Millipore Sigma Vibrant Logo
 
 

Cell Analysis On-Demand Webinars


Listen and watch our cell analysis webinars to learn about the latest technologies, experimental tips and troubleshooting strategies for simple cell counts, multidimensional cell analysis, imaging flow cytometry and more.

Automation of the In Vitro Micronucleus Assay using Imaging Flow Cytometry

Oct2017
  • Presenter:  Matthew. A Rodrigues, PhD., Research Scientist with Amnis-Merck KGaA, Darmstadt, Germany
  • Abstract
    In this webinar, adaptation of the in vitro MN assay to an imaging flow cytometry-based method will be described. Using the ISX Mark II imaging flow cytometer, images of micronucleated mono- and binucleated cells as well as polynucleated cells can be captured at a high flow rate and automatically identified and scored in the Image Data Exploration and Analysis Software (IDEAS®) that accompanies the ISX. A data analysis template created specifically for this application allows for the determination of both genotoxicity and cytotoxicity following treatment with known clastogens and aneugens. This work is the first demonstration of fully automated method for performing the in vitro MN assay on an imaging flow cytometry platform.


ToxTracker Assay for High-throughput Screening of Genetic Toxicity

June | 2017
  • Presenter: Giel Hendriks, CEO, Toxys
  • Abstract
    With the increasing production of new chemicals for a wide range of applications in health care, food and cosmetics comes the demand for rapid and reliable approaches for toxicity assessment. Novel innovative in vitro systems should ideally not only identify toxic properties of chemicals, but also provide insight into the type of cellular damage inflicted in order to more reliably predict human health hazard of novel compounds.

    ToxTracker is a mammalian stem cell-based reporter assay that detects activation of specific cellular signalling pathways upon exposure to unknown compounds (Hendriks et al, Tox Sci 2016). ToxTracker contains six different GFP-tagged reporters that allows discrimination between induction of different types of DNA damage, various cellular anti-oxidant responses and activation of the unfolded protein response. The ToxTracker reporter cell lines are combined in 96-well plates, exposed to the test samples, typically for 24h, and induction of the fluorescent reporters is examined by flow cytometry (Merck Guava® easyCyte).

    The integrative approach of the ToxTracker assay provides a powerful tool for in vitro cancer hazard screening of chemicals by unveiling activation of specific cellular signalling pathways upon exposure and deliver insight into the underlying mechanism of toxicity.

    In this webcast you will learn:
    • A novel approach to toxicity screening
    • How the unique microcapillary fluidics design of the Guava® flow cytometers enables direct sampling, conferring the distinct advantage of absolute cell counts, and permitting direct correlation between cellular responses and cytotoxicity


Monitoring Immune Function by Imaging Flow Cytometry

June | 2017
  • Presenter: Orla Maguire, Ph.D., Roswell Park Cancer Institute, Buffalo, NY
  • Abstract
    The immune system plays a critical role not only in fending off pathogen attack, but also in cancer surveillance, and more recently as a tool in immunotherapy-based treatments. Immune cell functions are tightly regulated by essential transcription factors such as NF-κB and NFAT. Monitoring immune cell activity – including phenotyping immune cell subsets, tracking cell proliferation, and measuring cytokine production – can provide insights into the overall status of immune function in patients, particularly those undergoing immunosuppression after transplants, enduring cancer treatment, or suffering from autoimmune disease or other pathologies that affect the immune system. Imaging flow cytometry (IFC) has emerged as a useful and efficient tool for studying the signaling pathways in immunophenotypically defined subpopulations of immune cells. This technique enables quantitative image analysis of the intracellular localization of the signaling intermediaries NF-κB and NFAT as parameters of immune activity. This webinar will introduce viewers to the process of using IFC to determine subcellular localization of biomarkers, including a discussion of how IFC can help to assess the activity of transcription factors, or the drug-induced stimulation or inhibition thereof, in clinical samples.

    What will you learn?
    During the webinar, viewers will:
    • Learn the process of using IFC to study transcription-factor signaling
    • Discover how IFC can help to determine the effects of drug inhibition or stimulation on immune function
    • Gain insight into preclinical and clinical applications of IFC
    • Be able to ask questions during the live broadcast!


Circulating Tumour Cells: Liquid Biopsy by High-throughput, High-resolution Imaging Flow Cytometry

March | 2017
  • Presenter: Dr. David Jamieson, Research Associate Northern Institute for Cancer Research, University of Newcastle upon Tyne
  • Abstract
    Circulating tumour cells (CTCs) have the potential to act as a source of tumour tissue for the measurement of pharmacodynamic biomarkers in early phase clinical trials. This webcast will describe the sample processing, data collection and data analysis used by Dr. David Jamieson to characterise CTCs by imaging flow cytometry.


Live Cell Imaging Reveals How Cells Find and Maintain Their Division Sites

Nov | 2016
  • Presenter: PJ Buske, PhD Postdoctoral Research Fellow Lab of Dr. Dyche Mullins, HHMI/UCSF
  • Abstract
    Knowing when and where to divide is essential to cell survival and to guarantee proper allotment of the cellular contents in order to produce viable daughter cells. In bacteria, the division site is marked by the formation of a cytoskeletal ring (Z ring) made up of filaments comprised of the highly conserved tubulin homolog FtsZ. Once assembled, the Z ring serves as a platform for assembly of the division machinery. To date, how anchoring of FtsZ filaments to the membrane occurs has been poorly understood.

    In this webinar, Dr. PJ Buske will describe how dual-color, continuous live-cell fluorescent imaging using the CellASIC® Onix2 system has allowed the Mullins lab at UCSF to track co-localization of FtsZ and anchor proteins over multiple division cycles. Dr. Buske will discuss how this technology can be used to elucidate key protein interactions with FtsZ to drive filament dynamics and other processes such as cell wall synthesis and DNA replication.


Autophagy in the Hematopoietic System

Nov | 2016
  • Presenter: Dr. Anna Katharina (Katja) A Simon, Kennedy Institute of Rheumatology, University of Oxford
  • Abstract
    Autophagy is essential in determining cell fate in hematopoietic cells. Firstly the removal of mitochondria from erythroblasts allows survival and their final maturation step into mature red blood cells. Secondly autophagy is essential to strike the fine balance between quiescence, self-renewal, and differentiation of hematopoietic stem cells. Thirdly declining levels of autophagy in ageing macrophages contributes to senescent features. And lastly we have shown that memory T cells, the cells that mediate long-term protection after vaccination, cannot form in the absence of autophagy as they fail to undergo a necessary metabolic switch. We have also established different ways to detect autophagy in primary immune cells and rare stem cells using the ImageStream® imaging flow cytometer and traditional flow cytometry. With these techniques we have been able to contribute to the description of the first immune deficiencies in which the autophagy/lysoosomal pathway seems to be involved.

    In the long term, understanding molecular mechanisms that can manipulate cell fate of hematological stem cells and their progeny will be essential in the context of hematological malignancies, regenerative medicine and immune responses.

    In this webinar you will learn:
    • How autophagy influences hematopoietic cell survival and cell fate.
    • Macrophage senescence is associated with decreased autophagy.
    • The best ways to measure autophagy in primary cells.
    • How autophagy contributes to immune deficiencies.


Analysis of Extracellular Vesicles Including Exosomes by Imaging Flow Cytometry

Jun | 2016
  • Presenter: André Görgens, Ph.D. University Hospital-Essen, Essen, Germany
  • Abstract
    Extracellular vesicles (EVs) such as exosomes (70 nm – 160 nm in diameter) and microvesicles (100 nm – 1,000 nm diameter) can be harvested from cell-culture supernatants and from all bodily fluids. Current standard techniques to visualize, quantify, and characterize EVs are electron microscopy, nanoparticle tracking analyses, and dynamic light scattering. To further characterize and discriminate EVs, more exact high-throughput technologies to analyze their surface are highly desired. Although conventional flow cytometry is limited to measuring particles down to approximately 300 nm – 500 nm, a relatively new flow-cytometric method—called “imaging flow cytometry”—allows for the analysis of EVs smaller than 300 nm. This webinar will introduce viewers to the challenges, limitations, and pitfalls of flow cytometry-based EV analysis, and to the imaging flow cytometry methodology. Also covered will be techniques for analyzing exosomes, microvesicles, and apoptotic bodies in unprocessed samples, how imaging flow cytometry can be used to evaluate or reevaluate EV isolation techniques, and the advantages and disadvantages of using this method.


Quantitating Critical Subcellular Events in Significant Populations Using Imaging Flow Cytometry

Mar | 2016
  • Presenters: Olivier Thaunat, M.D., Assistant Professor, Hôpital Edouard Herriot, Hospices Civils de Lyon, University of Lyon; and Christopher Parris, Ph.D., Director, Division of Biosciences-Life Sciences, Brunel University London, College of Health and Life Sciences
  • Abstract
    During the activation of humoral immune responses, B cells acquire antigen for subsequent presentation to cognate T cells. Imaging flow cytometry reveals that antigen polarization is preserved during B cell division, promoting asymmetric antigen segregation among progeny. The generation of progeny with differential capacities for antigen presentation may have implications for somatic hypermutation and class switching during affinity maturation and as B cells commit to effector cell fates.

    Rigorous methods for the detection of DNA-damaged foci in eukaryotic cells are fundamental to DNA repair studies, which delineate mechanisms and may identify differences in DNA repair capacity among cell types. The Parris lab performed the first analyses to demonstrate the use of imaging flow cytometry for the detection of ã-H2AX foci in cells exposed to ionizing radiation that induces DNA double strand breaks. This webinar presents data validating the enhancement of foci quantitation and image resolution employing imaging flow cytometry, using cell lines derived from normal individuals. In this webinar, Dr. Thaunat’s work demonstrates that imaging flow cytometry (IFC) is key to the identification of polarized distribution of antigen on B cells that persists in vivo. Dr. Parris will describe how IFC with multimagnification and extended depth of field enhances quantitation of DNA damage in cells, providing a quantitative alternative to traditional low-throughput in situ microscopy methods for the detection of ã-H2AX foci.

High-Throughput Analysis of Viral Composition, Entry Kinetics, and Conformational Change

Nov | 2015
  • Presenter: Dr. Hector Aguilar-Carreno, Assistant Professor, Paul G. Allen School for Global Animal Health, Washington State University
  • Abstract
    Viral fusion with the cell membrane is essential for infection by enveloped viruses. For BSL4 (biosafety level 4) human pathogens such as the deadly Nipah virus (NiV), mechanistic studies of the fusion process are particularly challenging, as cell-cell fusion assays do not fully recapitulate the variables of viral entry into cells. In this webinar, our panelist will describe how his lab uses flow cytometry and surrogate virus particles to transform technical and biosafety challenges into a new model for study of virus-cell fusion. Following the presentation, attendees will have an opportunity to ask questions concerning their research and receive answers in real-time.

Innovation in Cancer treatment: Using a Novel ‘Theranostic’ Agent to Enhance the Diagnostic and Therapeutic Properties of Chemotherapy

Oct | 2015
  • Presenter: Dr. Anthony J. Berdis, Cleveland State University
  • Abstract
    According to the American Cancer Society, there will be more than 1.7 million new cases of cancers diagnosed in 2015. More than 50% of these patients will be treated with DNA damaging agents as part of their therapy. Unfortunately, many will not respond favorably to these treatments for several reasons that include ineffective DNA repair and the subsequent misreplication of unrepaired DNA lesions.

    The Berdis lab has taken an innovative approach to combat the latter complication by developing a series of non-natural nucleoside analogs that selectively and potently inhibit the ability of specialized DNA polymerases to replicate certain DNA lesions. Dr. Anthony Berdis will discuss a specific nucleoside analog designated 5-Endosine which shows unprecedented specificity for targeting terminal deoxynucleotidyl transferase (TdT) and pol eta, two specialized DNA polymerases involved in replicating DNA lesions in cancers such as acute lymphoblastic leukemia.

    He will discuss how 5-Endosine can be used as a “theranostic agent” – compounds that possess both therapeutic and diagnostic activities. Combining these properties provides a way to accurately measure the therapeutic activity of the drug in real time and ultimately represents a new area in personalized medicine that may lead to more effective treatments and patient responses to chemotherapy.

Characterizing Cell Morphology Using Imaging Flow Cytometry

Sept | 2015
  • Presenters: Kathleen E. McGrath, Ph.D., University of Rochester, Rochester, NY; and David Archer, Ph.D., Emory University, Atlanta, GA
  • Abstract
    Defining the morphological characteristics—such as the size, shape, or structure—of different cell types has played a key role in assessing the progression and status of various diseases. Imaging flow cytometry combines the visual analysis capabilities of microscopy with flow cytometry to provide researchers with a powerful tool to study cell morphology and the potential effects of therapeutic interventions. In this webinar, we will explore ways to characterize cell morphology and how this information can be applied to better understand blood disorders, including how erythrocyte morphology is used in the analysis of sickle cell disease and to identify functional intermediates in erythropoiesis.

3D cancer tissue engineering and therapeutic response using flow cytometry

July | 2015
  • Presenters: Jennifer Munson, Ph.D., Asst. Prof of Biomedical Engineering, University of Virginia; and Katherine Gillis, Application Scientist, MilliporeSigma
  • Abstract
    In this webcast we look at how to explore the tumor microenvironment in vitro using human-derived 3D models and flow cytometry. Multiparametric flow cytometry has proven to be a valuable tool both for assessing subpopulations in 3D tissue engineered samples that model the tumor microenvironment, and for determining the mechanism and sequence by which anti-cancer compounds/drugs modulate cellular stress, death and survival.

    Dr. Jennifer Munson will explain how her lab at the University of Virginia uses microcapillary flow cytometry to phenotype the cellular constituents of human-derived 3D models of the tumor microenvironment. Dr. Munson will also discuss the critical biophysical attributes including mechanical stiffness and interstitial flow that such models can recapitulate, and will demonstrate how tissue engineering can yield outcomes similar to animal models.

    Then, Katherine Gillis from MilliporeSigma will show data from a powerful multiplexed approach using caspase activation, mitochondrial potential changes, annexin V-based response and cell death markers in a single assay to assess the effects of anticancer therapeutics. She will explain how microcapillary fluidics enable extraction of this data from fewer cells to facilitate drug discovery and mechanism based studies. Such studies provide an enhanced understanding of the mechanism and sequence by which anticancer compounds modulate cellular stress, death and proliferative processes.

    In this webcast you will learn:
    • How biophysical elements in the tumor microenvironment contribute to progression and therapeutic responses.
    • How to explore the tumor microenvironment in vitro using human-derived 3D models and flow cytometry.
    • Methods for determining the mechanism and sequence by which anticancer compounds modulate proliferation, apoptosis, and cell death.

Strategies for Enrichment, Quantitation, and Characterization of Extracellular Vesicles (EVs) for Cancer Research

June | 2015
  • Presenters: Kamala Tyagarajan, Ph.D.; Amedeo Cappione, Ph.D.; and Haley Pugsley, Ph.D.
  • Abstract
    Due to their size and the variable nature of the biofluids in which they are typically suspended, disparate approaches to collection, isolation, and analysis of extracellular vesicles have led to efforts to enhance accuracy and correlation of data. In an effort to establish standards for workflow, sample collection, and data analysis of extracellular vesicles, the International Society for Extracellular Vesicles (ISEV) has published best practices for standardizing and reducing workflow, and eliminating contamination of EV preparations with dead-cell-derived vesicles. These recommendations state the absolute necessity to quantify the proportion of dead cells present in culture prior to preparing samples for enrichment and isolation of EV, necessitating the use of a cell analysis platform capable of returning quantitative data from cell health assays. Workshop participants will learn about the capabilities of the Muse benchtop cell analyzer which couples simple, intuitive software with assays fully optimized for rapid and consistent assessment of cell health parameters including count, viability, apoptotic fraction, autophagy, oxidative stress, DNA damage, and other activation of signaling pathways.

    Critical to understanding the precise functions of extracellular vesicles such as exosomes in tumor progression is reliable isolation of pure fractions. The current gold standard for exosome enrichment is differential ultracentrifugation, which requires specific, costly instrumentation, is lengthy, and is labor intensive. We present a rapid, ultrafiltration-based approach for microvesicle isolation from biological samples, optimizing the process using an infrared spectrometer that permits simultaneous monitoring of protein quantitation and analysis of total lipid content during exosome fractionation.

    Finally, we will show how imaging flow cytometry improves the analysis and characterization of these microvesicles in a quantitative and reproducible manner above and beyond the current methods of analysis.

Sourcing Niche Cell Populations: Techniques for Isolating and Characterizing Progenitor Cells

June | 2015
  • Presenters: Donald G. Phinney, Ph.D., The Scripps Research Institute, Jupiter, FL; and Stefano Da Sacco, Ph.D., Children’s Hospital Los Angeles, Los Angeles, CA
  • Abstract
    Understanding stem/progenitor cells—which can develop into various types of tissue—are key to developing novel approaches for regenerative medicine. Techniques enabling the isolation of specific cell populations have advanced our ability to identify and characterize niche cell types which may be used to replace or engineer cells/tissue. The use of cell surface markers to isolate specific cell populations is one common method for separating cells; however, isolating live cells based on their RNA expression is a powerful new way enabling the study of small cell niches in nongenetically modified animal models and human tissue. In this webinar, we will hear from experts about different ways to isolate and characterize specific cell populations, including the nephrogenic progenitors that give rise to mature functional renal cells and mesenchymal stem cells which can differentiate into multiple cell types.

Simplified Methods for Investigating Deregulation of the Cell Cycle and Apoptosis in Cancer Progression, and the Implications for Anti-Cancer Therapies

June | 2015
  • Presenters: Dr. Kai Stoeber, Shionogi Ltd.; and Dr. Kamala Tyagarajan, MilliporeSigma
  • Abstract
    The complexities of cancer research demand diverse approaches that include studies of the critical events that propel the tumor cell and its progeny into uncontrolled expansion and invasion. This evolution of normal cells into cancer cells is facilitated by loss of fidelity in the processes that replicate, repair and segregate the genome, such as the regulation of cell cycle, apoptosis and cell proliferation. Recent advances in our understanding of these critical events suggest molecular mechanisms for cellular transformation, and may help to identify potential targets for improved cancer therapies. The DNA replication initiation machinery, a component of the cell cycle engine which serves as a relay station connecting signaling networks with DNA synthesis, is a promising target for diagnostic and therapeutic interventions. In this webinar Dr. Kai Stoeber (Shionogi Ltd.) will introduce methods for studying DNA replication initiation factors in human tissues and body fluids and provide an overview of their clinical utility for cancer detection, prognosis and prediction of therapeutic response to cell cycle phase-specific drugs. He will also discuss the emerging concept of targeting the replication initiation machinery for cancer therapy. Dr. Kamala Tyagarajan (MilliporeSigma) will present data from studies performed on a novel flow cytometry platform that quantitates the actions of anti-cancer compounds on apoptosis/cell death and cell cycle arrest in parallel. She will also discuss simplified methods and assays for accurate evaluation of cell cycle distribution and Annexin V-based apoptosis detection, caspase activation and cell death.

Imaging Flow Cytometry for the Detection of Sub-Visible Particles in Therapeutic Protein Formulations

Apr | 2015
  • Presenters: David B. Volkin, Ph.D., Distinguished Professor, Department of Pharmaceutical Chemistry, University of Kansas; and Christine Probst, Application Scientist, Amnis Corporation — Part of MilliporeSigma
  • Abstract
    Development of safe and effective protein therapeutics relies on the accurate assessment of the amount, type, and size distribution of particle impurities within the formulation. Precise characterization of particles between 1–10 ìm is increasingly in demand due to potential immunogenicity or loss of potency. However, observations at this size range have tested the limits of established analytical methods. One particular challenge has been the accurate discrimination of aggregated protein particles from silicone oil droplets, which form during drug storage in siliconized syringes.

    While there are various ways to characterize particles in this size range, imaging flow cytometry offers some unique advantages in convenience of a platform technology. Imaging flow cytometry simultaneously collects fluorescent, bright-field, and side-scatter imagery for each particle in suspension, allowing particles to be identified using specific fluorescent stains and/or morphological properties. In this webinar, you will learn how certain imaging flow cytometry techniques can facilitate detection and classification of sub-10 ìm protein aggregates and silicone oil droplets.

    David B. Volkin, Ph.D., from the School of Pharmacy at the University of Kansas, will discuss the use of micro-flow imaging analysis as a tool to improve the identification of protein aggregates and microparticle detection.

    Christine Probst, M.S., application scientist at Amnis Corporation, part of MilliporeSigma, will provide an overview of imaging flow cytometry technology and will describe the development of a simple staining approach to fluorescently label protein aggregates and silicone oil droplets. Additionally, she will present data evaluating the Amnis® brand ImageStream®X and FlowSight® imaging flow cytometer performance for characterization of these particles, from which it will be demonstrated that the combination of fluorescence staining and imaging flow cytometry analysis greatly improves the specificity and sensitivity of protein aggregate and silicone oil detection.

Understanding Infectious Disease Through Dynamic Live Cell Analysis

Nov | 2014
  • Presenters: Dr. Andrew Ball, Ph.D., R&D Manager MilliporeSigma; and Sergio Rodriguez, Bente Lab, 3rd Year Graduate Student, Department of Microbiology & Immunology, University of Texas
  • Abstract
    The ability to monitor kinetic events in a host immune response is fundamental to furthering our understanding of infectious disease pathways. For example, nairoviruses, such as the dangerous Crimean-Congo Hemorrhagic Fever virus, are often transmitted by tick vectors. Not only do ticks attach to the host skin, but they also produce compounds that subvert host immune response.  Despite decades of research, management of diseases caused by nairoviruses has been difficult, in part because so little is known about interactions of the virus, tick, and host at the initial site of infection. Recent advances in microscopy, software and culture systems have led to a new era of live cell biology, based on quantitative mapping of live cell movement, morphology, and host-pathogen interaction, which can elucidate such mechanisms at the infection site.

A Closer Look at Cell Death with Imaging Flow Cytometry

Nov | 2014
  • Presenters: Irene La-Beck, Pharm.D., Texas Tech University Health Sciences Center; Sabine Pietkiewicz, Ph.D., Otto von Guericke University, Germany; and Haley R. Pugsley, Ph.D., MilliporeSigma
  • Abstract
    Apoptosis, necrosis, and autophagy are all different forms of programmed cell death (PCD) and are essential for regulating homeostasis and eliminating undesirable cells. Distinguishing between different forms of PCD can be challenging because these processes depend upon changes in morphology and subcellular structure that are often masked in studies of heterogeneous cell populations. The use of imaging flow cytometry has been a method to overcome this problem and has led to new discoveries in the field of PCD.

Finding the miRNAs that Matter with Microcapillary Flow Cytometry

Sept | 2014
  • Presenters: Sukhinder Sandhu, Ph.D., Swift BioSciences; and Daniel C. Pregibon, Ph.D., FireFly BioWorks
  • Abstract
    To elucidate the biological roles of microRNAs (miRNAs) and validate their utility as biomarkers, robust methods are needed to profile large sample cohorts. In this webinar, you will learn how guava easyCyte™ microcapillary flow cytometers enable miRNA profiling with exceptional throughput and high signal-to-noise ratios. Specifically, the guava easyCyte™ flow cytometers enable the new SmartRNAplex™ miRNA profiling assays, which can measure up to 68 targets simultaneously in a single well...
    More...

Many Roads to Cell Death: Gaining a Practical Understanding of Apoptosis, Necrosis, and Autophagy

June | 2014
  • Presenters: John Abrams, Ph.D., University of Texas Southwestern Medical Center, Dallas, TX and William G. Telford, Ph.D., National Institutes of Health.Bethesda, MD
  • Abstract
    Cell death is, ironically, an essential part of life. In recent years, the study and understanding of cell death pathways has been dramatically transformed by the insights gained into non-apoptotic pathways, including necro-apoptosis and autophagy, together with a deeper understanding of the mechanism of the apoptotic cascade. New discoveries have been enabled by cutting-edge technologies, particularly in the realm of cytometry and cell-death–specific markers. In this webinar, the latest insights into cell death pathways will be discussed, including the molecular markers and cellular changes that characterize each pathway. Viewers will also learn practical cytometry-based strategies for dissecting cell death pathways, and how to use the data to better understand the pathophysiology of diseases such as cancer as well as to uncover new targets for drug discovery and development.

    During this webinar, the speakers will:
    • Review the latest insights into the different cell death pathways
    • Present their own recent data and research on cell death mechanisms and impacts
    • Describe techniques to detect and dissect cell death pathways
    • Answer your questions live and in real time!

Using imaging flow cytometry to study dendritic cell biology and function

April | 2014
  • Presenters: Juan J. Garcia-Vallejo, MD, Ph.D., VU University Medical Center
    Edith Janssen, Ph.D., Cincinnati Children’s Hospital Medical Center
  • Abstract
    Dendritic cells (DCs) are the most efficient antigen-presenting cells in the immune system. Upon recognition of pathogens, DCs become activated and migrate to lymphoid structures where they present relevant antigens to lymphocytes. The uptake of pathogens and the mechanisms leading to proper processing and presentation of relevant antigens are crucial to an immune response.

    In this webinar, Dr. Juan Garcia-Vallejo and Dr. Edith Janssen discuss how they have used imaging flow cytometry to gain a greater understanding of DC biology and function.

    Before they begin Robert Smith-McCollum of MilliporeSigma will give a brief introduction to imaging flow cytometry.

    Dr. Garcia-Vallejo will then talk about his research into the C-type lectin receptors (CLRs) expressed by DCs and the mechanisms leading to CLR-mediated internalization, intracellular routing and antigen processing. He will describe a series of imaging flow cytometry-based methods he developed to study antigen/receptor internalization as well as tracking of antigens/receptors through different intracellular compartments.

    Finally Dr. Janssen will outline her studies comparing dendritic cell functionality in primary dendritic cells from normal mice and mice susceptible to various autoimmune diseases as well as primary and cultured DCs from healthy human donors. She will discuss how imaging flow cytometry can be used to assess the type and mechanism of endocytosis as well as the trafficking of endocytosed materials in phagosomes.

Advances in controlling cell environment during dynamic live cell imaging

Dec | 2013
  • Presenters: Kurt Thorn, Ph.D. Assistant Professor Department of Biochemistry & Biophysics, UCSF and Andrew Ball, Ph.D. R&D Manager MilliporeSigma
  • Abstract
    To image living cells with a microscope over time, they must stay alive throughout the duration of the experiment. Generally, this requires maintaining the cellular microenvironment within the physiological range. In turn, this means controlling the temperature, humidity, media composition, and atmosphere (CO2 concentration) around your cells so that they survive being imaged. The first speaker, Dr. Kurt Thorn (University of California, San Francisco; UCSF) will discuss a variety of hardware approacheflimes that can be used to provide such environmental control. Following Dr. Thorn’s talk, Dr. Andrew Ball (MilliporeSigma) will review a specific workflow involving microfluidic chambers used with a fluorescent microscope and open source image analysis software to provide insights for live cell dynamic applications such as cancer cell autophagy, host-pathogen interactions, neuron growth, and cell migration.

Exploring granulocyte function with imaging flow cytometry

Oct | 2013
  • Presenters: Brian K. McFarlin, Assistant Professor, University of North Texas and David Basiji, Senior Director of Marketing for Cellular Analysis, MilliporeSigma
  • Abstract
    Advances in camera-based flow cytometry have resulted in the creation of new, lower cost benchtop image-based cytometers such as the Amnis® FlowSight instrument. In this webinar, we will demonstrate how the FlowSight® system’s increased detection sensitivity and imaging capabilities can enhance a traditional flow cytometry assay for granulocyte function. Current flow cytometry based methods of assessing granulocyte function provide minimal meaningful data other than a confirmation of granulocyte activation. In contrast, the FlowSight® system enabled us to gain new insight regarding how various patient treatments affect the time course of change in granulocyte function. Also, by leveraging the unique quantitative imaging upgrade on the FlowSight® instrument, we have been able to collect unique information beyond what is possible with traditional flow cytometry plots.

Synergistic cytotoxicity of pre-transplant drugs used in hematopoietic stem cell transplantation

Oct | 2013
  • Presenters: Ben C. Valdez, Ph.D., Associate Professor, The University of Texas and Mark Santos, R&D Manager, MilliporeSigma
  • Abstract
    Hematopoietic stem cell transplantation (HSCT) is used for treatment of hematologic malignancies. The success of HSCT largely depends on the efficacy of pre-transplant/conditioning therapy. Dr. Ben Valdez’s laboratory aims to design efficacious and safe conditioning regimens by identifying HSCT drugs that provide synergistic cytotoxicity when combined. Most recently, his team reported the synergism of two nucleoside analogs, gemcitabine (Gem) and clofarabine (Clo), in killing multiple myeloma (MM) cell lines and cells derived from MM patients. Drug cytotoxicity was monitored by Annexin V assay using the Muse® Cell Analyzer and synergism was determined by the Chou and Talalay method. The mechanism of cytotoxicity was determined by Western blot analysis, PCR and flow cytometry. The synergistic cytotoxicity of Gem and Clo could be attributed to at least five interrelated mechanisms: (1) Gem-mediated activation/phosphorylation of deoxycytidine kinase, which initially phosphorylates nucleoside analogs, (2) inhibition of DNA synthesis and repair, (3) nucleolar stress through inhibition of rRNA production, (4) decrease in mitochondrial membrane potential, and (5) induction of apoptosis. The preclinical results provide a rationale for clinical trials incorporating [Gem+Clo] combinations as part of conditioning therapy for high-risk patients with MM undergoing HSCT. Dr. Valdez will be covering the key aspects of his study during the first part of the talk. Following Dr. Valdez’s talk, Mark Santos will provide a brief overview of the underlying technology and capabilities of the Muse® Cell Analyzer.

Expanding cell analysis in cancer research with imaging flow cytometry

April | 2013
  • Presenters: Andrew Filby, Ph.D., Deputy Head of Facility FACS Laboratory, London Research Institute, Cancer Research UK
    John Hazin, Doctoral Candidate, Division of Translational Immunology, German Cancer Research Center (DKFZ) 
    David Basiji, Ph.D., Sr. Director, Cellular Analysis, MilliporeSigma
  • Abstract
    Imaging flow cytometry combines the speed, sensitivity, and phenotyping capabilities of flow cytometry with the detailed imagery and functional insights of microscopy. This unique combination has enabled a broad range of applications for cancer cell analysis that would be impossible using either technique alone. 

    In this webinar, Dr. Andrew Filby of The London Research Institute will describe the recent development of a novel method for reporting the cell cycle position and division history of asynchronously dividing cells. This method allowed detailed analysis of cell cycle inhibitor function down to the mitotic stage across multiple division rounds. 

    John Hazin of the German Cancer Research Center will describe how imaging flow cytometry expanded understanding of cell cycle and cell adhesion molecules in cancer biology in his laboratory. Hazin and his colleagues identified a subset of cells in which the transmembrane cell-adhesion molecule L1CAM (L1) forms polarized membrane domains. They further investigated the co-localization of other proteins with L1, as well as nuclear translocation of an intracellular L1 fragment believed to participate in signaling functions. 

    These examples demonstrate the practical use and the power of imaging flow cytometry. 

    What You Will Learn in This Webinar:
    • How imaging flow cytometry facilitates analysis of highly heterogeneous samples and rare cell sub-populations
    • How imaging flow cytometry allows simultaneous phenotypic and functional studies
    • How imaging flow cytometry advances research of trans-membrane cell-adhesion molecules and analyzes their functionality in signaling complexes


    Who Should Attend:
    • Cell biologists
    • Cellular immunologists
    • Cancer cell biologists
    • Cancer drug developers
    • Membrane protein biochemists
    • Cell signaling scientists
    • Stem cell biologists


    A live Q&A session will follow the presentations, offering you a chance to pose questions to our expert panelists.