Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Anal Chem ; 88(20): 9902-9907, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27598856

ABSTRACT

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 system's 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.

2.
Anal Chem ; 86(13): 6355-62, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24927449

ABSTRACT

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.


Subject(s)
Cell Separation/instrumentation , Chemical Fractionation/instrumentation , DNA/isolation & purification , Magnets/chemistry , Proteins/isolation & purification , RNA, Viral/isolation & purification , Animals , Cell Line , Equipment Design , Green Fluorescent Proteins/isolation & purification , HIV/genetics , HIV/isolation & purification , Humans , Luminescent Proteins/isolation & purification , RNA, Viral/genetics , Red Fluorescent Protein
3.
Anal Chem ; 86(15): 7258-63, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25032783

ABSTRACT

We present a simple method, called fluorescence-based assessment of plasma-induced hydrophilicity (FAPH), that enables spatial mapping of the local hydrophilicity of surfaces normally inaccessible by traditional contact angle measurement techniques. The method leverages the change in fluorescence of a dye, Nile Red, which is adsorbed on an oxygen plasma-treated surface, and its correlation with the contact angle of water. Using FAPH, we explored the effect of microchannel geometries on the penetration distance of oxygen plasma into a microchannel and found that entrance effects prevent uniform treatment. We showed that these variations have a significant impact on cell culture, and thus the design of cell-based microfluidic assays must consider this phenomenon to obtain repeatable and homogeneous results.


Subject(s)
Microfluidics/instrumentation , Oxazines/chemistry , Adsorption , Fluorescence , Hydrophobic and Hydrophilic Interactions , Plasma Gases
4.
Anal Chem ; 83(4): 1408-17, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21261280

ABSTRACT

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.


Subject(s)
Endothelial Cells/metabolism , Microfluidic Analytical Techniques/instrumentation , Microtechnology/methods , Polystyrenes/chemistry , Diffusion , Dimethylpolysiloxanes/chemistry , Epoxy Compounds/chemistry , Equipment Design , Humans , Printing , Reproducibility of Results , Surface Properties , Temperature , Time Factors
5.
SLAS Technol ; 26(1): 80-91, 2021 02.
Article in English | MEDLINE | ID: mdl-32552316

ABSTRACT

Over the past decade, induced pluripotent stem cells (iPSCs) have become a major focus of stem cell and developmental biology research, offering researchers a clinically relevant source of cells that are amenable to genetic engineering approaches. Though stem cells are promising for both research and commercial endeavors, iPSC-based assays require tedious protocols that include complex treatments, expensive reagents, and specialized equipment that limit their integration into academic curricula and cell biology research groups. Expanding on existing Kit-On-A-Lid-Assay (KOALA) technologies, we have developed a self-contained, injection molded, pipette-less iPSC culture and differentiation platform that significantly reduces associated costs and labor of stem cell maintenance and differentiation. The KOALA kit offers users the full range of iPSC culture necessities, including cell cryopreservation, media exchanges, differentiation, endpoint analysis, and a new capability, cell passaging. Using the KOALA kit, we were able to culture ~20,000 iPSCs per microchannel for at least 7 days, while maintaining stable expression of stemness markers (SSEA4 and Oct4) and normal iPSC phenotype. We also adapted protocols for differentiating iPSCs into neuroepithelial cells, cardiomyocytes, and definitive endodermal cells, a cell type from each germ layer of human development.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation , Culture Media , Humans , Lab-On-A-Chip Devices , Myocytes, Cardiac
6.
Lab Chip ; 18(3): 496-504, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29309079

ABSTRACT

Microscale cell-based assays have demonstrated unique capabilities in reproducing important cellular behaviors for diagnostics and basic biological research. As these assays move beyond the prototyping stage and into biological and clinical research environments, there is a need to produce microscale culture platforms more rapidly, cost-effectively, and reproducibly. 'Rapid' injection molding is poised to meet this need as it enables some of the benefits of traditional high volume injection molding at a fraction of the cost. However, rapid injection molding has limitations due to the material and methods used for mold fabrication. Here, we characterize advantages and limitations of rapid injection molding for microfluidic device fabrication through measurement of key features for cell culture applications including channel geometry, feature consistency, floor thickness, and surface polishing. We demonstrate phase contrast and fluorescence imaging of cells grown in rapid injection molded devices and provide design recommendations to successfully utilize rapid injection molding methods for microscale cell-based assay development in academic laboratory settings.


Subject(s)
Cell Culture Techniques/instrumentation , Computer-Aided Design , Equipment Design/methods , Microfluidic Analytical Techniques/instrumentation , Animals , Cattle , Cell Line , Microscopy, Fluorescence
7.
Lab Chip ; 18(22): 3446-3458, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30334061

ABSTRACT

Rare cell populations provide a patient-centric tool to monitor disease treatment, response, and resistance. However, understanding rare cells is a complex problem, which requires cell isolation/purification and downstream molecular interrogation - processes challenged by non-target populations, which vary patient-to-patient and change with disease. As such, cell isolation platforms must be amenable to a range of sample types while maintaining high efficiency and purity. The multiplexed technology for automated extraction (mTAE) is a versatile magnetic bead-based isolation platform that facilitates positive, negative, and combinatorial selection with integrated protein staining and nucleic acid isolation. mTAE is validated by isolating circulating tumor cells (CTCs) - a model rare cell population - from breast and prostate cancer patient samples. Negative selection yielded high efficiency capture of CTCs while positive selection yielded higher purity with an average of only 95 contaminant cells captured per milliliter of processed whole blood. With combinatorial selection, an overall increase in capture efficiency was observed, highlighting the potential significance of integrating multiple capture approaches on a single platform. Following capture (and staining), on platform nucleic acid extraction enabled the detection of androgen receptor-related transcripts from CTCs isolated from prostate cancer patients. The flexibility (e.g. negative, positive, combinatorial selection) and capabilities (e.g. isolation, protein staining, and nucleic acid extraction) of mTAE enable users to freely interrogate specific cell populations, a capability required to understand the potential of emerging rare cell populations and readily adapt to the heterogeneity presented across clinical samples.


Subject(s)
Cell Separation/instrumentation , Analytic Sample Preparation Methods , Cell Line , Equipment Design , Humans , Neoplastic Cells, Circulating/pathology , Systems Integration
8.
SLAS Technol ; 22(3): 348-357, 2017 06.
Article in English | MEDLINE | ID: mdl-28298147

ABSTRACT

Although average survival rates for lung cancer have improved, earlier and better diagnosis remains a priority. One promising approach to assisting earlier and safer diagnosis of lung lesions is bronchoalveolar lavage (BAL), which provides a sample of lung tissue as well as proteins and immune cells from the vicinity of the lesion, yet diagnostic sensitivity remains a challenge. Reproducible isolation of lung epithelia and multianalyte extraction have the potential to improve diagnostic sensitivity and provide new information for developing personalized therapeutic approaches. We present the use of a recently developed exclusion-based, solid-phase-extraction technique called SLIDE (Sliding Lid for Immobilized Droplet Extraction) to facilitate analysis of BAL samples. We developed a SLIDE protocol for lung epithelial cell extraction and biomarker staining of patient BALs, testing both EpCAM and Trop2 as capture antigens. We characterized captured cells using TTF1 and p40 as immunostaining biomarkers of adenocarcinoma and squamous cell carcinoma, respectively. We achieved up to 90% (EpCAM) and 84% (Trop2) extraction efficiency of representative tumor cell lines. We then used the platform to process two patient BAL samples in parallel within the same sample plate to demonstrate feasibility and observed that Trop2-based extraction potentially extracts more target cells than EpCAM-based extraction.


Subject(s)
Bronchoalveolar Lavage Fluid/cytology , Epithelial Cells/chemistry , Immunohistochemistry/methods , Lung Neoplasms/diagnosis , Specimen Handling/methods , Biomarkers, Tumor/analysis , Cell Line, Tumor , Humans , Immunohistochemistry/instrumentation , Specimen Handling/instrumentation
9.
Clin Cancer Res ; 23(3): 746-756, 2017 02.
Article in English | MEDLINE | ID: mdl-27401243

ABSTRACT

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; 1-11. ©2016 AACR.

10.
Mol Biol Cell ; 27(13): 1981-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27146113

ABSTRACT

Aneuploidy, an abnormal chromosome number that deviates from a multiple of the haploid, has been recognized as a common feature of cancers for >100 yr. Previously, we showed that the rate of chromosome missegregation/chromosomal instability (CIN) determines the effect of aneuploidy on tumors; whereas low rates of CIN are weakly tumor promoting, higher rates of CIN cause cell death and tumor suppression. However, whether high CIN inhibits tumor initiation or suppresses the growth and progression of already initiated tumors remained unclear. We tested this using the Apc(Min/+) mouse intestinal tumor model, in which effects on tumor initiation versus progression can be discriminated. Apc(Min/+) cells exhibit low CIN, and we generated high CIN by reducing expression of the kinesin-like mitotic motor protein CENP-E. CENP-E(+/-);Apc(Min/+) doubly heterozygous cells had higher rates of chromosome missegregation than singly heterozygous cells, resulting in increased cell death and a substantial reduction in tumor progression compared with Apc(Min/+) animals. Intestinal organoid studies confirmed that high CIN does not inhibit tumor cell initiation but does inhibit subsequent cell growth. These findings support the conclusion that increasing the rate of chromosome missegregation could serve as a successful chemotherapeutic strategy.


Subject(s)
Chromosome Segregation/genetics , Chromosome Segregation/physiology , Neoplasms/metabolism , Aneuploidy , Animals , Cell Death , Cell Line, Tumor/metabolism , Cell Transformation, Neoplastic/genetics , Chromosomal Instability/genetics , Chromosomal Instability/physiology , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes , Colorectal Neoplasms/metabolism , Kinesins/genetics , Mice , Mice, Inbred C57BL , Mitosis , Neoplasms/genetics , Spindle Apparatus/metabolism
11.
ACS Appl Mater Interfaces ; 8(24): 15040-5, 2016 Jun 22.
Article in English | MEDLINE | ID: mdl-27249333

ABSTRACT

Analyte isolation is an important process that spans a range of biomedical disciplines, including diagnostics, research, and forensics. While downstream analytical techniques have advanced in terms of both capability and throughput, analyte isolation technology has lagged behind, increasingly becoming the bottleneck in these processes. Thus, there exists a need for simple, fast, and easy to integrate analyte separation protocols to alleviate this bottleneck. Recently, a new class of technologies has emerged that leverages the movement of paramagnetic particle (PMP)-bound analytes through phase barriers to achieve a high efficiency separation in a single or a few steps. Specifically, the passage of a PMP/analyte aggregate through a phase interface (aqueous/air in this case) acts to efficiently "exclude" unbound (contaminant) material from PMP-bound analytes with higher efficiency than traditional washing-based solid-phase extraction (SPE) protocols (i.e., bind, wash several times, elute). Here, we describe for the first time a new type of "exclusion-based" sample preparation, which we term "AirJump". Upon realizing that much of the contaminant carryover stems from interactions with the sample vessel surface (e.g., pipetting residue, wetting), we aim to eliminate the influence of that factor. Thus, AirJump isolates PMP-bound analyte by "jumping" analyte directly out of a free liquid/air interface. Through careful characterization, we have demonstrated the validity of AirJump isolation through comparison to traditional washing-based isolations. Additionally, we have confirmed the suitability of AirJump in three important independent biological isolations, including protein immunoprecipitation, viral RNA isolation, and cell culture gene expression analysis. Taken together, these data sets demonstrate that AirJump performs efficiently, with high analyte yield, high purity, no cross contamination, rapid time-to-isolation, and excellent reproducibility.


Subject(s)
Solid Phase Extraction/methods , Gene Expression Profiling , Immunoprecipitation , Proteins/isolation & purification , RNA, Viral/isolation & purification , Reproducibility of Results
12.
PLoS One ; 11(7): e0159397, 2016.
Article in English | MEDLINE | ID: mdl-27459545

ABSTRACT

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.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Neoplastic Cells, Circulating/metabolism , Humans , Immunophenotyping/methods , Immunophenotyping/standards , Neoplasm Metastasis , Neoplasm Staging , Reproducibility of Results , Sensitivity and Specificity
13.
J Lab Autom ; 20(2): 146-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25424385

ABSTRACT

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.


Subject(s)
Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/instrumentation , High-Throughput Screening Assays/methods , Microfluidics/instrumentation , Microfluidics/methods , Immunochemistry/instrumentation , Immunochemistry/methods
14.
Lab Chip ; 15(11): 2364-78, 2015 Jun 07.
Article in English | MEDLINE | ID: mdl-25906246

ABSTRACT

This tutorial review offers protocols, tips, insight, and considerations for practitioners interested in using micromilling to create microfluidic devices. The objective is to provide a potential user with information to guide them on whether micromilling would fill a specific need within their overall fabrication strategy. Comparisons are made between micromilling and other common fabrication methods for plastics in terms of technical capabilities and cost. The main discussion focuses on "how-to" aspects of micromilling, to enable a user to select proper equipment and tools, and obtain usable microfluidic parts with minimal start-up time and effort. The supplementary information provides more extensive discussion on CNC mill setup, alignment, and programming. We aim to reach an audience with minimal prior experience in milling, but with strong interests in fabrication of microfluidic devices.


Subject(s)
Equipment Design/methods , Microfluidic Analytical Techniques/instrumentation , Microtechnology/methods , Plastics
15.
PLoS One ; 10(12): e0143631, 2015.
Article in English | MEDLINE | ID: mdl-26630135

ABSTRACT

Viral load (VL) measurements are critical to the proper management of HIV in developing countries. However, access to VL assays is limited by the high cost and complexity of existing assays. While there is a need for low cost VL assays, performance must not be compromised. Thus, new assays must be validated on metrics of limit of detection (LOD), accuracy, and dynamic range. Patient plasma samples from the Joint Clinical Research Centre in Uganda were de-identified and measured using both an existing VL assay (Abbott RealTime HIV-1) and our assay, which combines low cost reagents with a simplified method of RNA isolation termed Exclusion-Based Sample Preparation (ESP).71 patient samples with VLs ranging from <40 to >3,000,000 copies/mL were used to compare the two methods. We demonstrated equivalent LOD (~50 copies/mL) and high accuracy (average difference between methods of 0.08 log, R2 = 0.97). Using expenditures from this trial, we estimate that the cost of the reagents and consumables for this assay to be approximately $5 USD. As cost is a significant barrier to implementation of VL testing, we anticipate that our assay will enhance access to this critical monitoring test in developing countries.


Subject(s)
HIV Infections/economics , HIV Infections/virology , HIV-1/genetics , Molecular Diagnostic Techniques/economics , Serologic Tests/economics , Specimen Handling/economics , Viral Load , HIV Infections/blood , HIV Seropositivity , HIV-1/classification , HIV-1/isolation & purification , Humans , Molecular Diagnostic Techniques/methods , RNA, Messenger/genetics , Reagent Kits, Diagnostic , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Serologic Tests/methods
16.
J Mol Diagn ; 16(3): 297-304, 2014 May.
Article in English | MEDLINE | ID: mdl-24613822

ABSTRACT

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.


Subject(s)
Filtration/instrumentation , HIV Infections/diagnosis , HIV/isolation & purification , RNA, Viral/isolation & purification , Base Sequence , Equipment Design , Filtration/methods , HIV/genetics , HIV Infections/virology , Humans , Limit of Detection , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Surface Tension , Viral Load
17.
J Lab Autom ; 19(3): 267-74, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24401822

ABSTRACT

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.


Subject(s)
Analytic Sample Preparation Methods/instrumentation , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Molecular Diagnostic Techniques/instrumentation , RNA, Viral/analysis , Robotics/instrumentation , Analytic Sample Preparation Methods/economics , Benchmarking , Diagnostic Errors/prevention & control , HIV/isolation & purification , HIV/metabolism , HIV Infections/blood , HIV Infections/diagnosis , HIV Infections/economics , HIV Infections/virology , Health Care Costs , Humans , Lab-On-A-Chip Devices/economics , Microfluidic Analytical Techniques/economics , Molecular Diagnostic Techniques/economics , Proof of Concept Study , RNA, Viral/blood , RNA, Viral/isolation & purification , RNA, Viral/metabolism , Real-Time Polymerase Chain Reaction/economics , Real-Time Polymerase Chain Reaction/instrumentation , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/economics , Reverse Transcriptase Polymerase Chain Reaction/instrumentation , Robotics/economics , Viral Load , Waxes/chemistry
18.
Lab Chip ; 13(3): 424-31, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23229806

ABSTRACT

Microscale methods for cell-based assays typically rely on macroscopic reagent handling and fluidic loading protocols that are technically challenging and do not scale with the number of assays favorably. Here, we demonstrate a microfluidic platform technology called "Kit-On-A-Lid-Assay" (KOALA), that enables the creation of self-contained microfluidic cell-based assays, integrating all the steps required to perform cell-based assays. The KOALA platform allows the pre-packaging of reagents, cryopreservation of cell suspensions, thawing of cell suspensions, culture of cells, and operation of whole cell-based assays. The operation of the KOALA platform is user-friendly and consists of bringing together a lid containing the microchannels, and a base containing the pre-packaged reagents, thereby causing fluidic exchange in all the channels simultaneously. We demonstrate that the KOALA cell-based assays can be simply operated from start to finish without any external laboratory equipment.


Subject(s)
Biological Assay/instrumentation , Microfluidics/instrumentation , 3T3 Cells , Animals , Biological Assay/methods , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cryopreservation , Equipment Design , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunochemistry , Mice , Microfluidics/methods
19.
Lab Chip ; 13(3): 391-6, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23223939

ABSTRACT

Isolation and characterization of a specific subset of cells from a large heterogeneous population is necessary for studying rare subpopulations of cells. Existing methods require transfer or wash steps that risk causing loss of the rare cell population of interest. Integrated methods reduce loss, making these methods especially useful for reliable isolation of rare cell populations. In this report, we demonstrate the VerIFAST, a device that builds upon the simplified workflow of the Immiscible Filtration Assisted by Surface Tension (IFAST) to integrate a method for cellular isolation with methods for extra- and intracellular staining. First, a front-end purification step allows cells and unwanted particulates to passively settle out of the operational path of the paramagnetic particles, resulting in good efficiency of capture (>80%) and purity (>70%) with a single virtual wall traverse. Second, a Sieve Chamber is used downstream of the isolation chamber that removes excess unbound paramagnetic particles (PMPs) and performs complex multi-step washing procedures without centrifugation or transfer steps. Further, cellular staining can be performed in the device and is demonstrated for extracellular epithelial cell adhesion molecule (EpCAM), intracellular pan-cytokeratins, and Ki-67.


Subject(s)
Cell Separation/methods , Filtration/instrumentation , Staining and Labeling/methods , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Equipment Design , Filtration/methods , Flow Cytometry , Humans , Immunohistochemistry , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Surface Tension
20.
Lab Chip ; 12(13): 2317-21, 2012 Jul 07.
Article in English | MEDLINE | ID: mdl-22592853

ABSTRACT

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.


Subject(s)
Microfluidic Analytical Techniques/instrumentation , Oxygen/chemistry , Dimethylpolysiloxanes/chemistry , Hydrophobic and Hydrophilic Interactions , Polystyrenes/chemistry , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL