skip to main content


Search for: All records

Creators/Authors contains: "Kurabayashi, Katsuo"

Note: When clicking on a Digital Object Identifier (DOI) number, you will be taken to an external site maintained by the publisher. Some full text articles may not yet be available without a charge during the embargo (administrative interval).
What is a DOI Number?

Some links on this page may take you to non-federal websites. Their policies may differ from this site.

  1. Free, publicly-accessible full text available October 1, 2024
  2. null (Ed.)
  3. null (Ed.)
  4. Despite widespread concern regarding cytokine storms leading to severe morbidity in COVID-19, rapid cytokine assays are not routinely available for monitoring critically ill patients. We report the clinical application of a digital protein microarray platform for rapid multiplex quantification of cytokines from critically ill COVID-19 patients admitted to the intensive care unit (ICU) at the University of Michigan Hospital. The platform comprises two low-cost modules: (i) a semi-automated fluidic dispensing/mixing module that can be operated inside a biosafety cabinet to minimize the exposure of the technician to the virus infection and (ii) a 12–12–15 inch compact fluorescence optical scanner for the potential near-bedside readout. The platform enabled daily cytokine analysis in clinical practice with high sensitivity (<0.4 pg mL −1 ), inter-assay repeatability (∼10% CV), and rapid operation providing feedback on the progress of therapy within 4 hours. This test allowed us to perform serial monitoring of two critically ill patients with respiratory failure and to support immunomodulatory therapy using the selective cytopheretic device (SCD). We also observed clear interleukin-6 (IL-6) elevations after receiving tocilizumab (IL-6 inhibitor) while significant cytokine profile variability exists across all critically ill COVID-19 patients and to discover a weak correlation between IL-6 to clinical biomarkers, such as ferritin and C-reactive protein (CRP). Our data revealed large subject-to-subject variability in patients' response to COVID-19, reaffirming the need for a personalized strategy guided by rapid cytokine assays. 
    more » « less
  5. Recent advances in transcriptomic analysis at single-cell resolution reveal cell-to-cell heterogeneity in a biological sample with unprecedented resolution. Partitioning single cells in individual micro-droplets and harvesting each cell's mRNA molecules for next-generation sequencing has proven to be an effective method for profiling transcriptomes from a large number of cells at high throughput. However, the assays to recover the full transcriptomes are time-consuming in sample preparation and require expensive reagents and sequencing cost. Many biomedical applications, such as pathogen detection, prefer highly sensitive, reliable and low-cost detection of selected genes. Here, we present a droplet-based microfluidic platform that permits seamless on-chip droplet sorting and merging, which enables completing multi-step reaction assays within a short time. By sequentially adding lysis buffers and reactant mixtures to micro-droplet reactors, we developed a novel workflow of single-cell reverse transcription loop-mediated-isothermal amplification (scRT-LAMP) to quantify specific mRNA expression levels in different cell types within one hour. Including single cell encapsulation, sorting, lysing, reactant addition, and quantitative mRNA detection, the fully on-chip workflow provides a rapid, robust, and high-throughput experimental approach for a wide variety of biomedical studies. 
    more » « less
  6. We report on system integration of plasmonic nanoparticles and a few-layered molybdenum disulfide (M0S2) photoconductive nanochannel sheet on a silicon substrate. Plasma-assisted electrostatic bonding and van der Waals bonding are employed to create a high-sensitivity photoelectronic biosensor for immunological analysis. 
    more » « less
  7. null (Ed.)
    Abstract Digital protein assays have great potential to advance immunodiagnostics because of their single-molecule sensitivity, high precision, and robust measurements. However, translating digital protein assays to acute clinical care has been challenging because it requires deployment of these assays with a rapid turnaround. Herein, we present a technology platform for ultrafast digital protein biomarker detection by using single-molecule counting of immune-complex formation events at an early, pre-equilibrium state. This method, which we term “pre-equilibrium digital enzyme-linked immunosorbent assay” (PEdELISA), can quantify a multiplexed panel of protein biomarkers in 10 µL of serum within an unprecedented assay incubation time of 15 to 300 seconds over a 104 dynamic range. PEdELISA allowed us to perform rapid monitoring of protein biomarkers in patients manifesting post-chimeric antigen receptor T-cell therapy cytokine release syndrome, with ∼30-minute sample-to-answer time and a sub–picograms per mL limit of detection. The rapid, sensitive, and low-input volume biomarker quantification enabled by PEdELISA is broadly applicable to timely monitoring of acute disease, potentially enabling more personalized treatment. 
    more » « less
  8. Abstract

    The ability to detect low‐abundance proteins in human body fluids plays a critical role in proteomic research to achieve a comprehensive understanding of protein functions and early‐stage disease diagnosis to reduce mortality rates. Ultrasensitive (sub‐fM), rapid, simple “mix‐and‐read” plasmonic colorimetric biosensing of large‐size (≈180 kDa) proteins in biofluids using an ultralow‐noise multilayer molybdenum disulfide (MoS2) photoconducting channel is reported here. With its out‐of‐plane structure optimized to minimize carrier scattering, the multilayer MoS2channel operated under near‐infrared illumination enables the detection of a subtle plasmonic extinction shift caused by antigen‐induced nanoprobe aggregation. The demonstrated biosensing strategy allows quantifying carcinoembryonic antigen in unprocessed whole blood with a dynamic range of 106, a sample‐to‐answer time of 10 min, and a limit of detection of 0.1–3 pg mL−1, which is ≈100‐fold more sensitive than the clinical‐standard enzyme‐linked immunosorbent assays. The biosensing methodology can be broadly used to realize timely personalized diagnostics and physiological monitoring of diseases in point‐of‐care settings.

     
    more » « less