Fluidigm
  • Products
      • Applications
      • Mass Cytometry
      • Imaging Mass Cytometry
      • Genomics and Gene Expression
      • Single-Cell Genomics
      • Research Areas
      • Immune Profiling
      • Immuno-Oncology
      • Ag-Genomics
      • Cancer Research
      • System Solutions
      • Instruments
      • Maxpar Antibodies and Assays
      • Advanta Assays and Kits
      • Microfluidic IFCs
      • Software
      • Catalog
  • Stories
    • See all Stories >
    • The Influence of Inflammation on Mental HealthRead More

    • Machine learning and infectious disease: how we can better understand vaccine efficacy at the patient levelRead More

    • The intestinal stem cell response Investigating transcriptomic changes at the microscale levelRead More

    • DNA Fingerprinting at a Genetics BiorepositoryRead More

  • Resources
      • Learn
      • Fluidigm Technologies
      • Customer Stories
      • Publications
      • Webinars
      • Tools
      • Maxpar Panel Designer
      • D3 Assay Design
      • C1 Open App
      • Script Hub
  • Support
      • By Instrument
      • Access Array
      • Biomark HD
      • C1
      • EP1
      • Flow Conductor
      • Helios and CyTOF
      • Hyperion Imaging System
      • Juno
      • Polaris
      • Additional Support
      • Training
      • Service Plans
      • Contact Us
  • About Us
      • Our Story
      • Careers
      • Events
      • Investors
      • Legal
      • Press
SIGN IN
Forgot password? Register

Sign in to your account to access order status, past orders and discussions.

If you don't have an account, please sign up and start one today!

  • Reset password

    Enter your e-mail address and we'll email you a reset link.

    Back
ORDER
  • Breast cancer disparities and tumor biology

    Defining spatial characteristics using Imaging Mass Cytometry

    Spotlight

    How Imaging Mass Cytometry transformed one lab’s discovery of the intrinsic behavior of a tumor

    Download spotlight

    Breast cancer mortality is more than 40% higher for African American women in the US than Caucasian women. Yet, the cancer’s incidence across race is very similar.

    Melissa Davis, PhD, is Scientific Director of the International Center for the Study of Breast Cancer Subtypes (ICSBCS) and Assistant Professor of Cell and Developmental Biology at Weill Cornell Medical College. She is working to uncover the various components that contribute to cancer mortality disparities between races.

    While multiple factors play a role in cancer survival, tumor characteristics are believed to account for almost a quarter of racial differences. Early research on variations in tumor biology among breast cancer patients of different races labeled hormone receptor status as a differentiating factor. To take this further, Davis explains, “The triple-negative subtype in breast cancer lacks expression of hormone receptors that are the basis of common targeted therapies and has the highest rate in non-Hispanic black patients. It is important that we identify what molecular markers underly this subtype beyond hormone receptor status or genomic profiling, which can be impractical in a clinical setting.”

    A look into African history explains much about the origins of the triple-negative subtype and what other related phenotypes might be present in women with this diagnosis. Davis focuses on the African diaspora, commonly used to describe the mass dispersion of African people during the transatlantic slave trade, recruiting patients for sample collection from Africa, the Caribbean islands and North America.

    Africa has the lowest incidence of breast cancer in the world, yet the highest mortality rate due to the prevalence of triple-negative diagnoses. This component of African ancestry spreads to black Americans in the US and black South Africans, with similar trends in West Africa, all coinciding with the transatlantic slave trade across the Americas and the Caribbean.

    “The triple-negative subtype in breast cancer lacks expression of hormone receptors that are the basis of common targeted therapies and has the highest rate in non-Hispanic black patients. It is important that we identify what molecular markers underly this subtype beyond hormone receptor status or genomic profiling, which can be impractical in a clinical setting.”
    Watch Melissa Davis’ webinar

    Exploring the DARC phenotype

    Davis and her lab aim to find molecular markers and functional differences that might explain the frequency of the triple-negative subtype in these populations. They are currently focused on one particular allele, the Duffy-null allele of the Duffy antigen receptor for chemokines (DARC) gene, that arose in sub-Saharan Africa and plays a role in immune response regulation. It was fixed into this population because it confers resistance to malaria.

    Its global distribution is vast, with over 70% of Africans carrying the allele in America alone, and tracks with movement along the transatlantic slave trade. Duffy-null carriers are also erythrocytic-null, expressing the gene only in endothelial cells. This is an important feature because it results in recruitment of immune cells into inflamed areas through the circulatory system. In a tumor, this would influence the microenvironment in an organ site, such as in breast cancer.

    Recently, the Davis lab discovered that tumors expressing this gene also exhibit higher levels of inflammatory chemokines and infiltrating immune cells. Owing to this chemokine recruitment, a high level of DARC expression in a tumor is associated with significant survival benefits in all subtypes.

    Imaging DARC with immunohistochemistry

    In the effort to uncover how the DARC gene phenotype bestows a survival advantage, a look at gene signatures correlated with DARC status identified a gene subset that tended to be more highly expressed in DARC-positive tumors. Davis used these as a template to learn more at the single-cell level with traditional immunohistochemistry (IHC).

    The goal was to determine whether these markers and the patterns of these markers correlate synchronously across the tumor space by compartmentalizing features and quantifying cell types. However, IHC quickly became its own challenge, with the inability to multiplex marker staining on single sections. The initial work was tedious but yielded results after months of simple cell identification. The team saw a higher rate of infiltrating CD3+ cells. Davis focused in on 20-plus markers, but with IHC, this would become exponentially more difficult. The solution was Imaging Mass Cytometry™ (IMC™).

    “If we were going to describe and characterize a tumor phenotype based on heterogeneity, we needed a better platform. We turned to IMC with the Hyperion™ Imaging System at the Englander Institute for Precision Medicine to define spatial distinctions characterizing the DARC immune tumor type,” said Davis. “We were able to multiplex up to 30 markers as our target number of conjugated antibodies on a single slide and acquire this data in a way where we could quantify the markers and do computational analyses.”

    Moving forward with IMC

    A first step was careful consideration of the regions of interest (ROIs). With a need to compare populations, it was important to collaborate with a pathologist who could demarcate areas of interest to capture the stroma as well as the solid tumor space and to ensure that the captured area was comparable across different patient populations.

    IMC comparison of the tumor types showed that in the DARC-negative tumors, immune cell markers isolated to the stromal area with less infiltration into solid tumor space. Conversely, images of DARC-positive tumors showed more infiltration into the solid tumor space and increased association with endothelial cells and vascular structure. Differences in tumor architecture suggest that infiltration occurs because DARC-positive tumors are more vascularized. These findings blend nicely with prior bulk-tumor RNA-seq data indicating that the spatial distribution of immune cells is variable between tumors.

    “These images provided a view of the distribution of markers and relationships of cell populations across DARC-positive and DARC-negative tumors. Quantifying behavior at a single-cell level with IMC offers more information about what is happening across the architecture and connects these findings to clinical implications in breast cancer,” clarifies Davis.

    IMC made it easy to identify subpopulations of cells present only in a DARC-high vs. a DARC-low environment by combining all ROIs to view non-overlapping subsets. Davis found one subset of cells in DARC-positive tumors that co-expressed immune cell and mesenchymal markers, mapping these vimentin-positive immune cells as infiltrating into the solid tumor area and indicating a distinct DARC-regulated tumor microenvironment. Further study will include functional characterization of these subpopulations using models of DARC tumors in mice and ex vivo patient-derived organoids.

    Initial clinical implications of these phenotypes for survival advantage are promising. Next steps in this project will be to correlate DARC phenotypes in tissues with clinical outcome and clinical annotation and to stratify populations by subtype.

    Imaging Mass Cytometry empowered Davis not only to discover what cell populations are present in these tumors and where they are located but, simultaneously, also to detail the intricate behavior and interactions occurring among cells and how these actions affect clinical outcome in triple-negative breast cancer patients.

    “Why is this important in the context of disparities? We know from research on human evolution that immune cells originating from people of African descent compared to European descent have completely different responses to the same bacterial pathogen. This indicates that evolutionary consequences or natural events, like the types of pathogens present in Europe or in Africa, have elicited unique immune responses,” Davis concludes.

    “And we can apply this to other diseases, such as cancer, that are highly reliant on an immune response. Consider implications on therapeutics, and whether tumor cell subtypes could identify subpopulations in which immunotherapies are effective and predict treatment response based on these patterns.”

    Important links:

    • Watch Melissa Davis’ webinar on this topic
    • See how Imaging Mass Cytometry supports cancer research

    Complete the form below to request more information about Imaging Mass Cytometry.

    For Research Use Only. Not for use in diagnostic procedures.

    • Related Applications

      • Imaging Mass Cytometry
    • Related Products

      • Hyperion Imaging System
  • Research links cell signaling, location and patient prognosis in brain tumors

    How CyTOF technology enabled a look into the heterogeneity of glioblastoma

    Spotlight

    The Ihrie Lab at Vanderbilt develops new approach to associate cell type to overall survival

    Download spotlight

    Sometimes the smallest things can help you see the bigger picture. That’s the idea behind one professor’s research into brain cancer. By revealing the abnormal cells that are most aggressive within brain tumors, we can find answers about which cell features may lead to which clinical outcomes.

    Rebecca Ihrie, PhD, Associate Professor of Cell and Developmental Biology at Vanderbilt University, recently talked with us about glioblastoma, one of the most common and malignant forms of primary brain tumor.

    Although glioblastoma has been a heavy focus of study for many years, prognosis has not improved much in recent decades. Current standard treatments center on the alkylating agent temozolomide in addition to radiotherapy. While use of this agent extends survival to some extent, overall prognosis and survival outcomes remain poor.

    Mass cytometry is a primary research approach used in the Ihrie Lab, being particularly informative for glioblastoma investigation. “When we think about everything going on within a cell, DNA and RNA profiling, which have been widely used in brain tumors and many other tissues, are certainly very informative. But when we think about the functional repertoire of a cell, this information is incomplete,” explains Ihrie. “Mass cytometry allows us to complement what we have learned at the DNA and RNA level to better understand beyond copy number analysis, sequence analysis and transcript analysis how cells are dynamically responding to their neighbors and what their signaling capability at the protein level is.”

    The complexity of glioblastoma is evident within tumors, which can contain multiple cancer lineage cells resembling different stages of neural stem cell differentiation; neurons, astrocytes, and glia (the resident cells of the brain); various immune cells; and endothelial cells. Three major subtypes of glioblastoma have been classified, distinguished primarily by alterations in growth factor receptor gene copy number as well as transcriptional signatures that resemble different progenitor cells within the brain.

    Ihrie is interested in understanding these heterogeneous mixtures of cells and how they vary across patients. She discusses two recent questions her lab and collaborators are currently focused on: How can we identify highly malignant phenotypes across glioblastoma, finding the most aggressive cells that drive recurrence but that might be rare and variable across patients? And how can we do discovery work in smaller cohorts of patients? “We really wanted to leverage all of the information generated from approaches like mass cytometry, where we can obtain millions of cells and lots of data on specific parameters to figure out which of these parameters can help us find the cells that are driving the worst outcomes and then identify their targetable features,” Ihrie says.

    “Mass cytometry allows us to complement what we have learned at the DNA and RNA level to better understand … how cells are dynamically responding to their neighbors and what their signaling capability at the protein level is,” says Ihrie.
    Watch Rebecca Ihrie’s webinar

    Stratifying patients with RAPID

    Risk Assessment Population IDentification (RAPID), an unsupervised and automated machine learning algorithm, identifies phenotypically distinct cell populations and determines whether these populations stratify patient survival. The algorithm couples the per-cell data obtained using mass cytometry to continuous features like progression-free survival, measured in days or weeks, and overall survival, which in glioblastoma has a median of about 15 months.

    Initially, the team analyzed over 2 million glioblastoma cells from 28 patients by mass cytometry, using a panel of 40 different parameters to find immune, endothelial and other infiltrating cells within brain tumors.

    The team assigned all cells to clusters based on phenotype and determined which clusters were meaningfully associated with a better or worse outcome. RAPID analysis identified both glioblastoma-negative prognostic (GNP) cells, whose abundance is predictive of especially bad outcome, and glioblastoma-positive prognostic (GPP) cells, whose abundance indicates a long overall survival. She notes that since these survival differences had not been discovered through other data methods at the genomic DNA or RNA level, “seeing something that stratified survival was very exciting, because it gives us an opportunity to better understand the biology of these tumors and think about how to target them.”

    The in-depth data gleaned from their mass cytometry experiments revealed distinct and mixed phenotypes with opposing expression patterns that further distinguished the two prognostic groups. “Access to the high-dimensional data from CyTOF® was fantastic for us to be able to do additional ongoing discovery work in these tumors and to understand these phenotypes,” adds Ihrie. “But we want to be able to translate this to approaches that neuropathologists typically use.”

    Translating CyTOF findings to neuropathology

    Ihrie wondered if brain tumor location could affect cancer cells and immune cells within the tumor. The goal was to apply the phenotypes discovered from suspension mass cytometry and pinpoint their location within individual tumors or across tumor classifications.

    Based on MRI images, the team asked whether glioblastoma tumors contacting specific structures within the brain have better or worse outcomes. At initial diagnosis, about 50% of patients had a tumor that contacts the lateral ventricles and the stem cell niche that resides there. These patients had poorer overall survival. A smaller subset had a tumor that contacts the other major stem cell niche of the brain, the dentate gyrus—but surprisingly, contact with this niche did not correlate with survival differences.

    Overlaying their originally determined cell phenotypes with tumor location classification, they found a cluster of cells that is enriched in noncontacting tumors with protein expression levels very closely resembling the glioma-positive prognostic phenotype.

    Other known predictors of outcome, such as the size of the tumor, the ability of the surgeon to resect it and bulk DNA and transcriptomic profiles, did not explain the effects of ventricular contact and raised novel questions about the biology behind these differences in outcome. Moving forward, Ihrie is interested in coupling MRI images, a routine visualization for glioblastoma patients, to this type of information to help decide on treatment options.

    Connecting the dots on the influence of tumor location on potential tumor prognosis

    One challenge with traditional imaging approaches is the inability to view and analyze all parameters simultaneously. “Wouldn't it be nice if we could measure everything together? That's something we are working on now, together with the Bursky Center for Human Immunology & Immunotherapy at Washington University in St. Louis using Imaging Mass Cytometry™,” Ihrie says. “We are eager to take all the parameters we used on dissociated tumors in suspension and apply them across the tissue to get more information, including spatial data for better classification of patients.”

    Ihrie next used Imaging Mass Cytometry to identify and associate cell-to-cell variation and distribution within different tumors to phenotypes. The team developed and validated a brain-centered imaging panel that included lineage-specific transcription factors, phosphoproteins and other cancer-centric markers that overlap with the panel used for earlier RAPID studies.

    In glioblastoma samples, images revealed a nonrandom distribution of signaling, with separate clusters of cells signaling together. Further study could isolate these signaling groups to decipher their role in the tumor and whether distance from the stem cell niche might come into play.

    Particularly in glioblastoma, where transcriptomic and other genomic analyses have not readily allowed stratification of patients into different groups based on outcome, mass cytometry measurements at the protein level that include phosphorylation events have helped classify prognostic subgroups. The custom workflow, RAPID, is now published, and it generates simple stratification assays that use high-dimensional data to compare findings in fixed tissue and suspension.

    “Ultimately, we are very excited to find that stem cell niche contact changes the cancer cell and immune cell features and is linked to some of the differences in prognosis that we found before. And importantly, these phenotypes are not uniformly distributed through the tissue. The ability to go back and forth between what we can discover in suspension and in fixed tissue has been an incredibly useful tool for us,” concludes Ihrie.

    Important links:

    • Rebecca Ihrie’s webinar on this topic: Linking cellular signaling, location and patient prognosis in brain tumors

    References:

    • Leelatian, N., Sinnaeve, J., Mistry, A.M. et al. “Unsupervised machine learning reveals risk stratifying glioblastoma tumor cells.” eLife 9 (2020): e56879.
    • Mistry, A.M., Hale, A.T., Chambless, L.B. et al. “Influence of glioblastoma contact with the lateral ventricle on survival: a meta-analysis.” Journal of Neuro-oncology 131 (2017): 125–133.
    • Hakobyan, S. et al. “Complement biomarkers as predictors of disease progression in Alzheimer’s disease.” Journal of Alzheimer's Disease 54 (2016): 707–716
    • Mistry, A.M., Wooten, D.J., Davis, L.T. et al. “Ventricular-subventricular zone contact by glioblastoma is not associated with molecular signatures in bulk tumor data.” Scientific Reports 9 (2019): 1842.

    Complete the form below to request more information about mass cytometry or Imaging Mass Cytometry.

    For Research Use Only. Not for use in diagnostic procedures.

    • Related Applications

      • Mass Cytometry
  • Moving forward with mass cytometry in the midst of the pandemic

    A quick pivot to COVID-19 research with CyTOF

    Spotlight

    Mass cytometry assists research on role of immune system in disorders: A COVID-19 update

    Download spotlight

    Lori Turner, PhD, a post-doc in the NIHR Cambridge BRC Cell Phenotyping Hub and the Department of Psychiatry at the University of Cambridge, had been using the Maxpar® Direct™ Immune Profiling Assay™ in a potentially groundbreaking search for treatments for depression that act through the immune system.

    But with the onset of the COVID-19 pandemic, the multi-site Antidepressant Trial With P2X7 Antagonist JNJ-54175446 (ATP), the first clinical study by the Wellcome Trust Consortium, came to a halt in March 2020. While the team waited for the ATP trial to restart, its members decided to repurpose their entire workflow to do a COVID-19 study. “We had everything sitting there ready to go. We thought, this is a perfect opportunity to immunophenotype COVID-19,” says Turner.

    The team got back to work using that same workflow staining, fixing and freezing COVID-19 samples and then thawing them later for analysis. The ability to work with frozen samples was a defining factor of the new study, since more samples were coming in than could be analyzed fresh. The team also purified white blood cells and performed immunophenotyping on them.

    The study gave Turner a unique opportunity to compare flow versus mass cytometry data for quite a large number of patients, since their new analysis used both. She found that across the various types of immune cells—naive B cells, memory B cells, gamma delta T cells, MAIT cell, and CD4 naive central memory cells, to name a few—there was “a remarkable correlation” in the data. “I was really impressed to see that,” says Turner.

    Moving forward, Turner is considering using the workflow in other studies as well, especially as things have changed with the pandemic. “We are not recalling patients into the hospital, and so we’re planning lots of multi-site studies in the future,” she notes. By validating the workflow for general use, it will become very easy for the lab to adopt quickly. “You add the whole blood to the [Maxpar Direct Assay] tube, stain it for 30 minutes, add a stabilizer for 10 minutes, and then stick it at –80 °C. Or stick it on dry ice and ship it to a central site where samples can be analyzed.” With several trials using the workflow in planning stages, Turner thinks it’s going to make a lot of their work much simpler going forward.

    Listen to Lori Turner’s story

    The Influence of Inflammation on Mental Health

    How a new clinical trial using CyTOF® could change treatment options for depression

    As an immunologist with a background in host-pathogen interactions, Turner explores immune system function and dysregulation in various psychiatric disorders.

    With depression . . . we see a spectrum of symptoms. We believe that different symptoms correlate with numbers of certain immune cells and changes occurring in these cells.”

    Before ATP was put on hold, Turner was coordinating the biomarker studies in the clinical trial as part of the Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer’s Disease (NIMA). Rather than target the nervous system, as do many current medicines for depression and neurodegeneration, the NIMA consortium focused on the immune system. Several studies have already linked the immune system to psychiatric and neurodegenerative disorders, revealing increased levels of inflammation in patients with depression1,2 and Alzheimer’s disease3. By bringing together academia and pharmaceutical companies, NIMA investigates whether targeting the immune system could become an effective alternative treatment.

    Immune cell biomarkers in depression

    Certain immune cell subsets are increased in depression while others are decreased, and these changes relate to the level of severity of specific symptoms. Turner focuses on whether new treatments could influence these changes and subsequently affect symptoms. “With depression, some people feel anxiety or fatigue for example, and some people don’t—we see a spectrum of symptoms. We believe that different symptoms correlate with numbers of certain immune cells and changes occurring in these cells. This is what we’ve set out to identify,” explains Turner.

    In the Phase 2 clinical trial, the research team intends to test an anti-inflammatory that crosses the blood-brain barrier to block activity of the P2X7 receptor, present on various immune cells and linked to stress-related depression. The group will perform immune profiling by mass cytometry to monitor the immune system before, during and after treatment. With any observed response, further study could identify immune biomarkers related to that response. Patients will also complete additional blood tests, questionnaires and magnetic resonance imaging brain scans throughout the trial to more comprehensively understand anti-inflammatory effects on the immune system and the brain.

    Neuro-immunophenotyping with CyTOF

    Turner came into mass cytometry through her work with the flow cytometry core facility at Cambridge. While her early research used flow cytometry, her more recent work at the Cambridge Biomedical Research Centre (BRC), part of the National Institute for Health Research (NIHR), requires larger panels that can only be analyzed by mass cytometry. In the first phases of the study, Turner directed preclinical work to assess depressed participants and healthy controls by mass cytometry, using the Fluidigm customizable human immune monitoring kit. Initial data focused on the P2X7 receptor and potential associated biomarkers.

    “Since standardization is extremely important for a multi-site clinical trial, we needed a pre-validated, as-is kit that included the markers we were focused on and that could be used across multiple sites …”

    For the clinical trial, the team moved to the Maxpar Direct Immune Profiling Assay. “Since standardization is extremely important for a multi-site clinical trial, we needed a pre-validated, as-is kit that included the markers we were focused on and that could be used across multiple sites since the trial will be carried out across five centers in the UK,” says Turner. “The idea of being able to add a whole blood sample directly to a pre-prepared assay tube, reducing site-to-site variation, was a huge factor for us. And the inclusion of Maxpar Pathsetter™ software allows us to quickly generate a report that goes into our online database, which is then shared with the clinical team for complete quality control.”

    As the trial moves forward, the group is optimistic about the opportunity to identify related biomarkers that indicate response to the treatment. Good results could then initiate an expanded trial with more and different types of participants. Turner notes that the Maxpar Direct Immune Profiling Assay and Maxpar Pathsetter software can evolve with the study, allowing addition of new markers to investigate other aspects of immune cell subsets. For a list of this and other clinical trials employing mass cytometry, go to clinicaltrials.gov.

    Learn more about the award-winning Maxpar Direct Immune Profiling Assay and Maxpar Pathsetter software.
    Learn more

    Useful links:

    • Read a multi-site study publications:
      • Bagwell, C.B. et al. “Multi-site reproducibility of a human immunophenotyping assay in whole blood and peripheral blood mononuclear cells preparations using CyTOF technology coupled with Maxpar Pathsetter, an automated data analysis system.” Cytometry Part B Clinical Cytometry 98 (2019): 146–160
      • Geanon, D. et al. “A streamlined CyTOF workflow to facilitate standardized multi-site immune profiling of COVID-19 patients.” medRxiv (2020): doi. org/10.1101/2020.06.26.20141341
    • View results and download white paper:
      • Deep Immune Profiling with the Maxpar Direct Immune Profiling System
    • Download poster:
      • The Maxpar Direct Immune Profiling Assay and Maxpar Pathsetter Analysis

    References:

    • Chamberlain, S. et al. “Treatment-resistant depression and peripheral C-reactive protein.” British Journal of Psychiatry 214 (2019): 11–19. doi:10.1192/bjp.2018.66
    • Lynall, M. et al. “Peripheral blood cell immunophenotyping reveals distinct subgroups of inflamed depression.” bioRxiv (2019): doi: 10.1101/706309
    • Hakobyan, S. et al. “Complement biomarkers as predictors of disease progression in Alzheimer’s disease.” Journal of Alzheimer's Disease 54 (2016): 707–716

    Complete the form below to request more information about the Maxpar Direct Immune Profiling System or mass cytometry.

    For Research Use Only. Not for use in diagnostic procedures.

    • Related Applications

      • Mass Cytometry
    • Related Products

      • Helios, a CyTOF System
  • Battling pathogens, from HIV to SARS-CoV-2

    UCSF researcher focuses on immune system responses

    Spotlight

    The critical role CyTOF plays in helping Nadia Roan understand viral persistence and clearance mechanisms

    Download Spotlight

    Although treatments for HIV infections keep the disease at bay, neither an effective vaccine nor a cure for HIV currently exists. With a long-standing interest in studying the interactions between immune cells and microbial pathogens, Nadia Roan, PhD, is working to better understand how HIV establishes initial infection and how it can persist in the face of antiretroviral therapy.

    Understanding host-pathogen interactions centers on immune responses. In the case of HIV, key cells that orchestrate immune responses are the same ones that are attacked by the virus. These cells, CD4+ T cells, are the main targets of HIV infection. Which of the many different CD4+ T cells are the most susceptible to infection, and the first to become infected? How does HIV manipulate, modify or remodel infected CD4+ T cells to quickly spread, and survive in the form of a long-lived reservoir in the presence of antiretroviral drugs? Answers to these questions not only could help prevent the spread of HIV but could also further our understanding of the role CD4+ T cells play in immune homeostasis.

    Roan, an Associate Professor at the University of California, San Francisco, and a scientist at Gladstone Institutes, focuses her research on the mechanisms of HIV transmission and the possibilities of a cure. The research lab headed by Roan uses various ex vivo models of HIV infection. By isolating immune cells from blood and various tissues of healthy individuals and exposing the cells to a reporter HIV virus, the lab can characterize cell types, activation status, infection susceptibility and more.

    A recent paper from Roan’s lab published in eLife depicts how a newly infected cell changes its regulation and signaling following HIV infection. Using an ex vivo genital infection model and CyTOF® to model HIV transmission through the female reproductive tract, the group found down-regulation of receptors that could disrupt proper signaling and up-regulation of survival factors to help infected cells live long enough to spread the virus to other cells. They further used bioinformatics approaches to predict the original states of infected cells before viral-induced remodeling to define the CD4+ T cells most susceptible to HIV.

    Supported by CyTOF

    In much of her early work Roan used flow cytometry to study immune cell features and responses, but she recently shifted to mass cytometry to more easily characterize and directly compare immune cell subsets. “One common use of mass cytometry is to phenotype all the major immune subsets in a single sample,” Roan explains, “without the need to split the sample to be stained by multiple smaller phenotyping panels. And while we do use mass cytometry for that, more often we take advantage of the technology to really dive deeply into one particular cellular subset, such as CD4+ T cells.”

    Along these lines, the lab has designed multiple T and NK cell panels, as well as panels focused on characterizing expression levels of viral intracellular sensors and restriction factors. One use of a T cell panel designed by the Roan Lab was to specifically study HIV latency during antiretroviral therapy, in particular to characterize the phenotypes of the in vivo HIV reservoir. By understanding the phenotypes of cells latently infected with HIV and the mechanisms that allow the persistence of these cells, Roan hopes that novel approaches can be designed to target these cells as a strategy to cure HIV.

    “The high-dimensional nature of CyTOF datasets allows various pseudotime analytical approaches, which can be used to predict cell states prior to cellular changes, including that caused by viral reactivation.”

    HIV persists in infected individuals on antiretroviral therapy due to a small reservoir of long-lived CD4+ T cells, and potentially other immune cells, containing quiescent HIV. Even though HIV researchers have known for quite some time about the existence of this reservoir, no biomarkers that can specifically detect this cell population have been identified. Combining CyTOF-based phenotyping of infected cells and predicted precursor cells as determined by single-cell linkage using distance estimation (PP-SLIDE), Roan could trace back the original states of latently infected cells. This was accomplished by establishing an atlas of different CD4+ cell types and then using bioinformatics approaches to identify the original phenotypes of the latent cells induced to come out of latency by reactivation. This approach of charting the in vivo latent reservoir was recently accepted for publication in eLife.

    “The high-dimensional nature of CyTOF datasets allows various pseudotime analytical approaches, which can be used to predict cell states prior to cellular changes, including that caused by viral reactivation,” Roan says. “Another advantage of CyTOF is the ability to work with fixed cells. This becomes very important in our research, where we collect longitudinal human specimens and then want to analyze them all at once to limit batch effects.”

    Applying host-pathogen knowledge to SARS-CoV-2

    Since Roan and her lab members already had in-depth knowledge about T cell behavior and response to viral infection, early reports of T cell depletion in severe COVID-19 cases intrigued them. “We already know T cells to be very important for viral infections, and in the case of severe COVID-19, these were the ones that were markedly depleted.”

    Given the development of a wide variety of T cell-specific panels from their HIV research, the group was already primed to adapt to studying the functional features of T cells specific to SARS-CoV-2. In addition, the team modified an intracellular cytokine stimulation assay they built for identifying cytomegalovirus-specific and HIV-specific T cells, whereby cells responding to peptide pools can be identified and deep-phenotyped by CyTOF.

    “A better understanding of mild and asymptomatic cases opens the possibility of vaccines or treatments that promote less severe symptoms after contracting SARS-CoV-2.”

    Collaborating with UCSF colleague Dr. Sulggi Lee, MD, PhD, who established a patient cohort, COVID-19 Host Immune Response Tracking Genesis Report (CHIRP), Roan has been able to examine the SARS-CoV-2 specific immune response that occurs in individuals who recover from mild or asymptomatic COVID-19. A better understanding of mild and asymptomatic cases opens the possibility of vaccines or treatments that promote less severe symptoms after contracting SARS-CoV-2.

    In a paper recently published in Cell Reports Medicine, Roan and colleagues performed longitudinal analyses on a group of convalesced individuals who had recovered from mild SARS-CoV-2 infection and were never hospitalized. They used CyTOF and developed a tailored T cell panel to characterize SARS-CoV-2 specific CD4+ and CD8+ T cells and to measure their longevity. The team tested patients up to 69 days post-infection, when they were still able to see a clear population of SARS-CoV-2 specific T cells. Those cells were capable of expanding in response to IL-7, suggesting their ability to homeostatically proliferate.

    Ongoing studies by the Roan Lab will compare these mild and asymptomatic cases to more severe cases of hospitalized individuals to define effective vs. ineffective or pathological immune responses against the virus. Longitudinal analyses of these patient cohorts will be an important aspect of the studies to better understand what responses associate with full and rapid recovery. Roan can take advantage of CyTOF to generate these data and directly correlate responses across individuals showing various degrees of symptoms.

    With the pandemic still ongoing, the hope is that SARS-CoV-2 will ultimately be easier to manage than something like HIV. “Traditional approaches that are being implemented for SARS-CoV-2 should work in some capacity, even if it’s not lifelong immunity. And the more we understand what we’re dealing with using CyTOF and other tools to get as much information as possible, the closer we can get to improved disease management and control,” adds Roan.

    Want to Learn More?

    Submit the form below to have a representative contact you with more information about Helios™, a CyTOF system or Maxpar® antibody panels for use on mass cytometry systems.

    For Research Use Only. Not for use in diagnostic procedures.



    References:

    • Neidleman, J., Luo, X., Frouard, J. et al. “Phenotypic analysis of the unstimulated in vivo HIV CD4 T cell reservoir.” eLife (2020): Accepted.
    • Neidleman, J., Luo, X., Frouard, J. et al. “SARS-CoV-2-specific T cells exhibit phenotypic features of robust helper function, lack of terminal differentiation, and high proliferative potential.” Cell Reports Medicine (2020): 100081.
    • Ma, T., Luo, X., George, A.F. et al. “HIV efficiently infects T cells from the endometrium and remodels them to promote systemic viral spread. 2020.” eLife 9 (2020): e55487.
    • Cavrois, M., Banerjee, T., Mukherjee, G. et al. “Mass cytometric analysis of HIV entry, replication, and remodeling in tissue CD4+ T cells.” Cell Reports 20 (2017): 984–998. 10.1016/j.celrep.2017.06.087
    • Related Applications

      • Mass Cytometry
    • Tags

      • HIV
      • Nadia Roan
      • SARS-CoV-2
      • host-pathogen interactions
      • mechanisms of HIV transmission
  • Enabling better insight

    High-dimensional single-cell analysis enhances precision medicine

    Spotlight

    How a dual-mode technology enables better insight

    Download Spotlight

    Xianting Ding, PhD, is a professor in the School of Biomedical Engineering and deputy director of the Institute for Personalized Medicine at Shanghai Jiao Tong University. The Ding lab focuses on two main objectives: detecting disease-correlated biomarkers to enable early diagnoses and manipulating these targets to develop effective and personalized combinatorial drug therapies. “We believe that the exploration of immunity, metabolism and cell activity in clinical specimens is important for optimizing treatment response and improving patients’ survival time,” he says.

    Biological systems are made up of complex layers of molecules interacting to generate a healthy homeostasis with the ability to respond to infiltrating disease. A dysfunctional system can cause aberrant expression and other signaling errors. Ding points out that research on SARS-CoV-2, for example, shows evidence that immune system abnormalities are highly correlated with disease progression. Likewise, he and his team aim to correlate abnormal immune profiles with clinical phenomics of other diseases by applying high-dimensional single-cell analysis.

    "Compared to other high-multiplex technologies, IMC excels at providing both spatial information and high-multiplexity."

    Given that available samples, typically blood or tissue, tend to be small or acquired sparingly, Ding must apply various techniques to comprehensively gather enough data to formulate solutions. The group uses a combination of CyTOF® in mass cytometry and Imaging Mass Cytometry™ (IMC™) and fluorescence-activated cell sorting (FACS), Orbitrap™ for mass spectrometry and liquid chromatography-mass spectrometry (LC-MS) to study the different aspects of a disease.

    The lab recently published work on colon cancer, where IMC allowed them to map more complex characteristics and related immune cell networks. These types of studies require labeling more markers than allowed by conventional techniques. With IMC, the team can increase its marker count to meet this demand using as many as 37 markers in one panel.

    “IMC helps us understand the cell population distribution in a diseased area in contrast to a non-diseased area,” he explains. “Compared to other high-multiplex technologies, IMC excels at providing both spatial information and high multiplexity. It allows us to see spatial context more intuitively and enables accurate detection, treatment and prognosis of the disease by analyzing the immune system in the peripheral blood.”

    Taking advantage of dual mode

    One successful approach in generating an in-depth view of tissue architecture and spatial distribution of cells in addition to an overall immune cell profile is the dual-mode capability of the Hyperion™ Imaging System, which enables analysis of both suspension and tissue. Ding discussed his use of mass cytometry and IMC for an in-depth characterization of the immune cell subsets and protein profiles involved in signaling pathways in the peripheral blood and skin of patients with psoriasis (PS).

    Based on the behavior of the immune cell subsets as seen by CyTOF, Ding was able to hypothesize how cell migration affects the progression of the disease.

    The group analyzed peripheral blood mononuclear cell (PBMC) samples isolated from four patients who were newly diagnosed with graft-versus-host disease (GVHD) and four samples from healthy controls. Based on a combination of surface markers, they used CyTOF to simultaneously analyze T cell and B cell subsets using mass cytometry. Results showed that the frequencies of total T and B cells were statistically significantly decreased in GVHD samples, while frequencies of specific CD8+ T effector memory cells were increased.

    Following CyTOF mass cytometry analysis, two samples of PS lesions were obtained and stained for 31 immune markers for Imaging Mass Cytometry. In the IMC images, the team observed colocalization of immune cell subsets, validating their expected cell distribution and indicating the stability and reliability of the technique. Using dual-mode capability on the Hyperion Imaging System, the team identified 15 major immune cell populations in T cell lineages and characterized various other T cell populations simultaneously.

    Based on the behavior of the immune cell subsets as seen by CyTOF, Ding was able to hypothesize how cell migration affects the progression of the disease. For example, a reduction of lymphocytes in peripheral blood suggests that they may migrate to the lesion area. That hypothesis can be verified with IMC data. Ding adds that the use of single-cell mass cytometry allows systemic-level characterization of lymphocyte subpopulations and dysregulated signaling pathways in the blood, allowing him to identify abnormalities of different immune cell subsets.

    The use of both suspension and imaging modes on the Hyperion Imaging System has the potential to benefit many research areas, including oncology, immunology, immunophenotyping and more. Mass cytometry data can be used to predict the occurrence and progression of diseases, and IMC data represents the mechanism in lesions or other diseased tissue. Further study using these technologies has prompted Ding to develop novel probes for IMC to support successful imaging acquisition and offer new opportunities for diagnosing malignancies in the future.

    References:

    • Guo, R. et al. “Lymphocyte mass cytometry identifies a CD3-CD4+ cell subset with a potential role in psoriasis.” JCI Insight 4 (2019): e125306.
    • Yu, Y. et al. “Metal-labeled aptamers as novel nanoprobes for Imaging Mass Cytometry analysis.” Analytical Chemistry 92 (2020): 6,312–6,320.
    • Zhang, T. et al. “Immunocyte profiling using single-cell mass cytometry reveals EpCAM+ CD4+ T cells abnormal in colon cancer.” Frontiers in Immunology 10 (2019): 01571.

    Other Useful Links:

    • Imaging Mass Cytometry
    • Mass cytometry
    • Explore more applications using dual mode with Imaging Mass Cytometry

    Sign up to receive more about mass cytometry and Imaging Mass Cytometry

    For Research Use Only. Not for use in diagnostic procedures.

    • Related Applications

      • Imaging Mass Cytometry
      • Mass Cytometry
  • CyTOF Reveals Immune Disruption

    Discovering a hallmark phenotype of COVID-19

    Spotlight

    Mass Cytometry Key in Coronavirus Science Publication

    Impaired interferon response found in severe cases

    Download Spotlight

    As a specialist in the management of systemic autoximmune diseases, Benjamin Terrier, MD, PhD, regularly works with patient samples to learn how the activation state and proportions of immune cell subsets change with disease severity.

    Terrier is Professor of Medicine in the Internal Medicine Department of Cochin Hospital in Paris. Given his in-depth understanding of how the immune system can react to various diseases, confirmation of the novel coronavirus in France prompted a shift in his focus toward the management of COVID-19 infections. Terrier and his team observed patients with progressive worsening of the disease from cytokine storm and questioned the mechanism involved in disease severity. They initiated an investigation of the immunological characteristics associated with disease deterioration consistently observed between 8 and 12 days from the onset of symptoms.

    Because of the urgent need for more information about COVID-19 disease progression, the study covered a cross-sectional analysis of patients admitted due to disease worsening around day 10. And while comorbidities and potential autoimmune predispositions are known to correlate to worsening disease, the team specifically examined patients with no or very mild or controlled comorbidities so they could focus on the modifications provoked by the virus itself. A unique phenotype was identified, displaying impaired interferon type I response characterized by low interferon production and activity in severe and critically ill patients. The phenotype was also associated with a persistent blood virus load and an exacerbated inflammatory response.

    Terrier collaborates with the Pitié-Salpêtrière Cytometry (CyPS) to apply mass cytometry and the Maxpar® Direct™ Immune Profiling Assay™ in his current studies, and now to COVID-19 immune profiling. “Analyzing the immune response of patients with mild-to-moderate disease, severe disease and critical disease by mass cytometry allows us to cover the innate and adaptive immune responses simultaneously. Not only could we analyze immune cells, but also cytokine production and the transcriptional signatures in these patients. Essentially, we could look at everything in the whole blood,” Terrier explains.

    And while sampling whole blood might not be the perfect way to analyze a disease that tends to be localized in the lung, it is clearly a much less invasive approach to collecting samples from patients. Between ongoing collaborations with the CyPS facility and with groups at both the Imagine and Pasteur Institutes working on interferon type I signaling, Terrier was well-positioned for a quick start with an integrated approach.

    Easily transitioning CyTOF to COVID-19

    “We used the Maxpar Direct Immune Profiling Assay because we wanted a global approach to analyze the different immune cell types from both the innate and adaptive immune response. Most previous works on immune perturbations in autoimmune disease only analyzed the B cell compartment, the T cell compartment, or the regulatory T cells. A more comprehensive view is complicated using conventional flow cytometry, involving many different tubes to analyze one sample from a single patient,” says Terrier. “So, we wanted to use a more global approach facilitated by mass cytometry.”

    Because Terrier had been using mass cytometry for the analysis of perturbations in different autoimmune diseases and associations with disease outcome and response to therapy, he was able to quickly adapt his CyTOF® mass cytometry assays to COVID-19 studies. He recognized that this type of global analysis would align well with similar disruptions of the innate response and apparent abnormalities of the adaptive response observed in COVID-19.

    The study, now published in Science (Hadjadj, J. et al.), included the use of the Maxpar Direct Immune Profiling Assay with the addition of PD-1 and Tim-3 immune checkpoint inhibitors to the assay’s standard backbone 30-marker panel. Using this expanded version of the assay panel, the team was able to work quickly to generate quality data, analyze the results and submit a preprint publication within one month. With the success of this panel, the team plans to add at least 12 markers to develop a new panel for further characterization of the cells, including the definition, differentiation and activation status of the immune cell populations.

    “Also, a very important advantage to us was the ability to automate our analysis with Maxpar Pathsetter™ software. This is significant time savings and generates data that is much more reproducible and less impacted by the subjectivity that occurs with manual analysis,” he adds.

    Expanding current studies with new panels

    Terrier notes that further longitudinal follow-up of patients is planned, and additional comorbidity studies are already underway. He and his team are interested in how different comorbidities might affect the observed defect in interferon production across patients with similar severity levels of COVID-19. In addition, the group would like to examine how each cell population assessed differs across disease severity, and to better understand the immune system perturbation that was observed.

    Terrier points out that getting this global perspective of immune response using mass cytometry worked so well that he plans to expand its use in his current studies on autoimmune disease, adding in new markers to increase immune cell characterization and determine differences between various autoimmune diseases. By generating a baseline signature, it may be possible to identify specific changes occurring in response to therapy and determine predictive characteristics for this response, potentially offering a path to better disease management.

    Want to Learn More?

    Submit the form below to have a representative contact you with more information about the Maxpar Direct Immune Profiling System or Helios™, a CyTOF system.

    For Research Use Only. Not for use in diagnostic procedures.



    References:

    • Hadjadj, J. et al. "Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients." Science (2020): doi:10.1126/science.abc6027
    • Related Applications

      • Mass Cytometry
    • Related Products

      • Helios, a CyTOF System
  • Extending global understanding of COVID-19

    Achieving multi-omic insights with microfluidics and CyTOF technology

    Spotlight

    IMC a key to early data on SARS-CoV-2

    Immune response in respiratory cases focus of researchers in China

    Download Spotlight

    “The host's immune response to a pathogen is an ongoing theme in infectious disease research, contributing to disease progression and a patient's symptoms and outcome,” says Dr. Dexi Chen, PhD, MD, of the Beijing Institute of Hepatology and You’an Hospital. “As such, our current research focuses on patients’ immune response to 2019-nCoV in the lungs of severe COVID-19 patients.”

    In December 2019, an outbreak of COVID-19 was reported in Wuhan, Hubei Province, China. The disease spread in China, followed by reports of increasing cases globally. Initially, most patients who died in the hospital died of severe pneumonia and acute respiratory distress syndrome (ARDS). Chen and his team worked quickly to understand COVID-19 pathology in critical patients, determined to help direct treatments for the disease. To date, this prolific lab has published four studies using mass cytometry to investigate COVID-19.

    While their initial pathological findings from a COVID-19 patient who had developed ARDS showed the characteristic diffuse alveolar damage and obvious mononuclear cell infiltration in the lung, the types and subsets of infiltrating immune cells in the lung tissues were not clear. The group used Imaging Mass Cytometry™ (IMC™) to analyze the immune cell clusters in lung tissue after biopsy of two patients with COVID-19: one who had died from ARDS and the other of severe pneumonia (Zhang et al.).

    'Imaging Mass Cytometry with CyTOF technology is our method of choice for these studies because the limited samples are very valuable. CyTOF allows more than 30 markers in one panel, and we don’t have to worry about color compensation or background autofluorescence issues,' says Dr. Dexi Chen, PhD, MD.

    Results showed that ARDS in the first patient was the result of an increased infiltration of immune cells—specifically T cell lymphocyte and macrophage subsets—as well as more focal infiltration of natural killer cells. The added complication of bacterial pneumonia in the second patient led to a different distribution, shown by cluster infiltration of neutrophils and macrophages, diffuse infiltration of T cell subsets and scattered infiltration of natural killer cells and dendritic cells. This newfound data suggests the specific cell types that play a major role in lung injury.

    “Imaging Mass Cytometry with CyTOF® technology is our method of choice for these studies because the limited samples are very valuable. CyTOF allows more than 30 markers in one panel, and we don’t have to worry about color compensation or background autofluorescence issues,” explains Chen.

    Moving forward with these new findings, the team examined gene expression levels associated with the modified immune response observed during disease progression. With the need to move swiftly to determine the immune response of COVID-19 patients before and after treatment, they chose the Biomark™ HD system and 96.96 Dynamic Array™ integrated fluidic circuits (IFCs) for gene expression (GE) (Ouyang et al.). Chen adds that in a short period of time, one 96.96 IFC for gene expression can process 9,216 reactions from 96 samples and assays, using less sample and reagent to achieve high-quality, consistent results. "This is critical for us to answer these questions quickly,” he says.

    Using Juno™ and Biomark HD to examine the innate immune status and immune-related gene expression levels in 11 patients, the group found decreased T cell proportions and down-regulated gene expression involved in T cell activation and differentiation in severe COVID-19 patients, indicating suppression of the T cell immune response.

    The team then applied mass cytometry to analyze PBMC from patients with different disease progression, observing obvious differences in composition of immune cells in severe patients (Wang et al.). CyTOF technology revealed a disturbed immune system homeostasis with dysfunction in T cell subsets, dendritic cells and macrophages that were excessively activated at first and became exhausted as the disease progressed to critical stages. Further study with CyTOF and cytokine assays on additional patients confirmed these findings, showing a decrease in T cells, B cells and NK cells along with a progressive decrease of interleukin-2 (IL-2) in plasma (Shi et al.). Association of immune cell suppression with IL-2 could serve as a warning of disease deterioration in patients with COVID-19 pneumonia.

    Such a combination of approaches to uncover the diverse mechanisms activated in the COVID-19 response strengthens our understanding of the disease and its progression. These studies demonstrate the unique changes in immune response across varying disease progression as well as patient response to treatment. Further experiments could be performed to support this data, increasing patient numbers and expanding the antibodies used to discern more targeted cell type behaviors.

    SARS-CoV-2 has displayed distinct properties throughout this pandemic, yet it also shares some similarities with SARS-CoV and MERS-CoV. These findings have helped extend global understanding of the SARS-CoV-2 infection mechanism, and they provide a basis for future novel immune therapeutic strategies.

    Want to Learn More?

    Submit the form below to request a representative contact you or to sign up to get future information about mass cytometry and Imaging Mass Cytometry.

    For Research Use Only. Not for use in diagnostic procedures.



    References:

    • Ouyang, Y. et al. “Down-regulated gene expression spectrum and immune responses changed during the disease progression in COVID-19 patients.” Clinical Infectious Diseases (2020): ciaa462.
    • Shi, H. et al. “The inhibition of IL-2/IL-2R gives rise to CD8+ T cell and lymphocyte decrease through JAK1-STAT5 in critical patients with COVID-19 pneumonia.” Cell Death & Disease 11 (2020): 429.
    • Wang, W. et al. “High-dimensional immune profiling by mass cytometry revealed immunosuppression and dysfunction of immunity in COVID-19 patients.” Cellular & Molecular Immunology 17 (2020): 650–652.
    • Zhang, Y. et al. “Inflammatory response cells during acute respiratory distress syndrome in patients with coronavirus disease 2019 (COVID-19).” Annals of Internal Medicine (2020): 10.7326/L20-0227.

    Other Useful Links:

    • Imaging Mass Cytometry
    • Mass cytometry
    • Biomark HD System
    • COVID research solutions
    • Related Applications

      • Imaging Mass Cytometry
      • Mass Cytometry
    • Related Products

      • Biomark HD
      • Juno
  • COVID-19, cardiovascular disease and mass cytometry

    A lesson in how CyTOF technology can empower flexibility in research

    Spotlight

    University of Virginia researcher focuses on role of immune cell subsets in chronic inflammatory diseases

    Download Spotlight

    Junior Investigator Hema Kothari, PhD, is one of many researchers who have been able to pivot their research to study mechanisms that influence the risk of severe COVID-19 progression. Dr. Kothari is a Research Assistant Professor in Dr. Coleen McNamara’s lab at the University of Virginia (UVA) in Charlottesville and focuses on understanding the role of immune cell subsets in cardiovascular disease and other chronic inflammatory diseases.

    In an effort to develop approaches that better enable personalized treatments for immunomodulatory and anti-inflammatory therapies, the lab created a mass cytometry-based pipeline for in-depth analysis of human peripheral blood mononuclear cells (PBMC) to identify specific immune phenotypes that associate with disease severity and response to therapy. Work is ongoing using these customized panels and study populations including subjects with rheumatoid arthritis, coronary artery disease and other inflammatory illnesses.

    Recently, IL-1β, a proinflammatory cytokine investigated by Kothari, has emerged as a key mediator of the cytokine storm linked to high morbidity and mortality from COVID-19. In addition, blocking the IL-1 receptor with anakinra has entered clinical trials in COVID-19 patients.

    Upon the realization that her current study could easily translate to and possibly accelerate COVID-19 research, Kothari refocused her efforts on the specific immune cell subsets targeted by IL-1β and IL-1β-induced signaling pathways in humans.

    Cardiovascular problems are a known issue with cytokine storm onset in COVID-19 patients and correlate with disease severity and mortality. Applying her knowledge from cardiovascular research to SARS-CoV-2 activated inflammation, Kothari has initiated a new study that is funded by the UVA Manning Fund for COVID-19 Research. The study’s goals are to phenotype PBMC samples collected from COVID-19 patients at UVA by mass cytometry to identify circulating immune cell subtypes that are most responsive to IL-1β stimulation and immune phenotypes that correlate with disease severity, and to develop a customized diagnostic biomarker assay for early identification of those at risk of a cytokine storm for improved patient outcomes.

    With high interindividual heterogeneity in response, when some patients get better and some progress to critical stages of the disease, the ability to look at broad intracellular signaling supports a greater understanding of what is occurring upstream of cytokine production. This will allow earlier identification and possible prediction of which patients are at a higher risk of cytokine storm and which are most likely to benefit from anakinra therapy.

    Uniquely CyTOF

    Kothari works with CyTOF® technology for the majority of her projects because it offers comprehensive immune cell analysis, the ability to more deeply profile immune cell subtypes and the flexibility to adjust and expand panels easily. With flow cytometry or other conventional cell analysis methods, it can be challenging to expand a panel to more than 20 markers. That ceiling tends to be just enough to identify different immune cell types in the PBMC sample but does not allow the opportunity to ask more in-depth questions about what these cells are doing.

    “CyTOF is really helpful because you can identify all cell subsets as well as the specific proteins that you are interested in while avoiding challenges with fluorophore spectrum overlap,” explains Kothari. “For example, we were able to use a 37-marker panel in the COVID-19 study. I am interested in looking for cell surface receptors such as IL-1 and IL-6 receptors, intracellular signaling proteins activated downstream of IL-1β and IL-6, intracellular cytokines and other markers across different immune cell types. The power of CyTOF is that you can have a lot of different markers in a panel and analyze them all at once.”

    Furthermore, CyTOF allows more flexibility in experimental approach as the metal-tagged antibodies used are compatible with harsh cell fixation and permeabilization treatments. The compatibility of CyTOF with methanol treatment is key for performing analyses such as phosphoflow in the high-parameter space, mentions Kothari. Many fluorophores used in high-parameter fluorescence cytometry can be destroyed when cells are treated with methanol.

    Further exploration takes many directions

    With their CyTOF based approach already in place, the team can easily look for IL-1β-mediated signaling pathways across different immune cell subtypes and identify immune cell targets for IL-1β in humans, for more general cardiovascular implications as well as for COVID-19 specifically. One of the key findings uniquely enabled by CyTOF and advanced computational analysis was the discovery that CCR6-positive T cells are a major target of IL-1β in humans. Next steps could include looking for downstream effects of this interaction and whether it plays a role in IL-1β-mediated inflammatory mechanisms in cardiovascular diseases or in other inflammatory diseases. Kothari would also like to take these findings back into murine systems and investigate the impact of CCR6-positive T cells on atherosclerosis.

    Kothari is able to expand her investigations in many directions using the lab’s current approach involving custom panel development and CyTOF technology to identify mechanisms, predict disease outcomes and characterize therapeutic response.

    One therapy does not fit all. With this in mind, Kothari ultimately would like to apply her findings to developing precision and personalized approaches for the treatment of cardiovascular disease.

    Learn more about Fluidigm’s response to the COVID-19 pandemic.




    Sign up to receive mass cytometry updates.

    * You may withdraw your consent anytime in every email you will receive from us.
    To learn more about how we use your information, read our Privacy Policy.

    For Research Use Only. Not for use in diagnostic procedures.

    • Related Applications

      • Mass Cytometry
  • Coupling immunology and microfluidics

    How one lab explores the immune system one T cell at a time

    Spotlight

    Coupling immunology and microfluidics: How one lab explores the immune system one T cell at a time

    There is an art to moving from complex scientific concept to simple question. How can we connect a single molecule to a disease state? How can our understanding of a disorder help us discover effective immunosuppressive drugs? What can we model in the laboratory that might impact the efficacy of a new vaccine? Dr. Luc Teyton, MD, PhD, has routinely pursued answers to scientific questions like these throughout his career as a clinical immunologist-turned-professor who now challenges his students to do the same.

    Putting these basic questions into practice, researchers in the Teyton lab at the renowned Scripps Research institute in La Jolla, California, primarily focus on autoimmunity by investigating the association between human leukocyte antigen (HLA) molecules and various immune conditions using genomics, transcriptomics and proteomics. By approaching research one simple question at a time, Teyton is able to look broadly at a scientific hypothesis instead of limiting himself to a particular technique or discipline. “Rather, we collaborate with specialists in various fields to perform whatever is needed to give us different parts of an answer," he says.

    Teyton’s pioneering approach to taking on any technique that makes a project successful has led the lab to adopt an array of methods from biochemistry, structural biology and biophysics to genetics and cell biology. However, a fundamental challenge in the study of the immune system involves isolating and analyzing small numbers of cells. As Teyton explains, “Sampling humans dictates the approaches you can use. Human cells are accessible from only three origins: saliva, blood and biopsies, which provide a very limited number of immune cells.” Teyton and his team have spent years developing enrichment tools and techniques that would allow them to isolate the needle in the haystack, such as the major histocompatibility complex (MHC) tetramer that enables labeling and sorting of single CD4 T cells based on their antigen recognition. “That being said, we usually end up with ten to a hundred cells of interest from each sample, leaving us no choice but to have recourse to single-cell approaches.”

    The microfluidics entourage

    Traditional manual analysis of single T cells to determine sequence and structure equated to a daunting amount of work and dismal efficiency. It took one post-doctoral fellow five years to sequence 250 cells from diabetic mice—work that now represents two experiments and two weeks of work, with efficiency of cell recovery reaching 60–70%, thanks to microfluidics.

    Fluidigm microfluidics technologies have transformed the time and workload of these single-cell experiments with automation and conservation of sample and reagents. Single-cell isolation and sample preparation upstream of next-generation sequencing and real-time PCR analysis can be done using the C1™ system, and bulk cell and tissue analysis is easily completed on the Biomark™ HD system. Researchers rely heavily on Biomark to quantitatively examine series of 96 genes of interest in single cells. They can also comprehensively sequence T cell receptor chains from the same cell to analyze gene expression patterns after preparation on C1. “We have been impressed by the quality of the data generated by C1 with a depth of interrogation that was on average beyond 10,000 genes per cell,” adds Teyton.

    Teyton describes as an example how microfluidics has allowed his lab to perform an in-depth analysis of the early anti-insulin response in the nonobese diabetic (NOD) mouse, a model of type 1 diabetes (T1D). Now the group can work on translating these observations to humans, particularly at-risk populations like first-degree relatives of a newly diagnosed T1D patient. Teyton hopes that his lab’s data on activation of an anti-insulin T cell response will explain the progression of autoimmunity in real time and offer an improved monitoring tool to replace tools that are unreliable and poorly predictive. Upon further study, the same approach could potentially be used to evaluate the efficacy of immune intervention during the pre-clinical phase of disease.

    Integration of microfluidics across hypotheses

    In addition to C1 and Biomark HD, Teyton’s lab recently acquired the Juno™ system, which can process and prepare all Fluidigm microfluidic devices. The lab will incorporate Juno into a project that extensively uses C1 for scRNA-seq, where they will examine immune cell populations in colonic biopsies of patients with inflammatory bowel diseases.

    Teyton would like to translate some of his approaches to the clinic for use in diagnosis and therapeutic monitoring. “To be successful, we have to validate the genomics data obtained using the Fluidigm suite of instruments by proteomics data to address protein expression for each cell we analyze,” he explains. “Ultimately, our success will depend on the efficiency of our collaboration and the integration of data from each platform. I believe that single-cell analysis has a huge potential in the clinic, but I am also aware that this potential is matched by challenges of a similar magnitude.”

    Sign up to receive mass cytometry updates.


    For Research Use Only. Not for use in diagnostic procedures.

    • Related Applications

      • Mass Cytometry
    • Related Products

      • Helios, a CyTOF System
  • Machine Learning and Infectious Disease

    Adriana Tomic on the role machine learning is playing in vaccine development

    Spotlight

    Machine learning and infectious disease: how we can better understand vaccine efficacy at the patient level

    Download Spotlight

    In one of the 20 most-read papers of 2019, Stanford University and University of Oxford researchers developed an automated machine learning system, Sequential Iterative Modeling “OverNight” (SIMON), that compares high-dimensional datasets from heterogeneous clinical studies focusing on vaccine response (Figure 1). The aim of the project is to build more accurate predictive models and accelerate analysis and discovery. “SIMON is a highly useful approach for data-driven hypothesis generation from disparate clinical datasets,” says Adriana Tomic, PhD, lead author on the paper.


    Figure 1. Graphical illustration of SIMON.

    Blending machine learning with systems immunology

    Taking a systems immunology approach to evaluate changes in immune cells and molecules during an infection or treatment can accelerate the understanding of an effective immune response. Integration of machine learning algorithms into systems immunology can further facilitate the discovery of unseen patterns in relevant immune cell subsets and genes. However, current approaches are challenged by heterogeneous or incomplete datasets. To overcome these obstacles and tailor machine learning to adapt to the variability of multiple clinical studies, Tomic and team developed SIMON open source software for the application of machine learning to high-dimensional clinical datasets. Put more simply, SIMON can predict if an individual will respond appropriately to a vaccine based on a set of immune system parameters.

    SIMON was developed to improve computational analysis of influenza vaccination responses in humans, a task that is typically unreliable due to smaller sample sizes and single-target studies. Instead, SIMON analyzes multiple parameters from numerous individuals across several studies to accurately capture biological variability and to increase statistical significance. The inclusion of machine learning helps minimize sample loss and identify algorithms that fit any given data distribution, maximizing predictive accuracy and other performance measurements.

    Initially, SIMON analyzed data from the Stanford Human Immune Monitoring Center (HIMC) in a novel project, FluPRINT. Five separate clinical studies of seasonal influenza vaccination were included, with various platforms and expanding parameters, including single-cell analysis at the gene and protein levels using mass cytometry to capture both immune system and individual variation. “By using SIMON, we identified subsets of immune cells not previously described to provide protection against the virus. These results are important for the development of the next generation of vaccines and have the capacity to fundamentally change vaccinology by application of machine learning to speed up discovery,” explains Tomic.

    The data generated from this study was collected in an open-access FluPRINT database, meant to enable large-scale studies exploring the cellular and molecular basis of successful antibody responses to influenza vaccines. The resource can be used to uncover new markers and mechanisms that are important for influenza vaccine immunogenicity.

    The move to any infectious disease

    The SIMON systems immunology approach used in the FluPRINT project to predict flu vaccine response could also become crucial for understanding other infectious diseases, such as SARS-CoV-2 (Figure 2), and developing a vaccine to stop the COVID-19 pandemic. “Generating a similar dataset from ongoing clinical trials on SARS-CoV-2 will be critical for understanding this virus and generating an efficient vaccine,” says Tomic.


    Figure 2. Coronavirus morphology.

    Evaluating interindividual variation in response to an infection or a vaccine aligns with clinical presentation of COVID-19, where such a wide and unpredictable range of immune responses is observed from patient to patient. SIMON could assist in determining which patients might progress to critical stages of the disease based on their immune profiles and which would respond to certain treatments. To this end, Tomic is part of a University of Oxford team that has now identified a SARS-CoV-2 vaccine candidate and is working toward the first clinical testing phase.

    The FluPRINT project is described in detail at fluprint.com and SIMON is available for download at genular.org.

    Read the official release about the new vaccine candidate against COVID-19: ovg.ox.ac.uk/news/covid-19-vaccine-development

    Sign up to receive mass cytometry updates.


    * You may withdraw your consent anytime in every email you will receive from us.
    To learn more about how we use your information, read our Privacy Policy.

    For Research Use Only. Not for use in diagnostic procedures.

    • Related Applications

      • Mass Cytometry
    • Related Products

      • Helios, a CyTOF System
  • About Us
  • Careers
  • Contact
  • Legal
  • EEO Statement
  • Press
  • Investors
  • Feedback
  • Accessibility & Disability Statement
  • Facebook
  • Twitter
  • LinkedIn
  • Glassdoor