa S, Yano S, Kimura H, Yamamoto M, Toneri M, Murakami T, et al. Heterogeneous cell-cycle behavior in response to UVB irradiation by a population of single cancer cells visualized by time-lapse FUCCI imaging. Cell Cycle. 2015;14(12):1932-1937
  116. 116. Jovic D, Sakaue-Sawano A, Abe T, Cho CS, Nagaoka M, Miyawaki A, et al. Direct observation of cell cycle progression in living mouse embryonic stem cells on an extracellular matrix of E-cadherin. Springerplus. 2013;2:585
  117. 117. Holt JE, Lane SI, Jones KT. Time-lapse epifluorescence imaging of expressed cRNA to cyclin B1 for studying meiosis I in mouse oocytes. Methods in Molecular Biology. 2013;957:91-106
  118. 118. Shankaran SS, Mackay DR, Ullman KS. A time-lapse imaging assay to study nuclear envelope breakdown. Methods in Molecular Biology. 2013;931:111-122
  119. 119. Andrade R, Crisol L, Prado R, Boyano MD, Arluzea J, Arechaga J. Plasma membrane and nuclear envelope integrity during the blebbing stage of apoptosis: A time-lapse study. Biology of the Cell. 2009;102(1):25-35
  120. 120. Hernandez-Verdun D, Louvet E, Muro E. Time-lapse, photoactivation, and photobleaching imaging of nucleolar assembly after mitosis. Methods in Molecular Biology. 2013;1042:337-350
  121. 121. Drpic D, Barisic M, Pinheiro D, Maiato H. Selective tracking of template DNA strands after induction of mitosis with unreplicated genomes (MUGs) in drosophila S2 cells. Chromosome Research: An International Journal on the Molecular, Supramolecular and Evolutionary Aspects of Chromosome Biology. 2013;21(3):329-337
  122. 122. Schultz N, Onfelt A. Video time-lapse study of mitosis in binucleate V79 cells: Chromosome segregation and cleavage. Mutagenesis. 1994;9(2):117-123
  123. 123. Wong MN, Nguyen TP, Chen TH, Hsu JJ, Zeng X, Saw A, et al. Preferred mitotic orientation in pattern formation by vascular mesenchymal cells. American Journal of Physiology. Heart and Circulatory Physiology. 2012;303(12):H1411-H1417
  124. 124. Siegel AL, Kuhlmann PK, Cornelison DD. Muscle satellite cell proliferation and association: New insights from myofiber time-lapse imaging. Skeletal Muscle. 2011;1(1):7
  125. 125. Spear PC, Erickson CA. Interkinetic nuclear migration: A mysterious process in search of a function. Development, Growth & Differentiation. 2012;54(3):306-316
  126. 126. Spear PC, Erickson CA. Apical movement during interkinetic nuclear migration is a two-step process. Developmental Biology. 2012;370(1):33-41
  127. 127. Dong Z, Yang N, Yeo SY, Chitnis A, Guo S. Intralineage directional notch signaling regulates self-renewal and differentiation of asymmetrically dividing radial glia. Neuron. 2012;74(1):65-78
  128. 128. Namba T, Mochizuki H, Suzuki R, Onodera M, Yamaguchi M, Namiki H, et al. Time-lapse imaging reveals symmetric neurogenic cell division of GFAP-expressing progenitors for expansion of postnatal dentate granule neurons. PLoS One. 2011;6(9):e25303
  129. 129. Huang S, Law P, Francis K, Palsson BO, Ho AD. Symmetry of initial cell divisions among primitive hematopoietic progenitors is independent of ontogenic age and regulatory molecules. Blood. 1999;94(8):2595-2604
  130. 130. Neumann B, Walter T, Heriche JK, Bulkescher J, Erfle H, Conrad C, et al. Phenotypic profiling of the human genome by time-lapse microscopy reveals cell division genes. Nature. 2010;464(7289):721-727
  131. 131. Kosodo Y, Toida K, Dubreuil V, Alexandre P, Schenk J, Kiyokage E, et al. Cytokinesis of neuroepithelial cells can divide their basal process before anaphase. The EMBO Journal. 2008;27(23):3151-3163
  132. 132. Krishan A. Cytochalasin-B: Time-lapse cinematographic studies on its effects on cytokinesis. The Journal of Cell Biology. 1972;54(3):657-664
  133. 133. Kogo H, Fujimoto T. Concentration of caveolin-1 in the cleavage furrow as revealed by time-lapse analysis. Biochemical and Biophysical Research Communications. 2000;268(1):82-87
  134. 134. Guizetti J, Mantler J, Muller-Reichert T, Gerlich DW. Correlative time-lapse imaging and electron microscopy to study abscission in HeLa cells. Methods in Cell Biology. 2010;96:591-601
  135. 135. Sattler CA, Sawada N, Sattler GL, Pitot HC. Electron microscopic and time lapse studies of mitosis in cultured rat hepatocytes. Hepatology. 1988;8(6):1540-1549
  136. 136. Cone CD Jr, Tongier M Jr. Mitotic synchronization of L-strain fibroblasts with 5-aminouracil as determined by time-lapse cinephotography. NASA TN D-5021. Technical Note United States National Aeronautics and Space Administration. 1969:1-27
  137. 137. Stoll EA, Habibi BA, Mikheev AM, Lasiene J, Massey SC, Swanson KR, et al. Increased re-entry into cell cycle mitigates age-related neurogenic decline in the murine subventricular zone. Stem Cells. 2011;29(12):2005-2017
  138. 138. Absher M, Ryan US. Comparison of pulmonary endothelial cell and fibroblast proliferation using time-lapse cinematographic analysis. Tissue & Cell. 1981;13(4):645-650
  139. 139. Bedford JS, Mitchell JB. Mitotic accumulation of HeLa cells during continuous irradiation. Observations using time-lapse cinemicrography. Radiation Research. 1977;70(1):173-186
  140. 140. Collyn-d’Hooghe M, Hemon D, Gilet R, Curtis SB, Valleron AJ, Malaise EP. Comparative effects of 60Co gamma-rays and neon and helium ions on cycle duration and division probability of EMT 6 cells. A time-lapse cinematography study. International Journal of Radiation Biology and Related Studies in Physics, Chemistry, and Medicine. 1981;39(3):297-306
  141. 141. Sigoillot FD, Huckins JF, Li F, Zhou X, Wong ST, King RW. A time-series method for automated measurement of changes in mitotic and interphase duration from time-lapse movies. PLoS One. 2011;6(9):e25511
  142. 142. Dykstra B, Ramunas J, Kent D, McCaffrey L, Szumsky E, Kelly L, et al. High-resolution video monitoring of hematopoietic stem cells cultured in single-cell arrays identifies new features of self-renewal. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(21):8185-8190
  143. 143. Chu K, Teele N, Dewey MW, Albright N, Dewey WC. Computerized video time lapse study of cell cycle delay and arrest, mitotic catastrophe, apoptosis and clonogenic survival in irradiated 14-3-3sigma and CDKN1A (p21) knockout cell lines. Radiation Research. 2004;162(3):270-286
  144. 144. Dover R, Potten CS. Heterogeneity and cell cycle analyses from time-lapse studies of human keratinocytes in vitro. Journal of Cell Science. 1988;89(Pt 3):359-364
  145. 145. Zielke-Temme B, Hopwood L. Time-lapse cinemicrographic observations of heated G1-phase Chinese hamster ovary cells. I. Division probabilities and generation times. Radiation Research. 1982;92(2):320-331
  146. 146. d’Hooghe MC, Hemon D, Valleron AJ, Malaise EP. Comparative effects of ionizing radiations on cycle time and mitotic duration. A time-lapse cinematography study. Radiation Research. 1980;81(3):384-392
  147. 147. Colly-d’Hooghe M, Valleron AJ, Malaise EP. Time-lapse cinematography studies of cell cycle and mitosis duration. Experimental Cell Research. 1977;106(2):405-407
  148. 148. Coronado D, Godet M, Bourillot PY, Tapponnier Y, Bernat A, Petit M, et al. A short G1 phase is an intrinsic determinant of naive embryonic stem cell pluripotency. Stem Cell Research. 2013;10(1):118-131
  149. 149. Kinzel V, Bonheim G, Richards J. Phorbol ester-induced G2 delay in HeLa cells analyzed by time lapse photography. Cancer Research. 1988;48(7):1759-1762
  150. 150. Sundaram M, Guernsey DL, Rajaraman MM, Rajaraman R. Neosis: A novel type of cell division in cancer. Cancer Biology & Therapy. 2004;3(2):207-218
  151. 151. Yu B, Kim HW, Gong M, Wang J, Millard RW, Wang Y, et al. Exosomes secreted from GATA-4 overexpressing mesenchymal stem cells serve as a reservoir of anti-apoptotic microRNAs for cardioprotection. International Journal of Cardiology. 2015;182:349-360
  152. 152. Nagy G, Pinter G, Kohut G, Adam AL, Trencsenyi G, Hornok L, et al. Time-lapse analysis of cell death in mammalian and fungal cells. DNA and Cell Biology. 2010;29(5):249-259
  153. 153. Zahm JM, Baconnais S, Monier S, Bonnet N, Bessede G, Gambert P, et al. Chronology of cellular alterations during 7-ketocholesterol-induced cell death on A7R5 rat smooth muscle cells: Analysis by time lapse-video microscopy and conventional fluorescence microscopy. Cytometry Part A: The Journal of the International Society for Analytical Cytology. 2003;52(2):57-69
  154. 154. Forrester HB, Vidair CA, Albright N, Ling CC, Dewey WC. Using computerized video time lapse for quantifying cell death of X-irradiated rat embryo cells transfected with c-myc or c-Ha-ras. Cancer Research. 1999;59(4):931-939
  155. 155. Hurwitz C, Tolmach LJ. Time lapse cinemicrographic studies of x-irradiated HeLa S3 cells. I. Cell progression and cell disintegration. Biophysical Journal. 1969;9(4):607-633
  156. 156. Hwang SY, Cho SH, Cho DY, Lee M, Choo J, Jung KH, et al. Time-lapse, single cell based confocal imaging analysis of caspase activation and phosphatidylserine flipping during cellular apoptosis. Biotechnic & Histochemistry: Official Publication of the Biological Stain Commission. 2011;86(3):181-187
  157. 157. Saijo S, Nagata K, Masuda J, Matsumoto I, Kobayashi Y. Discrimination of early and late apoptotic cells by NBD-phosphatidylserine-labelling and time-lapse observation of phagocytosis of apoptotic cells by macrophages. Journal of Biochemistry. 2007;141(3):301-307
  158. 158. Wallberg F, Tenev T, Meier P. Time-lapse imaging of necrosis. Methods in Molecular Biology. 2013;1004:17-29
  159. 159. Cai Q, Zakaria HM, Sheng ZH. Long time-lapse imaging reveals unique features of PARK2/parkin-mediated mitophagy in mature cortical neurons. Autophagy. 2012;8(6):976-978
  160. 160. Oh DJ, Lee GM, Francis K, Palsson BO. Phototoxicity of the fluorescent membrane dyes PKH2 and PKH26 on the human hematopoietic KG1a progenitor cell line. Cytometry. 1999;36(4):312-318
  161. 161. Herman B, Albertini DF. A time-lapse video image intensification analysis of cytoplasmic organelle movements during endosome translocation. The Journal of Cell Biology. 1984;98(2):565-576
  162. 162. Farnum CE, Turgai J, Wilsman NJ. Visualization of living terminal hypertrophic chondrocytes of growth plate cartilage in situ by differential interference contrast microscopy and time-lapse cinematography. Journal of Orthopaedic Research: Official Publication of the Orthopaedic Research Society. 1990;8(5):750-763
  163. 163. Barasa A, Godina G, Buffa P, Pasquali-Ronchetti I. Biochemical lesions of respiratory enzymes and configurational changes of mitochondria in vivo. I. The effect of fluoroacetate: A study by phase-contrast microscopy and time-lapse cinemicrography. Zeitschrift für Zellforschung und Mikroskopische Anatomie. 1973;138(2):187-210
  164. 164. Sison M, Chakrabortty S, Extermann J, Nahas A, James Marchand P, Lopez A, et al. 3D time-lapse imaging and quantification of mitochondrial dynamics. Scientific Reports. 2017;7:43275
  165. 165. Gonzalez S, Fernando R, Berthelot J, Perrin-Tricaud C, Sarzi E, Chrast R, et al. In vivo time-lapse imaging of mitochondria in healthy and diseased peripheral myelin sheath. Mitochondrion. 2015;23:32-41
  166. 166. Spacek J. Dynamics of the Golgi method: A time-lapse study of the early stages of impregnation in single sections. Journal of Neurocytology. 1989;18(1):27-38
  167. 167. Pampalona J, Januschke J, Sampaio P, Gonzalez C. Time-lapse recording of centrosomes and other organelles in Drosophila neuroblasts. Methods in Cell Biology. 2015;129:301-315
  168. 168. Bang C, Cheng J. Dynamic interplay of spectrosome and centrosome organelles in asymmetric stem cell divisions. PLoS One. 2015;10(4):e0123294
  169. 169. Wiese C, Mayers JR, Albee AJ. Analysis of centrosome function and microtubule dynamics by time-lapse microscopy in Xenopus egg extracts. Methods in Molecular Biology. 2009;586:89-113
  170. 170. Braun A, Caesar NM, Dang K, Myers KA. High-resolution time-lapse imaging and automated analysis of microtubule dynamics in living human umbilical vein endothelial cells. Journal of Visualized Experiments: JoVE. 2016;114:54265
  171. 171. Sullivan KF, Shelby RD. Using time-lapse confocal microscopy for analysis of centromere dynamics in human cells. Methods in Cell Biology. 1999;58:183-202
  172. 172. Snapp EL, Lajoie P. Time-lapse imaging of membrane traffic in living cells. Cold Spring Harbor Protocols. 2011;2011(11):1362-1365
  173. 173. Verkuyl JM, Matus A. Time-lapse imaging of dendritic spines in vitro. Nature Protocols. 2006;1(5):2399-2405
  174. 174. Pearson RA, Luneborg NL, Becker DL, Mobbs P. Gap junctions modulate interkinetic nuclear movement in retinal progenitor cells. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2005;25(46):10803-10814
  175. 175. Kosodo Y, Suetsugu T, Suda M, Mimori-Kiyosue Y, Toida K, Baba SA, et al. Regulation of interkinetic nuclear migration by cell cycle-coupled active and passive mechanisms in the developing brain. The EMBO Journal. 2011;30(9):1690-1704
  176. 176. Abe S, Seitoku E, Iwadera N, Hamba Y, Yamagata S, Akasaka T, et al. Estimation of biocompatibility of nano-sized ceramic particles with osteoblasts, osteosarcomas and hepatocytes by static and time-lapse observation. Journal of Biomedical Nanotechnology. 2016;12(3):472-480
  177. 177. Schwamborn R, Dussmann H, Konig HG, Prehn JHM. Time-lapse imaging of p65 and IkappaBalpha translocation kinetics following Ca(2+)-induced neuronal injury reveals biphasic translocation kinetics in surviving neurons. Molecular and Cellular Neurosciences. 2017;80:148-158
  178. 178. Jokhadar SZ, Sustar V, Svetina S, Batista U. Time lapse monitoring of CaCo-2 cell shapes and shape dependence of the distribution of integrin beta1 and F-actin on their basal membrane. Cell Communication & Adhesion. 2009;16(1-3):1-13
  179. 179. Shimojo H, Isomura A, Ohtsuka T, Kori H, Miyachi H, Kageyama R. Oscillatory control of Delta-like1 in cell interactions regulates dynamic gene expression and tissue morphogenesis. Genes & Development. 2016;30(1):102-116
  180. 180. Trinkle-Mulcahy L, Andrews PD, Wickramasinghe S, Sleeman J, Prescott A, Lam YW, et al. Time-lapse imaging reveals dynamic relocalization of PP1gamma throughout the mammalian cell cycle. Molecular Biology of the Cell. 2003;14(1):107-117
  181. 181. Webb CD, Graumann PL, Kahana JA, Teleman AA, Silver PA, Losick R. Use of time-lapse microscopy to visualize rapid movement of the replication origin region of the chromosome during the cell cycle in Bacillus subtilis. Molecular Microbiology. 1998;28(5):883-892
  182. 182. Georgescu W, Osseiran A, Rojec M, Liu Y, Bombrun M, Tang J, et al. Characterizing the DNA damage response by cell tracking algorithms and cell features classification using high-content time-lapse analysis. PLoS One. 2015;10(6):e0129438
  183. 183. Mazo-Vargas A, Park H, Aydin M, Buchler NE. Measuring fast gene dynamics in single cells with time-lapse luminescence microscopy. Molecular Biology of the Cell. 2014;25(22):3699-3708
  184. 184. Villalta JI, Galli S, Iacaruso MF, Antico Arciuch VG, Poderoso JJ, Jares-Erijman EA, et al. New algorithm to determine true colocalization in combination with image restoration and time-lapse confocal microscopy to MAP kinases in mitochondria. PLoS One. 2011;6(4):e19031
  185. 185. Wang MD, Axelrod D. Time-lapse total internal reflection fluorescence video of acetylcholine receptor cluster formation on myotubes. Developmental Dynamics: An Official Publication of the American Association of the Anatomists. 1994;201(1):29-40
  186. 186. Swedlow JR, Sedat JW, Agard DA. Multiple chromosomal populations of topoisomerase II detected in vivo by time-lapse, three-dimensional wide-field microscopy. Cell. 1993;73(1):97-108
  187. 187. Hiraoka Y, Minden JS, Swedlow JR, Sedat JW, Agard DA. Focal points for chromosome condensation and decondensation revealed by three-dimensional in vivo time-lapse microscopy. Nature. 1989;342(6247):293-296
  188. 188. Owens DF, Kriegstein AR. Patterns of intracellular calcium fluctuation in precursor cells of the neocortical ventricular zone. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 1998;18(14):5374-5388
  189. 189. Stricker SA. Time-lapse confocal imaging of calcium dynamics in starfish embryos. Developmental Biology. 1995;170(2):496-518
  190. 190. Dussmann H, Perez-Alvarez S, Anilkumar U, Papkovsky DB, Prehn JH. Single-cell time-lapse imaging of intracellular O2 in response to metabolic inhibition and mitochondrial cytochrome-c release. Cell Death & Disease. 2017;8(6):e2853
  191. 191. Hoffman H, Frank ME. Time-lapse photomicrography of cell growth and division in Escherichia coli. Journal of Bacteriology. 1965;89:212-216
  192. 192. Hoffman H, Frank ME. Time-lapse photomicrography of lashing, flexing, and snapping movements in Escherichia coli and Corynebacterium microcultures. Journal of Bacteriology. 1965;90(3):789-795
  193. 193. Shapiro JA, Hsu C. Escherichia coli K-12 cell-cell interactions seen by time-lapse video. Journal of Bacteriology. 1989;171(11):5963-5974
  194. 194. Hoffman H, Frank ME. Time-lapse photomicrography of the formation of a free spherical granule in an Escherichia coli cell end. Journal of Bacteriology. 1963;86:1075-1078
  195. 195. Penil Cobo M, Libro S, Marechal N, D’Entremont D, Penil Cobo D, Berkmen M. Visualizing bacterial colony morphologies using time-lapse imaging chamber MOCHA. Journal of Bacteriology. 2018;200(2):e00413-17
  196. 196. Ungphakorn W, Lagerback P, Nielsen EI, Tangden T. Automated time-lapse microscopy a novel method for screening of antibiotic combination effects against multidrug-resistant gram-negative bacteria. Clinical Microbiology and Infection: The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases. 2018;24(7):778.e777-778.e714
  197. 197. Durham NN, Noller EC, Burger MW, Best GK. Time-lapse cinematography of vancomycin--treated microbial cells. Canadian Journal of Microbiology. 1967;13(4):417-421
  198. 198. Ungphakorn W, Malmberg C, Lagerback P, Cars O, Nielsen EI, Tangden T. Evaluation of automated time-lapse microscopy for assessment of in vitro activity of antibiotics. Journal of Microbiological Methods. 2017;132:69-75
  199. 199. Louvet JN, Carrion C, Stalder T, Alrhmoun M, Casellas M, Potier O, et al. Vancomycin sorption on activated sludge gram(+) bacteria rather than on EPS; 3D confocal laser scanning microscopy time-lapse imaging. Water Research. 2017;124:290-297
  200. 200. Schmidt GW, Frey O, Rudolf F. The CellClamper: A convenient microfluidic device for time-lapse imaging of yeast. Methods in Molecular Biology. 2018;1672:537-555
  201. 201. Kumar A, Mendoza M. Time-lapse fluorescence microscopy of budding yeast cells. Methods in Molecular Biology. 2016;1369:1-8
  202. 202. Kron SJ. Digital time-lapse microscopy of yeast cell growth. Methods in Enzymology. 2002;351:3-15
  203. 203. Heymann JB, Cheng N, Newcomb WW, Trus BL, Brown JC, Steven AC. Dynamics of herpes simplex virus capsid maturation visualized by time-lapse cryo-electron microscopy. Nature Structural Biology. 2003;10(5):334-341
  204. 204. Lemay P, Collyn-D’Hooghe M. Flow cytophotometric and time-lapse cinematographic study of human cells infected by adenovirus type 2 wild-type and two DNA-negative temperature-sensitive mutants. The Journal of General Virology. 1984;65(Pt 8):1419-1423
  205. 205. Wright HT Jr, Kasten FH, McAllister RM. Human cytomegalovirus, observations of intracellular lesion development as revealed by phase contrast, time-lapse cinematography. Proceedings of the Society for Experimental Biology and Medicine Society for Experimental Biology and Medicine. 1968;127(4):1032-1036
  206. 206. Smith KM, Brown RM Jr, Walne PL. Ultrastructural and time-lapse studies on the replication cycle of the blue-green algal virus LPP-1. Virology. 1967;31(2):329-337
  207. 207. Duprex WP, Rima BK. Using green fluorescent protein to monitor measles virus cell-to-cell spread by time-lapse confocal microscopy. Methods in Molecular Biology. 2002;183:297-307
  208. 208. Riddle PN. Time-lapse cinemicroscopy. Methods in Molecular Biology. 1990;5:415-446
  209. 209. Kulesa PM, Kasemeier-Kulesa JC. Construction of a heated incubation chamber around a microscope stage for time-lapse imaging. CSH Protocols. 2007;2007:pdb prot4792
  210. 210. Tuttle C. Analysis of equipment and methods for time lapse motion photomicrography. Journal of Biological Photography. 1992;60(2):54-55
  211. 211. Constable FL, Moffat MA. A glass tissue culture chamber for use in time-lapse cinematography. Journal of Clinical Pathology. 1958;11(5):455-457
  212. 212. Fischler HA. Design of a phase contrast time-lapse cinemicrographic unit. Journal of the Biological Photographic Association. 1966;34(2):53-82
  213. 213. Shand FL. An inexpensive high intensity lamp unit for phase-contrast time-lapse cinemicrography. Journal of the Royal Microscopical Society. 1967;86(4):437-440
  214. 214. Dawson M, Johnstone AJ, Matthews JE. An electronic light source and modified Bolex H-16 camera for time-lapse cinemicrography. Journal of Microscopy. 1970;91(2):139-143
  215. 215. Dawson M, Matthews JE. Apparatus for time-lapse cinemicrography. Journal of Microscopy. 1972;96(1):97-103
  216. 216. Peters JH. Device for time lapse studies on living cells in vitro (author’s transl). Microscopica Acta. 1979;81(3):217-225
  217. 217. Vesely P, Maly M, Cumpelik J, Pluta M, Tuma V. Improved spatial and temporal resolution in an apparatus for time-lapse phase contrast cine micrography of cells in vitro. Journal of Microscopy. 1982;125(Pt 1):67-76
  218. 218. Allen TD. Time lapse video microscopy using an animation control unit. Journal of Microscopy. 1987;147(Pt 2):129-135
  219. 219. Endlich B, Radford IR, Forrester HB, Dewey WC. Computerized video time-lapse microscopy studies of ionizing radiation-induced rapid-interphase and mitosis-related apoptosis in lymphoid cells. Radiation Research. 2000;153(1):36-48
  220. 220. Armstrong WD, Wilt JC, Pritchard ET. Vacuolation in the human amnion cell studies by time—Lapse photography and electron microscopy. American Journal of Obstetrics and Gynecology. 1968;102(7):932-948
  221. 221. Choi BH, Cho KH, Lapham LW. Effects of methylmercury on human fetal neurons and astrocytes in vitro: A time-lapse cinematographic, phase and electron microscopic study. Environmental Research. 1981;24(1):61-74
  222. 222. Balfour BM, Goscicka T, MacKenzie JL, Gautam A, Tate M, Clark J. Combined time-lapse cinematography and immuno-electron microscopy. The Anatomical Record. 1990;226(4):509-514
  223. 223. LeSage AJ, Kron SJ. Design and implementation of algorithms for focus automation in digital imaging time-lapse microscopy. Cytometry. 2002;49(4):159-169
  224. 224. Brieu N, Navab N, Serbanovic-Canic J, Ouwehand WH, Stemple DL, Cvejic A, et al. Image-based characterization of thrombus formation in time-lapse DIC microscopy. Medical Image Analysis. 2012;16(4):915-931
  225. 225. Concha ML, Adams RJ. Oriented cell divisions and cellular morphogenesis in the zebrafish gastrula and neurula: A time-lapse analysis. Development. 1998;125(6):983-994
  226. 226. Aletta JM, Greene LA. Growth cone configuration and advance: A time-lapse study using video-enhanced differential interference contrast microscopy. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 1988;8(4):1425-1435
  227. 227. Montgomery PO, Bonner WA, Roberts F. Ultra-violet flying spot time lapse motion picture observations of living cells. Texas Reports on Biology and Medicine. 1957;15(3):386-395
  228. 228. Malide D, Metais JY, Dunbar CE. In vivo clonal tracking of hematopoietic stem and progenitor cells marked by five fluorescent proteins using confocal and multiphoton microscopy. Journal of Visualized Experiments: JoVE. 2014;90:e51669
  229. 229. Noctor SC. Time-lapse imaging of fluorescently labeled live cells in the embryonic mammalian forebrain. Cold Spring Harbor Protocols. 2011;2011(11):1350-1361
  230. 230. Stadtfeld M, Varas F, Graf T. Fluorescent protein-cell labeling and its application in time-lapse analysis of hematopoietic differentiation. Methods in Molecular Medicine. 2005;105:395-412
  231. 231. Ellenberg J, Lippincott-Schwartz J, Presley JF. Two-color green fluorescent protein time-lapse imaging. BioTechniques. 1998;25(5):838-42-8344-6
  232. 232. Nakamura M, Miyamoto K, Hayashi K, Awaad A, Ochiai M, Ishimura K. Time-lapse fluorescence imaging and quantitative single cell and endosomal analysis of peritoneal macrophages using fluorescent organosilica nanoparticles. Nanomedicine: Nanotechnology, Biology and Medicine. 2013;9(2):274-283
  233. 233. Cova L, Bigini P, Diana V, Sitia L, Ferrari R, Pesce RM, et al. Biocompatible fluorescent nanoparticles for in vivo stem cell tracking. Nanotechnology. 2013;24(24):245603
  234. 234. Yun SW, Leong C, Bi X, Ha HH, Yu YH, Tan YL, et al. A fluorescent probe for imaging symmetric and asymmetric cell division in neurosphere formation. Chemical Communications. 2014;50(56):7492-7494
  235. 235. Lee GM, Fong SS, Oh DJ, Francis K, Palsson BO. Characterization and efficacy of PKH26 as a probe to study the replication history of the human hematopoietic KG1a progenitor cell line. In Vitro Cellular & Developmental Biology. Animal. 2002;38(2):90-96
  236. 236. Heye RR, Kiebler EW, Arnzen RJ, Tolmach LJ. Multiplexed time-lapse photomicrography of cultured cells. Journal of Microscopy. 1982;125(Pt 1):41-50
  237. 237. Kallman RF, Blevins N, Coyne MA, Prionas SD. Novel instrumentation for multifield time-lapse cinemicrography. Computers and Biomedical Research, An International Journal. 1990;23(2):115-129
  238. 238. Hamant O, Das P, Burian A. Time-lapse imaging of developing meristems using confocal laser scanning microscope. Methods in Molecular Biology. 2014;1080:111-119
  239. 239. Boisset JC, Andrieu-Soler C, van Cappellen WA, Clapes T, Robin C. Ex vivo time-lapse confocal imaging of the mouse embryo aorta. Nature Protocols. 2011;6(11):1792-1805
  240. 240. Nowotschin S, Ferrer-Vaquer A, Hadjantonakis AK. Imaging mouse development with confocal time-lapse microscopy. Methods in Enzymology. 2010;476:351-377
  241. 241. Megason SG. In toto imaging of embryogenesis with confocal time-lapse microscopy. Methods in Molecular Biology. 2009;546:317-332
  242. 242. Rupp PA, Kulesa PM. High-resolution, intravital 4D confocal time-lapse imaging in avian embryos using a Teflon culture chamber design. CSH Protocols. 2007;2007:pdb prot4790
  243. 243. Carvalho L, Heisenberg CP. Imaging zebrafish embryos by two-photon excitation time-lapse microscopy. Methods in Molecular Biology. 2009;546:273-287
  244. 244. Rebollo E, Gonzalez C. Time-lapse imaging of embryonic neural stem cell division in drosophila by two-photon microscopy. Current Protocols in Stem Cell Biology. 2010; 13(1):1H.2.1-1H.2.9
  245. 245. Pakan JM, McDermott KW. A method to investigate radial glia cell behavior using two-photon time-lapse microscopy in an ex vivo model of spinal cord development. Frontiers in Neuroanatomy. 2014;8:22
  246. 246. Gerlich D, Ellenberg J. 4D imaging to assay complex dynamics in live specimens. Nature Cell Biology. 2003;(Suppl):S14-S19
  247. 247. Dierickx P, Vermunt MW, Muraro MJ, Creyghton MP, Doevendans PA, van Oudenaarden A, et al. Circadian networks in human embryonic stem cell-derived cardiomyocytes. EMBO Reports. 2017;18(7):1199-1212
  248. 248. Kuo HL, Ho PC, Huang SS, Chang NS. Chasing the signaling run by tri-molecular time-lapse FRET microscopy. Cell Death Discovery. 2018;4:45
  249. 249. Majdi JA, Qian H, Li Y, Langsner RJ, Shea KI, Agrawal A, et al. The use of time-lapse optical coherence tomography to image the effects of microapplied toxins on the retina. Investigative Ophthalmology & Visual Science. 2014;56(1):587-597
  250. 250. Pan YT, Wu ZL, Yuan ZJ, Wang ZG, Du CW. Subcellular imaging of epithelium with time-lapse optical coherence tomography. Journal of Biomedical Optics. 2007;12(5):050504
  251. 251. Pan YT, Wu Q, Wang ZG, Brink PR, Du CW. High-resolution imaging characterization of bladder dynamic morphophysiology by time-lapse optical coherence tomography. Optics Letters. 2005;30(17):2263-2265
  252. 252. Gabriel M, Balle D, Bigault S, Pornin C, Getin S, Perraut F, et al. Time-lapse contact microscopy of cell cultures based on non-coherent illumination. Scientific Reports. 2015;5:14532
  253. 253. Schiefer J, Kampe K, Dodt HU, Zieglgansberger W, Kreutzberg GW. Microglial motility in the rat facial nucleus following peripheral axotomy. Journal of Neurocytology. 1999;28(6):439-453
  254. 254. Maurice DM, Zhao J, Nagasaki T. A novel microscope system for time-lapse observation of corneal cells in a living mouse. Experimental Eye Research. 2004;78(3):591-597
  255. 255. Youssef S, Gude S, Radler JO. Automated tracking in live-cell time-lapse movies. Integrative Biology: Quantitative Biosciences From Nano to Macro. 2011;3(11):1095-1101
  256. 256. Dewan MA, Ahmad MO, Swamy MN. Tracking biological cells in time-lapse microscopy: An adaptive technique combining motion and topological features. IEEE Transactions on Bio-Medical Engineering. 2011;58(6):1637-1647
  257. 257. Kirby BB, Takada N, Latimer AJ, Shin J, Carney TJ, Kelsh RN, et al. In vivo time-lapse imaging shows dynamic oligodendrocyte progenitor behavior during zebrafish development. Nature Neuroscience. 2006;9(12):1506-1511
  258. 258. Chen X, Zhou X, Wong ST. Automated segmentation, classification, and tracking of cancer cell nuclei in time-lapse microscopy. IEEE Transactions on Bio-Medical Engineering. 2006;53(4):762-766
  259. 259. Meijering E, Dzyubachyk O, Smal I. Methods for cell and particle tracking. Methods in Enzymology. 2012;504:183-200
  260. 260. Dzyubachyk O, van Cappellen WA, Essers J, Niessen WJ, Meijering E. Advanced level-set-based cell tracking in time-lapse fluorescence microscopy. IEEE Transactions on Medical Imaging. 2010;29(3):852-867
  261. 261. Jaqaman K, Loerke D, Mettlen M, Kuwata H, Grinstein S, Schmid SL, et al. Robust single-particle tracking in live-cell time-lapse sequences. Nature Methods. 2008;5(8):695-702
  262. 262. Huth J, Buchholz M, Kraus JM, Schmucker M, von Wichert G, Krndija D, et al. Significantly improved precision of cell migration analysis in time-lapse video microscopy through use of a fully automated tracking system. BMC Cell Biology. 2010;11:24
  263. 263. Schoenauer Sebag A, Plancade S, Raulet-Tomkiewicz C, Barouki R, Vert JP, Walter T. A generic methodological framework for studying single cell motility in high-throughput time-lapse data. Bioinformatics. 2015;31(12):i320-i328
  264. 264. Bray MA, Carpenter AE. CellProfiler tracer: Exploring and validating high-throughput, time-lapse microscopy image data. BMC Bioinformatics. 2015;16:368
  265. 265. Brandes S, Mokhtari Z, Essig F, Hunniger K, Kurzai O, Figge MT. Automated segmentation and tracking of non-rigid objects in time-lapse microscopy videos of polymorphonuclear neutrophils. Medical Image Analysis. 2015;20(1):34-51
  266. 266. Barbier de Reuille P, Routier-Kierzkowska AL, Kierzkowski D, Bassel GW, Schupbach T, Tauriello G, et al. MorphoGraphX: A platform for quantifying morphogenesis in 4D. eLife. 2015;4:05864
  267. 267. Mankowski WC, Winter MR, Wait E, Lodder M, Schumacher T, Naik SH, et al. Segmentation of occluded hematopoietic stem cells from tracking. Conference proceedings: Annual International Conference of the IEEE Engineering in Medicine and Biology Society IEEE Engineering in Medicine and Biology Society Annual Conference. 2014;2014:5510-5513
  268. 268. Tarnawski W, Kurtcuoglu V, Lorek P, Bodych M, Rotter J, Muszkieta M, et al. A robust algorithm for segmenting and tracking clustered cells in time-lapse fluorescent microscopy. IEEE Journal of Biomedical and Health Informatics. 2013;17(4):862-869
  269. 269. Storr A, Venetis C, Cooke S, Kilani S, Ledger W. Time-lapse algorithms and morphological selection of day-5 embryos for transfer: A preclinical validation study. Fertility and Sterility. 2018;109(2):276-83.e3
  270. 270. Liu Y, Feenan K, Chapple V, Matson P. Assessing efficacy of day 3 embryo time-lapse algorithms retrospectively: Impacts of dataset type and confounding factors. Human Fertility. 2018:1-9. https://doi.org/10.1080/14647273.2018.1425919
  271. 271. Reignier A, Lammers J, Barriere P, Freour T. Can time-lapse parameters predict embryo ploidy? A systematic review. Reproductive Biomedicine Online. 2018;36(4):380-387
  272. 272. Swain JE. Could time-lapse embryo imaging reduce the need for biopsy and PGS? Journal of Assisted Reproduction and Genetics. 2013;30(8):1081-1090
  273. 273. Herrero J, Meseguer M. Selection of high potential embryos using time-lapse imaging: The era of morphokinetics. Fertility and Sterility. 2013;99(4):1030-1034
  274. 274. Ergin EG, Caliskan E, Yalcinkaya E, Oztel Z, Cokelez K, Ozay A, et al. Frequency of embryo multinucleation detected by time-lapse system and its impact on pregnancy outcome. Fertility and Sterility. 2014;102(4):1029-33.e1
  275. 275. Desai N, Goldberg JM, Austin C, Falcone T. Are cleavage anomalies, multinucleation, or specific cell cycle kinetics observed with time-lapse imaging predictive of embryo developmental capacity or ploidy? Fertility and Sterility. 2018;109(4):665-674
  276. 276. Milewski R, Ajduk A. Time-lapse imaging of cleavage divisions in embryo quality assessment. Reproduction. 2017;154(2):R37-R53
  277. 277. Chawla M, Fakih M, Shunnar A, Bayram A, Hellani A, Perumal V, et al. Morphokinetic analysis of cleavage stage embryos and its relationship to aneuploidy in a retrospective time-lapse imaging study. Journal of Assisted Reproduction and Genetics. 2015;32(1):69-75
  278. 278. Campbell A, Fishel S, Laegdsmand M. Aneuploidy is a key causal factor of delays in blastulation: Author response to ‘a cautionary note against aneuploidy risk assessment using time-lapse imaging’. Reproductive Biomedicine Online. 2014;28(3):279-283
  279. 279. Daughtry BL, Chavez SL. Time-lapse imaging for the detection of chromosomal abnormalities in primate preimplantation embryos. Methods in Molecular Biology. 2018;1769:293-317
  280. 280. Castello D, Motato Y, Basile N, Remohi J, Espejo-Catena M, Meseguer M. How much have we learned from time-lapse in clinical IVF? Molecular Human Reproduction. 2016;22(10):719-727
  281. 281. Sterckx S, Cockbain J, Pennings G. Patenting time-lapse microscopy: The European story. Reproductive Biomedicine Online. 2014;28(2):146-150
  282. 282. Sterckx S, Cockbain J, Pennings G. Patenting medical diagnosis methods in Europe: Stanford University and time-lapse microscopy. Reproductive Biomedicine Online. 2017;34(2):166-168
  283. 283. Urbano LF, Masson P, VerMilyea M, Kam M. Automatic tracking and motility analysis of human sperm in time-lapse images. IEEE Transactions on Medical Imaging. 2017;36(3):792-801
  284. 284. Wei F, Rong XX, Xie RY, Jia LT, Wang HY, Qin YJ, et al. Cytokine-induced killer cells efficiently kill stem-like cancer cells of nasopharyngeal carcinoma via the NKG2D-ligands recognition. Oncotarget. 2015;6(33):35023-35039
  285. 285. Schiraldi C, Zappavigna S, D’ Agostino A, Porto S, Gaito O, Lusa S, et al. Nanoparticles for the delivery of zoledronic acid to prostate cancer cells: A comparative analysis through time lapse video-microscopy technique. Cancer Biology & Therapy. 2014;15(11):1524-1532
  286. 286. Lin HD, Fong CY, Biswas A, Choolani M, Bongso A. Human Wharton’s jelly stem cells, its conditioned medium and cell-free lysate inhibit the growth of human lymphoma cells. Stem Cell Reviews. 2014;10(4):573-586
  287. 287. Bago JR, Pegna GJ, Okolie O, Mohiti-Asli M, Loboa EG, Hingtgen SD. Electrospun nanofibrous scaffolds increase the efficacy of stem cell-mediated therapy of surgically resected glioblastoma. Biomaterials. 2016;90:116-125
  288. 288. Bago JR, Alfonso-Pecchio A, Okolie O, Dumitru R, Rinkenbaugh A, Baldwin AS, et al. Therapeutically engineered induced neural stem cells are tumour-homing and inhibit progression of glioblastoma. Nature Communications. 2016;7:10593
  289. 289. Yasin R, Van Beers G, Riddle PN, Brown D, Widdowson G, Thompson EJ. An abnormality of cell behaviour in human dystrophic muscle cultures: A time-lapse study. Journal of Cell Science. 1979;38:201-210
  290. 290. Weiger MC, Vedham V, Stuelten CH, Shou K, Herrera M, Sato M, et al. Real-time motion analysis reveals cell directionality as an indicator of breast cancer progression. PLoS One. 2013;8(3):e58859
  291. 291. Tsujioka T, Matsuoka A, Tohyama Y, Tohyama K. Approach to new therapeutics: Investigation by the use of MDS-derived cell lines. Current Pharmaceutical Design. 2012;18(22):3204-3214
  292. 292. Yano S, Tazawa H, Hashimoto Y, Shirakawa Y, Kuroda S, Nishizaki M, et al. A genetically engineered oncolytic adenovirus decoys and lethally traps quiescent cancer stem-like cells in S/G2/M phases. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2013;19(23):6495-6505
  293. 293. Turani M, Banfalvi G, Peter A, Kukoricza K, Kiraly G, Talas L, et al. Antibiotics delay in vitro human stem cell regrowth. Toxicology In Vitro: An International Journal Published in Association with BIBRA. 2015;29(2):370-379
  294. 294. Pulkkinen JO, Elomaa L, Joensuu H, Martikainen P, Servomaa K, Grenman R. Paclitaxel-induced apoptotic changes followed by time-lapse video microscopy in cell lines established from head and neck cancer. Journal of Cancer Research and Clinical Oncology. 1996;122(4):214-218
  295. 295. Nakamura Y, Ishigaki Y. Immunostaining and time-lapse analysis of vinblastine-induced paracrystal formation in human A549 cells. Oncology Letters. 2014;8(6):2387-2392
  296. 296. Furukawa T, Inoue H, Sugita K, Eguchi M, Sakakibara H, Sugiyama S, et al. Long-term cinemicrography of erythroblasts from a patient with congenital dyserythropoietic anemia type III: Direct observation of dysplastic erythroblast formation. Blood Cells. 1993;19(2):493-506. Discussion 7-8
  297. 297. Xiang Z, Block M, Lofman C, Nilsson G. IgE-mediated mast cell degranulation and recovery monitored by time-lapse photography. The Journal of Allergy and Clinical Immunology. 2001;108(1):116-121
  298. 298. Barretto RP, Ko TH, Jung JC, Wang TJ, Capps G, Waters AC, et al. Time-lapse imaging of disease progression in deep brain areas using fluorescence microendoscopy. Nature Medicine. 2011;17(2):223-228
  299. 299. Dieterlen-Lievre F, Jaffredo T. Decoding the hemogenic endothelium in mammals. Cell Stem Cell. 2009;4(3):189-190
  300. 300. Slocum HK, Parsons JC, Winslow EO, Broderick L, Minderman H, Toth K, et al. Time-lapse video reveals immediate heterogeneity and heritable damage among human ileocecal carcinoma HCT-8 cells treated with raltitrexed (ZD1694). Cytometry. 2000;41(4):252-260

Written By

John L. Collins, Bart van Knippenberg, Kai Ding and Alexander V. Kofman

Submitted: 08 August 2018 Reviewed: 29 August 2018 Published: 05 November 2018