Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David J. Guckenberger is active.

Publication


Featured researches published by David J. Guckenberger.


Analytical Chemistry | 2011

Rapid Prototyping of Arrayed Microfluidic Systems in Polystyrene for Cell-Based Assays

Edmond W. K. Young; Erwin Berthier; David J. Guckenberger; Eric K. Sackmann; Casey Lamers; Ivar Meyvantsson; Anna Huttenlocher; David J. Beebe

Microfluidic cell-based systems have enabled the study of cellular phenomena with improved spatiotemporal control of the microenvironment and at increased throughput. While poly(dimethylsiloxane) (PDMS) has emerged as the most popular material in microfluidics research, it has specific limitations that prevent microfluidic platforms from achieving their full potential. We present here a complete process, ranging from mold design to embossing and bonding, that describes the fabrication of polystyrene (PS) microfluidic devices with similar cost and time expenditures as PDMS-based devices. Emphasis was placed on creating methods that can compete with PDMS fabrication methods in terms of robustness, complexity, and time requirements. To achieve this goal, several improvements were made to remove critical bottlenecks in existing PS embossing methods. First, traditional lithographic techniques were adapted to fabricate bulk epoxy molds capable of resisting high temperatures and pressures. Second, a method was developed to emboss through-holes in a PS layer, enabling creation of large arrays of independent microfluidic systems on a single device without need to manually create access ports. Third, thermal bonding of PS layers was optimized in order to achieve quality bonding over large arrays of microsystems. The choice of materials and methods was validated for biological function in two different cell-based applications to demonstrate the versatility of our streamlined fabrication process.


Molecular Biology of the Cell | 2016

High rates of chromosome missegregation suppress tumor progression but do not inhibit tumor initiation

Lauren M. Zasadil; Eric M.C. Britigan; Sean D. Ryan; Charanjeet Kaur; David J. Guckenberger; David J. Beebe; Amy R. Moser; Beth A. Weaver

Expression of a truncated allele of the Apc tumor suppressor causes intestinal tumors with a low rate of chromosomal instability (CIN). Increasing the rate of CIN suppresses tumor growth without inhibiting tumor initiation in both the small intestine and colon, suggesting that increasing CIN is a useful chemotherapeutic strategy.


The Journal of Molecular Diagnostics | 2014

HIV Viral RNA Extraction in Wax Immiscible Filtration Assisted by Surface Tension (IFAST) Devices

Scott M. Berry; Alex J. LaVanway; Hannah M. Pezzi; David J. Guckenberger; Meghan A. Anderson; Jennifer M. Loeb; David J. Beebe

The monitoring of viral load is critical for proper management of antiretroviral therapy for HIV-positive patients. Unfortunately, in the developing world, significant economic and geographical barriers exist, limiting access to this test. The complexity of current viral load assays makes them expensive and their access limited to advanced facilities. We attempted to address these limitations by replacing conventional RNA extraction, one of the essential processes in viral load quantitation, with a simplified technique known as immiscible filtration assisted by surface tension (IFAST). Furthermore, these devices were produced via the embossing of wax, enabling local populations to produce and dispose of their own devices with minimal training or infrastructure, potentially reducing the total assay cost. In addition, IFAST can be used to reduce cold chain dependence during transportation. Viral RNA extracted from raw samples stored at 37°C for 1 week exhibited nearly complete degradation. However, IFAST-purified RNA could be stored at 37°C for 1 week without significant loss. These data suggest that RNA isolated at the point of care (eg, in a rural clinic) via IFAST could be shipped to a central laboratory for quantitative RT-PCR without a cold chain. Using this technology, we have demonstrated accurate and repeatable measurements of viral load on samples with as low as 50 copies per milliliter of sample.


Clinical Cancer Research | 2017

Integrated Analysis of Multiple Biomarkers from Circulating Tumor Cells Enabled by Exclusion-Based Analyte Isolation.

Jamie M. Sperger; Lindsay N. Strotman; Allison Welsh; Benjamin P. Casavant; Zachery Chalmers; Sacha Horn; Erika Heninger; Stephanie M. Thiede; Jacob T Tokar; Benjamin K. Gibbs; David J. Guckenberger; Lakeesha Carmichael; Scott M. Dehm; Philip J. Stephens; David J. Beebe; Scott M. Berry; Joshua M. Lang

Purpose: There is a critical clinical need for new predictive and pharmacodynamic biomarkers that evaluate pathway activity in patients treated with targeted therapies. A microscale platform known as VERSA (versatile exclusion-based rare sample analysis) was developed to integrate readouts across protein, mRNA, and DNA in circulating tumor cells (CTC) for a comprehensive analysis of the androgen receptor (AR) signaling pathway. Experimental Design: Utilizing exclusion-based sample preparation principles, a handheld chip was developed to perform CTC capture, enumeration, quantification, and subcellular localization of proteins and extraction of mRNA and DNA. This technology was validated across integrated endpoints in cell lines and a cohort of patients with castrate-resistant prostate cancer (CRPC) treated with AR-targeted therapies and chemotherapies. Results: The VERSA was validated in cell lines to analyze AR protein expression, nuclear localization, and gene expression targets. When applied to a cohort of patients, radiographic progression was predicted by the presence of multiple AR splice variants and activity in the canonical AR signaling pathway. AR protein expression and nuclear localization identified phenotypic heterogeneity. Next-generation sequencing with the FoundationOne panel detected copy number changes and point mutations. Longitudinal analysis of CTCs identified acquisition of multiple AR variants during targeted treatments and chemotherapy. Conclusions: Complex mechanisms of resistance to AR-targeted therapies, across RNA, DNA, and protein endpoints, exist in patients with CRPC and can be quantified in CTCs. Interrogation of the AR signaling pathway revealed distinct patterns relevant to tumor progression and can serve as pharmacodynamic biomarkers for targeted therapies. Clin Cancer Res; 23(3); 746–56. ©2016 AACR.


Analytical Chemistry | 2014

Efficient sample preparation from complex biological samples using a sliding lid for immobilized droplet extractions.

Benjamin P. Casavant; David J. Guckenberger; David J. Beebe; Scott M. Berry

Sample preparation is a major bottleneck in many biological processes. Paramagnetic particles (PMPs) are a ubiquitous method for isolating analytes of interest from biological samples and are used for their ability to thoroughly sample a solution and be easily collected with a magnet. There are three main methods by which PMPs are used for sample preparation: (1) removal of fluid from the analyte-bound PMPs, (2) removal of analyte-bound PMPs from the solution, and (3) removal of the substrate (with immobilized analyte-bound PMPs). In this paper, we explore the third and least studied method for PMP-based sample preparation using a platform termed Sliding Lid for Immobilized Droplet Extractions (SLIDE). SLIDE leverages principles of surface tension and patterned hydrophobicity to create a simple-to-operate platform for sample isolation (cells, DNA, RNA, protein) and preparation (cell staining) without the need for time-intensive wash steps, use of immiscible fluids, or precise pinning geometries. Compared to other standard isolation protocols using PMPs, SLIDE is able to perform rapid sample preparation with low (0.6%) carryover of contaminants from the original sample. The natural recirculation occurring within the pinned droplets of SLIDE make possible the performance of multistep cell staining protocols within the SLIDE by simply resting the lid over the various sample droplets. SLIDE demonstrates a simple easy to use platform for sample preparation on a range of complex biological samples.


PLOS ONE | 2016

High Specificity in Circulating Tumor Cell Identification Is Required for Accurate Evaluation of Programmed Death-Ligand 1

Jennifer L. Schehr; Zachery D. Schultz; Jay Warrick; David J. Guckenberger; Hannah M. Pezzi; Jamie M. Sperger; Erika Heninger; Anwaar Saeed; Ticiana Leal; Kara Mattox; Anne M. Traynor; Toby C. Campbell; Scott M. Berry; David J. Beebe; Joshua M. Lang

Background Expression of programmed-death ligand 1 (PD-L1) in non-small cell lung cancer (NSCLC) is typically evaluated through invasive biopsies; however, recent advances in the identification of circulating tumor cells (CTCs) may be a less invasive method to assay tumor cells for these purposes. These liquid biopsies rely on accurate identification of CTCs from the diverse populations in the blood, where some tumor cells share characteristics with normal blood cells. While many blood cells can be excluded by their high expression of CD45, neutrophils and other immature myeloid subsets have low to absent expression of CD45 and also express PD-L1. Furthermore, cytokeratin is typically used to identify CTCs, but neutrophils may stain non-specifically for intracellular antibodies, including cytokeratin, thus preventing accurate evaluation of PD-L1 expression on tumor cells. This holds even greater significance when evaluating PD-L1 in epithelial cell adhesion molecule (EpCAM) positive and EpCAM negative CTCs (as in epithelial-mesenchymal transition (EMT)). Methods To evaluate the impact of CTC misidentification on PD-L1 evaluation, we utilized CD11b to identify myeloid cells. CTCs were isolated from patients with metastatic NSCLC using EpCAM, MUC1 or Vimentin capture antibodies and exclusion-based sample preparation (ESP) technology. Results Large populations of CD11b+CD45lo cells were identified in buffy coats and stained non-specifically for intracellular antibodies including cytokeratin. The amount of CD11b+ cells misidentified as CTCs varied among patients; accounting for 33–100% of traditionally identified CTCs. Cells captured with vimentin had a higher frequency of CD11b+ cells at 41%, compared to 20% and 18% with MUC1 or EpCAM, respectively. Cells misidentified as CTCs ultimately skewed PD-L1 expression to varying degrees across patient samples. Conclusions Interfering myeloid populations can be differentiated from true CTCs with additional staining criteria, thus improving the specificity of CTC identification and the accuracy of biomarker evaluation.


Lab on a Chip | 2012

Induced hydrophobic recovery of oxygen plasma-treated surfaces

David J. Guckenberger; Erwin Berthier; Edmond W. K. Young; David J. Beebe

Plasma treatment is a widely used method in microfabrication laboratories and the plasticware industry to functionalize surfaces for device bonding and preparation for mammalian cell culture. However, spatial control of plasma treatment is challenging because it typically requires a tedious masking step that is prone to alignment errors. Currently, there are no available methods to actively revert a surface from a treated hydrophilic state to its original hydrophobic state. Here, we describe a method that relies on physical contact treatment (PCT) to actively induce hydrophobic recovery of plasma-treated surfaces. PCT involves applying brushing and peeling processes with common wipers and tapes to reverse the wettability of hydrophilized surfaces while simultaneously preserving hydrophilicity of non-contacted surfaces. We demonstrate that PCT is a user-friendly method that allows 2D and 3D surface patterning of hydrophobic regions, and the protection of hydrophilic surfaces from unwanted PCT-induced recovery. This method will be useful in academic and industrial settings where plasma treatment is frequently used.


Journal of Laboratory Automation | 2015

High-Density Self-Contained Microfluidic KOALA Kits for Use by Everyone

David J. Guckenberger; Erwin Berthier; David J. Beebe

Cell-based assays are essential tools used by research labs in a wide range of fields, including cell biology, toxicology, and natural product discovery labs. However, in some situations, the need for cell-based assays does not justify the costs of maintaining cell culture facilities and retaining skilled staff. The kit-on-a-lid assay (KOALA) technology enables accessible low-cost and prepackageable microfluidic platforms that can be operated with minimal infrastructure or training. Here, we demonstrate and characterize high-density KOALA methods for high-throughput applications, achieving an assay density comparable to that of a 384-well plate and usability by hand with no liquid-handling equipment. We show the potential for high-content screening and complex assays such as quantitative immunochemistry assays requiring multiple steps and reagents.


Journal of Laboratory Automation | 2014

A Combined Fabrication and Instrumentation Platform for Sample Preparation

David J. Guckenberger; Peter C. Thomas; Jacob Rothbauer; Alex J. LaVanway; Meghan A. Anderson; Dan Gilson; Kevin Fawcett; Tristan Berto; Kevin Barrett; David J. Beebe; Scott M. Berry

While potentially powerful, access to molecular diagnostics is substantially limited in the developing world. Here we present an approach to reduced cost molecular diagnostic instrumentation that has the potential to empower developing world communities by reducing costs through streamlining the sample preparation process. In addition, this instrument is capable of producing its own consumable devices on demand, reducing reliance on assay suppliers. Furthermore, this instrument is designed with an “open” architecture, allowing users to visually observe the assay process and make modifications as necessary (as opposed to traditional “black box” systems). This open environment enables integration of microfluidic fabrication and viral RNA purification onto an easy-to-use modular system via the use of interchangeable trays. Here we employ this system to develop a protocol to fabricate microfluidic devices and then use these devices to isolate viral RNA from serum for the measurement of human immunodeficiency virus (HIV) viral load. Results obtained from this method show significantly reduced error compared with similar nonautomated sample preparation processes.


Analytical Chemistry | 2016

Magnetic System for Automated Manipulation of Paramagnetic Particles.

David J. Guckenberger; Hannah M. Pezzi; Mary C. Regier; Scott M. Berry; Kevin Fawcett; Kevin Barrett; David J. Beebe

The simple, rapid magnetic manipulation of paramagnetic particles (PMPs) paired with the wide range of available surface chemistries has strongly positioned PMPs in the field of analyte isolation. One recent technology, sliding lid for immobilized droplet extractions (SLIDE), presents a simple, rapid alternative to traditional PMP isolation protocols. Rather than remove fluid from PMP-bound analyte, SLIDE directly removes the PMPs from the fluid. SLIDE collects the PMPs on a hydrophobic, removable surface, which allows PMPs to be captured from one well and then transferred and released into a second well. Despite several key advantages, SLIDE remains limited by its passive magnetic manipulation that only allows for a one-time capture-and-release of PMPs, preventing wash steps and limiting purity. Furthermore, the strategy employed by SLIDE constrains the position of the wells, thereby limiting throughput and integration into automated systems. Here, we introduce a new, mechanically and operationally simplistic magnetic manipulation system for integration with the SLIDE technology to overcome the previously stated limitations. This magnetic system is compatible with nearly any plate design, can be integrated into automated workflows, enables high-throughput formats, simplifies mechanical requirements, and is amenable to a range of analytes. Using this magnetic system, PMPs can be collected, released, and resuspended throughout multiple wells regardless of proximity. We demonstrate this systems capabilities to isolate whole cells, mRNA, and DNA, demonstrating up to a 28-fold improvement of purity via the multiwash protocols enabled by this magnetic technology.

Collaboration


Dive into the David J. Guckenberger's collaboration.

Top Co-Authors

Avatar

David J. Beebe

Wisconsin Alumni Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Scott M. Berry

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Erwin Berthier

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Hannah M. Pezzi

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Joshua M. Lang

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Alex J. LaVanway

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Anna Huttenlocher

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jamie M. Sperger

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin P. Casavant

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge