Abstract
Shared research resource facilities, also known as core laboratories (Cores), are responsible for generating a significant and growing portion of the research data in academic biomedical research institutions. Cores represent a central repository for institutional knowledge management, with deep expertise in the strengths and limitations of technology and its applications. They inherently support transparency and scientific reproducibility by protecting against cognitive bias in research design and data analysis, and they have institutional responsibility for the conduct of research (research ethics, regulatory compliance, and financial accountability) performed in their Cores. The Association of Biomolecular Resource Facilities (ABRF) is a FASEB-member scientific society whose members are scientists and administrators that manage or support Cores. The ABRF Research Groups (RGs), representing expertise for an array of cutting-edge and established technology platforms, perform multicenter research studies to determine and communicate best practices and community-based standards. This review provides a summary of the contributions of the ABRF RGs to promote scientific rigor and reproducibility in Cores from the published literature, ABRF meetings, and ABRF RGs communications.
Keywords: shared resource, core laboratories, multicenter research studies, collaborative research, community based standards
INTRODUCTION
Advances in biomedical research–an increasingly complex collaboration of both basic and clinical science–are driven by improvements, innovations, and breakthroughs in technology. The application of technology requires significant expertise, as well as implementation of best practices for scientific rigor and transparency, and the acquisition of equipment, instruments, and reagents to make it possible.1−4 This increasing sophistication, combined with significant financial investment, has been the impetus for the growth of Shared Research Resource Laboratories (Cores) in academia, government, and industry.5−7 Shared research resources are highly valued for making efficient use of research funds and broadening access to advanced technologies.8−11 Working with Cores, the research community can more effectively promote rigorous research practices, quality technical training, and collaborative research.6 The Association of Biomolecular Resource Facilities (ABRF) is an international scientific society dedicated to advancing shared research resource core laboratories through research, communication, and education.12 ABRF represents over 700 members from 19 countries working within or in support of Cores in government, academia, research, industry, and commercial settings, representing a collaborative knowledge base to advance the adoption of more rigorous, reproducible, and transparent research practices.
Recently, well-publicized allegations of the inability to reproduce published biomedical research13−16 have elicited discussions and examination within the research community and among public stakeholders.17−21 Federal agencies that support research activities, including the U.S. National Institutes of Health (NIH) and the National Science Foundation, have implemented new policies to address these concerns and improve communication of critical experimental details within the research community and to the public.22−24 The National Academy of Sciences also released guidelines on Research Reproducibility and Replicability in Science.25 In response, the ABRF Committee for Core Rigor and Reproducibility conducted a survey to gain information on how NIH initiatives on advancing scientific rigor and reproducibility influenced current shared resource services and new technology development and to identify the challenges and opportunities related to implementation of new reporting requirements.26 The survey results supported that core scientists are deeply invested in supporting transparency and scientific reproducibility. By employing best practices, protecting against cognitive bias in research design, employing full transparency in reporting experimental details and authentication of key resources, Cores directly addresses 2 of the 4 areas of improvement identified by the NIH.22, 26 As responsible stewards for the majority of research data produced at many institutions, with 94% of all core scientists trained in the operation and performance of instrument and equipment in their laboratories, Cores have a fundamental role in scientific excellence.5−7, 26 Recognizing this, federal granting agencies have already made significant investments in Cores through direct and indirect mechanisms, with the goal of providing cutting-edge technologies and expertise to individual scientific investigators.27
Collectively, and synergized through membership in ABRF, core scientists contribute to best practices, data reproducibility, and consensus guidelines for standard operating procedures, data standards, and quality analysis and quality control (QC).28, 29 ABRF was founded on these premises,30 its mission to both define and support best practices and scientific excellence and reproducibility in biomedical discovery through research, communication, and education. The hallmarks of ABRF, distinguishing it from all other professional scientific societies, are the ABRF-sponsored multisite Research Group (RG) studies. RG study participants prepare more effectively for change in the analytical environment and stay current with best practices for scientific rigor and reproducibility. In conjunction with the ABRF web-based discussion forum (http://list.abrf.org/), both contribute to the education of resource core directors, scientists, administrators, customers, and interested members of the scientific community.
Because scientific research advances are driven by the temporal nature of technology, scientific rigor is the foundation that drives data quality. Reviewed here are the ABRF RG contributions and current efforts in supporting these tenets, spanning protein primary structural analysis to single-cell transcriptomics.
ABRF RGs
The concept of developing test samples for Cores to use for methodology and proficiency assessment was initiated in 1986 at the Sixth International Conference on Methods in Protein Sequence Analysis Research Resource Facility Satellite Meeting (which later evolved formally into what we now know as the ABRF). A group of attendee scientists with an interest in assessing the reproducibility and rigor among laboratories performing Edman sequencing and amino acid analysis distributed test samples to 103 laboratories, thus establishing the first ABRF RG.31 Today, the ABRF RGs are organized by ABRF members who are experts in their respective disciplines and who design studies relevant to the prevailing state of the technology to inform scientific rigor in research performed across national and international academic, government, and industry laboratories. These objectives are met by designing and distributing study materials through several means: 1) provide a protocol for self-evaluation of scientific accuracy and procedural efficiency; 2) contribute improvements to instrumentation, methods, and instrument operation/maintenance by identifying limitations or common problems encountered in laboratories in the field; 3) determine best practices for standard operating procedures, and 4) define realistic expectations for both core scientists and users.32 Through participation in the multisite RG studies, participants are more effective at addressing inevitable changes, improvements, and upgrades in the analytical environment and remain current with best practices for scientific rigor and reproducibility.33
The first decade of ABRF RG studies focused on the advancement of technologies and education of the scientific community on the best methods and strategies for the hot topic of research focus at the time—protein primary structure and function analysis. These early RGs [Edman Sequencing RG (ESRG), Protein Sequencing RG (PSRG), and related Internal Sequencing RG studies] provided rich detail on the scientific rigor demanded for the cutting-edge technologies of this era.34−53 A critical adjunct to the ESRG was the Proteomic Standards RG (sPRG), which focused on designing and producing peptide standards.54−57 The sPRG produced 3 synthetic peptides that were registered and certified as reference standards, which is important work done in collaboration with the National Institute of Standards and Technology (NIST). A significant asset to the scientific community at large, these peptides are commercially available.58
A cornerstone of quantitative protein characterization, amino acid analysis was an early methodology and technology focus for ABRF. The amino acid analysis RG members detailed advances in the detection and sensitivity of amino acid analysis, tracing technology platform evolution, and ion-based chromatographic methods to mass spectrometry analyses.59−74 And the PSRG was instrumental in bringing improvements to synthetic chemistry methods, most significantly leading efforts for research and methods for introducing fluorenylmethyloxycarbonyl chloride amino acid chemistry (FMOC) to the forefront.75−90
Later RG efforts and publications focused on determining best practices for analyzing post-translational modifications (Carbohydrate Analysis RG, Glycoprotein RG)91, 92 and establishing quality measurement standards for analyzing molecular interactions (protein-protein; protein-ligand) (Molecular Interactions RG).93−97
Individually and as a valuable resource in aggregate, the ABRF RG studies remain highly referenced for technology applications and methodology. New studies continue, driven by new technologies and the need for robust methods and protocols and building on the success of the early RGs. The current ABRF RGs encompass 4 broad, technology-based categories integral to modern bioresearch: 1) genomics, 2) bioinformatics, 3) mass spectrometry (proteomics, metabolomics), and 4) imaging (microscopy and flow cytometry) and are reviewed here.
RESULTS
Genomics
The field of genomics has exploded over the past 40 yr, with the rapid development of next-generation technologies spawning multiple generations of sequencing platforms, each improving on cost, speed, and data quality, even portability, as exemplified by recent sequencing data performed on the International Space Station.98 Although these technology advances have significantly impacted our understanding of biology and disease, the sheer volume of new data that these technologies produce have created tremendous challenges related to data sharing and data management. As a consequence, standardized procedures and data formats as well as comprehensive quality management considerations are at the forefront.99
The current Genomics RGs (GRGs) represent 5 overlapping subspecialties representative of critical genomics based technology and applications: 1) next-generation sequencing (NGS), 2) Genome Editing RG (GERG), 3) DNA Sequencing RG (DSRG), 4) GRG, and 5) Metagenomics RG (MGRG). The predecessor genomics-related RGs researched methods and applications for microarrays, fragment analysis genotyping, and nucleic acid analysis [Nucleic Acids RG (NARG)], finally merging to form the current GRG. The mission of all the genomics-related RGs is to analyze, establish, and disseminate standards and best practices for Cores in the fast-evolving field of genomics technologies and applications.
Next-Gen Sequencing RG
The NGSRG is dedicated to educating ABRF members and scientific community in support of scientific rigor and data reproducibility, identifying optimal methods and strategies for NGS projects as well as performance evaluation of the ever-evolving NGS platforms. To that end, the 2018 ABRF-NGS study was a coordinated multi-RG effort by 4 RGs [NGSRG, DSRG, GRG, and Genomics Bioinformatics RG (GBIRG)] and over 20 Cores. The long-term goals of the study are to optimize the detection of genetic variation with the latest sequencing tools, establish a community resource for self-evaluation and self-improvement, and improve sequencing performance while evaluating existing and emerging protocols and platforms. Previous RG studies focused on RNA sequencing (RNA-seq), using standard Microarray Quality Control Consortium (MAQC) total RNA samples combined with External RNA Controls Consortium synthetic spike-in RNA.100, 101 This RNA profiling phase of the ABRF-NGS Study included the Illumina HiSeq 2000/2500 and MiSeq (Illumina, San Diego, CA), Roche 454 GS FLX+ (Roche, Basel, Switzerland), Life Technologies Ion Torrent PGM and Proton (Thermo Fisher Scientific, Waltham, MA, USA), and Pacific Biosciences PacBio RS (Pacific Biosciences, Menlo Park, CA) platforms. The Phase 1 ABRF-NGS Study (2012–2015) assessed sequencing accuracy, absolute and relative expression levels, RNA splice junction detection, and differential expression detection between samples. In a similar fashion, the current Phase 2 ABRF-NGS Study (2016–2020), performed in collaboration with the NIST Genome in a Bottle Consortium,102 the U.S. Food and Drug Administration’s Sequencing Quality Control Consortium103 and other sequencing community stakeholders will utilize standardized reference genomic DNA samples.
High-throughput RNA-seq greatly expands the potential for genomics discoveries, but the wide variety of platforms, protocols, and performance capabilities has created the need for comprehensive reference data. Here we describe the ABRF-NGS study on RNA-seq: Replicate experiments across 15 laboratory sites were performed using reference RNA standards to test 4 protocols (poly-A–selected, ribo-depleted, size-selected, and degraded) across 5 sequencing platforms (Illumina HiSeq, Life Technologies PGM and Proton, Pacific Biosciences RS, and Roche 454).104 The results showed high intraplatform (Spearman rank R > 0.86) and interplatform (R > 0.83) concordance for expression measures across the deep-count platforms but highly variable efficiency and cost for splice junction and variant detection between all platforms. For intact RNA, gene expression profiles from ribosomal ribonucleic acid (rRNA) depletion and poly-A enrichment were similar. In addition, rRNA depletion enabled effective analysis of degraded RNA samples, new approaches for correcting batch effects in large-scale studies105 and more efficient detection of splice isoforms.106 These RG studies provide a broad foundation for cross-platform standardization, evaluation, and improvement of RNA-seq107, 108 and have led to other standards in genomics, metagenomics, and multiomics.109
Genome Editing RG
With increasing demand for CRISPR/Cas9 technology, new or existing Cores have adapted their services to fit this technology into their workflows. In the GERG 2017 survey,110 plasmid format and lipofection delivery were favored among cell culture users. The 2018 GERG study evaluated the reproducibility of indel formation rates by comparing guide RNA format and delivery methods across participating labs. Various configurations of guide RNA and Cas9 components can be used for editing cells, including 1) a plasmid expressing both the guide RNA and Cas9, 2) Cas9 protein combined with a synthetic single guide RNA, and 3) Cas9 combined with a synthetic 2-part guide RNA. The study evaluated the cutting efficiency at 3 different guide RNA targets based on the guide RNA format using 2 delivery methods (lipofection or nucleofection transfection methods). The study results suggest the ribonucleoprotein (RNP) format for the guide RNA and Cas9 is gaining in popularity in combination with nucleofection delivery. The GERG RG is finalizing the results of the study to determine the most reproducible method (Sergison, E., Regan, M., Delventhal, K., Gurumurthy, C., Kmiec, E., Pruett-Miller, S., Dahlem, T., Marsischky, G., unpublished results).
DNA Sequencing RG
The DSRG conducts studies to assess the capabilities of DNA sequencing technologies, protocols, kits, and reagents; provides a means of self-evaluation for sequencing technologies to evaluate their own performance; disseminate the findings while still relevant; and promote communication and cooperation among laboratories that perform Sanger sequencing and NGS. The ABRF NARG, a precursor to the DSRG, first conducted a comparison of DNA sequencing technologies in 1995.111–113 In the late 1990s, the ABRF DSRG established a series of long-term studies, called in aggregate The Never Ending Story, designed to evaluate the performance of Sanger sequencing instruments.108 The DSRG 1997 study involved sending a guanine-cytosine (GC)-rich DNA template to Cores that offered DNA sequencing as a service.114 The objective was to evaluate whether chemical additives, altered thermocycling conditions, and analysis methods could improve the sequence obtained from a GC-rich template. A total of 134 Cores participated in this study. The study demonstrated that manual review and editing of data generated by automated sequencing instruments had the most impact in improving sequencing accuracy, using the automated analysis algorithms available at that time. Continuation of this study resulted in definition and dissemination of best practices for sequencing high GC templates.115
The second in The Never Ending Story series assessed the state-of-the-art in DNA sequencing to create a publically available quality control resource. Sequencing groups submitted unedited sequence for a common standard template [i.e., bacterial plasmid cloning vector pGEM template with the M13 (−20) forward primer] using the instruments, reagents, and protocols common in their core and also included all metadata (e.g., chemistry type and concentration, instrument type and run conditions, and analysis algorithms and methods). Data were collected electronically and analyzed based on sequencing accuracy, read length, signal sensitivity, enzyme dilution tolerance, and ease of use. The aim of establishing this readily updatable, online benchmark resource for self-evaluation was to promote high standards and reproducible results for DNA sequencing in core laboratories. More than 300 Cores participated in this study.116
The Never Ending Story continued with DSRG studies in 1999, 2000, and 2001, evaluating the outcomes of different DNA sequencing methods on sequencing both standard and difficult templates to expand the web-based quality control resource and produce a defined standard test array of difficult-to-sequence templates.117 The Never Ending Story continued in 2003, 2005, 2006, and 2008, with DSRG studies that evaluated and defined best practices for sequencing through difficult-to-sequence DNA templates containing extensive repetitive sequences.118 In 2019, The Never Ending Story was rebooted with a DSRG study on Sanger-based sequencing best practices in response to the release of new Sanger sequencing dye terminator chemistries. This study was a cross-site evaluation of Sanger sequencing chemistries, designed to evaluate the performance of sequencing both standard and difficult-to-sequence templates using a variety of legacy and new-to-market Sanger sequencing chemistries and provide guidance to Cores in regard to modifying existing sequencing protocols to further the quality and robustness of their Sanger sequencing production pipelines.119−129
Recent DSRG studies expanded the rigorous assessment of sequencing reagents to NGS modalities as well, with an eye toward implications for the selection of kits for specific experimental contexts. Multisite projects included the following: a robust evaluation of small RNA NGS library sequencing kits with respect to accurate representation of microRNAs (miRNA) differential expression, retention of small RNA and other noncoding RNAs (ncRNAs), technical replication, conservation of material, and ease of use across 11 different ABRF member sites (Herbert ZT, Thimmapuram J, Xie S, et al., unpublished results); an assessment of ribosomal reduction protocols for human/mouse/rat RNA upstream of NGS sequencing in order to determine effectiveness at retaining messenger RNA (mRNA) and ncRNA components of a total RNA specimen for sequencing while minimizing the intervening and noninformative rRNA and transfer RNA (tRNA) components130; and a comparison of methods designed for high throughput enzymatic fragmentation of DNA prior to next-gen library generation to assess resulting library complexity, bias in sequence and genomic context, and site-to-site consistency (DSRG posters and presentations). These and an array of RG studies with broad participation have driven improvements in quality, accuracy, and reproducibility for DNA and RNA sequencing provided by Cores.131−147
Genomics RG
The GRG members provide both academic and industrial scientists useful information and guidance in the use of various microarray, NGS, and other genomic platforms and applications in their research. Earlier RGs that focused on microarrays, fragment analysis genotyping, and nucleic acid analysis (NARG) have merged into the GRG. These RGs performed foundational studies on the evaluation and application of microarrays148−160 and fragment analysis.161, 162
More recent GRG studies have focused on single-cell genomics, a field that is rapidly evolving in both platform technology and analytical methodology. To assess variability and determine best practices across various leading platform technologies, the 2017 GRG study evaluated the key technologies for single-cell RNA-seq. The platforms chosen were Takara’s iCell8 (Mountain View, CA), Fluidigm’s C1 AutoPrep (South San Francisco, CA), 10X Genomics Chromium Controller (Pleasanton, CA), and Illumina/BioRad’s ddSEQ joint venture. A well-characterized triple-negative breast cancer cell line was distributed to multiple labs for analysis in duplicate across all platform technologies to assess correlation with bulk RNA-seq data, assess reproducibility, and evaluate concordance of expression results using mock vs. drug exposure. Results further understanding of the limitations of each technology, furthering more rigorous experimental design in platform selection (Fournier, C., Reyero-Vinas, N., Ashton, J., Jen, J., Boswell, S., Chittur, S., Mason, C., Rehrauer, H., Steen, R., unpublished results). The 2018 GRG study, performed in collaboration with the American Natural History Museum, assessed accuracy and reproducibility of several long-read sequencing platforms, evaluating PacBio Sequel, Oxford Nanopore Technologies GridION, 10X genomics linked read technology, and Illumina’s MatePair chemistry. An outcome goal of this study was the creation of a full genome reference of an endangered parrot for conservatory purposes (Fournier, C., Reyero-Vinas, N., Ashton, J., Jen, J., Boswell, S., Chittur, S, Mason, C., Rehrauer, H., Steen, R., unpublished results).
Metagenomics RG
The MGRG was created in response to the rapidly growing field of metagenomics by a team of scientists with backgrounds in microbiology, genetics and genomics, bioinformatics, oceanography, geochemistry, planetary sciences, climate research, and extremophile research.163
Early ABRF studies on metagenomics were performed by the DSRG and GRG.164 International reference samples used and characterized by the MGRG are used to assess the performance of impact of protocols like whole-genome amplification165 and help individual laboratories compare their local results with those of the larger research community or improve clinical implementation of “precision metagenomics”166. The tested, titrated bacteria/fungal mixtures are now continuously used in a wide range of environments167 for both genetic and epigenetic applications.168 Reference standards facilitate the development of much needed peripheral reagents including high-performance DNA extraction kits, complex enzyme mixes for microbial lysis, nucleic acid-free sample concentrators, and bioinformatic pipelines. Improvements in metagenomic methods will ultimately benefit from the availability of standardized reference samples that represent the range of organisms potentially present in samples from the field.163
The MGRG has initiated a novel microbiome project Extreme Microbiome Project169 to characterize organisms from extreme environments around the world. Further goals of the MGRG include assembling microbial standards and characterize shortcomings of current metagenomic techniques170 and comparison to older techniques in microbiome research like 16S rRNA sequencing171 including optimizing DNA extraction protocols, library synthesis methods for different NGS platforms and developing best approaches for bioinformatics. The RG also works very closely with the appropriate vendors of the field to help advance this technology for metagenomics while developing low-input RNA-seq to further enable metatranscriptomics.172, 173 The development of a bacterial counting platform (similar to simple low-cost mammalian cell counters), in partnership with specific vendors, is needed for research and clinical metagenomic quality control requirements. This enumeration device will be validated against standard techniques such as flow cytometry, microscopic, and light scattering chromatography (LSC) techniques.
Bioinformatics
Advances in the technologies and informatics used to generate and process large biologic data sets (“omics data”) are promoting a critical shift in the study of biomedical sciences and a need for interdisciplinary data integration strategies to support a better understanding of biologic systems. Analysis of genomics, transcriptomics, and proteomics data are still primarily analyzed individually with distinct approaches generating monothematic rather than integrated knowledge. Computational methods for data management, algorithms for statistical pattern inference and recognition, and data integration are necessary for the integrated or constructionist view of biology. Coupled with the increase in metabolomics, epigenomics, and pharmacogenomics data needs, the ABRF members have formed 2 RGs to exemplify and evaluate computational methods for bioinformatics (data exchange and management) and quantitative mathematical modeling to meet current needs in data analysis and sharing.
Genomics Bioinformatics RG
The goals of the GBIRG are to 1) provide collaborative bioinformatics and bio-information technology (bio-IT) support for ABRF genomics-focused RG studies, 2) explore collaborations with the ABRF proteomics bioinformatics focused RGs, and 3) investigate questions of interest to all genomics-focused bioinformatics and bio-IT Cores by creating surveys of bioinformatics and bio-IT Core management and funding models, conducting studies of computational biology analysis tools and data management methods issues, and identifying best practices. Most recently, GBIRG has supported a multiplatform assessment of transcriptome profiling using RNA-seq with the ABRF Next-generation Sequencing Study group.104
Proteome Informatics RG
The mission of the ABRF Proteome Informatics RG (iPRG) is to educate ABRF members and the scientific community on best application and practice of bioinformatics toward accurate and comprehensive analysis of proteomics data. The iPRG members actively support and participate in the development and advancement of new algorithms, software tools, and strategies for proteome informatics with the goal of both educating and introducing these technologies to the membership. The iPRG research studies174−178 have typically started by generating ground truth data sets to challenge and benchmark commonly used algorithms and statistical methods in proteomics. Several of these data sets have seen reuse independently from the iPRG study for which they were generated, demonstrating the lasting value of reference data from well-designed experiments.179–181 All past studies, including the data sets and their documentation, are archived and available on the ABRF website.182
Proteomics, Metabolomics, and Proteomics Standards RGs
Edman sequencing dominated the early RG studies, the only method at the time for determining the primary structure of proteins. With the advent of mass spectrometric instruments, methods evolved to provide rapid and sensitive qualitative and quantitative analysis of biomolecules (proteins, peptides, oligosaccharides, metabolites, lipids, DNA, and RNA). The PSRG and Protein Identification RG supplanted the ESRG.
Significant advances in mass spectrometry (MS) platform technology and informatics data analysis increased sensitivity and throughput, supplanting earlier chemistry-based technologies.183 The PSRG studies focused on N- and C-terminal sequence analysis of proteins by any technology yielding information about the termini of proteins.184−197
The current Proteomics, Metabolomics, and Proteomics Standards RGs studies are designed to examine reproducibility and other capabilities of emerging techniques to establish best practices for scientific excellence. The Mass Spectrometry RG, which preceded the current Proteomics, Glycoprotein, and Metabolomics RGs (MRGs), contributed the Delta Mass utility website still employed today (Delta Mass). However, our current knowledge that more than 21,000 human genes198 may code 1 million199 or more proteoforms leaves ample room for development.200 New methodologies, coupled with transcriptomics data and MS/MS peptide sequence analysis at the subpicomole level have enabled a multiomics approach to protein identification for biologic and disease studies within a given biologic sample. Recent advances in mass spectrometry have clearly revolutionized the studies of post-translational modifications and include the development of specific strategies to preferentially enrich modified amino acids via covalent modifications incorporating affinity tags.
Proteomics RG
The PRG is dedicated to sharing knowledge about the analysis of proteins in support of scientific rigor and data reproducibility. The 2018 PRG study is focused on enabling mass spectrometry–based proteomics laboratories to use Data-Independent Acquisition (DIA) technology. For this study, the PRG reached out to laboratories around the world. Sixty-four laboratories from 16 countries (and 20 U.S. states) were provided with a test sample set, protocols, and resources to facilitate the use of data independent acquisition (DIA). Forty-one participants (64% of sample recipients) deposited the raw data for PRG to analyze. Almost half of the participants in the study were new to DIA (Jagtap, P., Herring, L., Midha, M., Martin, R., Neely, B., Phinney, B., Shan, B., Stemmer, P., Wang, Y., unpublished results). The previous 2 studies conducted by the PRG were based on detection and quantitation of low-abundance proteins in a highly complex sample. In the 2016 PRG study, data submitted from study participants was used to measure intralaboratory variation in liquid chromatography-mass spectrometry (LC-MS) performance to determine the types of QC procedures implemented in proteomics laboratories and identify the elements of system design/setup that correlate with variability. The results showed variability in the identification of lower-abundance spiked-in proteins from different laboratories and demonstrated a significant advantage of performing fractionation on complex samples to detect proteins at an extremely low concentration. Unexpectedly, when identification files provided by the participants were reanalyzed for validation of self-reported values, significant differences in participant-reported and study-validated values were found.201
The 2017 PRG study was a member-only study on the quantification of unidentified low-abundance proteins with mass spectrometry spectra (MS1) data and bioinformatics tools. In this follow-up of the 2016 PRG study, retention time and accurate mass of peptides with relatively high concentration (500 fmol in 25 ug cell lysate) were employed to quantify the same peptides in samples with low concentration (20 fmol in 25 μg cell lysate). Four software programs (2 open source and 2 commercial) were used to analyze data set from Orbitrap Fusion, Q Exactive, and Orbitrap Velos instruments (Thermo Fisher Scientific). All evaluated software programs extracted quantitative information from MS1 spectra that did not yield peptide spectral matches in samples with low concentrations of spike-in proteins. False quantification of peptides in the zero spike-in sample was observed. This was attributed to carryover between runs and misassignment of noise in the signal.202
Proteomics Standards RG
The mission of the sPRG is to promote and support the development and use of standards in proteomics and committed to identifying and implementing technical standards for accuracy, clarity, and consistency, supporting ABRF’s commitment to scientific rigor and reproducibility. Examples of technical standards include, but are not limited to, reference materials, data sets, conditions, and procedures that give proteomics researchers and analysts’ independent criteria to evaluate their abilities to produce predictable, consistent results.54−58 The sPRG strongly supports ongoing efforts for standardization of the recording and reporting of proteomics experiments. The 2018 sPRG Study delivered a new heavy-labeled phosphopeptide standard and a designed study to test the ability of proteomics labs to detect endogenous phosphopeptides (Herren, A., Lee, K., Searle, B., Patel, B., Leib, R., Chien, A., Hawke, D., Koller, A., Isovev, G., Neeley, B., unpublished results).
Metabolomics RG
Metabolomics is the comprehensive profiling of metabolites and other small molecules. The large structural diversity of these compounds makes both comprehensive profiling and identification challenging. With nuclear magnetic resonance (NMR) and MS being the major platforms used, there are a variety of approaches, including untargeted profiling, targeted approaches, and fluxomics. Challenges include identification of metabolites, how they change in relation to a biologic perturbation (e.g., drug, diet, disease) and the biologic significance of these changes.
The goals of the MRG are 1) to educate research scientists and resource facilities in the analytical approaches and management of data resulting from comprehensive metabolite studies and 2) to promote the science and standardization of metabolomic analyses for a variety of applications.203 The 2013 MRG study assessed the ability of laboratories to conduct successful untargeted and targeted metabolomics analyses. The study sample was human plasma spiked with different amounts of metabolite standards in 2 groups to emulate a typical metabolomics study. Results from the 2013 MRG study highlighted current challenges in the field that include missing benchmarks for comparing different methodological approaches and analytical platforms to enable cross-laboratory reproducibility.204
The 2016 MRG study focused on metabolomics data analysis methods—bioinformatics and statistical approaches for pre- and postprocessing of global profiling data sets, and assessed interlaboratory reproducibility, a major concern that we tried to assess by providing study participants with already-acquired mass spectrometry data. The task was to detect feature level differences between 2 groups to shed light on the contribution of data pre- and postprocessing methods on metabolomics analysis results. A major conclusion from this study identifies proper data preprocessing as a critical step of the data analysis workflow when using untargeted metabolomics profiling.205
We believe that consolidation of validated metabolomics methodologies and benchmarking standards of use and reporting will further augment routine and widespread practice of this powerful and cost-effective technology with many applications in the life and biomedical sciences.
Light Microscopy and Flow Cytometry RGs
Two diverse technology areas focused on capturing images of cell populations, single cells, cells represented in tissues and organelles, entire organisms, and imaging at the molecular level (which may or may not be within a cell) are represented in the Light Microscopy RG (LMRG) and Flow Cytometry RG (FCRG). Few technologies are more widespread in modern biologic laboratories than optical imaging techniques using light emitted through fluorescence or bioluminescence, and new imaging techniques include optical coherence tomography, multiphoton microscopy, total internal reflection fluorescence provide ever-increasing ability to monitor biologic phenomena with higher resolution, specificity, dimensionality, complexity, and scale, all while maintaining viability and biologic relevance. These imaging modalities are increasingly multiparametric and rely heavily on computational approaches, which are in many cases nearly as important as the optics, not only for automating and optimizing image acquisition but also for visualizing and analyzing the data.
Light Microscopy RG
Relative to the other technologies represented by the ABRF, Light Microscopy and Flow Cytometry are unique in their involvement of the individual researcher and therefore unique in their challenges to reproducibility. Light Microscopy Cores typically train individual researchers who then use the core instruments independently to collect their data, in contrast to other types of Cores in which trained Core staff perform or oversee most data collection. Light Microscopy Core users may range widely in expertise, from novice undergraduate and graduate students to more highly experienced postdocs and staff scientists. Moreover, many research laboratories may have their own microscope systems (potentially including “advanced” instrumentation such as confocal and superresolution systems) and may conduct their entire study without ever setting foot in a Core. Finally, light microscopy is, by its nature, an interactive technology in which the user is intimately involved in the data collection process. Microscope users select which regions of a specimen to image and have expansive control over the appearance of those images. Experienced microscopists are well aware of the ethical pitfalls of having such control; equally important, however, are its impacts on reproducibility.206
Given the highly individualized and highly decentralized nature of light microscopy–based research, improving reproducibility necessitates providing both Core and individual microscope users with easily accessible standards for characterizing their instruments and protocols. The LMRG promotes scientific exchange between researchers using microscopy in Cores to increase our general knowledge and expertise and conducts multisite experiments to establish light microscopy standards. The first 2 LMRG studies focused on developing quality assurance tests for light microscopes, including the characterization of objective lenses, the accuracy of spectral detection and separation, illumination stability and uniformity, and characterization of the point spread function.207−210
A third LMRG study of microscope resolution, distortion, intensity quantification, and signal-to-noise as a function of depth in 3D is being finalized, with the goal of creating a 3D biologically relevant test slide and imaging protocol to assess 1) system resolution and distortions in 2D and 3D, 2) dependence of intensity quantification and image signal-to-noise of the microscope on imaging depth, and 3) dependence of the microscope sensitivity on imaging depth. The test sample consists of a mixture of fluorescence microspheres imbedded in a 120-um-thick layer of CyGel (Cy10500; BioStatus, Loughborough, United Kingdom) with a refractive index of 1.37 closely matched to biologic tissue. Double-sided adhesive 18-mm square spacers with a well (9-mm diameter, 120-μm deep) were used for the sample preparation (70327-8s; Electron Microscopy Sciences, Hatfield, PA, USA). The mixture of microspheres includes 1 μm Orange, 2.5 μm Green 20% brightness, 2.5 μm Green 100% brightness, 6 μm Far Red 36%, 6 μm Far Red, and 15 μm Blue Core/Orange Ring (Kubow, K., Abrams, B., Ammer, A., Arvanitis, C., Callahan, L., Cole, R., Dragavon, J., Itano, M., Mezzano, V., Pengo, T., Powers, J., Sanders, M., Wee, E., unpublished results).
All 3 LMRG studies have resulted in microscopy standards as well as easily accessible protocols for the characterization and verification of microscope systems. The next LMRG study will focus on reproducibility issues in image analysis. As with the acquisition of microscope images, their subsequent analysis would also typically be performed entirely by the end-user and therefore presents a major reproducibility challenge.
Flow Cytometry RG
The FCRG was formed with the goal of providing key information related to the art of flow cytometry, including its cross-technology applications to genomics, proteomics, and other Core-related areas.211, 212 Flow cytometry is a broad term describing the rapid measurement of large numbers of cells individually using light-scatter and fluorescence detection to analyze cell characteristics (size, intracellular pH, membrane potential) and intracellular cellular components (DNA, protein, calcium, cell surface receptors) and distinguish based on these parameters for isolating cell populations, critical for downstream analysis of cells. Mass cytometry has recently emerged as a form of flow cytometry where lanthanides are used as the label on cells that are measured in a time of flight mass cytometer. Cell sorting uses flow cytometry to separate cells based on protein expression labeled with fluorescent tags and has many downstream applications such as functional assays. In other experimental workflows, cells sorted will be assessed for gene expression, such as single-cell analysis (10X Genomics, Fluidigm C1). The first FCRG study investigated the impact of cell sorting on cell health using a range of instrument types and configurations across multiple sites, as assessed by cell viability, proliferation, and gene expression. Variables such as sorting vs. not sorting, high vs. low pressure, and the presence vs. absence of UV light were analyzed with RNA-seq and microarray in Jurkat cells, primary mouse B cells, and mouse ES cells. Information garnered through this multisite study provided guidelines for sorting cells upstream of other technologies including genomics and proteomics.213 A second study surveyed flow cytometry Cores for sorter cleaning practices; a subset of study participants submitted sheath fluid samples for testing. The study results showed that the majority of sorters had significant endotoxin contamination, little to no RNase, with bacterial concentrations quite variable. There was no correlation found between sorter cleanliness and any surveyed variables, including sorter age, cleaning practices, date of last preventive maintenance, sheath source, or known recent contamination. Because a number of sorters assayed in the sorter cleanliness study were contaminated with endotoxin, effectiveness of an H2O2 cleaning procedure was tested to assess removal of the endotoxin from the sorters. Sheath fluid samples collected before and after cleaning were tested with an Limulus amebocyte lysate (LAL) quantitation kit and it was determined that the contamination was only partially mitigated. Also, the endotoxin levels reached precleaning levels within a few weeks of the sterilization. The results of the second study will be published in combination with the endotoxin removal results (Thornton, S., Bowen, S., Bispo, C., Hassel, C., Abshari, M., Adams, D., Bergeron, A., Brundage, K., Cochran, M., Del Rio Guerra, R., Dwyer, K., Harley, R., Holmes, L., Loof, N., Meyer, M., Niziolek, Z., Saluk, A., unpublished results).
Increasingly, investigators ask Flow Cytometry Cores to sort fixed cells for RNA isolation either in bulk or at the single-cell level. The 2018 FCRG study performed a systematic evaluation of the reported fixation methods prior to sorting to determine the impact on the purity, quality, and RNA yield from sorted cells (Thornton, S., Bowen, S., Bispo, C., Hassel, C., Abshari, M., Adams, D., Bergeron, A., Brundage, K., Cochran, M., Del Rio Guerra, R., Dwyer, K., Harley, R., Holmes, L., Loof, N., Meyer, M., Niziolek, Z., Saluk, A., unpublished results).
Interest groups
The ABRF supports a fifth category of Interest Networks, which facilitate discussion and review for key technology areas of interest that may be considered for RG status. The Antibody Technology Interest Network is dedicated to sharing its collective knowledge about generating, producing, purifying, fragmenting, and conjugating antibodies, with ABRF members and within the larger scientific community. The Antibody Technology Interest Network holds online sessions and workshops to facilitate the discussion of antibody use and application, immunization strategies, fusion protocols, screening strategies, antibody production purification and labeling, antibody applications (e.g., Flow Cytometry, Microscopy, Chromatin immunoprecipitation combined with sequencing (ChIP-Seq), in vivo, in vitro applications), and laboratory organizational and fiscal structures.
CONCLUSIONS
The research enterprise includes numerous stakeholders: universities and other research institutions that educate, employ, and train researchers; the federal and industrial sponsors of research, journal, and book publishers; and scientific societies.1, 5, 6, 23 These stakeholders can act in ways that either support or undermine the integrity of research.23 As presented here, ABRF sponsorship of multicenter research studies promote community-based standards and provide a sustainable framework for sharing best practices in methodology, standard operating procedures, and data management.26 The ABRF RGs, led by highly trained scientists, provide an extremely valuable yet undervalued service to the research community by helping to provide clarity on approaches and best practices for experimental design and data analysis. The ABRF RGs collectively address enigmatic technology-based problems to answer complex biologic questions, ranging from determining best practices for cytometric cell sorting for optimal RNA extraction to partnering with instrumentation manufacturers to improve MS methods for protein and metabolome studies.
At an institutional level, Cores generate the majority of research data at many institutions so their role in maintaining needed expertise and generating quality data is considerable and represents a central repository for knowledge management, with deep expertise and knowledge of the strengths and limitations of the technology and applications.3−7 Core science inherently supports transparency and scientific reproducibility through unbiased acquisition, minimizing interoperable variability and promoting transparent processes and reporting (detailed experimental materials and methods) for publications and grants.5−7 Data provenance is assured—detailing who performed what experiment on which instrument, instrument standardization and maintenance, quality assurance/QC (required controls, standards, documentation and tracking of buffers, reagents, components, lot numbers, version, expiration dates), location of source data and shared data (curation in compliance with Data Storage Standards for Research Core Laboratories, OMB Circular A-110 and NIH GDS Policy)—and essential for research integrity.23−27 Recognizing this, federal granting agencies have already made significant investments in Cores through a variety of direct and indirect mechanisms, with the goal of providing cutting-edge technologies and expert consultation to individual scientific investigators.27
The shared goals of the NIH, other research stakeholders, and research institutions are more likely to be achieved when core scientists and research scientists work together to identify and minimize risk to research data, thereby improving research quality, rigor, and reproducibility. The ABRF supports data management policies and deposition of data/methods to prevent digital meddling, either through repositories specific for techniques (i.e., https://flowrepository.org) or general data repositories sponsored by the Center for Open Science (https://osf.io). Engaging RG core scientists as technology editors and reviewers26 will promote and support rigorous, transparent, and reproducible research and the responsible conduct of research. Scientists, funding sponsors, and institutions are encouraged to foster an atmosphere of quality data management and to give credit for data sharing by the data author.214 This ensures that the available data set follows a set of guiding principles to make data findable, accessible, interoperable, and reusable (FAIR).215 FAIR Guiding Principles instruct that the data and metadata meet criteria of findability, accessibility, interoperability, and reusability are standard practice.214– 216 Changes by scientific journals to improve reporting transparency includes providing generous length limits for methods section, and the use of a checklist during editorial processing to ensure the reporting of key methodological, and analytical information to reviewers and readers.16, 19, 20
ACKNOWLEDGMENTS
The authors thank the members of the current ABRF Research Groups (see Appendix) and all the ABRF members who have served on the ABRF Research Groups for their time, scientific expertise, and body of published literature dedicated to advancing technology over the last 30 yr. Sincere thanks to Ron Niece and George Grills, who offered insightful and critical contributions to this manuscript. N.C.F. is supported by P30 CA016086 Cancer Center Core Support Grant to the UNC Lineberger Comprehensive Cancer Center. K.L.K. is supported in part by the National Cancer Institute of the U.S. National Institutes of Health (NIH) under award number P30 CA086862. S.M.M. is supported in part by the National Cancer Institute of the NIH Cancer Center Grant P30 CA068485. S.M.M. is supported by P30 CA016087 Cancer Center Core Support Grant at the New York University Langone Laura and Isaac Perlmutter Comprehensive Cancer Center. F.W.-G. is supported by P30 CA008748 Cancer Center Core Support Grant at the Memorial Sloan Kettering Cancer Center. K.S.C. is supported in part by the National Institute of General Medical Sciences of the NIH IDeA program under award numbers P30 GM114736 and P20 GM103446. The authors declare no conflicts of interest.
Footnotes
This article includes supplemental data. Please visit http://www.fasebj.org to obtain this information.
REFERENCES
- 1.Berns KI, Bond EC, Manning FJ, eds. Resource Sharing in Biomedical Research. Institute of Medicine, Washington, DC: The National Academies Press, 1996. [PubMed] [Google Scholar]
- 2.Bennett LM, Gadlin H. Collaboration and team science: from theory to practice. J Investig Med. 2012;60:768–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.National Institutes of Health; Association of Biomolecular Research Facilities . Workshop on enhancing efficiency of research core facilities. J Biomol Tech. 2015;26:1–3.25649473 [Google Scholar]
- 4.Chang MC, Birken S, Grieder F, Anderson J. U.S. National Institutes of Health core consolidation-investing in greater efficiency. J Biomol Tech. 2015;26:1–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Angeletti RH, Bonewald LF, De Jongh K, Niece R, Rush J, Stults J. Research technologies: fulfilling the promise. FASEB J. 1999;13:595–601. [DOI] [PubMed] [Google Scholar]
- 6.Federation of American Societies for Experimental Biology (FASEB) Science-Policy-Advocacy-and-Communications Maximizing shared research resources. Part II: Survey findings and analysis. Available at: https://www.faseb.org/Portals/2/PDFs/opa/2017/Maximizing%20Shared%20Research%20Resources%20-%20Part%20II.pdf Retrieved January 4, 2019.
- 7.Meder D, Morales M, Pepperkok R, Schlapbach R, Tiran A, Van Minnebruggen G. Institutional core facilities: prerequisite for breakthroughs in the life sciences: core facilities play an increasingly important role in biomedical research by providing scientists access to sophisticated technology and expertise. EMBO Rep. 2016;17:1088–1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Gould J. Core facilities: shared support. Nature. 2015;519:495–496. [DOI] [PubMed] [Google Scholar]
- 9.Haley R. A framework for managing core facilities within the research enterprise. J Biomol Tech. 2009;20:226–230. [PMC free article] [PubMed] [Google Scholar]
- 10.Farber GK, Weiss L. Core facilities: maximizing the return on investment. Sci Transl Med. 2011;3:95cm21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hockberger P, Weiss J, Rosen A, Ott A. Building a sustainable portfolio of core facilities: a case study. J Biomol Tech. 2018;29:79–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.ABRF Mission Statement Association of Biomolecular Resource Facilities Available at: https://abrf.org/mission-statement-0. Accessed January 18, 2019.
- 13.The Economist Trouble at the lab. 2013;23–27.
- 14.Freedman LP, Inglese J. The increasing urgency for standards in basic biologic research. Cancer Res. 2014;74:4024–4029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Begley CG, Ioannidis JP. Reproducibility in science: improving the standard for basic and preclinical research. Circ Res. 2015;116:116–126. [DOI] [PubMed] [Google Scholar]
- 16.Nosek BA, Alter G, Banks GC, et al. . SCIENTIFIC STANDARDS. Promoting an open research culture. Science. 2015;348:1422–1425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Freedman LP, Cockburn IM, Simcoe TS. The economics of reproducibility in preclinical research. PLoS Biol. 2015;13:e1002165; erratum: 16, e1002626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Goodman SN, Fanelli D, Ioannidis JP. What does research reproducibility mean? Sci Transl Med. 2016;8:341ps12. [DOI] [PubMed] [Google Scholar]
- 19.McNutt M. Journals unite for reproducibility. Science. 2014;346:679. [DOI] [PubMed] [Google Scholar]
- 20.Reality check on reproducibility. Nature. 2016;533:437. [DOI] [PubMed] [Google Scholar]
- 21.Munafò MR, Nosek BA, Bishop DVM, et al. . A manifesto for reproducible science. Nature Human Behavior. 2017;1:0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Collins FS, Tabak LA. Policy: NIH plans to enhance reproducibility. Nature. 2014;505(7485):612–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.National Academies of Sciences, Engineering, and Medicine Fostering Integrity in Research, Washington, DC: The National Academies Press (US) National Academy of Sciences, 2017. [Google Scholar]
- 24.Steneck NH. ORI Introduction To the Responsible Conduct of Research, Washington, DC: Available at: https://ori.hhs.gov/ori-introduction-responsible-conduct-research: 2004. [Google Scholar]
- 25.National Academies of Sciences, Engineering and Medicine Reproducibility and Replicability in Science, Washington, DC: The National Academies Press, 2019. [PubMed] [Google Scholar]
- 26.Knudtson KL, Carnahan RH, Hegstad-Davies RL, et al. . Survey on scientific shared resource rigor and reproducibility. J Biomol Tech. 2019;30:36–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chang M, Grieder FB. Sharing core facilities and research resources--an investment in accelerating scientific discoveries. J Biomol Tech. 2016;27:2–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ritter NM, Fowler E. Analytical laboratory quality: part I. General quality practices. J Biomol Tech. 2001;12:4–10. [PMC free article] [PubMed] [Google Scholar]
- 29.Tabb DL. Quality assessment for clinical proteomics. Clin Biochem. 2013;46:411–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Niece RL. History and activities of the ABRF. In Flickinger MC, Drew SW (eds): The Encyclopedia of Bioprocess Technology: Fermentation, Biocatalysis & Bioseparation. Hoboken, NJ: John Wiley& Sons, Inc., 1999:2090–2092. [Google Scholar]
- 31.Niece RL, Williams KR, Wadsworth CL, et al. . A synthetic peptide for evaluating protein sequencer and amino acid analyzer performance in core facilities: design and results. In Hugli T. (ed): Techniques in Protein Chemistry. San Diego, CA: Academic Press, 1989:89–110. [Google Scholar]
- 32.Association of Biomolecular Resource Facilities ABRF RG Handbook. Available at: https://abrf.org/sites/default/files/temp/Resources/rg_handbook_2010_03-08-11.pdf.
- 33.Association of Biomolecular Resource Facilities ABRF RG study guidelines. Available at: https://abrf.org/sites/default/files/temp/Resources/research_group_study_participation_guidelines_2010.pdf.
- 34.Speicher DW, Grant GA, Niece RL, Blacher RW, Fowler AV, Williams KR. Design, characterization and results of ABRF-89SEQ: a test sample for evaluating protein sequencer performance in protein microchemistry core facilities. In Hugli TE. (ed): Current Research in Protein Chemistry. San Diego, CA: Academic Press, 1989:159–166. [Google Scholar]
- 35.Beach CM, Hathaway GM, Hayes TK, Smith AJ, Niece RL. Instrumentation used in protein and nucleic acid resource facilities: a survey of users. ABRF News. 1991;2(Suppl):1–7. [Google Scholar]
- 36.Niece RL, Ericsson LH, Fowler AV, et al. . Amino acid analysis and sequencing - what is state-of-the-art? In Jörnvall H, Höög J-O, Gustavsson A-M (eds): Methods in Protein Sequence Analysis. Basel, Switzerland: Birkhäuser Verlag, 1991:133–141. [Google Scholar]
- 37.Yuksel KU, Grant GA, Mende-Muller LM, Niece RL, Williams KR, Speicher DW. Protein sequencing from polyvinylidene difluoride membranes: design and characterization of a test sample (ABRF-90SEQ) and evaluation of results. In Villafranca JJ. (ed): Techniques in Protein Chemistry II. San Diego, CA: Academic Press, 1991:151–162. [Google Scholar]
- 38.Crimmins DL, Grant GA, Mende-Muller LM, et al. . Evaluation of protein sequencing core facilities: design, characterization, and results form a test sample (ABRF-91SEQ). In Angeletti RH. (ed): Techniques in Protein Chemistry III. San Diego, CA: Academic Press, 1992:35. [Google Scholar]
- 39.Mische SM, Yuksel KU, Mende-Muller LM, Matsudaira P, Crimmins DL, Andrews PC. Protein sequencing of post-translationally modified peptides and proteins: design, characterization and results of ABRF-92SEQ. In Angeletti RH. (ed): Techniques in Protein Chemistry IV. San Diego, CA: Academic Press, 1993:453–461. [Google Scholar]
- 40.Rush J, Andrews PC, Crimmins DL, et al. . A synthetic peptide for evaluating protein sequencing capabilities: design of ABRF-93SEQ and results. In Crabb JW. (ed): Techniques in Protein Chemistry V. San Diego, CA: Academic Press, 1994:133–141. [Google Scholar]
- 41.Gambee JE, Andrews PC, Grant GA, Merrill B, Mische SM, Rush J. Assignment of cysteine and tryptophan residues during protein sequencing: results of ABRF-94SEQ. In Crabb JW. (ed): Techniques in Protein Chemistry VI. San Diego, CA: Academic Press, 1995:209–217. [Google Scholar]
- 42.DeJongh KS, Fernandez J, Gambee JE, et al. . Design and analysis of ABRF-95SEQ, a recombinant protein with sequence heterogeneity. In Marshak DR. (ed): Techniques in Protein Chemistry VII. San Diego, CA: Academic Press, 1996:347–358. [Google Scholar]
- 43.Williams K, Hellman U, Kobayashi R, Lane W, Mische S, Speicher D. Internal protein sequencing of SDS page-separated proteins: a collaborative ABRF study. In Marshak DR. (ed): Techniques in Protein Chemistry VIII. San Diego, CA: Academic Press; 1997;99–109 [Google Scholar]
- 44.Fernandez J, Admon A, DeJongh K, et al. . Evaluation of ABRF-96SEQ: a sequence assignment exercise. In Marshak DR. (ed): Techniques in Protein Chemistry VIII. San Diego, CA: Academic Press; 1997:69–78. [Google Scholar]
- 45.Mische S, Hellman U, Speicher D, Williams K. 1997. Internal protein sequencing. In Flickinger MC, Drew SW (eds): The Encyclopedia of Bioprocess Technology: Fermentation, Biocatalysis & Bioseparation. New York, NY: John Wiley& Sons, 1997:2099–2013. [Google Scholar]
- 46.Stone K, Fernandez J, Admon A, et al. . ABRF-97SEQ: sequencing results of a low-level sample. J Biomol Tech. 1999;10:26–32. [Google Scholar]
- 47.Henzel WJ, Admon A, Carr SA, et al. . ABRF-98SEQ: evaluation of peptide sequencing at high sensitivity. J Biomol Tech. 2000;11:92–99. [PMC free article] [PubMed] [Google Scholar]
- 48.Buckel SD, Cook RG, Crawford JM, et al. . ABRF-2002ESRG, a difficult sequence: analysis of a PVDF-bound known protein with a heterogeneous amino-terminus. J Biomol Tech. 2002;13:246–257. [PMC free article] [PubMed] [Google Scholar]
- 49.Buckel SD, Cook RG, Crawford JM, et al. . ABRF-ESRG’03: analysis of a PVDF-bound known protein with a homogeneous amino-terminus. J Biomol Tech. 2003;14:278–288. [PMC free article] [PubMed] [Google Scholar]
- 50.Brune D, Crawford JM, Cook RG, et al. . Edman sequencing research group 2004 study: modified amino acids IN EDMAN sequencing. J Biomol Tech. 2005;16:272–284. [Google Scholar]
- 51.Brune D, Denslow ND, Kobayashi R, et al. . ABRF ESRG 2005 study: identification of seven modified amino acids by Edman sequencing. J Biomol Tech. 2006;17:308–326. [PMC free article] [PubMed] [Google Scholar]
- 52.Thoma RS, Smith JS, Sandoval W, et al. . The ABRF Edman Sequencing Research Group 2008 study: investigation into homopolymeric amino acid N-terminal sequence tags and their effects on automated Edman degradation. J Biomol Tech. 2009;20:216–225. [PMC free article] [PubMed] [Google Scholar]
- 53.Brune DC, Hampton B, Kobayashi R, et al. . ABRF ESRG 2006 study: Edman sequencing as a method for polypeptide quantitation. J Biomol Tech. 2007;18:306–320. [PMC free article] [PubMed] [Google Scholar]
- 54.Biringer RG, Hao Z, Hühmer AFR. P150-T utilization of ABRF sPRG protein standard for developing optimized experimental strategies for ETD based protein identification. J Biomol Tech. 2007;18:52. [Google Scholar]
- 55.Colangelo C, Dufresne CP, Farmar JG, et al. . ABRF-sPRG2011 study: development of a comprehensive standard for analysis of post-translational modifications. J Biomol Tech. 2011;22(Suppl):S20. [Google Scholar]
- 56.Dufresne CP, Ivanov AR, Koller A, et al. . ABRF-sPRG 2013 study: development and characterization of a proteomics normalization standard consisting of 1000 stable isotope labeled peptides and a qualitative stability study of peptides from the ABRF-sPRG 2012 study. J Biomol Tech. 2013;24(Suppl):S69–S70. [Google Scholar]
- 57.Dufresne C, Hawke D, Ivanov AR, et al. . ABRF research group development and characterization of a proteomics normalization standard consisting of 1,000 stable isotope labeled peptides. J Biomol Tech. 2014;25(Suppl):S1. [Google Scholar]
- 58.Colangelo C, Chung L, Dufresne C, et al. . Development and characterization of SpikeMixTM ABRF (cross-species standard) consisting of 1,000 stable isotope labeled peptides. Available at: https://www.jpt.com/fileadmin/user_upload/AppNote_Proteomics_SpikeMix_ABRF_SIL_Peptides.pdf.
- 59.Cohen SA, Strydom DJ. Amino acid analysis utilizing phenylisothiocyanate derivatives. Anal Biochem. 1988;174:1–16. [DOI] [PubMed] [Google Scholar]
- 60.Crabb JW, Ericsson LH, Atherton D, Smith AJ, Kutny R. 1990. A collaborative amino acid analysis study from the Association of Biomolecular Resource Facilities. In Villafranca JJ. (ed): Current Research in Protein Chemistry. Academic Press, San Diego, p. 49–61. [Google Scholar]
- 61.Tarr GE, Paxton RJ, Pan Y-CE, Ericsson LH, Crabb JW. Amino acid analysis 1990: the third collaborative study from the Association of Biomolecular Resource Facilities (ABRF). In Villafranca JJ. (ed): Techniques in Protein Chemistry II. San Diego, CA: Academic Press, 1991:139–150. [Google Scholar]
- 62.Strydom DJ, Tarr GE, Pan Y-CE, Paxton RJ. Collaborative trial analyses of ABRF-91AAA. In Hogue-Angeletti R. (ed): Techniques in Protein Chemistry III. San Diego, CA: Academic Press, 1992:261–274. [Google Scholar]
- 63.Strydom DJ, Andersen TT, Apostol I, Fox JW, Paxton RJ, Crabb JW. Cysteine and tryptophan amino acid analysis of ABRF92-AAA. In Hogue-Angeletti R. (ed): Techniques in Protein Chemistry IV. San Diego, CA: Academic Press, 1993:279–288. [Google Scholar]
- 64.Strydom DJ, Cohen SA. Sensitive analysis of cystine/cysteine using 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate derivatives. In Angeletti RH. (ed): Techniques in Protein Chemistry IV. San Diego, CA: Academic Press, 1993:299–306. [Google Scholar]
- 65.Cohen SA, Michaud DP. Synthesis of a fluorescent derivatizing reagent, 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate, and its application for the analysis of hydrolysate amino acids via high-performance liquid chromatography. Anal Biochem. 1993;211:279–287. [DOI] [PubMed] [Google Scholar]
- 66.Hong JL. Determination of amino acids by pre-column derivatization with 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate and high performance liquid chromatography with ultraviolet detection. J Chromatog. 1994;670:59–66. [Google Scholar]
- 67.Strydom DJ, Cohen SA. Comparison of amino acid analyses by phenylisothiocyanate and 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate precolumn derivatization. Anal Biochem. 1994;222:19–28. [DOI] [PubMed] [Google Scholar]
- 68.Cohen SA, De Antonis KM. Applications of amino acid derivatization with 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate. Analysis of feed grains, intravenous solutions and glycoproteins. J Chromatogr A. 1994;661:25–34. [DOI] [PubMed] [Google Scholar]
- 69.Yüksel KÜ, Andersen TT, Apostol I, et al. . The hydrolysis process and the quality of amino acid analysis: ABRF-94AAA collaborative trial. In Crabb JW. (ed): Techniques in Protein Chemistry VI. San Diego, CA: Academic Press, 1994:185–192. [Google Scholar]
- 70.Yüksel KÜ, Andersen TT, Apostol I, et al. . Amino acid analysis of phospho-peptides: ABRF-93AAA. In Crabb JW. (ed): Techniques in Protein Chemistry V. San Diego, CA: Academic Press, 1994:231–240. [Google Scholar]
- 71.Mahrenholz AM, Denslow ND, Andersen TT, et al. . Amino acid analysis-recovery from PVDF membranes: ABRF '95AAA collaborative trial. In Marshak DR. (ed): Techniques in Protein Chemistry VII. San Diego, CA: Academic Press, 1996:323–330. [Google Scholar]
- 72.Schegg KM, Denslow ND, Andersen TT, et al. . 1997. Quantitation and identification of proteins by amino acid analysis. In Marshak DR. (ed): Techniques in Protein Chemistry VIII. San Diego, CA: Academic Press, 1996:207–216. [Google Scholar]
- 73.Van Wandelen C, Cohen SA. Using quaternary high-performance liquid chromatography eluent systems for separating 6-aminoquinolyl-N-10. hydroxysuccinimidyl carbamate-derivatized amino acid mixtures. J Chromatog. 1997;763:11–22. [Google Scholar]
- 74.Hunziker P, Andersen TT, Bao Y, et al. . Identification of proteins electroblotted to polyvinylidene difluoride membrane by combined amino acid analysis and bioinformatics: an ABRF multicenter study. J Biomol Tech. 1999;10:129–136. [PMC free article] [PubMed] [Google Scholar]
- 75.Bonewald LF, Bibbs L, Kates SA, et al. . Study on the synthesis and characterization of peptides containing phosphorylated tyrosine. J Pept Res. 1999;53:161–169. [DOI] [PubMed] [Google Scholar]
- 76.Bonewald LF, Bibbs L, Kates SA, et al. . 1997 ABRF Peptide synthesis research committee study on a peptide containing a phosphorylated tyrosine. In Tam JP, and Kaumaya PTP, (eds): Peptides: Frontiers of Peptide Science. Dordrecht: The Netherlands: Kluver Academic Publishers, 1999:705–706. [Google Scholar]
- 77.Bonewald LF, Angeletti RH. A guide to peptide synthesis and peptide facilities for biomolecular research. In Flickinger MC, Drew SW (eds): Encyclopedia of Bioprocess Technology. New York, NY: John Wiley & Sons, 1999:705–706. [Google Scholar]
- 78.Angeletti RH, Bibbs L, Bonewald LF, et al. . 1998. A multicenter study of racemization during standard solid-phase peptide synthesis. In Ramage R, Epton R (eds): Peptides 1996. Proceedings of the 24th EPS. Kingswinford, UK: Mayflower Scientific Ltd; 1998:207–208. [Google Scholar]
- 79.Angeletti RH, Bonewald LF, Fields GB. Six year study of peptide synthesis. In Fields G. (ed): Methods in Enzymology, Vol. 289 Cambridge, MA: Academic Press (Elsevier) 1997:607–717 [DOI] [PubMed] [Google Scholar]
- 80.Angeletti RH, Bibbs L, Bonewald LF, et al. . Analysis of racemization during “standard” solid phase peptide synthesis: a multicenter study. In Marshak DR. (ed): Techniques in Protein Chemistry VIII. San Diego, CA: Academic Press, Inc., 1997:875–890. [Google Scholar]
- 81.Angeletti RH, Bibbs L, Bonewald LF, et al. . Formation of a disulfide bond in a octreotide-like peptide: a multicenter study. In Marshak DR. (ed): Techniques in Protein Chemistry VII. San Diego, CA: Academic Press, 1996:261–274. [Google Scholar]
- 82.Fields GB, Bibbs L, Bonewald LF, et al. . Multi-center study of post-assembly problems in solid phase peptide synthesis. In Kaumaya PTP, and Hodges RS (eds): Peptides: Chemistry, Structure and Biology. Kingswinford, UK: Mayflower Scientific Ltd, 1996:52–54. [Google Scholar]
- 83.Fields GB, Angeletti RH, Bonewald LF, et al. . Correlation of cleavage techniques with side-reactions following solid-phase peptide synthesis. In Crabb JW. (ed): Techniques in Protein Chemistry VI. San Diego, CA: Academic Press, 1995:539–546. [Google Scholar]
- 84.Fields GB, Angeletti RH, Carr SA, et al. . Variable success of peptide-resin cleavage and deprotection following solid-phase synthesis. In Crabb JW. (ed): Techniques in Protein Chemistry V. San Diego, CA: Academic Press, 1994:501–507. [Google Scholar]
- 85.Fields GB, Carr SA, Marshak DR, et al. . Evaluation of a peptide synthesis as practiced in 53 different laboratories. In Hogue-Angeletti R. (ed): Techniques in Protein Chemistry IV. San Diego, CA: Academic Press, 1993:229–237. [Google Scholar]
- 86.Smith AJ, Young JD, Carr SA, Marshak DR, Williams LC, Williams KR. State-of-the-art peptide synthesis: comparative characterization of a 16-mer synthesized in 31 different laboratories. In Hogue-Angeletti R. (ed): Techniques in Protein Chemistry III. San Diego, CA: Academic Press, 1992:219–229. [Google Scholar]
- 87.Bibbs L, Ambulos NP Jr, Kates SA, et al. . Strategies for the synthesis of labeled peptides. J Biomol Tech. 2000;11:155–165. [PMC free article] [PubMed] [Google Scholar]
- 88.Medzihradszky K, Ambulos NP Jr, Khatri A, et al. . Mass spectrometry analysis for the determination of side reactions for cyclic peptides prepared from an Fmoc/tBu/Dmab protecting group strategy. Lett Pept Sci. 2001;8:1–12. [Google Scholar]
- 89.Ambulos NP Jr, Bibbs L, Bonewald LF, et al. . Methods for analysis of synthetic peptides. In Albericia F, Kates SA (eds): Solid phase Synthesis: A Practical Guide. New York, NY: Marcel Dekker Publications, 2000:751-789. [Google Scholar]
- 90.Bibbs L, Ambulos NP Jr, Kates SA, et al. Strategies for the synthesis of labeled peptides. In Barany G, Fields G (eds): Peptides for the New Millennium. Proceedings of the 16th American Peptide Symposium, June 26–July 1, 1999, Minneapolis, Minnesota, USA. 2000. [PMC free article] [PubMed] [Google Scholar]
- 91.Townsend RR, Manzi A, Merkle RK, et al. . Quantitative monosaccharide analysis: a multi-center study. ABRF News. 1997;8:14–21. [Google Scholar]
- 92.Arnott D, Gawinowicz MA, Grant RA, et al. . ABRF-PRG03: phosphorylation site determination. J Biomol Tech. 2003;14:205–215. [PMC free article] [PubMed] [Google Scholar]
- 93.Myszka DG, Abdiche YN, Arisaka F, et al. . The ABRF-MIRG’02 study: assembly state, thermodynamic, and kinetic analysis of an enzyme/inhibitor interaction. J Biomol Tech. 2003;14:247–269. [PMC free article] [PubMed] [Google Scholar]
- 94.Yadav SP, Bergqvist S, Doyle ML, et al. . Conclusions from the MIRG 2010 benchmark study: molecular interactions in a three component system and presentation of 2011 survey results on label-free technologies. J Biomol Tech. 2011;22(Suppl):S23–S24. [Google Scholar]
- 95.Yamniuk AP, Edavettal SC, Bergqvist S, et al. . ABRF-MIRG benchmark study: molecular interactions in a three-component system. J Biomol Tech. 2012;23:101–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yadav SP, Bergqvist S, Doyle ML, Neubert TA, Yamniuk AP. MIRG survey 2011: snapshot of rapidly evolving label-free technologies used for characterizing molecular interactions. J Biomol Tech. 2012;23:94–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Yamniuk AP, Newitt JA, Doyle ML, et al. . Development of a model protein interaction pair as a benchmarking tool for the quantitative analysis of 2-site protein-protein interactions. J Biomol Tech. 2015;26:125–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Castro-Wallace SL, Chiu CY, John KK, et al. . Nanopore DNA sequencing and genome assembly on the international Space station. Sci Rep. 2017;7:18022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Endrullat C, Glökler J, Franke P, Frohme M. Standardization and quality management in next-generation sequencing. Appl Transl Genomics. 2016;10:2–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Tighe S, Baldwin D, Grills G, Mason C. The ABRF-next generation sequencing study: a five-platform, cross-site, cross-protocol examination of RNA sequencing. J Biomol Tech. 2013;24(Suppl):S67. [Google Scholar]
- 101.Li S, Tighe S, Nicolet C, Baldwin D, Grills G; ABRF-NGS Consortium . Mason CE. The ABRF next generation sequencing study: multi-platform and cross-methodological reproducibility of transcriptome profiling by RNA-seq. J Biomol Tech. 2014;25(Suppl):S1–S2. [Google Scholar]
- 102.Zook JM, Catoe D, McDaniel J, et al. . Extensive sequencing of seven human genomes to characterize benchmark reference materials. Sci Data. 2016;3:160025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.SEQC/MAQC-III Consortium A comprehensive assessment of RNA-seq accuracy, reproducibility and information content by the Sequencing Quality Control Consortium. Nat Biotechnol. 2014;32:903–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Li S, Tighe SW, Nicolet CM, et al. . Multi-platform assessment of transcriptome profiling using RNA-seq in the ABRF next-generation sequencing study. Nat Biotechnol. 2014;32:915–925; erratum: 1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Li S, Łabaj PP, Zumbo P, et al. . Detecting and correcting systematic variation in large-scale RNA sequencing data. Nat Biotechnol. 2014;32:888–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Chhangawala S, Rudy G, Mason CE, Rosenfeld JA. The impact of read length on quantification of differentially expressed genes and splice junction detection. Genome Biol. 2015;16:131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Mason CE, Afshinnekoo E, Tighe S, Wu S, Levy S. International standards for genomes, transcriptomes, and metagenomes. J Biomol Tech. 2017;28:8–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Grills G, Leviten D, Hall L, et al. Effects of different DNA sequencing methods evaluated using a web based quality control resource: the ABRF DNA sequence research group 2001 standard template study. Available at: ABRF Website 2002; Never_Ending_Story.pdf.
- 109.Herbert ZT, Kershner JP, Butty VL, et al. . Cross-site comparison of ribosomal depletion kits for Illumina RNAseq library construction. BMC Genomics. 2018;19:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Delventhal K, Sergison E, Pruett-Miller S, Dahlem T, Gurumurthy C, Kmiec E, Marsischky G. CRISPR/Cas9 Methods: Preferences from the Field. 2018. Available at: https://abrf.org/sites/default/files/gerg_survey_poster_2018_final_0.pdf.
- 111.Pon RT, Buck GA, Niece RL, Robertson M, Smith AJ, Spicer E. A survey of nucleic acid services in core laboratories. Biotechniques. 1994;17:526–534. [PubMed] [Google Scholar]
- 112.Naeve CW, Buck GA, Niece RL, Pon RT, Robertson M, Smith AJ. Accuracy of automated DNA sequencing: a multi-laboratory comparison of sequencing results. Biotechniques. 1995;19:448–453. [PubMed] [Google Scholar]
- 113.Pon RT, Buck GA, Hager KM, et al. . Multi-facility survey of oligonucleotide synthesis and an examination of the performance of unpurified primers in automated DNA sequencing. Biotechniques. 1996;21:680–685. [PubMed] [Google Scholar]
- 114.Adams PS, Dolejsi MK, Grills GS, et al. . Effects of DMSO, thermocycling and editing on a template with a 72% GC rich area. Microb Comp Genomics. 1997;2:198. [Google Scholar]
- 115.Adams PS, Dolejsi MK, Grills G, et al. . An analysis of techniques used to improve the accuracy of automated DNA sequencing of a GC-rich template. J Biomol Tech. 1998;9:9–18. [Google Scholar]
- 116.Grills GS, Adams PS, Dolejsi MK, et al. . Analysis of the effects of different DNA sequencing methods on sequencing quality, creation of a quality control resource, and assessment of the current state of the art: results from the 1998 ABRF DNA Sequence Research Committee study. Microb Comp Genomics. 1998;3:C-88. [Google Scholar]
- 117.Grills GS, Adams PS, Dolejsi MK, et al. . Evaluation of the effects of different DNA sequencing methods on sequencing standard and difficult templates, expansion of a web based quality control resource, and establishing a test array of sequencing templates: the ABRF DNA Sequencing Research Committee 1999 standard and difficult template studies. J Biomol Tech. 1999;10:97. [Google Scholar]
- 118.Thannhauser T, Adams PS, Dolejsi MK, Grills G, Hardin S, Robertson M. Effects of different DNA sequencing methods on accuracy, quality and expansion of a web-based sequencing resource: results of the ABRF DNA sequencing group 1999 study. ABRF 2000: from singular to global analyses of biological systems. J Biomol Tech. 2000;11:56. [Google Scholar]
- 119.Hager KM, Fox JW, Gunthorpe M, Lilley KS, Yeung A. Survey of current trends in DNA synthesis core facilities. J Biomol Tech. 1999;10:187–193. [PMC free article] [PubMed] [Google Scholar]
- 120.Albertson H. Microarrays in a shared facility - practical and scientific experiences from the huntsman cancer Institute. J Biomol Tech. 1999;10:105. [Google Scholar]
- 121.Buck GA, Fox JW, Gunthorpe M, et al. . Design strategies and performance of custom DNA sequencing primers. Biotechniques. 1999;27:528–536. [DOI] [PubMed] [Google Scholar]
- 122.Naeve CW, Adams PS. Nucleic acids. In Flickinger MC, Drew SW (eds): Encyclopedia of Bioprocess Technology. New York, NY: John Wiley & Sons, 1999:2089–2120. [Google Scholar]
- 123.Leviten D, Grills G, Hardin S, Robertson M, Thannhauser T. Results from the DNA sequencing research group’s general survey of core facilities: a detailed summary of the composition and configuration of the average DNA sequencing core laboratory. ABRF 2000: from singular to global analyses of biological systems. J Biomol Tech. 2000;11:56. [Google Scholar]
- 124.Thannhauser T, Grills G, Hardin S, Leviten D, Robertson M, Van Ee JI. Preliminary results from the DNA sequencing research group 2000 study, part 1: factors that affect the sequencing performance of a standard BAC template. ABRF 2000: from singular to global analyses of biological systems. J Biomol Tech. 2000;11:30. [Google Scholar]
- 125.Thannhauser T, Grills G, Hardin S, Leviten D, Robertson M, Van Ee JI. Preliminary results from the DNA sequencing research group 2000 study part 2: a comparison of methods used to isolate BAC templates in preparation for sequencing. ABRF 2000: from singular to global analyses of biological systems. J Biomol Tech. 2000;11:34. [Google Scholar]
- 126.Van Ee JI, Grills G, Hardin S, Leviten D, Robertson M, Thannhauser T. Preliminary results from the DNA sequencing research group 2000 study: an evaluation of methods used to sequence and isolate bacterial artificial chomosomes (BACs). ABRF 2000: from singular to global analyses of biological systems. J Biomol Tech. 2000;11:56. [Google Scholar]
- 127.Robertson MA, Hall L, Hawes J, et al. . Preliminary results from the DNA sequencing research group 2001 study: factors that affect the sequencing and detection of mixed base sequences in PCR products. ABRF 2001: the new biology: technologies for resolving macromolecular communications. J Biomol Tech. 2000;11:225. [Google Scholar]
- 128.Thannhauser T, Hall LS, Hawes J, et al. . Update on the continuation of the DNA sequencing research group 2000 study: an evaluation of methods used to sequence and isolate bacterial artificial chromosomes. ABRF 2001: the new biology: technologies for resolving macromolecular communications. J Biomol Tech. 2000;11:189. [Google Scholar]
- 129.Robertson MA. Advances in DNA sequencing. ABRF 2001: the new biology: technologies for resolving macromolecular communications. J Biomol Tech. 2000;11:230. [Google Scholar]
- 130.Herbert ZT, Kershner JP, Butty VL, et al. . Cross-site comparison of ribosomal depletion kits for Illumina RNAseq library construction. BMC Genomics. 2018;19:199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Jackson-Machelski E, Hunter TC, Leviten D, Pershad R, Spicer D. DNA sequencing roundtable discussion. ABRF 2002: biomolecular technologies: tools for discovery in proteomics and genomics. J Biomol Tech. 2001;12:91. [Google Scholar]
- 132.Gunthorpe ME, Fox JW, Hager KM, Lilley KS, Scaringe S, Yeung AT. Use of mass spectrometry, capillary electrophoresis, and gel electrophoresis for quality analysis of synthetic oligonucleotides: perspectives from the ABRF Nucleic Acid Research Group. J Biomol Tech. 2001;12:16–24. [Google Scholar]
- 133.Leviten D, Hawes J, Hunter T, et al. . DNA sequencing research group (DSRG) single nucleotide polymorphism (SNP) study 2002 results. ABRF 2002: biomolecular technologies: tools for discovery in proteomics and genomics. J Biomol Tech. 2001;12:135. [Google Scholar]
- 134.Hawes J, Bartley D, Hunter TC, et al. . DNA sequencing research group (DSRG) 2001-2002 general survey: a snapshot of the composition and configuration of the current DNA sequencing core facility. ABRF 2002: biomolecular technologies: tools for discovery in proteomics and genomics. J Biomol Tech. 2001;12:139. [Google Scholar]
- 135.Hawes JW, Grills GS, Jackson-Machelski E, et al. . DSRG 2003 study: evaluation of methods for analysis of difficult DNA repeat sequences. ABRF 2003: translating biology using proteomics and functional genomics. J Biomol Tech. 2003;14:55. [Google Scholar]
- 136.Wiebe GJ, Pershad R, Adam D, et al. . DNA sequencing research group presentation and open discussion: the development and launch of the web-based DNA sequencing troubleshooting resource. J Biomol Tech. 2005;16:73. [Google Scholar]
- 137.Hawes JW, Knudtson KL, Escobar H, et al. . Evaluation of methods for sequence analysis of highly repetitive DNA templates. J Biomol Tech. 2006;17:138–144. [PMC free article] [PubMed] [Google Scholar]
- 138.Kieleczawa J, Adam D, Bintzler D, et al. . Identification of optimal protocols for sequencing difficult templates: results of the 2008 ABRF DNA sequencing research group difficult template study 2008. J Biomol Tech. 2009;20:116–127. [PMC free article] [PubMed] [Google Scholar]
- 139.Wiebe GJ, Pershad R, Escobar H, et al. . DNA Sequencing Research Group (DSRG) 2003 - a general survey of core DNA sequencing facilities. J Biomol Tech. 2003;14:231–237. [PMC free article] [PubMed] [Google Scholar]
- 140.Pershad R, Hawes J, Hunter T, et al. . The state of the art of DNA sequencing laboratories. ABRF 2003: translating biology using proteomics and functional genomics. J Biomol Tech. 2003;14:55. [Google Scholar]
- 141.Hunter TC, Escobar H, Grills G, et al. . DNA sequencing research group roundtable discussion. ABRF 2003: translating biology using proteomics and functional genomics. J Biomol Tech. 2003;14:105. [Google Scholar]
- 142.Lin H, Escobar H, Hawes JW, et al. . DSRG-2004 - a web-based, user-interactive DNA sequencing troubleshooting resource. ABRF 2004: integrating technologies in proteomics and genomics. J Biomol Tech. 2004;15:12. [Google Scholar]
- 143.Pershad R, Lyons R, Grills G. Data tracking and quality control in high throughput DNA sequencing: the ABRF DNA Sequencing Research Group (DSRG) 2004 applied technology and informatics tutorial. ABRF 2004: integrating technologies in proteomics and genomics. J Biomol Tech. 2004;15:82. [Google Scholar]
- 144.Escobar H, Hawes JW, Lin H, et al. . DNA Sequencing Research Group (DSRG) presentation and open discussion: a web-based DNA sequencing troubleshooting resource. ABRF 2004: integrating technologies in proteomics and genomics. J Biomol Tech. 2004;15:84. [Google Scholar]
- 145.Lin H, Adam D, Escobar H, et al. DNA Sequencing Research Group . DNA sequencing troubleshooting guide. ABRF 2005: biomolecular technologies: discovery to hypothesis. J Biomol Tech. 2005;16:74. [Google Scholar]
- 146.Adams PS, Bintzler D, Bodi K, et al. . Comparison of commercially available target enrichment methods for next generation sequencing with Illumina platform J Biomol Tech. 2010;21:S17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Kieleczawa J. LIMS primer. J Biomol Tech. 2010;21:S8. [PMC free article] [PubMed] [Google Scholar]
- 148.Grills GS. Problems and pleasures of implementing microarray technologies. J Biomol Tech. 1999;10:105. [Google Scholar]
- 149.Massimi A. Implementation of a custom cDNA microarray facility at AECOM. J Biomol Tech. 1999;10:105. [Google Scholar]
- 150.Sandmeyer S. A users-eye view of the affymetrix gene expression system: the early days. J Biomol Tech. 1999;10:105. [Google Scholar]
- 151.Grills GS, Griffin C, Heck D, et al. . The state of the art of microarray analysis: a profile of microarray laboratories. J Biomol Tech. 2000;11:30. [Google Scholar]
- 152.Grills GS, Burngarner R, Delrow J, et al. . The state of the art of microarray research: results of the ABRF Microarray Research Group studies. 12th International Genome Sequencing and Analysis Conference 2000; Available at: https://abrf.org/sites/default/files/temp/RGs/GRG/marg_survey_2000_poster.pdf.
- 153.Grills GS, Griffin C, Massimi A, Lilley K, Knudtson K, VanEe JI. A current profile of microarray laboratories: results of the 2000-2001 ABRF Microarray Research Group survey of laboratories using microarray technologies. J Biomol Tech. 2000;11:225. [Google Scholar]
- 154.Grills GS, Griffin C, Lilley K, Massimi A, Bao Y, VanEe JI. A profile of microarray laboratories. Proceedings of the Northwest Microarray Conference 2000. Available at: https://abrf.org/sites/default/files/temp/RGs/GRG/marg_poster_2001.pdf.
- 155.Bumgarner R. DNA arrays: technologies, applications, and data analysis. J Biomol Tech. 2000;11:35. [Google Scholar]
- 156.Knudtson KL, Griffin C, Brooks A, et al. . Factors contributing to variability in DNA microarray results: the ABRF microarray research group 2002 study. J Biomol Tech. 2001;12:108. [Google Scholar]
- 157.Brooks AI, Knudtson KL, Griffin C, et al. . Assessing the performance and utility of oligonucleotide libraries for transcript profiling via microarray analysis: 2002-2003 Microarray Research Group study. J Biomol Tech. 2003;14:101. [Google Scholar]
- 158.Li J, Auer H, Brooks A, et al. . Microarray Research Group study: evaluation of small sample nucleic acid amplification technologies for gene expression profiling. J Biomol Tech. 2005;16:73. [Google Scholar]
- 159.Viale A, Hester S, Khitrov G, et al. . The effects of data analysis methods on microarray results as evaluated by Quantitative PCR. J Biomol Tech. 2006;17:70–72. [Google Scholar]
- 160.Viale A, Li J, Tiesman J, Hester S, et al. . Big results from small samples: evaluation of amplification protocols for gene expression profiling. J Biomol Tech. 2007;18:150–161. [PMC free article] [PubMed] [Google Scholar]
- 161.Bartley DA, Ballard LW, Bao Y, Bintzler D, Grills GS, Kasch L. Fragment analysis: a changing field and a new research group. J. Biomol. Tech. 1999; 10(2):101) (Bartley DA, Adams PS, Ballard LW, Bao Y, Bintzler D, Grills GS, Kasch L, Morrison P, Terrell C. The effects on mobility of various protocols, reagents and equipment used in fragment analysis. J Biomol Tech. 2000;11:34. [Google Scholar]
- 162.Yeung AT, Holloway BP, Adams PS, Shipley GL. Evaluation of dual-labeled fluorescent DNA probe purity versus performance in real-time PCR. Biotechniques. 2004;36:266–270, 272, 274–275. [DOI] [PubMed] [Google Scholar]
- 163.Tighe S, Baldwin D, Green S, Reyero N; ABRF MGRG/XMP Consortium . Next generation sequencing and the extreme microbiome project (XMP). J Next Generation Sequencing and Applications. 2015;2:1–2. [Google Scholar]
- 164.Thimmapuram J, Wright C, Albert I, et al. . A methodology study for metagenomics using next generation sequencers. J Biomol Tech. 2012;23(Suppl):S16–S17. [Google Scholar]
- 165.Ahsanuddin S, Afshinnekoo E, Gandara J, et al. . Assessment of repli-G multiple displacement whole genome amplification (WGA) techniques for metagenomic applications. J Biomol Tech. 2017;28:46–55; erratum: 96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Afshinnekoo E, Chou C, Alexander N, Ahsanuddin S, Schuetz AN, Mason CE. Precision metagenomics: rapid metagenomic analyses for infectious disease diagnostics and public health surveillance. J Biomol Tech. 2017;28:40–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.MetaSUB International Consortium The metagenomics and metadesign of the subways and urban biomes (MetaSUB) international consortium inaugural meeting report. Microbiome. 2016;4:24; erratum: 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.McIntyre ABR, Alexander N, Grigorev K, et al. . Single-molecule sequencing detection of N6-methyladenine in microbial reference materials. Nat Commun. 2019;10:579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Tighe S, Afshinnekoo E, Rock TM, et al. . Genomic methods and microbiological technologies for profiling novel and extreme environments for the extreme microbiome project (XMP). J Biomol Tech. 2017;28:31–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mason CE, Tighe S. Focus on metagenomics. J Biomol Tech. 2017;28:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Tessler M, Neumann JS, Afshinnekoo E, et al. . Large-scale differences in microbial biodiversity discovery between 16S amplicon and shotgun sequencing. Sci Rep. 2017;7:6589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Nicolet C, Paulson A, Shanker S, et al. . Evaluation of RNA amplification kits at subnanogram input amounts of total RNA for RNA-seq. J Biomol Tech. 2013;24(Suppl):S70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Forgetta V, Leveque G, Dias J, et al. . Sequencing of the Dutch elm disease fungus genome using the Roche/454 GS-FLX titanium system in a comparison of multiple genomics core facilities. J Biomol Tech. 2013;24:39–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Askenazi M, Bandeira N, Chalkley RJ, et al. . iPRG 2011: a study on the identification of electron transfer dissociation (ETD) mass spectra. J Biomol Tech. 2011;22:S20. [Google Scholar]
- 175.Chalkley RJ, Bandeira N, Chambers MC, et al. ; Proteome Informatics Research Group . Proteome informatics research group (iPRG)_2012: a study on detecting modified peptides in a complex mixture. Mol Cell Proteomics. 2014;13:360–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Bandeira N, Chambers MC, Clauser KR, et al. . iPRG 2012 study: detecting modified peptides in a complex mixture. J Biomol Tech. 2012;23:S24–S25. [Google Scholar]
- 177.Choi M, Eren-Dogu ZF, Colangelo C, et al. ABRF Proteome Informatics Research Group . ABRF proteome informatics research group (iPRG) 2015 study: detection of differentially abundant proteins in label-free quantitative LC-MS/MS experiments. J Proteome Res. 2017;16:945–957. [DOI] [PubMed] [Google Scholar]
- 178.Lee JY, Choi H, Colangelo CM, et al. . ABRF proteome informatics research group (iPRG) 2016 study: inferring proteoforms from bottom-up proteomics data. J Biomol Tech. 2018;29:39–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Fannes T, Vandermarliere E, Schietgat L, Degroeve S, Martens L, Ramon J. Predicting tryptic cleavage from proteomics data using decision tree ensembles. J Proteome Res. 2013;12:2253–2259. [DOI] [PubMed] [Google Scholar]
- 180.Weisser H, Choudhary JS. Targeted feature detection for data-dependent shotgun proteomics. J Proteome Res. 2017;16:2964–2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Matthew T, Fredrik E, Yasset P-R, et al. . A protein standard that emulates homology for the characterization of protein inference algorithms. J Proteome Res. 2018;17:1879–1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Proteome Informatics Research Group (iPRG) (2019). ABRF Association of Biomolecular Research. Available at: https://abrf.org/research-group/proteome-informatics-research-group-iprg.
- 183.Urban P. Quantitative mass spectrometry: an overview. Philos trans A math phys Eng Sci. 2016;374:2015038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Arnott DP, Gawinowicz M, Grant RA, et al. . Proteomics in mixtures: study results of ABRF-PRG02. J Biomol Tech. 2002;13:179–186. [PMC free article] [PubMed] [Google Scholar]
- 185.Arnott D, Gawinowicz MA, Kowalak JA, et al. . ABRF-PRG04: differentiation of protein isoforms. J Biomol Tech. 2007;18:124–134. [PMC free article] [PubMed] [Google Scholar]
- 186.Turck CW, Falick AM, Kowalak JA, et al. ; Association of Biomolecula Resource Facilities Proteomics Research Group . The association of biomolecular resource facilities proteomics research group 2006 study: relative protein quantitation. Mol Cell Proteomics. 2007;6:1291–1298. [DOI] [PubMed] [Google Scholar]
- 187.Falick AM, Kowalak JA, Lane WS, et al. . ABRF-PRG05: de novo peptide sequence determination. J Biomol Tech. 2008;19:251–257. [PMC free article] [PubMed] [Google Scholar]
- 188.Andacht TM, Bunger MK, Bystrom CE, et al. . PRG-2011: defining the interaction between users and suppliers of proteomics services/facilities. J Biomol Tech. 2011;22(Suppl):S51. [Google Scholar]
- 189.Kolarich D, Orlando R, Zaia J. gPRG: toward consensus on glycan analysis: reliable methods and reproducibility. J Biomol Tech. 2011;22(Suppl):S22. [Google Scholar]
- 190.Falick AM, Lane WS, Lilley KS, et al. . ABRF-PRG07: advanced quantitative proteomics study. J Biomol Tech. 2011;22:21–26. [PMC free article] [PubMed] [Google Scholar]
- 191.Friedman DB, Andacht TM, Bunger MK, et al. . The ABRF Proteomics Research Group studies: educational exercises for qualitative and quantitative proteomic analyses. Proteomics. 2011;11:1371–1381. [DOI] [PubMed] [Google Scholar]
- 192.Albers C, Hartmer R, Jabs W, Baessmann C, Kaspar S. N-terminal top-down protein sequencing of the ABRF-PSRG2012 study samples by ETD-UHR-TOF mass spectrometry. J Biomol Tech. 2012;23(Suppl):S51. [Google Scholar]
- 193.Ivanov AR, Colangelo CM, Dufresne CP, et al. . Interlaboratory studies and initiatives developing standards for proteomics. Proteomics. 2013;13:904–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Jones K, Kim K, Patel B, et al. . Mass spectrometry data collection in parallel at multiple core facilities operating tripleTOF 5600 and Orbitrap elite/velos pro/Q exactive mass spectrometers. J Biomol Tech. 2013;24:S52. [Google Scholar]
- 195.Leymarie N, Griffin PJ, Jonscher K, et al. . Interlaboratory study on differential analysis of protein glycosylation by mass spectrometry: the ABRF glycoprotein research multi-institutional study 2012. Mol Cell Proteomics. 2013;12:2935–2951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Song E, Mayampurath A, Yu CY, Tang H, Mechref Y. Glycoproteomics: identifying the glycosylation of prostate specific antigen at normal and high isoelectric points by LC-MS/MS. J Proteome Res. 2014;13:5570–5580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Bennett KL, Wang X, Bystrom CE, et al. . The 2012/2013 ABRF proteomic research group study: assessing longitudinal intralaboratory variability in routine peptide liquid chromatography tandem mass spectrometry analyses. Mol Cell Proteomics. 2015;14:3299–3309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Willyard C. New human gene tally reignites debate. Nature. 2018;558:354–355. [DOI] [PubMed] [Google Scholar]
- 199.Aebersold R, Agar JN, Amster IJ, et al. . How many human proteoforms are there? Nat Chem Biol. 2018;14:206–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Ponomarenko EA, Poverennaya EV, Ilgisonis EV, et al. . The size of the human proteome: the width and depth. Int J Anal Chem. 2016;2016:7436849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Van Riper S, Chen E, Chien A, et al. An ABRF-PRG study: Identification of low abundance proteins in a highly complex protein sample. 2016. Proceedings of the 64th ASMS Conference on Mass Spectrometry and Allied Topics; San Antonio, TX [Google Scholar]
- 202.Jagtap P, Martin RB, Shan B, Stemmer PM, Wang Y MS1-based quantification of low abundance proteins that were not identified using a MS/MS database search approach. 2017. 28th Annual Conference of Association of Biomolecular Research Facilities, San Diego, CA [Google Scholar]
- 203.Asara JM, Tolstikov VV, Aronov P, et al. . Metabolomics research group 2011 study. J Biomol Tech. 2011;22(Suppl):S25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Cheema AK, Asara JM, Wang Y, Neubert TA, Tolstikov V, Turck CW. The ABRF metabolomics research group 2013 study: investigation of spiked compound differences in a human plasma matrix. J Biomol Tech. 2015;26:83–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Metabolomics Research Group (MRG) (2017) ABRF Association of Biomolecular Resource Facilities: https://abrf.org/sites/default/files/temp/RGs/MRG/mrg_session_2017.pdf.
- 206.Ferrando-May E, Hartmann H, Reymann J, et al. ; German BioImaging network . Advanced light microscopy core facilities: balancing service, science and career. Microsc Res Tech. 2016;79:463–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Cole RW, Jinadasa T, Brown CM. Measuring and interpreting point spread functions to determine confocal microscope resolution and ensure quality control. Nat Protoc. 2011;6:1929–1941. [DOI] [PubMed] [Google Scholar]
- 208.Cole RW, Thibault M, Bayles CJ, et al. . International test results for objective lens quality, resolution, spectral accuracy and spectral separation for confocal laser scanning microscopes. Microsc Microanal. 2013;19:1653–1668. [DOI] [PubMed] [Google Scholar]
- 209.Stack RF, Bayles CJ, Girard AM, et al. . Quality assurance testing for modern optical imaging systems. Microsc Microanal. 2011;17:598–606. [DOI] [PubMed] [Google Scholar]
- 210.Brown CM, Reilly A, Cole RW. A quantitative measure of field illumination. J Biomol Tech. 2015;26:37–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Bergeron A, Box A, Chittur S, et al. . The new ABRF flow cytometry research group (FCRG). J Biomol Tech. 2013;24(Suppl):S70. [Google Scholar]
- 212.Tighe S, DeLay M, Lopez P. Overview of the new flow cytometry RG and proposed cell sorting (FACS) microarray study. J Biomol Tech. 2013;24(Suppl):S27. [Google Scholar]
- 213.DeLay M, Lopez P, Bergeron A, et al. . Evaluating effects of cell sorting on cellular integrity. J Biomol Tech. 2014;25(Suppl):S1. [Google Scholar]
- 214.Bierer BE, Crosas M, Pierce HH. Data authorship as an incentive to data sharing. N Engl J Med. 2017;376:1684–1687. [DOI] [PubMed] [Google Scholar]
- 215.Wilkinson MD, Dumontier M, Aalbersberg IJ, et al. . The FAIR Guiding Principles for scientific data management and stewardship. Sci Data. 2016;3:160018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Freedman LP, Venugopalan G, Wisman R. Reproducibility2020: progress and priorities. F1000 Res. 2017;6:604. [DOI] [PMC free article] [PubMed] [Google Scholar]