1
|
Sims AC, Schäfer A, Okuda K, Leist SR, Kocher JF, Cockrell AS, Hawkins PE, Furusho M, Jensen KL, Kyle JE, Burnum-Johnson KE, Stratton KG, Lamar NC, Niccora CD, Weitz KK, Smith RD, Metz TO, Waters KM, Boucher RC, Montgomery SA, Baric RS, Sheahan TP. Dysregulation of lung epithelial cell homeostasis and immunity contributes to Middle East respiratory syndrome coronavirus disease severity. mSphere 2025:e0095124. [PMID: 39882872 DOI: 10.1128/msphere.00951-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Coronaviruses (CoV) emerge suddenly from animal reservoirs to cause novel diseases in new hosts. Discovered in 2012, the Middle East respiratory syndrome coronavirus (MERS-CoV) is endemic in camels in the Middle East and is continually causing local outbreaks and epidemics. While all three newly emerging human CoVs from the past 20 years (SARS-CoV, SARS-CoV-2, and MERS-CoV) cause respiratory disease, each CoV has unique host interactions that drive differential pathogeneses. To better understand the virus and host interactions driving lethal MERS-CoV infection, we performed a longitudinal multi-omics analysis of sublethal and lethal MERS-CoV infection in mice. Significant differences were observed in body weight loss, virus titers, and acute lung injury among lethal and sub-lethal virus doses. Virus-induced apoptosis of type I and II alveolar epithelial cells suggests that loss or dysregulation of these key cell populations was a major driver of severe disease. Omics analysis suggested differential pathogenesis was multi-factorial with clear differences among innate and adaptive immune pathways as well as those that regulate lung epithelial homeostasis. Infection of mice lacking functional T and B cells showed that adaptive immunity was important in controlling viral replication but also increased pathogenesis. In summary, we provide a high-resolution host response atlas for MERS-CoV infection and disease severity. Multi-omics studies of viral pathogenesis offer a unique opportunity to not only better understand the molecular mechanisms of disease but also to identify genes and pathways that can be exploited for therapeutic intervention all of which is important for our future pandemic preparedness.IMPORTANCEEmerging coronaviruses like SARS-CoV, SARS-CoV-2, and MERS-CoV cause a range of disease outcomes in humans from an asymptomatic, moderate, and severe respiratory disease that can progress to death but the factors causing these disparate outcomes remain unclear. Understanding host responses to mild and life-threatening infections provides insight into virus-host networks within and across organ systems that contribute to disease outcomes. We used multi-omics approaches to comprehensively define the host response to moderate and severe MERS-CoV infection. Severe respiratory disease was associated with dysregulation of the immune response. Key lung epithelial cell populations that are essential for lung function get infected and die. Mice lacking key immune cell populations experienced greater virus replication but decreased disease severity implicating the immune system in both protective and pathogenic roles in response to MERS-CoV. These data could be utilized to design new therapeutic strategies targeting specific pathways that contribute to severe disease.
Collapse
Affiliation(s)
- Amy C Sims
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jacob F Kocher
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adam S Cockrell
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Padraig E Hawkins
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Minako Furusho
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kara L Jensen
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer E Kyle
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Kristin E Burnum-Johnson
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Kelly G Stratton
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Natalie C Lamar
- AI & Data Analytics Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Carrie D Niccora
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Katrina M Waters
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Richard C Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephanie A Montgomery
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Timothy P Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
2
|
Devlin KL, Leach DT, Stratton KG, Lamichhane G, Lin VS, Beatty KE. Proteomic characterization of Mycobacterium tuberculosis subjected to carbon starvation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623260. [PMID: 39605331 PMCID: PMC11601416 DOI: 10.1101/2024.11.12.623260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Mycobacterium tuberculosis ( Mtb ) is the causative agent of tuberculosis (TB), the leading cause of infectious-disease related deaths worldwide. TB infections present as a spectrum from active to latent disease. In the human host, Mtb faces hostile environments, such as nutrient deprivation, hypoxia, and low pH. Under these conditions, Mtb can enter a dormant, but viable, state characterized by a lack of cell replication and increased resistance to antibiotics. These dormant Mtb pose a major challenge to curing infections and eradicating TB globally. In the current study, we subjected Mtb to carbon starvation (CS), a culture condition that induces growth stasis and mimics nutrient-starved conditions associated with dormancy in vivo . We provide a detailed analysis of the proteome in CS compared to replicating samples. We observed extensive proteomic reprogramming, with 36% of identified proteins significantly altered in CS. Many enzymes involved in oxidative phosphorylation and lipid metabolism were retained or upregulated in CS. The cell wall biosynthetic machinery was present in CS, although numerous changes in the abundance of peptidoglycan, arabinogalactan, and mycolic acid biosynthetic enzymes likely result in pronounced remodeling of the cell wall. Many clinically approved anti-TB drugs target cell wall biosynthesis, and we found that these enzymes were largely retained in CS. Lastly, we compared our results to those of other dormancy models and propose that CS produces a physiologically-distinct state of stasis compared to hypoxia in Mtb .
Collapse
|
3
|
Van Fossen EM, Kroll JO, Anderson LN, McNaughton AD, Herrera D, Oda Y, Wilson AJ, Nelson WC, Kumar N, Frank AR, Elmore JR, Handakumbura P, Lin VS, Egbert RG. Profiling sorghum-microbe interactions with a specialized photoaffinity probe identifies key sorgoleone binders in Acinetobacter pittii. Appl Environ Microbiol 2024; 90:e0102624. [PMID: 39248464 PMCID: PMC11497774 DOI: 10.1128/aem.01026-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/15/2024] [Indexed: 09/10/2024] Open
Abstract
Interactions between plants and soil microbial communities that benefit plant growth and enhance nutrient acquisition are driven by the selective release of metabolites from plant roots, or root exudation. To investigate these plant-microbe interactions, we developed a photoaffinity probe based on sorgoleone (sorgoleone diazirine alkyne for photoaffinity labeling, SoDA-PAL), a hydrophobic secondary metabolite and allelochemical produced in Sorghum bicolor root exudates. We applied SoDA-PAL to the identification of sorgoleone-binding proteins in Acinetobacter pittii SO1, a potential plant growth-promoting microbe isolated from sorghum rhizosphere soil. Competitive photoaffinity labeling of A. pittii whole cell lysates with SoDA-PAL identified 137 statistically enriched proteins, including putative transporters, transcriptional regulators, and a subset of proteins with predicted enzymatic functions. We performed computational protein modeling and docking with sorgoleone to prioritize candidates for experimental validation and then confirmed binding of sorgoleone to four of these proteins in vitro: the α/β fold hydrolase SrgB (OH685_09420), a fumarylacetoacetase (OH685_02300), a lysophospholipase (OH685_14215), and an unannotated hypothetical protein (OH685_18625). Our application of this specialized sorgoleone-based probe coupled with structural bioinformatics streamlines the identification of microbial proteins involved in metabolite recognition, metabolism, and toxicity, widening our understanding of the range of cellular pathways that can be affected by a plant secondary metabolite.IMPORTANCEHere, we demonstrate that a photoaffinity-based chemical probe modeled after sorgoleone, an important secondary metabolite released by sorghum roots, can be used to identify microbial proteins that directly interact with sorgoleone. We applied this probe to the sorghum-associated bacterium Acinetobacter pittii and showed that probe labeling is dose-dependent and sensitive to competition with purified sorgoleone. Coupling the probe with proteomics and computational analysis facilitated the identification of putative sorgoleone binders, including a protein implicated in a conserved pathway essential for sorgoleone catabolism. We anticipate that discoveries seeded by this workflow will expand our understanding of the molecular mechanisms by which specific metabolites in root exudates shape the sorghum rhizosphere microbiome.
Collapse
Affiliation(s)
- Elise M. Van Fossen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Jared O. Kroll
- Energy Processes and Materials Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Lindsey N. Anderson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Andrew D. McNaughton
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Daisy Herrera
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Yasuhiro Oda
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Andrew J. Wilson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - William C. Nelson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Neeraj Kumar
- Advanced Computing, Mathematics, and Data Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Andrew R. Frank
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Joshua R. Elmore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Pubudu Handakumbura
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Vivian S. Lin
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Robert G. Egbert
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| |
Collapse
|
4
|
Eisfeld AJ, Anderson LN, Fan S, Walters KB, Halfmann PJ, Westhoff Smith D, Thackray LB, Tan Q, Sims AC, Menachery VD, Schäfer A, Sheahan TP, Cockrell AS, Stratton KG, Webb-Robertson BJM, Kyle JE, Burnum-Johnson KE, Kim YM, Nicora CD, Peralta Z, N'jai AU, Sahr F, van Bakel H, Diamond MS, Baric RS, Metz TO, Smith RD, Kawaoka Y, Waters KM. A compendium of multi-omics data illuminating host responses to lethal human virus infections. Sci Data 2024; 11:328. [PMID: 38565538 PMCID: PMC10987564 DOI: 10.1038/s41597-024-03124-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Human infections caused by viral pathogens trigger a complex gamut of host responses that limit disease, resolve infection, generate immunity, and contribute to severe disease or death. Here, we present experimental methods and multi-omics data capture approaches representing the global host response to infection generated from 45 individual experiments involving human viruses from the Orthomyxoviridae, Filoviridae, Flaviviridae, and Coronaviridae families. Analogous experimental designs were implemented across human or mouse host model systems, longitudinal samples were collected over defined time courses, and global multi-omics data (transcriptomics, proteomics, metabolomics, and lipidomics) were acquired by microarray, RNA sequencing, or mass spectrometry analyses. For comparison, we have included transcriptomics datasets from cells treated with type I and type II human interferon. Raw multi-omics data and metadata were deposited in public repositories, and we provide a central location linking the raw data with experimental metadata and ready-to-use, quality-controlled, statistically processed multi-omics datasets not previously available in any public repository. This compendium of infection-induced host response data for reuse will be useful for those endeavouring to understand viral disease pathophysiology and network biology.
Collapse
Affiliation(s)
- Amie J Eisfeld
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Lindsey N Anderson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Shufang Fan
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Coronavirus and Other Respiratory Viruses Laboratory Branch (CRVLB), Coronavirus and Other Respiratory Viruses Division (CORVD), National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, GA, 30329, USA
| | - Kevin B Walters
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - Peter J Halfmann
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Danielle Westhoff Smith
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Larissa B Thackray
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Qing Tan
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Amy C Sims
- Department of Epidemiology, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
- Nuclear, Chemistry, and Biosciences Division; National Security Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Vineet D Menachery
- Department of Epidemiology, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
| | - Timothy P Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Adam S Cockrell
- Department of Epidemiology, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
- Solid Biosciences, Charlston, MA, 02139, USA
| | - Kelly G Stratton
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Bobbie-Jo M Webb-Robertson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Jennifer E Kyle
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Kristin E Burnum-Johnson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Young-Mo Kim
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Carrie D Nicora
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Zuleyma Peralta
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, 10029, USA
- Partillion Bioscience, Los Angeles, CA, 90064, USA
| | - Alhaji U N'jai
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Biological Sciences, Fourah Bay College, Freetown, Sierra Leone
- Department of Microbiology, College of Medicine and Allied Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
- Department of Medical Education, California University of Science and Medicine, Colton, CA, 92324, USA
| | - Foday Sahr
- Department of Microbiology, College of Medicine and Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY, 10029, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, North Carolina, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Thomas O Metz
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Richard D Smith
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 108-8639, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, 108-8639, Japan
| | - Katrina M Waters
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA.
| |
Collapse
|
5
|
Bramer LM, Dixon HM, Degnan DJ, Rohlman D, Herbstman JB, Anderson KA, Waters KM. Expanding the access of wearable silicone wristbands in community-engaged research through best practices in data analysis and integration. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2024; 29:170-186. [PMID: 38160278 PMCID: PMC10766083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Wearable silicone wristbands are a rapidly growing exposure assessment technology that offer researchers the ability to study previously inaccessible cohorts and have the potential to provide a more comprehensive picture of chemical exposure within diverse communities. However, there are no established best practices for analyzing the data within a study or across multiple studies, thereby limiting impact and access of these data for larger meta-analyses. We utilize data from three studies, from over 600 wristbands worn by participants in New York City and Eugene, Oregon, to present a first-of-its-kind manuscript detailing wristband data properties. We further discuss and provide concrete examples of key areas and considerations in common statistical modeling methods where best practices must be established to enable meta-analyses and integration of data from multiple studies. Finally, we detail important and challenging aspects of machine learning, meta-analysis, and data integration that researchers will face in order to extend beyond the limited scope of individual studies focused on specific populations.
Collapse
Affiliation(s)
- Lisa M Bramer
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd Richland, WA 99354, United States,
| | | | | | | | | | | | | |
Collapse
|
6
|
Sims EK, Kulkarni A, Hull A, Woerner SE, Cabrera S, Mastrandrea LD, Hammoud B, Sarkar S, Nakayasu ES, Mastracci TL, Perkins SM, Ouyang F, Webb-Robertson BJ, Enriquez JR, Tersey SA, Evans-Molina C, Long SA, Blanchfield L, Gerner EW, Mirmira RG, DiMeglio LA. Inhibition of polyamine biosynthesis preserves β cell function in type 1 diabetes. Cell Rep Med 2023; 4:101261. [PMID: 37918404 PMCID: PMC10694631 DOI: 10.1016/j.xcrm.2023.101261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/18/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023]
Abstract
In preclinical models, α-difluoromethylornithine (DFMO), an ornithine decarboxylase (ODC) inhibitor, delays the onset of type 1 diabetes (T1D) by reducing β cell stress. However, the mechanism of DFMO action and its human tolerability remain unclear. In this study, we show that mice with β cell ODC deletion are protected against toxin-induced diabetes, suggesting a cell-autonomous role of ODC during β cell stress. In a randomized controlled trial (ClinicalTrials.gov: NCT02384889) involving 41 recent-onset T1D subjects (3:1 drug:placebo) over a 3-month treatment period with a 3-month follow-up, DFMO (125-1,000 mg/m2) is shown to meet its primary outcome of safety and tolerability. DFMO dose-dependently reduces urinary putrescine levels and, at higher doses, preserves C-peptide area under the curve without apparent immunomodulation. Transcriptomics and proteomics of DFMO-treated human islets exposed to cytokine stress reveal alterations in mRNA translation, nascent protein transport, and protein secretion. These findings suggest that DFMO may preserve β cell function in T1D through islet cell-autonomous effects.
Collapse
Affiliation(s)
- Emily K Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Abhishek Kulkarni
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Audrey Hull
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Nationwide Children's Hospital Pediatric Residency Program, Columbus, OH 43205, USA
| | - Stephanie E Woerner
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Susanne Cabrera
- Department of Pediatrics, Section of Endocrinology and Diabetes, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Lucy D Mastrandrea
- Division of Pediatric Endocrinology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Batoul Hammoud
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| | - Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Teresa L Mastracci
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Susan M Perkins
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Fangqian Ouyang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Jacob R Enriquez
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah A Tersey
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Carmella Evans-Molina
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medicine and the Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - S Alice Long
- Benaroya Research Institute, Center for Translational Immunology, Seattle, WA 98101, USA
| | - Lori Blanchfield
- Benaroya Research Institute, Center for Translational Immunology, Seattle, WA 98101, USA
| | | | - Raghavendra G Mirmira
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA.
| | - Linda A DiMeglio
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
7
|
Bramer LM, Dixon HM, Degnan DJ, Rohlman D, Herbstman JB, Anderson KA, Waters KM. Expanding the access of wearable silicone wristbands in community-engaged research through best practices in data analysis and integration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560217. [PMID: 37873084 PMCID: PMC10592864 DOI: 10.1101/2023.09.29.560217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Wearable silicone wristbands are a rapidly growing exposure assessment technology that offer researchers the ability to study previously inaccessible cohorts and have the potential to provide a more comprehensive picture of chemical exposure within diverse communities. However, there are no established best practices for analyzing the data within a study or across multiple studies, thereby limiting impact and access of these data for larger meta-analyses. We utilize data from three studies, from over 600 wristbands worn by participants in New York City and Eugene, Oregon, to present a first-of-its-kind manuscript detailing wristband data properties. We further discuss and provide concrete examples of key areas and considerations in common statistical modeling methods where best practices must be established to enable meta-analyses and integration of data from multiple studies. Finally, we detail important and challenging aspects of machine learning, meta-analysis, and data integration that researchers will face in order to extend beyond the limited scope of individual studies focused on specific populations.
Collapse
Affiliation(s)
- Lisa M Bramer
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd Richland, WA 99354, United States
| | - Holly M Dixon
- Environmental and Molecular Toxicology, Oregon State University, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States
| | - David J Degnan
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd Richland, WA 99354, United States
| | - Diana Rohlman
- College of Health, Oregon State University, 103 SW Memorial Place, Corvallis, OR 97331, United States
| | - Julie B Herbstman
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York City, NY 10032, United States
| | - Kim A Anderson
- Environmental and Molecular Toxicology, Oregon State University, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States
| | - Katrina M Waters
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd Richland, WA 99354, United States
- Environmental and Molecular Toxicology, Oregon State University, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States
| |
Collapse
|
8
|
Coradetti ST, Adamczyk PA, Liu D, Gao Y, Otoupal PB, Geiselman GM, Webb-Robertson BJM, Burnet MC, Kim YM, Burnum-Johnson KE, Magnuson J, Gladden JM. Engineering transcriptional regulation of pentose metabolism in Rhodosporidium toruloides for improved conversion of xylose to bioproducts. Microb Cell Fact 2023; 22:144. [PMID: 37537586 PMCID: PMC10398944 DOI: 10.1186/s12934-023-02148-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/13/2023] [Indexed: 08/05/2023] Open
Abstract
Efficient conversion of pentose sugars remains a significant barrier to the replacement of petroleum-derived chemicals with plant biomass-derived bioproducts. While the oleaginous yeast Rhodosporidium toruloides (also known as Rhodotorula toruloides) has a relatively robust native metabolism of pentose sugars compared to other wild yeasts, faster assimilation of those sugars will be required for industrial utilization of pentoses. To increase the rate of pentose assimilation in R. toruloides, we leveraged previously reported high-throughput fitness data to identify potential regulators of pentose catabolism. Two genes were selected for further investigation, a putative transcription factor (RTO4_12978, Pnt1) and a homolog of a glucose transceptor involved in carbon catabolite repression (RTO4_11990). Overexpression of Pnt1 increased the specific growth rate approximately twofold early in cultures on xylose and increased the maximum specific growth by 18% while decreasing accumulation of arabitol and xylitol in fast-growing cultures. Improved growth dynamics on xylose translated to a 120% increase in the overall rate of xylose conversion to fatty alcohols in batch culture. Proteomic analysis confirmed that Pnt1 is a major regulator of pentose catabolism in R. toruloides. Deletion of RTO4_11990 increased the growth rate on xylose, but did not relieve carbon catabolite repression in the presence of glucose. Carbon catabolite repression signaling networks remain poorly characterized in R. toruloides and likely comprise a different set of proteins than those mainly characterized in ascomycete fungi.
Collapse
Affiliation(s)
- Samuel T. Coradetti
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Sandia National Laboratories, Livermore, CA USA
- Present Address: Agricultural Research Service, United States Department of Agriculture, Ithaca, NY USA
| | - Paul A. Adamczyk
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Sandia National Laboratories, Livermore, CA USA
| | - Di Liu
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Sandia National Laboratories, Livermore, CA USA
| | - Yuqian Gao
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Pacific Northwest National Laboratory, Richland, WA USA
| | - Peter B. Otoupal
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Sandia National Laboratories, Livermore, CA USA
| | - Gina M. Geiselman
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Sandia National Laboratories, Livermore, CA USA
| | | | | | - Young-Mo Kim
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Pacific Northwest National Laboratory, Richland, WA USA
| | - Kristin E. Burnum-Johnson
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Pacific Northwest National Laboratory, Richland, WA USA
| | - Jon Magnuson
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Pacific Northwest National Laboratory, Richland, WA USA
| | - John M. Gladden
- DOE Agile Biofoundry, 5885 Hollis Street, Fourth Floor, Emeryville, CA 94608 USA
- Sandia National Laboratories, Livermore, CA USA
- Joint BioEnergy Institute, Emeryville, CA USA
| |
Collapse
|
9
|
Nehmeh B, Haydous F, Akoury E. Mass calibrants for positive chemical ionization-high resolution mass spectrometry (CI-HRMS) for the identification of unknown compounds using accurate mass measurements. RSC Adv 2023; 13:14001-14009. [PMID: 37181506 PMCID: PMC10167574 DOI: 10.1039/d3ra01977b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/01/2023] [Indexed: 05/16/2023] Open
Abstract
Gas Chromatography-Electron Ionization-Mass Spectrometry (GC-EI-MS) is still the most routinely performed method for metabolite profiling as compared to other hyphenated techniques. But when it comes to identification of unknown compounds, information on the molecular weight is not readily available because the molecular ion is not always found with electron ionization (EI). Thus, the use of chemical ionization (CI) is envisaged that commonly produces the molecular ion; in combination with accurate mass measurement, this technique would further allow for calculation of sum formulas of those compounds. However, for proper accuracy of analysis, a mass calibrant is needed. We set out to find a commercially available reference material with mass peaks that would qualify the substance as mass calibrant under CI conditions. Six commercially available mass calibrants, FC 43, PFK, Ultramark 1621, Ultramark 3200F, Triton X-100, and PEG 1000, were tested under CI conditions to understand their fragmentation behavior. Our findings indicate that Ultramark 1621 and PFK best fit the expectations of a mass calibrant for HRMS analysis whereby PFK provided a fragmentation pattern similar to EI outcomes thus enabling the use of mass reference tables commonly provided within commercial mass spectrometers. On the other hand, Ultramark 1621 is a mixture of fluorinated phosphazines that shows stable fragment intensities.
Collapse
Affiliation(s)
- Bilal Nehmeh
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University Beirut 1102-2801 Lebanon
| | - Fatima Haydous
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University Beirut 1102-2801 Lebanon
| | - Elias Akoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University Beirut 1102-2801 Lebanon
| |
Collapse
|
10
|
Bilbao A, Munoz N, Kim J, Orton DJ, Gao Y, Poorey K, Pomraning KR, Weitz K, Burnet M, Nicora CD, Wilton R, Deng S, Dai Z, Oksen E, Gee A, Fasani RA, Tsalenko A, Tanjore D, Gardner J, Smith RD, Michener JK, Gladden JM, Baker ES, Petzold CJ, Kim YM, Apffel A, Magnuson JK, Burnum-Johnson KE. PeakDecoder enables machine learning-based metabolite annotation and accurate profiling in multidimensional mass spectrometry measurements. Nat Commun 2023; 14:2461. [PMID: 37117207 PMCID: PMC10147702 DOI: 10.1038/s41467-023-37031-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 02/24/2023] [Indexed: 04/30/2023] Open
Abstract
Multidimensional measurements using state-of-the-art separations and mass spectrometry provide advantages in untargeted metabolomics analyses for studying biological and environmental bio-chemical processes. However, the lack of rapid analytical methods and robust algorithms for these heterogeneous data has limited its application. Here, we develop and evaluate a sensitive and high-throughput analytical and computational workflow to enable accurate metabolite profiling. Our workflow combines liquid chromatography, ion mobility spectrometry and data-independent acquisition mass spectrometry with PeakDecoder, a machine learning-based algorithm that learns to distinguish true co-elution and co-mobility from raw data and calculates metabolite identification error rates. We apply PeakDecoder for metabolite profiling of various engineered strains of Aspergillus pseudoterreus, Aspergillus niger, Pseudomonas putida and Rhodosporidium toruloides. Results, validated manually and against selected reaction monitoring and gas-chromatography platforms, show that 2683 features could be confidently annotated and quantified across 116 microbial sample runs using a library built from 64 standards.
Collapse
Affiliation(s)
- Aivett Bilbao
- Pacific Northwest National Laboratory, Richland, WA, USA.
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA.
| | - Nathalie Munoz
- Pacific Northwest National Laboratory, Richland, WA, USA
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
| | - Joonhoon Kim
- Pacific Northwest National Laboratory, Richland, WA, USA
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
| | - Daniel J Orton
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Yuqian Gao
- Pacific Northwest National Laboratory, Richland, WA, USA
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
| | | | - Kyle R Pomraning
- Pacific Northwest National Laboratory, Richland, WA, USA
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
| | - Karl Weitz
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Meagan Burnet
- Pacific Northwest National Laboratory, Richland, WA, USA
| | | | - Rosemarie Wilton
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
- Argonne National Laboratory, Lemont, IL, USA
| | - Shuang Deng
- Pacific Northwest National Laboratory, Richland, WA, USA
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
| | - Ziyu Dai
- Pacific Northwest National Laboratory, Richland, WA, USA
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
| | - Ethan Oksen
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Aaron Gee
- Agilent Research Laboratories, Agilent Technologies, Santa Clara, CA, USA
| | - Rick A Fasani
- Agilent Research Laboratories, Agilent Technologies, Santa Clara, CA, USA
| | - Anya Tsalenko
- Agilent Research Laboratories, Agilent Technologies, Santa Clara, CA, USA
| | - Deepti Tanjore
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - James Gardner
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | - Joshua K Michener
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
- Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - John M Gladden
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
- Sandia National Laboratory, Livermore, CA, USA
| | - Erin S Baker
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Christopher J Petzold
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Young-Mo Kim
- Pacific Northwest National Laboratory, Richland, WA, USA
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
| | - Alex Apffel
- Agilent Research Laboratories, Agilent Technologies, Santa Clara, CA, USA
| | - Jon K Magnuson
- Pacific Northwest National Laboratory, Richland, WA, USA
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA
| | - Kristin E Burnum-Johnson
- Pacific Northwest National Laboratory, Richland, WA, USA.
- US Department of Energy, Agile BioFoundry, Emeryville, CA, USA.
| |
Collapse
|
11
|
Bramer LM, Hontz RD, Eisfeld AJ, Sims AC, Kim YM, Stratton KG, Nicora CD, Gritsenko MA, Schepmoes AA, Akasaka O, Koga M, Tsutsumi T, Nakamura M, Nakachi I, Baba R, Tateno H, Suzuki S, Nakajima H, Kato H, Ishida K, Ishii M, Uwamino Y, Mitamura K, Paurus VL, Nakayasu ES, Attah IK, Letizia AG, Waters KM, Metz TO, Corson K, Kawaoka Y, Gerbasi VR, Yotsuyanagi H, Iwatsuki-Horimoto K. Multi-omics of NET formation and correlations with CNDP1, PSPB, and L-cystine levels in severe and mild COVID-19 infections. Heliyon 2023; 9:e13795. [PMID: 36915486 PMCID: PMC9988701 DOI: 10.1016/j.heliyon.2023.e13795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 03/09/2023] Open
Abstract
The detailed mechanisms of COVID-19 infection pathology remain poorly understood. To improve our understanding of SARS-CoV-2 pathology, we performed a multi-omics and correlative analysis of an immunologically naïve SARS-CoV-2 clinical cohort from blood plasma of uninfected controls, mild, and severe infections. Consistent with previous observations, severe patient populations showed an elevation of pulmonary surfactant levels. Intriguingly, mild patients showed a statistically significant elevation in the carnosine dipeptidase modifying enzyme (CNDP1). Mild and severe patient populations showed a strong elevation in the metabolite L-cystine (oxidized form of the amino acid cysteine) and enzymes with roles in glutathione metabolism. Neutrophil extracellular traps (NETs) were observed in both mild and severe populations, and NET formation was higher in severe vs. mild samples. Our correlative analysis suggests a potential protective role for CNDP1 in suppressing PSPB release from the pulmonary space whereas NET formation correlates with increased PSPB levels and disease severity. In our discussion we put forward a possible model where NET formation drives pulmonary occlusions and CNDP1 promotes antioxidation, pleiotropic immune responses, and vasodilation by accelerating histamine synthesis.
Collapse
Affiliation(s)
- Lisa M Bramer
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Robert D Hontz
- U.S. Naval Medical Research Unit No. TWO (NAMRU-2), Singapore, Singapore
| | - Amie J Eisfeld
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Amy C Sims
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Young-Mo Kim
- Pacific Northwest National Laboratory, Richland, WA, USA
| | | | | | | | | | - Osamu Akasaka
- Emergency Medical Center, Fujisawa City Hospital 2-6-1 Fujisawa, Fujisawa, Japan
| | - Michiko Koga
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Takeya Tsutsumi
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Morio Nakamura
- Department of Pulmonary Medicine, Tokyo Saiseikai Central, Tokyo, Japan
| | - Ichiro Nakachi
- Pulmonary Division, Department of Internal Medicine, Utsunomiya Hospital, Utsunomiya, Japan
| | - Rie Baba
- Pulmonary Division, Department of Internal Medicine, Utsunomiya Hospital, Utsunomiya, Japan
| | - Hiroki Tateno
- Department of Pulmonary Medicine, Saitama City Hospital, Saitama, Japan
| | - Shoji Suzuki
- Department of Pulmonary Medicine, Saitama City Hospital, Saitama, Japan
| | - Hideaki Nakajima
- Department of Hematology and Clinical Immunology, University School of Medicine, Yokohama, Japan
| | - Hideaki Kato
- Department of Hematology and Clinical Immunology, University School of Medicine, Yokohama, Japan
| | | | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoshifumi Uwamino
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Mitamura
- Division of Infection Control, Eiju General Hospital, Tokyo, Japan
| | | | | | - Isaac K Attah
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Andrew G Letizia
- U.S. Naval Medical Research Unit No. TWO (NAMRU-2), Singapore, Singapore
| | | | - Thomas O Metz
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Karen Corson
- U.S. Naval Medical Research Unit No. TWO (NAMRU-2), Singapore, Singapore
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Department of Microbiology and Immunology, Japan.,International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | - Hiroshi Yotsuyanagi
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo
| | | |
Collapse
|
12
|
Carfagno A, Lin SC, Chafran L, Akhrymuk I, Callahan V, Po M, Zhu Y, Altalhi A, Durkin DP, Russo P, Vliet KA, Webb-Robertson BJ, Kehn-Hall K, Bishop B. Bioprospecting the American Alligator Peptidome for antiviral peptides against Venezuelan equine encephalitis virus. Proteomics 2023; 23:e2200237. [PMID: 36480152 DOI: 10.1002/pmic.202200237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
The innate immune protection provided by cationic antimicrobial peptides (CAMPs) has been shown to extend to antiviral activity, with putative mechanisms of action including direct interaction with host cells or pathogen membranes. The lack of therapeutics available for the treatment of viruses such as Venezuelan equine encephalitis virus (VEEV) underscores the urgency of novel strategies for antiviral discovery. American alligator plasma has been shown to exhibit strong in vitro antibacterial activity, and functionalized hydrogel particles have been successfully employed for the identification of specific CAMPs from alligator plasma. Here, a novel bait strategy in which particles were encapsulated in membranes from either healthy or VEEV-infected cells was implemented to identify peptides preferentially targeting infected cells for subsequent evaluation of antiviral activity. Statistical analysis of peptide identification results was used to select five candidate peptides for testing, of which one exhibited a dose-dependent inhibition of VEEV and also significantly inhibited infectious titers. Results suggest our bioprospecting strategy provides a versatile platform that may be adapted for antiviral peptide identification from complex biological samples.
Collapse
Affiliation(s)
- Amy Carfagno
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, USA
| | - Shih-Chao Lin
- School of Systems Biology, George Mason University, Manassas, Virginia, USA.,National Taiwan Ocean University, Keelung City, Taiwan
| | - Liana Chafran
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, USA
| | - Ivan Akhrymuk
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Victoria Callahan
- School of Systems Biology, George Mason University, Manassas, Virginia, USA
| | - Marynet Po
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, USA
| | - Yaling Zhu
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, USA
| | - Amaal Altalhi
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, USA
| | - David P Durkin
- Chemistry Department, U.S. Naval Academy, Annapolis, Maryland, USA
| | - Paul Russo
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, USA
| | - Kent A Vliet
- Department of Biology, University of Florida, Gainesville, Florida, USA
| | | | - Kylene Kehn-Hall
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA.,Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Barney Bishop
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, USA
| |
Collapse
|
13
|
Degnan DJ, Stratton KG, Richardson R, Claborne D, Martin EA, Johnson NA, Leach D, Webb-Robertson BJM, Bramer LM. pmartR 2.0: A Quality Control, Visualization, and Statistics Pipeline for Multiple Omics Datatypes. J Proteome Res 2023; 22:570-576. [PMID: 36622218 DOI: 10.1021/acs.jproteome.2c00610] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The pmartR (https://github.com/pmartR/pmartR) package was designed for the quality control (QC) and analysis of mass spectrometry data, tailored to specific characteristics of proteomic (isobaric or labeled), metabolomic, and lipidomic data sets. Since its initial release, the tool has been expanded to address the needs of its growing userbase and now includes QC and statistics for nuclear magnetic resonance metabolomic data, and leverages the DESeq2, edgeR, and limma-voom R packages for transcriptomic data analyses. These improvements have made progress toward a unified omics processing pipeline for ease of reporting and streamlined statistical purposes. The package's statistics and visualization capabilities have also been expanded by adding support for paired data and by integrating pmartR with the trelliscopejs R package for the quick creation of trellis displays (https://github.com/hafen/trelliscopejs). Here, we present relevant examples of each of these enhancements to pmartR and highlight how each new feature benefits the omics community.
Collapse
|
14
|
Itaconic acid production is regulated by LaeA in Aspergillus pseudoterreus. Metab Eng Commun 2022; 15:e00203. [PMID: 36065328 PMCID: PMC9440423 DOI: 10.1016/j.mec.2022.e00203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/08/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
The global regulator LaeA controls secondary metabolism in diverse Aspergillus species. Here we explored its role in regulation of itaconic acid production in Aspergillus pseudoterreus. To understand its role in regulating metabolism, we deleted and overexpressed laeA, and assessed the transcriptome, proteome, and secreted metabolome prior to and during initiation of phosphate limitation induced itaconic acid production. We found that secondary metabolite clusters, including the itaconic acid biosynthetic gene cluster, are regulated by laeA and that laeA is required for high yield production of itaconic acid. Overexpression of LaeA improves itaconic acid yield at the expense of biomass by increasing the expression of key biosynthetic pathway enzymes and attenuating the expression of genes involved in phosphate acquisition and scavenging. Increased yield was observed in optimized conditions as well as conditions containing excess nutrients that may be present in inexpensive sugar containing feedstocks such as excess phosphate or complex nutrient sources. This suggests that global regulators of metabolism may be useful targets for engineering metabolic flux that is robust to environmental heterogeneity. The Itaconic acid biosynthetic gene cluster is regulated by laeA. LaeA is required for production of itaconic acid. Overexpression of laeA attenuates genes involved in phosphate acquisition. Global regulator engineering increases robustness of itaconic acid production.
Collapse
|
15
|
Chion M, Carapito C, Bertrand F. Accounting for multiple imputation-induced variability for differential analysis in mass spectrometry-based label-free quantitative proteomics. PLoS Comput Biol 2022; 18:e1010420. [PMID: 36037245 PMCID: PMC9462777 DOI: 10.1371/journal.pcbi.1010420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/09/2022] [Accepted: 07/21/2022] [Indexed: 11/20/2022] Open
Abstract
Imputing missing values is common practice in label-free quantitative proteomics. Imputation aims at replacing a missing value with a user-defined one. However, the imputation itself may not be optimally considered downstream of the imputation process, as imputed datasets are often considered as if they had always been complete. Hence, the uncertainty due to the imputation is not adequately taken into account. We provide a rigorous multiple imputation strategy, leading to a less biased estimation of the parameters' variability thanks to Rubin's rules. The imputation-based peptide's intensities' variance estimator is then moderated using Bayesian hierarchical models. This estimator is finally included in moderated t-test statistics to provide differential analyses results. This workflow can be used both at peptide and protein-level in quantification datasets. Indeed, an aggregation step is included for protein-level results based on peptide-level quantification data. Our methodology, named mi4p, was compared to the state-of-the-art limma workflow implemented in the DAPAR R package, both on simulated and real datasets. We observed a trade-off between sensitivity and specificity, while the overall performance of mi4p outperforms DAPAR in terms of F-Score.
Collapse
Affiliation(s)
- Marie Chion
- Institut de Recherche Mathématique Avancée, UMR 7501, CNRS-Université de Strasbourg, Strasbourg, France
- Laboratoire de Spectrométrie de Masse Bio-Organique, Institut Pluridisciplinaire Hubert Curien, UMR 7178, CNRS-Université de Strasbourg, Strasbourg, France
- Laboratoire Mathématiques appliquées à Paris 5, UMR 8145, CNRS-Université Paris Cité, Paris, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse Bio-Organique, Institut Pluridisciplinaire Hubert Curien, UMR 7178, CNRS-Université de Strasbourg, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFi - FR2048, 67087 Strasbourg, France
| | - Frédéric Bertrand
- Institut de Recherche Mathématique Avancée, UMR 7501, CNRS-Université de Strasbourg, Strasbourg, France
- Laboratoire Informatique et Société Numérique, Université de Technologie de Troyes, Troyes, France
| |
Collapse
|
16
|
Moraes ECDS, Martins-Gonçalves R, da Silva LR, Mandacaru SC, Melo RM, Azevedo-Quintanilha I, Perales J, Bozza FA, Souza TML, Castro-Faria-Neto HC, Hottz ED, Bozza PT, Trugilho MRO. Proteomic Profile of Procoagulant Extracellular Vesicles Reflects Complement System Activation and Platelet Hyperreactivity of Patients with Severe COVID-19. Front Cell Infect Microbiol 2022; 12:926352. [PMID: 35937696 PMCID: PMC9354812 DOI: 10.3389/fcimb.2022.926352] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/20/2022] [Indexed: 01/08/2023] Open
Abstract
Background Extracellular vesicles (EVs) are a valuable source of biomarkers and display the pathophysiological status of various diseases. In COVID-19, EVs have been explored in several studies for their ability to reflect molecular changes caused by SARS-CoV-2. Here we provide insights into the roles of EVs in pathological processes associated with the progression and severity of COVID-19. Methods In this study, we used a label-free shotgun proteomic approach to identify and quantify alterations in EV protein abundance in severe COVID-19 patients. We isolated plasma extracellular vesicles from healthy donors and patients with severe COVID-19 by size exclusion chromatography (SEC). Then, flow cytometry was performed to assess the origin of EVs and to investigate the presence of circulating procoagulant EVs in COVID-19 patients. A total protein extraction was performed, and samples were analyzed by nLC-MS/MS in a Q-Exactive HF-X. Finally, computational analysis was applied to signify biological processes related to disease pathogenesis. Results We report significant changes in the proteome of EVs from patients with severe COVID-19. Flow cytometry experiments indicated an increase in total circulating EVs and with tissue factor (TF) dependent procoagulant activity. Differentially expressed proteins in the disease groups were associated with complement and coagulation cascades, platelet degranulation, and acute inflammatory response. Conclusions The proteomic data reinforce the changes in the proteome of extracellular vesicles from patients infected with SARS-CoV-2 and suggest a role for EVs in severe COVID-19.
Collapse
Affiliation(s)
- Emilly Caroline dos Santos Moraes
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Remy Martins-Gonçalves
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Luana Rocha da Silva
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Samuel Coelho Mandacaru
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Reynaldo Magalhães Melo
- Laboratory Protein Chemistry and Biochemistry and Laboratory of Gene Biology, Department of Cell Biology, University of Brasília, Brasília, Brazil
| | | | - Jonas Perales
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Fernando A. Bozza
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- D’Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Thiago Moreno Lopes Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Eugenio D. Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Patricia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Monique R. O. Trugilho
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Plancade S, Berland M, Blein-Nicolas M, Langella O, Bassignani A, Juste C. A combined test for feature selection on sparse metaproteomics data-an alternative to missing value imputation. PeerJ 2022; 10:e13525. [PMID: 35769140 PMCID: PMC9235818 DOI: 10.7717/peerj.13525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 05/11/2022] [Indexed: 01/18/2023] Open
Abstract
One of the difficulties encountered in the statistical analysis of metaproteomics data is the high proportion of missing values, which are usually treated by imputation. Nevertheless, imputation methods are based on restrictive assumptions regarding missingness mechanisms, namely "at random" or "not at random". To circumvent these limitations in the context of feature selection in a multi-class comparison, we propose a univariate selection method that combines a test of association between missingness and classes, and a test for difference of observed intensities between classes. This approach implicitly handles both missingness mechanisms. We performed a quantitative and qualitative comparison of our procedure with imputation-based feature selection methods on two experimental data sets, as well as simulated data with various scenarios regarding the missingness mechanisms and the nature of the difference of expression (differential intensity or differential presence). Whereas we observed similar performances in terms of prediction on the experimental data set, the feature ranking and selection from various imputation-based methods were strongly divergent. We showed that the combined test reaches a compromise by correlating reasonably with other methods, and remains efficient in all simulated scenarios unlike imputation-based feature selection methods.
Collapse
Affiliation(s)
- Sandra Plancade
- UR875 MIAT, Université fédérale de Toulouse, INRAE, Castanet-Tolosan, France
| | - Magali Berland
- Université Paris-Saclay, INRAE, MGP, Jouy en Josas, France
| | - Mélisande Blein-Nicolas
- Université Paris-Saclay, CNRS, INRAE, AgroParisTech, GQE-Le Moulon, Gif-sur-Yvette, France,Université Paris-Saclay, CNRS, INRAE, AgroParisTech, PAPPSO, Gif-sur-Yvette, France
| | - Olivier Langella
- Université Paris-Saclay, CNRS, INRAE, AgroParisTech, GQE-Le Moulon, Gif-sur-Yvette, France,Université Paris-Saclay, CNRS, INRAE, AgroParisTech, PAPPSO, Gif-sur-Yvette, France
| | - Ariane Bassignani
- Université Paris-Saclay, INRAE, MGP, Jouy en Josas, France,Université Paris-Saclay, CNRS, INRAE, AgroParisTech, PAPPSO, Gif-sur-Yvette, France
| | - Catherine Juste
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParis Tech, Jouy-en-Josas, France
| |
Collapse
|
18
|
Unfolded Protein Response Inhibition Reduces Middle East Respiratory Syndrome Coronavirus-Induced Acute Lung Injury. mBio 2021; 12:e0157221. [PMID: 34372702 PMCID: PMC8406233 DOI: 10.1128/mbio.01572-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tissue- and cell-specific expression patterns are highly variable within and across individuals, leading to altered host responses after acute virus infection. Unraveling key tissue-specific response patterns provides novel opportunities for defining fundamental mechanisms of virus-host interaction in disease and the identification of critical tissue-specific networks for disease intervention in the lung. Currently, there are no approved therapeutics for Middle East respiratory syndrome coronavirus (MERS-CoV) patients, and little is understood about how lung cell types contribute to disease outcomes. MERS-CoV replicates equivalently in primary human lung microvascular endothelial cells (MVE) and fibroblasts (FB) and to equivalent peak titers but with slower replication kinetics in human airway epithelial cell cultures (HAE). However, only infected MVE demonstrate observable virus-induced cytopathic effect. To explore mechanisms leading to reduced MVE viability, donor-matched human lung MVE, HAE, and FB were infected, and their transcriptomes, proteomes, and lipidomes were monitored over time. Validated functional enrichment analysis demonstrated that MERS-CoV-infected MVE were dying via an unfolded protein response (UPR)-mediated apoptosis. Pharmacologic manipulation of the UPR in MERS-CoV-infected primary lung cells reduced viral titers and in male mice improved respiratory function with accompanying reductions in weight loss, pathological signatures of acute lung injury, and times to recovery. Systems biology analysis and validation studies of global kinetic transcript, protein, and lipid data sets confirmed that inhibition of host stress pathways that are differentially regulated following MERS-CoV infection of different tissue types can alleviate symptom progression to end-stage lung disease commonly seen following emerging coronavirus outbreaks.
Collapse
|
19
|
Escobar EE, Venkat Ramani MK, Zhang Y, Brodbelt JS. Evaluating Spatiotemporal Dynamics of Phosphorylation of RNA Polymerase II Carboxy-Terminal Domain by Ultraviolet Photodissociation Mass Spectrometry. J Am Chem Soc 2021; 143:8488-8498. [PMID: 34053220 DOI: 10.1021/jacs.1c03321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The critical role of site-specific phosphorylation in eukaryotic transcription has motivated efforts to decipher the complex phosphorylation patterns exhibited by the carboxyl-terminal domain (CTD) of RNA polymerase II. Phosphorylation remains a challenging post-translational modification to characterize by mass spectrometry owing to the labile phosphate ester linkage and low stoichiometric prevalence, two features that complicate analysis by high-throughput MS/MS methods. Identifying phosphorylation sites represents one significant hurdle in decrypting the CTD phosphorylation, a problem exaggerated by a large number of potential phosphorylation sites. An even greater obstacle is decoding the dynamic phosphorylation pattern along the length of the periodic CTD sequence. Ultraviolet photodissociation (UVPD) is a high-energy ion activation method that provides ample backbone cleavages of peptides while preserving labile post-translational modifications that facilitate their confident localization. Herein, we report a quantitative parallel reaction monitoring (PRM) method developed to monitor spatiotemporal changes in site-specific Ser5 phosphorylation of the CTD by cyclin-dependent kinase 7 (CDK7) using UVPD for sequence identification, phosphosite localization, and differentiation of phosphopeptide isomers. We capitalize on the series of phospho-retaining fragment ions produced by UVPD to create unique transition lists that are pivotal for distinguishing the array of phosphopeptides generated from the CTD.
Collapse
|
20
|
Thompson AM, Stratton KG, Bramer LM, Zavoshy NS, McCue LA. Fourier transform ion cyclotron resonance mass spectrometry (FT-ICR-MS) peak intensity normalization for complex mixture analyses. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e9068. [PMID: 33590907 DOI: 10.1002/rcm.9068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 06/12/2023]
Abstract
RATIONALE Fourier transform ion cyclotron resonance mass spectrometry (FT-ICR-MS) is a preferred technique for analyzing complex organic mixtures. Currently, there is no consensus normalization approach, nor an objective method for selecting one, for quantitative analyses of FT-ICR-MS data. We investigate a method to evaluate and score the amount of bias various normalization approaches introduce into the data. METHODS We evaluate the ability of the Statistical Procedure for the Analysis of Normalization Strategies (SPANS) to guide the selection of appropriate normalization approaches for two different FT-ICR-MS data sets. Furthermore, we test the robustness of SPANS results to changes in SPANS parameter values and assess the impact of using various normalization approaches on downstream statistical analyses. RESULTS The normalization approach identified by SPANS differed for the two data sets. Normalization methods impacted the statistical significance of peaks differently, underscoring the importance of carefully evaluating potential methods. More consistent SPANS scores resulted when at least 120 significant peaks are used, where larger sets of peaks were obtained by increasing the p-value threshold. Interestingly, we show that total sum scaling and highest peak normalization, used in previous studies, underperformed relative to SPANS-recommended normalization approaches. CONCLUSIONS Although there is no single, best normalization method for all data sets, SPANS provides a mechanism to identify an appropriate normalization method for analyzing FT-ICR-MS data quantitatively. The number of peaks used in the background distributions of SPANS contributes more significantly to the reproducibility of results than the p-value thresholds used to obtain those peaks.
Collapse
Affiliation(s)
- Allison M Thompson
- Environmental and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Kelly G Stratton
- National Security Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Lisa M Bramer
- National Security Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Nicole S Zavoshy
- National Security Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Lee Ann McCue
- Environmental and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| |
Collapse
|
21
|
Pomraning KR, Dai Z, Munoz N, Kim YM, Gao Y, Deng S, Kim J, Hofstad BA, Swita MS, Lemmon T, Collett JR, Panisko EA, Webb-Robertson BJM, Zucker JD, Nicora CD, De Paoli H, Baker SE, Burnum-Johnson KE, Hillson NJ, Magnuson JK. Integration of Proteomics and Metabolomics Into the Design, Build, Test, Learn Cycle to Improve 3-Hydroxypropionic Acid Production in Aspergillus pseudoterreus. Front Bioeng Biotechnol 2021; 9:603832. [PMID: 33898398 PMCID: PMC8058442 DOI: 10.3389/fbioe.2021.603832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
Biological engineering of microorganisms to produce value-added chemicals is a promising route to sustainable manufacturing. However, overproduction of metabolic intermediates at high titer, rate, and yield from inexpensive substrates is challenging in non-model systems where limited information is available regarding metabolic flux and its control in production conditions. Integrated multi-omic analyses of engineered strains offers an in-depth look at metabolites and proteins directly involved in growth and production of target and non-target bioproducts. Here we applied multi-omic analyses to overproduction of the polymer precursor 3-hydroxypropionic acid (3HP) in the filamentous fungus Aspergillus pseudoterreus. A synthetic pathway consisting of aspartate decarboxylase, beta-alanine pyruvate transaminase, and 3HP dehydrogenase was designed and built for A. pseudoterreus. Strains with single- and multi-copy integration events were isolated and multi-omics analysis consisting of intracellular and extracellular metabolomics and targeted and global proteomics was used to interrogate the strains in shake-flask and bioreactor conditions. Production of a variety of co-products (organic acids and glycerol) and oxidative degradation of 3HP were identified as metabolic pathways competing with 3HP production. Intracellular accumulation of nitrogen as 2,4-diaminobutanoate was identified as an off-target nitrogen sink that may also limit flux through the engineered 3HP pathway. Elimination of the high-expression oxidative 3HP degradation pathway by deletion of a putative malonate semialdehyde dehydrogenase improved the yield of 3HP by 3.4 × after 10 days in shake-flask culture. This is the first report of 3HP production in a filamentous fungus amenable to industrial scale biomanufacturing of organic acids at high titer and low pH.
Collapse
Affiliation(s)
- Kyle R Pomraning
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Ziyu Dai
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Nathalie Munoz
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Young-Mo Kim
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Yuqian Gao
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Shuang Deng
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Joonhoon Kim
- Pacific Northwest National Laboratory, Richland, WA, United States.,Joint BioEnergy Institute, Emeryville, CA, United States
| | - Beth A Hofstad
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Marie S Swita
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Teresa Lemmon
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - James R Collett
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Ellen A Panisko
- Pacific Northwest National Laboratory, Richland, WA, United States
| | | | - Jeremy D Zucker
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Carrie D Nicora
- Pacific Northwest National Laboratory, Richland, WA, United States
| | | | - Scott E Baker
- Pacific Northwest National Laboratory, Richland, WA, United States
| | | | - Nathan J Hillson
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Jon K Magnuson
- Pacific Northwest National Laboratory, Richland, WA, United States.,Joint BioEnergy Institute, Emeryville, CA, United States
| |
Collapse
|
22
|
From Plants to Ants: Fungal Modification of Leaf Lipids for Nutrition and Communication in the Leaf-Cutter Ant Fungal Garden Ecosystem. mSystems 2021; 6:6/2/e01307-20. [PMID: 33758033 PMCID: PMC8547007 DOI: 10.1128/msystems.01307-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lipids are essential to all living organisms, as an energy source, as an important cellular structural component, and as a communication tool. In this study, we used global lipidomic methods to evaluate the lipids in leaf-cutter ant fungal gardens. Leaf-cutter ants and their coevolved fungal cultivar, Leucoagaricus gongylophorus, are a model mutualistic system. The fungus enzymatically digests fresh plant material that the ants cut and deliver, converting energy and nutrients from plants and providing them to the ants through specialized hyphal swellings called gongylidia. Using combined liquid chromatography, ion mobility spectrometry, and tandem mass spectrometry, we evaluated differences between the molecular species of lipids in the leaf-cutter ant fungal garden ecosystem. This lipidomic study characterized leaves that are fed to the gardens, gongylidia that are produced by the fungus to feed the ants, and spatially resolved regions of the fungal garden through stages of leaf degradation. Lipids containing alpha-linolenic acid (18:3) were enriched in leaves and the top of the gardens but not dominant in the middle or bottom regions. Gongylidia were dominated by lipids containing linoleic acid (18:2). To evaluate the communicative potential of the lipids in fungal gardens, we conducted a behavioral experiment that showed Atta leaf-cutter ants responded differently to 18:3 and 18:2 fatty acids, with aggression toward 18:3 and attraction for 18:2. This work demonstrates the role of lipids in both the transfer of energy and as an interkingdom communication tool in leaf-cutter ant fungal gardens. IMPORTANCE In this work, we examined the role of lipids in the mutualism between leaf-cutter ants and fungus. These ants cut fresh leaf material, which they provide to their fungal cultivar, that converts energy and nutrients from the plants and provides it to the ants in specialized hyphal swellings called gongylidia. This work constitutes the first example of a global lipidomics study of a symbiotic system and provides insights as to how the fungus modifies plant lipids into a usable source for the ants. Through a behavioral experiment, this work also demonstrates how lipids can be used as an interkingdom communication tool, in this case, as an attractant rather than as a repellant, which is more often seen. Author Video: An author video summary of this article is available.
Collapse
|
23
|
Gao Y, Fillmore TL, Munoz N, Bentley GJ, Johnson CW, Kim J, Meadows JA, Zucker JD, Burnet MC, Lipton AK, Bilbao A, Orton DJ, Kim YM, Moore RJ, Robinson EW, Baker SE, Webb-Robertson BJM, Guss AM, Gladden JM, Beckham GT, Magnuson JK, Burnum-Johnson KE. High-Throughput Large-Scale Targeted Proteomics Assays for Quantifying Pathway Proteins in Pseudomonas putida KT2440. Front Bioeng Biotechnol 2020; 8:603488. [PMID: 33425868 PMCID: PMC7793925 DOI: 10.3389/fbioe.2020.603488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/10/2020] [Indexed: 11/13/2022] Open
Abstract
Targeted proteomics is a mass spectrometry-based protein quantification technique with high sensitivity, accuracy, and reproducibility. As a key component in the multi-omics toolbox of systems biology, targeted liquid chromatography-selected reaction monitoring (LC-SRM) measurements are critical for enzyme and pathway identification and design in metabolic engineering. To fulfill the increasing need for analyzing large sample sets with faster turnaround time in systems biology, high-throughput LC-SRM is greatly needed. Even though nanoflow LC-SRM has better sensitivity, it lacks the speed offered by microflow LC-SRM. Recent advancements in mass spectrometry instrumentation significantly enhance the scan speed and sensitivity of LC-SRM, thereby creating opportunities for applying the high speed of microflow LC-SRM without losing peptide multiplexing power or sacrificing sensitivity. Here, we studied the performance of microflow LC-SRM relative to nanoflow LC-SRM by monitoring 339 peptides representing 132 enzymes in Pseudomonas putida KT2440 grown on various carbon sources. The results from the two LC-SRM platforms are highly correlated. In addition, the response curve study of 248 peptides demonstrates that microflow LC-SRM has comparable sensitivity for the majority of detected peptides and better mass spectrometry signal and chromatography stability than nanoflow LC-SRM.
Collapse
Affiliation(s)
- Yuqian Gao
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | | | - Nathalie Munoz
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Gayle J Bentley
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,National Renewable Energy Laboratory, Golden, CO, United States
| | - Christopher W Johnson
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,National Renewable Energy Laboratory, Golden, CO, United States
| | - Joonhoon Kim
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Jamie A Meadows
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Sandia National Laboratories, Livermore, CA, United States
| | - Jeremy D Zucker
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Meagan C Burnet
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Anna K Lipton
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Aivett Bilbao
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Daniel J Orton
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Young-Mo Kim
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Ronald J Moore
- Pacific Northwest National Laboratory, Richland, WA, United States
| | - Errol W Robinson
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Scott E Baker
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Bobbie-Jo M Webb-Robertson
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Adam M Guss
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - John M Gladden
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Sandia National Laboratories, Livermore, CA, United States
| | - Gregg T Beckham
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,National Renewable Energy Laboratory, Golden, CO, United States
| | - Jon K Magnuson
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| | - Kristin E Burnum-Johnson
- Department of Energy, Agile BioFoundry, Emeryville, CA, United States.,Pacific Northwest National Laboratory, Richland, WA, United States
| |
Collapse
|
24
|
Bradley R, Langley BO, Ryan JJ, Phipps J, Hanes DA, Stack E, Jansson JK, Metz TO, Stevens JF. Xanthohumol microbiome and signature in healthy adults (the XMaS trial): a phase I triple-masked, placebo-controlled clinical trial. Trials 2020; 21:835. [PMID: 33028396 PMCID: PMC7542976 DOI: 10.1186/s13063-020-04769-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Natural products may provide a source for the discovery and development of adjunctive pharmacological interventions to modulate the inflammatory pathways contributing to chronic disease. Xanthohumol, a flavonoid from the hops plant (Humulus lupulus), has antioxidant and anti-inflammatory properties and may act as a prebiotic to the intestinal microbiota. Xanthohumol is not currently approved as a drug by the US Food and Drug Administration (FDA), but is available as a dietary supplement and ingredient in medical foods. To formally test the safety of xanthohumol, a phase I clinical trial ("XMaS") was designed and approved under an Investigational New Drug application to the US FDA. The main objective is to examine the clinical safety and subjective tolerability of xanthohumol in healthy adults compared to placebo. Additional aims are to monitor biomarkers related to inflammation, gut permeability, bile acid metabolism, routes, and in vivo products of xanthohumol metabolism, and to evaluate xanthohumol's impact on gut microbial composition. METHODS The safety and tolerability of xanthohumol in healthy adults will be evaluated in a triple-masked, randomized, placebo-controlled trial. Participants will be randomized to either 24 mg/day of xanthohumol or placebo for 8 weeks. Blood cell counts, hepatic and renal function tests, electrolytes, and self-reported health-related quality of life measures will be collected every 2 weeks. Participants will be queried for adverse events throughout the trial. Xanthohumol metabolites in blood, urine, and stool will be measured. Biomarkers to be evaluated include plasma tumor necrosis factor-alpha, various interleukins, soluble CD14, lipopolysaccharide-binding protein, fecal calprotectin, and bile acids to assess impact on inflammatory and gut permeability-related mechanisms in vivo. Stool samples will be analyzed to determine effects on the gut microbiome. DISCUSSION This phase I clinical trial of xanthohumol will assess safety and tolerability in healthy adults, collect extensive biomarker data for assessment of potential mechanism(s), and provide comparison data necessary for future phase II trials in chronic disease(s). The design and robustness of the planned safety and mechanistic evaluations planned provide a model for drug discovery pursuits from natural products. TRIAL REGISTRATION ClinicalTrials.gov NCT03735420 . Registered on November 8, 2018.
Collapse
Affiliation(s)
- Ryan Bradley
- National University of Natural Medicine, Portland, USA.
| | | | | | - John Phipps
- National University of Natural Medicine, Portland, USA
| | | | - Emily Stack
- National University of Natural Medicine, Portland, USA
| | | | - Thomas O Metz
- Pacific Northwest National Laboratory, Richland, USA
| | | |
Collapse
|
25
|
Sigdel TK, Piehowski PD, Roy S, Liberto J, Hansen JR, Swensen AC, Zhao R, Zhu Y, Rashmi P, Schroeder A, Damm I, Sur S, Luo J, Yang Y, Qian WJ, Sarwal MM. Near-Single-Cell Proteomics Profiling of the Proximal Tubular and Glomerulus of the Normal Human Kidney. Front Med (Lausanne) 2020; 7:499. [PMID: 33072769 PMCID: PMC7533534 DOI: 10.3389/fmed.2020.00499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 07/21/2020] [Indexed: 01/21/2023] Open
Abstract
Molecular assessments at the single cell level can accelerate biological research by providing detailed assessments of cellular organization and tissue heterogeneity in both disease and health. The human kidney has complex multi-cellular states with varying functionality, much of which can now be completely harnessed with recent technological advances in tissue proteomics at a near single-cell level. We discuss the foundational steps in the first application of this mass spectrometry (MS) based proteomics method for analysis of sub-sections of the normal human kidney, as part of the Kidney Precision Medicine Project (KPMP). Using ~30-40 laser captured micro-dissected kidney cells, we identified more than 2,500 human proteins, with specificity to the proximal tubular (PT; n = 25 proteins) and glomerular (Glom; n = 67 proteins) regions of the kidney and their unique metabolic functions. This pilot study provides the roadmap for application of our near-single-cell proteomics workflow for analysis of other renal micro-compartments, on a larger scale, to unravel perturbations of renal sub-cellular function in the normal kidney as well as different etiologies of acute and chronic kidney disease.
Collapse
Affiliation(s)
- Tara K. Sigdel
- Division of MultiOrgan Transplantation, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Paul D. Piehowski
- Pacific Northwest National Laboratory, Biological Sciences Division, Richland, WA, United States
| | - Sudeshna Roy
- Division of MultiOrgan Transplantation, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Juliane Liberto
- Division of MultiOrgan Transplantation, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Joshua R. Hansen
- Pacific Northwest National Laboratory, Biological Sciences Division, Richland, WA, United States
| | - Adam C. Swensen
- Pacific Northwest National Laboratory, Biological Sciences Division, Richland, WA, United States
| | - Rui Zhao
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Ying Zhu
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Priyanka Rashmi
- Division of MultiOrgan Transplantation, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Andrew Schroeder
- Division of MultiOrgan Transplantation, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Izabella Damm
- Division of MultiOrgan Transplantation, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Swastika Sur
- Division of MultiOrgan Transplantation, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Jinghui Luo
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Yingbao Yang
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Wei-Jun Qian
- Pacific Northwest National Laboratory, Biological Sciences Division, Richland, WA, United States
| | - Minnie M. Sarwal
- Division of MultiOrgan Transplantation, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
26
|
Schwämmle V, Hagensen CE, Rogowska-Wrzesinska A, Jensen ON. PolySTest: Robust Statistical Testing of Proteomics Data with Missing Values Improves Detection of Biologically Relevant Features. Mol Cell Proteomics 2020; 19:1396-1408. [PMID: 32424025 PMCID: PMC8015005 DOI: 10.1074/mcp.ra119.001777] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/1919] [Revised: 04/18/2020] [Indexed: 12/11/2022] Open
Abstract
Statistical testing remains one of the main challenges for high-confidence detection of differentially regulated proteins or peptides in large-scale quantitative proteomics experiments by mass spectrometry. Statistical tests need to be sufficiently robust to deal with experiment intrinsic data structures and variations and often also reduced feature coverage across different biological samples due to ubiquitous missing values. A robust statistical test provides accurate confidence scores of large-scale proteomics results, regardless of instrument platform, experimental protocol and software tools. However, the multitude of different combinations of experimental strategies, mass spectrometry techniques and informatics methods complicate the decision of choosing appropriate statistical approaches. We address this challenge by introducing PolySTest, a user-friendly web service for statistical testing, data browsing and data visualization. We introduce a new method, Miss test, that simultaneously tests for missingness and feature abundance, thereby complementing common statistical tests by rescuing otherwise discarded data features. We demonstrate that PolySTest with integrated Miss test achieves higher confidence and higher sensitivity for artificial and experimental proteomics data sets with known ground truth. Application of PolySTest to mass spectrometry based large-scale proteomics data obtained from differentiating muscle cells resulted in the rescue of 10-20% additional proteins in the identified molecular networks relevant to muscle differentiation. We conclude that PolySTest is a valuable addition to existing tools and instrument enhancements that improve coverage and depth of large-scale proteomics experiments. A fully functional demo version of PolySTest and Miss test is available via http://computproteomics.bmb.sdu.dk/Apps/PolySTest.
Collapse
Affiliation(s)
- Veit Schwämmle
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark; VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense M, Denmark.
| | - Christina E Hagensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark; VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense M, Denmark
| | - Adelina Rogowska-Wrzesinska
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark; VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense M, Denmark
| | - Ole N Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark; VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense M, Denmark
| |
Collapse
|
27
|
Goeminne LJE, Sticker A, Martens L, Gevaert K, Clement L. MSqRob Takes the Missing Hurdle: Uniting Intensity- and Count-Based Proteomics. Anal Chem 2020; 92:6278-6287. [PMID: 32227882 DOI: 10.1021/acs.analchem.9b04375] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Missing values are a major issue in quantitative data-dependent mass spectrometry-based proteomics. We therefore present an innovative solution to this key issue by introducing a hurdle model, which is a mixture between a binomial peptide count and a peptide intensity-based model component. It enables dramatically enhanced quantification of proteins with many missing values without having to resort to harmful assumptions for missingness. We demonstrate the superior performance of our method by comparing it with state-of-the-art methods in the field.
Collapse
Affiliation(s)
- Ludger J E Goeminne
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Krijgslaan 281-S9, B9000 Ghent, Belgium.,VIB-UGent Center for Medical Biotechnology, VIB, Albert Baertsoenkaai 3, B9000 Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Albert Baertsoenkaai 3, B9000 Ghent, Belgium.,Bioinformatics Institute Ghent, Ghent University, Technologiepark 927, B9052 Ghent, Belgium
| | - Adriaan Sticker
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Krijgslaan 281-S9, B9000 Ghent, Belgium.,VIB-UGent Center for Medical Biotechnology, VIB, Albert Baertsoenkaai 3, B9000 Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Albert Baertsoenkaai 3, B9000 Ghent, Belgium.,Bioinformatics Institute Ghent, Ghent University, Technologiepark 927, B9052 Ghent, Belgium
| | - Lennart Martens
- VIB-UGent Center for Medical Biotechnology, VIB, Albert Baertsoenkaai 3, B9000 Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Albert Baertsoenkaai 3, B9000 Ghent, Belgium.,Bioinformatics Institute Ghent, Ghent University, Technologiepark 927, B9052 Ghent, Belgium
| | - Kris Gevaert
- VIB-UGent Center for Medical Biotechnology, VIB, Albert Baertsoenkaai 3, B9000 Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Albert Baertsoenkaai 3, B9000 Ghent, Belgium
| | - Lieven Clement
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Krijgslaan 281-S9, B9000 Ghent, Belgium.,Bioinformatics Institute Ghent, Ghent University, Technologiepark 927, B9052 Ghent, Belgium
| |
Collapse
|
28
|
Piehowski PD, Zhu Y, Bramer LM, Stratton KG, Zhao R, Orton DJ, Moore RJ, Yuan J, Mitchell HD, Gao Y, Webb-Robertson BJM, Dey SK, Kelly RT, Burnum-Johnson KE. Automated mass spectrometry imaging of over 2000 proteins from tissue sections at 100-μm spatial resolution. Nat Commun 2020; 11:8. [PMID: 31911630 PMCID: PMC6946663 DOI: 10.1038/s41467-019-13858-z] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/20/2019] [Indexed: 12/19/2022] Open
Abstract
Biological tissues exhibit complex spatial heterogeneity that directs the functions of multicellular organisms. Quantifying protein expression is essential for elucidating processes within complex biological assemblies. Imaging mass spectrometry (IMS) is a powerful emerging tool for mapping the spatial distribution of metabolites and lipids across tissue surfaces, but technical challenges have limited the application of IMS to the analysis of proteomes. Methods for probing the spatial distribution of the proteome have generally relied on the use of labels and/or antibodies, which limits multiplexing and requires a priori knowledge of protein targets. Past efforts to make spatially resolved proteome measurements across tissues have had limited spatial resolution and proteome coverage and have relied on manual workflows. Here, we demonstrate an automated approach to imaging that utilizes label-free nanoproteomics to analyze tissue voxels, generating quantitative cell-type-specific images for >2000 proteins with 100-µm spatial resolution across mouse uterine tissue sections preparing for blastocyst implantation. Imaging mass spectrometry is a powerful emerging tool for mapping the spatial distribution of biomolecules across tissue surfaces. Here the authors showcase an automated technology for deep proteome imaging that utilizes ultrasensitive microfluidics and a mass spectrometry workflow to analyze tissue voxels, generating quantitative cell-type-specific images.
Collapse
Affiliation(s)
- Paul D Piehowski
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ying Zhu
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lisa M Bramer
- National Security Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Kelly G Stratton
- National Security Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Rui Zhao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Daniel J Orton
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jia Yuan
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hugh D Mitchell
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Yuqian Gao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | | | - Sudhansu K Dey
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ryan T Kelly
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA. .,Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| | | |
Collapse
|
29
|
Proximity Labeling To Map Host-Pathogen Interactions at the Membrane of a Bacterium-Containing Vacuole in Chlamydia trachomatis-Infected Human Cells. Infect Immun 2019; 87:IAI.00537-19. [PMID: 31405957 PMCID: PMC6803327 DOI: 10.1128/iai.00537-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
Many intracellular bacteria, including the obligate intracellular pathogen Chlamydia trachomatis, grow within a membrane-bound bacterium-containing vacuole (BCV). Secreted cytosolic effectors modulate host activity, but an understanding of the host-pathogen interactions that occur at the BCV membrane is limited by the difficulty in purifying membrane fractions from infected host cells. Many intracellular bacteria, including the obligate intracellular pathogen Chlamydia trachomatis, grow within a membrane-bound bacterium-containing vacuole (BCV). Secreted cytosolic effectors modulate host activity, but an understanding of the host-pathogen interactions that occur at the BCV membrane is limited by the difficulty in purifying membrane fractions from infected host cells. We used the ascorbate peroxidase (APEX2) proximity labeling system, which labels proximal proteins with biotin in vivo, to study the protein-protein interactions that occur at the chlamydial vacuolar, or inclusion, membrane. An in vivo understanding of the secreted chlamydial inclusion membrane protein (Inc) interactions (e.g., Inc-Inc and Inc-eukaryotic protein) and how these contribute to overall host-chlamydia interactions at this unique membrane is lacking. We hypothesize some Incs organize the inclusion membrane, whereas other Incs bind eukaryotic proteins to promote chlamydia-host interactions. To study this, Incs fused to APEX2 were expressed in C. trachomatis L2. Affinity purification-mass spectrometry (AP-MS) identified biotinylated proteins, which were analyzed for statistical significance using significance analysis of the interactome (SAINT). Broadly supporting both Inc-Inc and Inc-host interactions, our Inc-APEX2 constructs labeled Incs as well as known and previously unreported eukaryotic proteins localizing to the inclusion. We demonstrate, using bacterial two-hybrid and coimmunoprecipitation assays, that endogenous LRRFIP1 (LRRF1) is recruited to the inclusion by the Inc CT226. We further demonstrate interactions between CT226 and the Incs used in our study to reveal a model for inclusion membrane organization. Combined, our data highlight the utility of APEX2 to capture the complex in vivo protein-protein interactions at the chlamydial inclusion.
Collapse
|
30
|
Ebner JN, Ritz D, von Fumetti S. Comparative proteomics of stenotopic caddisfly Crunoecia irrorata identifies acclimation strategies to warming. Mol Ecol 2019; 28:4453-4469. [PMID: 31478292 PMCID: PMC6856850 DOI: 10.1111/mec.15225] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022]
Abstract
Species' ecological preferences are often deduced from habitat characteristics thought to represent more or less optimal conditions for physiological functioning. Evolution has led to stenotopic and eurytopic species, the former having decreased niche breadths and lower tolerances to environmental variability. Species inhabiting freshwater springs are often described as being stenotopic specialists, adapted to the stable thermal conditions found in these habitats. Whether due to past local adaptation these species have evolved or have lost intra-generational adaptive mechanisms to cope with increasing thermal variability has, to our knowledge, never been investigated. By studying how the proteome of a stenotopic species changes as a result of increasing temperatures, we investigate if the absence or attenuation of molecular mechanisms is indicative of local adaptation to freshwater springs. An understanding of compensatory mechanisms is especially relevant as spring specialists will experience thermal conditions beyond their physiological limits due to climate change. In this study, the stenotopic species Crunoecia irrorata (Trichoptera: Lepidostomatidae, Curtis 1834) was acclimated to 10, 15 and 20°C for 168 hr. We constructed a homology-based database and via liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based shotgun proteomics identified 1,358 proteins. Differentially abundant proteins and protein norms of reaction revealed candidate proteins and molecular mechanisms facilitating compensatory responses such as trehalose metabolism, tracheal system alteration and heat-shock protein regulation. A species-specific understanding of compensatory physiologies challenges the characterization of species as having narrow tolerances to environmental variability if that characterization is based on occurrences and habitat characteristics alone.
Collapse
Affiliation(s)
- Joshua N. Ebner
- Geoecology Research GroupDepartment of Environmental SciencesUniversity of BaselBaselSwitzerland
| | - Danilo Ritz
- Proteomics Core FacilityBiozentrumUniversity of BaselBaselSwitzerland
| | - Stefanie von Fumetti
- Geoecology Research GroupDepartment of Environmental SciencesUniversity of BaselBaselSwitzerland
| |
Collapse
|
31
|
Hüttenhain R, Choi M, Martin de la Fuente L, Oehl K, Chang CY, Zimmermann AK, Malander S, Olsson H, Surinova S, Clough T, Heinzelmann-Schwarz V, Wild PJ, Dinulescu DM, Niméus E, Vitek O, Aebersold R. A Targeted Mass Spectrometry Strategy for Developing Proteomic Biomarkers: A Case Study of Epithelial Ovarian Cancer. Mol Cell Proteomics 2019; 18:1836-1850. [PMID: 31289117 PMCID: PMC6731088 DOI: 10.1074/mcp.ra118.001221] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 05/07/2019] [Indexed: 12/11/2022] Open
Abstract
Protein biomarkers for epithelial ovarian cancer are critical for the early detection of the cancer to improve patient prognosis and for the clinical management of the disease to monitor treatment response and to detect recurrences. Unfortunately, the discovery of protein biomarkers is hampered by the limited availability of reliable and sensitive assays needed for the reproducible quantification of proteins in complex biological matrices such as blood plasma. In recent years, targeted mass spectrometry, exemplified by selected reaction monitoring (SRM) has emerged as a method, capable of overcoming this limitation. Here, we present a comprehensive SRM-based strategy for developing plasma-based protein biomarkers for epithelial ovarian cancer and illustrate how the SRM platform, when combined with rigorous experimental design and statistical analysis, can result in detection of predictive analytes.Our biomarker development strategy first involved a discovery-driven proteomic effort to derive potential N-glycoprotein biomarker candidates for plasma-based detection of human ovarian cancer from a genetically engineered mouse model of endometrioid ovarian cancer, which accurately recapitulates the human disease. Next, 65 candidate markers selected from proteins of different abundance in the discovery dataset were reproducibly quantified with SRM assays across a large cohort of over 200 plasma samples from ovarian cancer patients and healthy controls. Finally, these measurements were used to derive a 5-protein signature for distinguishing individuals with epithelial ovarian cancer from healthy controls. The sensitivity of the candidate biomarker signature in combination with CA125 ELISA-based measurements currently used in clinic, exceeded that of CA125 ELISA-based measurements alone. The SRM-based strategy in this study is broadly applicable. It can be used in any study that requires accurate and reproducible quantification of selected proteins in a high-throughput and multiplexed fashion.
Collapse
Affiliation(s)
- Ruth Hüttenhain
- ‡Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland.
| | - Meena Choi
- §Khoury College of Computer Sciences, Northeastern University, Boston, MA
| | | | - Kathrin Oehl
- ‖Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Ching-Yun Chang
- **Department of Statistics, Purdue University, West Lafayette, IN
| | - Anne-Kathrin Zimmermann
- ‖Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Susanne Malander
- ¶Department of Surgery and Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Håkan Olsson
- ¶Department of Surgery and Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Silvia Surinova
- ‡Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Timothy Clough
- **Department of Statistics, Purdue University, West Lafayette, IN
| | - Viola Heinzelmann-Schwarz
- ‡‡Gynecological Cancer Center, University Hospital Basel, University of Basel, Basel, Switzerland; §§Ovarian Cancer Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Peter J Wild
- ¶¶Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Daniela M Dinulescu
- ‖‖Department of Pathology, Division of Women's and Perinatal Pathology Brigham and Women's Hospital Harvard Medical School, Boston, MA
| | - Emma Niméus
- ¶Department of Surgery and Oncology, Clinical Sciences, Lund University, Lund, Sweden; ‡‡‡Department of Surgery, Skånes University hospital, Lund, Sweden
| | - Olga Vitek
- §Khoury College of Computer Sciences, Northeastern University, Boston, MA; **Department of Statistics, Purdue University, West Lafayette, IN
| | - Ruedi Aebersold
- ‡Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland; §§§Faculty of Science, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
32
|
McClure RS, Wendler JP, Adkins JN, Swanstrom J, Baric R, Kaiser BLD, Oxford KL, Waters KM, McDermott JE. Unified feature association networks through integration of transcriptomic and proteomic data. PLoS Comput Biol 2019; 15:e1007241. [PMID: 31527878 PMCID: PMC6748406 DOI: 10.1371/journal.pcbi.1007241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 07/02/2019] [Indexed: 11/18/2022] Open
Abstract
High-throughput multi-omics studies and corresponding network analyses of multi-omic data have rapidly expanded their impact over the last 10 years. As biological features of different types (e.g. transcripts, proteins, metabolites) interact within cellular systems, the greatest amount of knowledge can be gained from networks that incorporate multiple types of -omic data. However, biological and technical sources of variation diminish the ability to detect cross-type associations, yielding networks dominated by communities comprised of nodes of the same type. We describe here network building methods that can maximize edges between nodes of different data types leading to integrated networks, networks that have a large number of edges that link nodes of different-omic types (transcripts, proteins, lipids etc). We systematically rank several network inference methods and demonstrate that, in many cases, using a random forest method, GENIE3, produces the most integrated networks. This increase in integration does not come at the cost of accuracy as GENIE3 produces networks of approximately the same quality as the other network inference methods tested here. Using GENIE3, we also infer networks representing antibody-mediated Dengue virus cell invasion and receptor-mediated Dengue virus invasion. A number of functional pathways showed centrality differences between the two networks including genes responding to both GM-CSF and IL-4, which had a higher centrality value in an antibody-mediated vs. receptor-mediated Dengue network. Because a biological system involves the interplay of many different types of molecules, incorporating multiple data types into networks will improve their use as models of biological systems. The methods explored here are some of the first to specifically highlight and address the challenges associated with how such multi-omic networks can be assembled and how the greatest number of interactions can be inferred from different data types. The resulting networks can lead to the discovery of new host response patterns and interactions during viral infection, generate new hypotheses of pathogenic mechanisms and confirm mechanisms of disease.
Collapse
Affiliation(s)
- Ryan S. McClure
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA, United States of America
| | - Jason P. Wendler
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA, United States of America
| | - Joshua N. Adkins
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA, United States of America
| | - Jesica Swanstrom
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Ralph Baric
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Brooke L. Deatherage Kaiser
- Signatures Science and Technology Division, Pacific Northwest National Laboratory, Richland WA, United States of America
| | - Kristie L. Oxford
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA, United States of America
| | - Katrina M. Waters
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA, United States of America
| | - Jason E. McDermott
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA, United States of America
- Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Portland, OR, United States of America
| |
Collapse
|
33
|
Sharon G, Cruz NJ, Kang DW, Gandal MJ, Wang B, Kim YM, Zink EM, Casey CP, Taylor BC, Lane CJ, Bramer LM, Isern NG, Hoyt DW, Noecker C, Sweredoski MJ, Moradian A, Borenstein E, Jansson JK, Knight R, Metz TO, Lois C, Geschwind DH, Krajmalnik-Brown R, Mazmanian SK. Human Gut Microbiota from Autism Spectrum Disorder Promote Behavioral Symptoms in Mice. Cell 2019; 177:1600-1618.e17. [PMID: 31150625 PMCID: PMC6993574 DOI: 10.1016/j.cell.2019.05.004] [Citation(s) in RCA: 672] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 02/11/2019] [Accepted: 04/30/2019] [Indexed: 01/02/2023]
Abstract
Autism spectrum disorder (ASD) manifests as alterations in complex human behaviors including social communication and stereotypies. In addition to genetic risks, the gut microbiome differs between typically developing (TD) and ASD individuals, though it remains unclear whether the microbiome contributes to symptoms. We transplanted gut microbiota from human donors with ASD or TD controls into germ-free mice and reveal that colonization with ASD microbiota is sufficient to induce hallmark autistic behaviors. The brains of mice colonized with ASD microbiota display alternative splicing of ASD-relevant genes. Microbiome and metabolome profiles of mice harboring human microbiota predict that specific bacterial taxa and their metabolites modulate ASD behaviors. Indeed, treatment of an ASD mouse model with candidate microbial metabolites improves behavioral abnormalities and modulates neuronal excitability in the brain. We propose that the gut microbiota regulates behaviors in mice via production of neuroactive metabolites, suggesting that gut-brain connections contribute to the pathophysiology of ASD.
Collapse
Affiliation(s)
- Gil Sharon
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Nikki Jamie Cruz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Dae-Wook Kang
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ 85287, USA; Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85287, USA
| | - Michael J Gandal
- Center for Autism Research and Treatment, Program in Neurobehavioral Genetics, Semel Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, Semel Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bo Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Young-Mo Kim
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Erika M Zink
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Cameron P Casey
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Bryn C Taylor
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christianne J Lane
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Lisa M Bramer
- National Security Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Nancy G Isern
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - David W Hoyt
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Cecilia Noecker
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Michael J Sweredoski
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Annie Moradian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Elhanan Borenstein
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Department of Computer Science and Engineering, University of Washington, Seattle, WA 98195, USA; Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv 6997801, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Janet K Jansson
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Rob Knight
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Biongineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Thomas O Metz
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Daniel H Geschwind
- Center for Autism Research and Treatment, Program in Neurobehavioral Genetics, Semel Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, Semel Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ 85287, USA; Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85287, USA; School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ 85287, USA
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
34
|
Dickinson MS, Anderson LN, Webb-Robertson BJM, Hansen JR, Smith RD, Wright AT, Hybiske K. Proximity-dependent proteomics of the Chlamydia trachomatis inclusion membrane reveals functional interactions with endoplasmic reticulum exit sites. PLoS Pathog 2019; 15:e1007698. [PMID: 30943267 PMCID: PMC6464245 DOI: 10.1371/journal.ppat.1007698] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 04/15/2019] [Accepted: 03/12/2019] [Indexed: 11/18/2022] Open
Abstract
Chlamydia trachomatis is the most common cause of bacterial sexually transmitted infection, responsible for millions of infections each year. Despite this high prevalence, the elucidation of the molecular mechanisms of Chlamydia pathogenesis has been difficult due to limitations in genetic tools and its intracellular developmental cycle. Within a host epithelial cell, chlamydiae replicate within a vacuole called the inclusion. Many Chlamydia-host interactions are thought to be mediated by the Inc family of type III secreted proteins that are anchored in the inclusion membrane, but their array of host targets are largely unknown. To investigate how the inclusion membrane proteome changes over the course of an infected cell, we have adapted the APEX2 system of proximity-dependent biotinylation. APEX2 is capable of specifically labeling proteins within a 20 nm radius in living cells. We transformed C. trachomatis to express the enzyme APEX2 fused to known inclusion membrane proteins, allowing biotinylation and purification of inclusion-associated proteins. Using quantitative mass spectrometry against APEX2 labeled samples, we identified over 400 proteins associated with the inclusion membrane at early, middle, and late stages of epithelial cell infection. This system was sensitive enough to detect inclusion interacting proteins early in the developmental cycle, at 8 hours post infection, a previously intractable time point. Mass spectrometry analysis revealed a novel, early association between C. trachomatis inclusions and endoplasmic reticulum exit sites (ERES), functional regions of the ER where COPII-coated vesicles originate. Pharmacological and genetic disruption of ERES function severely restricted early chlamydial growth and the development of infectious progeny. APEX2 is therefore a powerful in situ approach for identifying critical protein interactions on the membranes of pathogen-containing vacuoles. Furthermore, the data derived from proteomic mapping of Chlamydia inclusions has illuminated an important functional role for ERES in promoting chlamydial developmental growth.
Collapse
Affiliation(s)
- Mary S. Dickinson
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, United States of America
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington, Seattle, WA, United States of America
| | - Lindsey N. Anderson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States of America
| | | | - Joshua R. Hansen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States of America
| | - Richard D. Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States of America
| | - Aaron T. Wright
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States of America
- The Gene and Linda Voiland College of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, United States of America
| | - Kevin Hybiske
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, United States of America
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington, Seattle, WA, United States of America
| |
Collapse
|
35
|
The M, Käll L. Integrated Identification and Quantification Error Probabilities for Shotgun Proteomics. Mol Cell Proteomics 2019; 18:561-570. [PMID: 30482846 PMCID: PMC6398204 DOI: 10.1074/mcp.ra118.001018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/05/2018] [Indexed: 02/02/2023] Open
Abstract
Protein quantification by label-free shotgun proteomics experiments is plagued by a multitude of error sources. Typical pipelines for identifying differential proteins use intermediate filters to control the error rate. However, they often ignore certain error sources and, moreover, regard filtered lists as completely correct in subsequent steps. These two indiscretions can easily lead to a loss of control of the false discovery rate (FDR). We propose a probabilistic graphical model, Triqler, that propagates error information through all steps, employing distributions in favor of point estimates, most notably for missing value imputation. The model outputs posterior probabilities for fold changes between treatment groups, highlighting uncertainty rather than hiding it. We analyzed 3 engineered data sets and achieved FDR control and high sensitivity, even for truly absent proteins. In a bladder cancer clinical data set we discovered 35 proteins at 5% FDR, whereas the original study discovered 1 and MaxQuant/Perseus 4 proteins at this threshold. Compellingly, these 35 proteins showed enrichment for functional annotation terms, whereas the top ranked proteins reported by MaxQuant/Perseus showed no enrichment. The model executes in minutes and is freely available at https://pypi.org/project/triqler/.
Collapse
Affiliation(s)
- Matthew The
- From the ‡Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Box 1031, 17121 Solna, Sweden
| | - Lukas Käll
- From the ‡Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Box 1031, 17121 Solna, Sweden
| |
Collapse
|
36
|
Bramer LM, Stratton KG, White AM, Bleeker AH, Kobold MA, Waters KM, Metz TO, Rodland KD, Webb-Robertson BJM. P-Mart: Interactive Analysis of Ion Abundance Global Proteomics Data. J Proteome Res 2019; 18:1426-1432. [PMID: 30667224 DOI: 10.1021/acs.jproteome.8b00840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The use of mass-spectrometry-based techniques for global protein profiling of biomedical or environmental experiments has become a major focus in research centered on biomarker discovery; however, one of the most important issues recently highlighted in the new era of omics data generation is the ability to perform analyses in a robust and reproducible manner. This has been hypothesized to be one of the issues hindering the ability of clinical proteomics to successfully identify clinical diagnostic and prognostic biomarkers of disease. P-Mart ( https://pmart.labworks.org ) is a new interactive web-based software environment that enables domain scientists to perform quality-control processing, statistics, and exploration of large-complex proteomics data sets without requiring statistical programming. P-Mart is developed in a manner that allows researchers to perform analyses via a series of modules, explore the results using interactive visualization, and finalize the analyses with a collection of output files documenting all stages of the analysis and a report to allow reproduction of the analysis.
Collapse
Affiliation(s)
- Lisa M Bramer
- Computing & Analytics Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States
| | - Kelly G Stratton
- Computing & Analytics Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States
| | - Amanda M White
- Computing & Analytics Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States
| | - Ameila H Bleeker
- Computing & Analytics Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States
| | - Markus A Kobold
- Computing & Analytics Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States
| | - Katrina M Waters
- Biological Sciences Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States
| | - Thomas O Metz
- Biological Sciences Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States
| | - Karin D Rodland
- Biological Sciences Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States.,Department of Cell, Developmental, and Cancer Biology , Oregon Health & Science University , Portland , Oregon 97221 , United States
| | - Bobbie-Jo M Webb-Robertson
- Computing & Analytics Division , Pacific Northwest National Laboratory , 902 Battelle Boulevard , Richland , Washington 99352 , United States
| |
Collapse
|
37
|
Stratton K, Webb-Robertson BJM, McCue LA, Stanfill B, Claborne D, Godinez I, Johansen T, Thompson AM, Burnum-Johnson KE, Waters KM, Bramer LM. pmartR: Quality Control and Statistics for Mass Spectrometry-Based Biological Data. J Proteome Res 2019; 18:1418-1425. [PMID: 30638385 PMCID: PMC6750869 DOI: 10.1021/acs.jproteome.8b00760] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Indexed: 02/06/2023]
Abstract
Prior to statistical analysis of mass spectrometry (MS) data, quality control (QC) of the identified biomolecule peak intensities is imperative for reducing process-based sources of variation and extreme biological outliers. Without this step, statistical results can be biased. Additionally, liquid chromatography-MS proteomics data present inherent challenges due to large amounts of missing data that require special consideration during statistical analysis. While a number of R packages exist to address these challenges individually, there is no single R package that addresses all of them. We present pmartR, an open-source R package, for QC (filtering and normalization), exploratory data analysis (EDA), visualization, and statistical analysis robust to missing data. Example analysis using proteomics data from a mouse study comparing smoke exposure to control demonstrates the core functionality of the package and highlights the capabilities for handling missing data. In particular, using a combined quantitative and qualitative statistical test, 19 proteins whose statistical significance would have been missed by a quantitative test alone were identified. The pmartR package provides a single software tool for QC, EDA, and statistical comparisons of MS data that is robust to missing data and includes numerous visualization capabilities.
Collapse
Affiliation(s)
- Kelly
G. Stratton
- National
Security Directorate, Pacific Northwest
National Laboratory, 902 Battelle Boulevard, Richland, Washington 99354, United States
| | - Bobbie-Jo M. Webb-Robertson
- National
Security Directorate, Pacific Northwest
National Laboratory, 902 Battelle Boulevard, Richland, Washington 99354, United States
| | - Lee Ann McCue
- Earth
& Biological Sciences Directorate, Pacific
Northwest National Laboratory, 902 Battelle Boulavard, Richland, Washington 99354, United States
| | - Bryan Stanfill
- National
Security Directorate, Pacific Northwest
National Laboratory, 902 Battelle Boulevard, Richland, Washington 99354, United States
| | - Daniel Claborne
- National
Security Directorate, Pacific Northwest
National Laboratory, 902 Battelle Boulevard, Richland, Washington 99354, United States
| | - Iobani Godinez
- National
Security Directorate, Pacific Northwest
National Laboratory, 902 Battelle Boulevard, Richland, Washington 99354, United States
| | - Thomas Johansen
- Department
of Statistics, Florida State University, 117 North Woodward Avenue, Tallahassee, Florida 32306, United States
| | - Allison M. Thompson
- Earth
& Biological Sciences Directorate, Pacific
Northwest National Laboratory, 902 Battelle Boulavard, Richland, Washington 99354, United States
| | - Kristin E. Burnum-Johnson
- Earth
& Biological Sciences Directorate, Pacific
Northwest National Laboratory, 902 Battelle Boulavard, Richland, Washington 99354, United States
| | - Katrina M. Waters
- Earth
& Biological Sciences Directorate, Pacific
Northwest National Laboratory, 902 Battelle Boulavard, Richland, Washington 99354, United States
| | - Lisa M. Bramer
- National
Security Directorate, Pacific Northwest
National Laboratory, 902 Battelle Boulevard, Richland, Washington 99354, United States
| |
Collapse
|
38
|
Handakumbura PP, Stanfill B, Rivas-Ubach A, Fortin D, Vogel JP, Jansson C. Metabotyping as a Stopover in Genome-to-Phenome Mapping. Sci Rep 2019; 9:1858. [PMID: 30755686 PMCID: PMC6372633 DOI: 10.1038/s41598-019-38483-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/21/2018] [Indexed: 01/01/2023] Open
Abstract
Predicting phenotypic expression from genomic and environmental information is arguably the greatest challenge in today's biology. Being able to survey genomic content, e.g., as single-nucleotide polymorphism data, within a diverse population and predict the phenotypes of external traits, represents the holy grail across genome-informed disciplines, from personal medicine and nutrition to plant breeding. In the present study, we propose a two-step procedure in bridging the genome to phenome gap where external phenotypes are viewed as emergent properties of internal phenotypes, such as molecular profiles, in interaction with the environment. Using biomass accumulation and shoot-root allometry as external traits in diverse genotypes of the model grass Brachypodium distachyon, we established correlative models between genotypes and metabolite profiles (metabotypes) as internal phenotypes, and between metabotypes and external phenotypes under two contrasting watering regimes. Our results demonstrate the potential for employing metabotypes as an integrator in predicting external phenotypes from genomic information.
Collapse
Affiliation(s)
- Pubudu P Handakumbura
- The Environmental Molecular Sciences Laboratory (EMSL), Pacific Northwest National Laboratory (PNNL), Washington, WA, 99352, USA.
| | - Bryan Stanfill
- Advanced Computing, Computing and Analytics Division, PNNL, Richland, WA, 99352, USA
| | - Albert Rivas-Ubach
- The Environmental Molecular Sciences Laboratory (EMSL), Pacific Northwest National Laboratory (PNNL), Washington, WA, 99352, USA
| | - Dan Fortin
- Advanced Computing, Computing and Analytics Division, PNNL, Richland, WA, 99352, USA
| | - John P Vogel
- US Department of Energy (DOE) Joint Genome Institute (JGI), Walnut Creek, CA, 94598, USA
| | - Christer Jansson
- The Environmental Molecular Sciences Laboratory (EMSL), Pacific Northwest National Laboratory (PNNL), Washington, WA, 99352, USA.
| |
Collapse
|
39
|
Pascovici D, Wu JX, McKay MJ, Joseph C, Noor Z, Kamath K, Wu Y, Ranganathan S, Gupta V, Mirzaei M. Clinically Relevant Post-Translational Modification Analyses-Maturing Workflows and Bioinformatics Tools. Int J Mol Sci 2018; 20:E16. [PMID: 30577541 PMCID: PMC6337699 DOI: 10.3390/ijms20010016] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/09/2018] [Accepted: 12/17/2018] [Indexed: 01/04/2023] Open
Abstract
Post-translational modifications (PTMs) can occur soon after translation or at any stage in the lifecycle of a given protein, and they may help regulate protein folding, stability, cellular localisation, activity, or the interactions proteins have with other proteins or biomolecular species. PTMs are crucial to our functional understanding of biology, and new quantitative mass spectrometry (MS) and bioinformatics workflows are maturing both in labelled multiplexed and label-free techniques, offering increasing coverage and new opportunities to study human health and disease. Techniques such as Data Independent Acquisition (DIA) are emerging as promising approaches due to their re-mining capability. Many bioinformatics tools have been developed to support the analysis of PTMs by mass spectrometry, from prediction and identifying PTM site assignment, open searches enabling better mining of unassigned mass spectra-many of which likely harbour PTMs-through to understanding PTM associations and interactions. The remaining challenge lies in extracting functional information from clinically relevant PTM studies. This review focuses on canvassing the options and progress of PTM analysis for large quantitative studies, from choosing the platform, through to data analysis, with an emphasis on clinically relevant samples such as plasma and other body fluids, and well-established tools and options for data interpretation.
Collapse
Affiliation(s)
- Dana Pascovici
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia.
| | - Jemma X Wu
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia.
| | - Matthew J McKay
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia.
| | - Chitra Joseph
- Department of Clinical Medicine, Macquarie University, Sydney, NSW 2109, Australia.
| | - Zainab Noor
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Karthik Kamath
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia.
| | - Yunqi Wu
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia.
| | - Shoba Ranganathan
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Vivek Gupta
- Department of Clinical Medicine, Macquarie University, Sydney, NSW 2109, Australia.
| | - Mehdi Mirzaei
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 2109, Australia.
- Department of Clinical Medicine, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
40
|
Peptide filtering differently affects the performances of XIC-based quantification methods. J Proteomics 2018; 193:131-141. [PMID: 30312678 DOI: 10.1016/j.jprot.2018.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 10/02/2018] [Accepted: 10/08/2018] [Indexed: 11/20/2022]
Abstract
In bottom-up proteomics, data are acquired on peptides resulting from proteolysis. In XIC-based quantification, the quality of the estimation of protein abundance depends on how peptide data are filtered and on which quantification method is used to express peptide intensity as protein abundance. So far, these two questions have been addressed independently. Here, we studied to what extent the relative performances of the quantification methods depend on the filters applied to peptide intensity data. To this end, we performed a spike-in experiment using Universal Protein Standard to evaluate the performances of five quantification methods in five datasets obtained after application of four peptide filters. Estimated protein abundances were not equally affected by filters depending on the computation mode and the type of data for quantification. Furthermore, we found that filters could have contrasting effects depending on the quantification objective. Intensity modeling proved to be the most robust method, providing the best results in the absence of any filter. However, the different quantification methods can achieve similar performances when appropriate peptide filters are used. Altogether, our findings provide insights into how best to handle intensity data according to the quantification objective and the experimental design. SIGNIFICANCE: We believe that our results are of major importance because they address, as far as we know for the first time, the crossed-effects of peptide intensity data filtering and XIC-based quantification methods on protein quantification. While previous papers have dealt with peptide filtering independently of the quantification method, here we combined four peptide filters (based on peptide sharing between proteins, retention time variability, peptides occurrence and peptide intensity profiles) with five XIC-based quantification methods representing different modes of calculating protein abundances from peptide intensities. For these different combinations, we analyzed the quality of protein quantification in terms of precision, accuracy and linearity of response to increasing protein concentration using a spike-in experiment. We showed that not only filters effect on the estimation of protein abundances depend on the quantification methods but also that quantification methods can reach similar performances when appropriate peptide filters are used. Also, depending on the quantification objective, i.e. absolute or relative, filters can have contrasting effects and we demonstrated that protein quantification by the peptide intensity modeling was the most robust method.
Collapse
|
41
|
Sadler NC, Webb-Robertson BJM, Clauss TR, Pounds JG, Corley R, Wright AT. High-Fat Diets Alter the Modulatory Effects of Xenobiotics on Cytochrome P450 Activities. Chem Res Toxicol 2018; 31:308-318. [PMID: 29688711 DOI: 10.1021/acs.chemrestox.8b00008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cytochrome P450 monooxygenase (P450) enzymes metabolize critical endogenous chemicals and oxidize nearly all xenobiotics. Dysregulated P450 activities lead to altered capacity for drug metabolism and cellular stress. The effects of mixed exposures on P450 expression and activity are variable and elusive. A high-fat diet (HFD) is a common exposure that results in obesity and associated pathologies including hepatotoxicity. Herein, we report the effects of cigarette smoke on P450 activities of normal weight and HFD induced obese mice. Activity-based protein profiling results indicate that HFD mice had significantly decreased P450 activity, likely instigated by proinflammatory chemicals, and that P450 enzymes involved in detoxification, xenobiotic metabolism, and bile acid synthesis were effected by HFD and smoke interaction. Smoking increased activity of all lung P450 and coexposure to diet effected P450 2s1. We need to expand our understanding of common exposures coupled to altered P450 metabolism to enhance the safety and efficacy of therapeutic drug dosing.
Collapse
Affiliation(s)
- Natalie C Sadler
- Chemical Biology & Exposure Sciences, Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 United States
| | - Bobbie-Jo M Webb-Robertson
- Chemical Biology & Exposure Sciences, Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 United States
| | - Therese R Clauss
- Chemical Biology & Exposure Sciences, Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 United States
| | - Joel G Pounds
- Chemical Biology & Exposure Sciences, Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 United States
| | - Richard Corley
- Chemical Biology & Exposure Sciences, Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 United States
| | - Aaron T Wright
- Chemical Biology & Exposure Sciences, Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 United States
| |
Collapse
|
42
|
MERS-CoV and H5N1 influenza virus antagonize antigen presentation by altering the epigenetic landscape. Proc Natl Acad Sci U S A 2018; 115:E1012-E1021. [PMID: 29339515 PMCID: PMC5798318 DOI: 10.1073/pnas.1706928115] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Convergent evolution dictates that diverse groups of viruses will target both similar and distinct host pathways to manipulate the immune response and improve infection. In this study, we sought to leverage this uneven viral antagonism to identify critical host factors that govern disease outcome. Utilizing a systems-based approach, we examined differential regulation of IFN-γ-dependent genes following infection with robust respiratory viruses including influenza viruses [A/influenza/Vietnam/1203/2004 (H5N1-VN1203) and A/influenza/California/04/2009 (H1N1-CA04)] and coronaviruses [severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome CoV (MERS-CoV)]. Categorizing by function, we observed down-regulation of gene expression associated with antigen presentation following both H5N1-VN1203 and MERS-CoV infection. Further examination revealed global down-regulation of antigen-presentation gene expression, which was confirmed by proteomics for both H5N1-VN1203 and MERS-CoV infection. Importantly, epigenetic analysis suggested that DNA methylation, rather than histone modification, plays a crucial role in MERS-CoV-mediated antagonism of antigen-presentation gene expression; in contrast, H5N1-VN1203 likely utilizes a combination of epigenetic mechanisms to target antigen presentation. Together, the results indicate a common mechanism utilized by H5N1-VN1203 and MERS-CoV to modulate antigen presentation and the host adaptive immune response.
Collapse
|
43
|
Webb-Robertson BJM, Bramer LM, Jensen JL, Kobold MA, Stratton KG, White AM, Rodland KD. P-MartCancer-Interactive Online Software to Enable Analysis of Shotgun Cancer Proteomic Datasets. Cancer Res 2017; 77:e47-e50. [PMID: 29092938 PMCID: PMC5679244 DOI: 10.1158/0008-5472.can-17-0335] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/19/2017] [Accepted: 07/12/2017] [Indexed: 11/16/2022]
Abstract
P-MartCancer is an interactive web-based software environment that enables statistical analyses of peptide or protein data, quantitated from mass spectrometry-based global proteomics experiments, without requiring in-depth knowledge of statistical programming. P-MartCancer offers a series of statistical modules associated with quality assessment, peptide and protein statistics, protein quantification, and exploratory data analyses driven by the user via customized workflows and interactive visualization. Currently, P-MartCancer offers access and the capability to analyze multiple cancer proteomic datasets generated through the Clinical Proteomics Tumor Analysis Consortium at the peptide, gene, and protein levels. P-MartCancer is deployed as a web service (https://pmart.labworks.org/cptac.html), alternatively available via Docker Hub (https://hub.docker.com/r/pnnl/pmart-web/). Cancer Res; 77(21); e47-50. ©2017 AACR.
Collapse
Affiliation(s)
| | - Lisa M Bramer
- Pacific Northwest National Laboratory, Richland, Washington
| | | | | | | | - Amanda M White
- Pacific Northwest National Laboratory, Richland, Washington
| | | |
Collapse
|
44
|
Ortega C, Frando A, Webb-Robertson BJ, Anderson LN, Fleck N, Flannery EL, Fishbaugher M, Murphree TA, Hansen JR, Smith RD, Kappe SHI, Wright AT, Grundner C. A Global Survey of ATPase Activity in Plasmodium falciparum Asexual Blood Stages and Gametocytes. Mol Cell Proteomics 2017; 17:111-120. [PMID: 29079720 DOI: 10.1074/mcp.ra117.000088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/03/2017] [Indexed: 01/12/2023] Open
Abstract
Effective malaria control and elimination in hyperendemic areas of the world will require treatment of the Plasmodium falciparum (Pf) blood stage that causes disease as well as the gametocyte stage that is required for transmission from humans to the mosquito vector. Most currently used therapies do not kill gametocytes, a highly specialized, non-replicating sexual parasite stage. Further confounding next generation drug development against Pf is the unknown metabolic state of the gametocyte and the lack of known biochemical activity for most parasite gene products in general. Here, we take a systematic activity-based proteomics approach to survey the activity of the large and druggable ATPase family in replicating blood stage asexual parasites and transmissible, non-replicating sexual gametocytes. ATPase activity broadly changes during the transition from asexual schizonts to sexual gametocytes, indicating altered metabolism and regulatory roles of ATPases specific for each lifecycle stage. We further experimentally confirm existing annotation and predict ATPase function for 38 uncharacterized proteins. By mapping the activity of ATPases associated with gametocytogenesis, we assign biochemical activity to a large number of uncharacterized proteins and identify new candidate transmission blocking targets.
Collapse
Affiliation(s)
- Corrie Ortega
- From the ‡Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, Washington 98109
| | - Andrew Frando
- From the ‡Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, Washington 98109.,§Department of Global Health, University of Washington, Seattle, Washington 98195
| | - Bobbie-Jo Webb-Robertson
- ¶Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Lindsey N Anderson
- ¶Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Neil Fleck
- From the ‡Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, Washington 98109
| | - Erika L Flannery
- From the ‡Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, Washington 98109
| | - Matthew Fishbaugher
- From the ‡Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, Washington 98109
| | - Taylor A Murphree
- ¶Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Joshua R Hansen
- ¶Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Richard D Smith
- ¶Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Stefan H I Kappe
- From the ‡Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, Washington 98109.,§Department of Global Health, University of Washington, Seattle, Washington 98195
| | - Aaron T Wright
- ¶Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352
| | - Christoph Grundner
- From the ‡Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, Washington 98109; .,§Department of Global Health, University of Washington, Seattle, Washington 98195
| |
Collapse
|
45
|
Resjö S, Brus M, Ali A, Meijer HJG, Sandin M, Govers F, Levander F, Grenville-Briggs L, Andreasson E. Proteomic Analysis of Phytophthora infestans Reveals the Importance of Cell Wall Proteins in Pathogenicity. Mol Cell Proteomics 2017; 16:1958-1971. [PMID: 28935716 DOI: 10.1074/mcp.m116.065656] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 09/19/2017] [Indexed: 11/06/2022] Open
Abstract
The oomycete Phytophthora infestans is the most harmful pathogen of potato. It causes the disease late blight, which generates increased yearly costs of up to one billion euro in the EU alone and is difficult to control. We have performed a large-scale quantitative proteomics study of six P. infestans life stages with the aim to identify proteins that change in abundance during development, with a focus on preinfectious life stages. Over 10 000 peptides from 2061 proteins were analyzed. We identified several abundance profiles of proteins that were up- or downregulated in different combinations of life stages. One of these profiles contained 59 proteins that were more abundant in germinated cysts and appressoria. A large majority of these proteins were not previously recognized as being appressorial proteins or involved in the infection process. Among those are proteins with putative roles in transport, amino acid metabolism, pathogenicity (including one RXLR effector) and cell wall structure modification. We analyzed the expression of the genes encoding nine of these proteins using RT-qPCR and found an increase in transcript levels during disease progression, in agreement with the hypothesis that these proteins are important in early infection. Among the nine proteins was a group involved in cell wall structure modification and adhesion, including three closely related, uncharacterized proteins encoded by PITG_01131, PITG_01132, and PITG_16135, here denoted Piacwp1-3 Transient silencing of these genes resulted in reduced severity of infection, indicating that these proteins are important for pathogenicity. Our results contribute to further insight into P. infestans biology, and indicate processes that might be relevant for the pathogen while preparing for host cell penetration and during infection. The mass spectrometry data have been deposited to ProteomeXchange via the PRIDE partner repository with the data set identifier PXD002446.
Collapse
Affiliation(s)
- Svante Resjö
- From the ‡Department of Plant Protection Biology, Swedish University of Agricultural Sciences, PO Box 102, SE-230 53 Alnarp, Sweden;
| | - Maja Brus
- From the ‡Department of Plant Protection Biology, Swedish University of Agricultural Sciences, PO Box 102, SE-230 53 Alnarp, Sweden
| | - Ashfaq Ali
- From the ‡Department of Plant Protection Biology, Swedish University of Agricultural Sciences, PO Box 102, SE-230 53 Alnarp, Sweden
| | - Harold J G Meijer
- §Laboratory of Phytopathology, Wageningen University and Research, The Netherlands
| | | | - Francine Govers
- §Laboratory of Phytopathology, Wageningen University and Research, The Netherlands
| | - Fredrik Levander
- ¶Department of Immunotechnology, Lund University, Sweden.,‖National Bioinformatics Infrastructure Sweden (NBIS), Lund University, Sweden
| | - Laura Grenville-Briggs
- From the ‡Department of Plant Protection Biology, Swedish University of Agricultural Sciences, PO Box 102, SE-230 53 Alnarp, Sweden
| | - Erik Andreasson
- From the ‡Department of Plant Protection Biology, Swedish University of Agricultural Sciences, PO Box 102, SE-230 53 Alnarp, Sweden
| |
Collapse
|
46
|
Kyle JE, Casey CP, Stratton KG, Zink EM, Kim YM, Zheng X, Monroe ME, Weitz KK, Bloodsworth KJ, Orton DJ, Ibrahim YM, Moore RJ, Lee CG, Pedersen C, Orwoll E, Smith RD, Burnum-Johnson KE, Baker ES. Comparing identified and statistically significant lipids and polar metabolites in 15-year old serum and dried blood spot samples for longitudinal studies. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2017; 31:447-456. [PMID: 27958645 PMCID: PMC5292309 DOI: 10.1002/rcm.7808] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/27/2016] [Accepted: 12/08/2016] [Indexed: 05/17/2023]
Abstract
RATIONALE The use of dried blood spots (DBS) has many advantages over traditional plasma and serum samples such as the smaller blood volume required, storage at room temperature, and ability to sample in remote locations. However, understanding the robustness of different analytes in DBS samples is essential, especially in older samples collected for longitudinal studies. METHODS Here we analyzed the stability of polar metabolites and lipids in DBS samples collected in 2000-2001 and stored at room temperature. The identified and statistically significant molecules were then compared to matched serum samples stored at -80°C to determine if the DBS samples could be effectively used in a longitudinal study following metabolic disease. RESULTS A total of 400 polar metabolites and lipids were identified in the serum and DBS samples using gas chromatograph/mass spectrometry (GC/MS), liquid chromatography (LC)/MS, and LC/ion mobility spectrometry-MS (LC/IMS-MS). The identified polar metabolites overlapped well between the sample types, though only one statistically significant metabolite was conserved in a case-control study of older diabetic males with low amounts of high-density lipoproteins and high body mass indices, triacylglycerides and glucose levels when compared to non-diabetic patients with normal levels, indicating that degradation in the DBS samples affects polar metabolite quantitation. Differences in the lipid identifications indicated that some oxidation occurs in the DBS samples. However, 36 statistically significant lipids correlated in both sample types. CONCLUSIONS The difference in the number of statistically significant polar metabolites and lipids indicated that the lipids did not degrade to as great of a degree as the polar metabolites in the DBS samples and lipid quantitation was still possible. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jennifer E. Kyle
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Cameron P. Casey
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Kelly G. Stratton
- National Security Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Erika M. Zink
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Young-Mo Kim
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Xueyun Zheng
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Matthew E. Monroe
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Karl K. Weitz
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Kent J. Bloodsworth
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Daniel J. Orton
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Yehia M. Ibrahim
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Ronald J. Moore
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Christine G. Lee
- Department of Medicine, Bone and Mineral Unit, Oregon Health and Science University, Portland, OR
- Research Service, Portland Veterans Affairs Medical Center, Portland, OR
| | - Catherine Pedersen
- Department of Medicine, Bone and Mineral Unit, Oregon Health and Science University, Portland, OR
| | - Eric Orwoll
- Department of Medicine, Bone and Mineral Unit, Oregon Health and Science University, Portland, OR
| | - Richard D. Smith
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | | | - Erin S. Baker
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| |
Collapse
|
47
|
Bryant AE, Aldape MJ, Bayer CR, Katahira EJ, Bond L, Nicora CD, Fillmore TL, Clauss TRW, Metz TO, Webb-Robertson BJ, Stevens DL. Effects of delayed NSAID administration after experimental eccentric contraction injury - A cellular and proteomics study. PLoS One 2017; 12:e0172486. [PMID: 28245256 PMCID: PMC5330483 DOI: 10.1371/journal.pone.0172486] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 02/06/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute muscle injuries are exceedingly common and non-steroidal anti-inflammatory drugs (NSAIDs) are widely consumed to reduce the associated inflammation, swelling and pain that peak 1-2 days post-injury. While prophylactic use or early administration of NSAIDs has been shown to delay muscle regeneration and contribute to loss of muscle strength after healing, little is known about the effects of delayed NSAID use. Further, NSAID use following non-penetrating injury has been associated with increased risk and severity of infection, including that due to group A streptococcus, though the mechanisms remain to be elucidated. The present study investigated the effects of delayed NSAID administration on muscle repair and sought mechanisms supporting an injury/NSAID/infection axis. METHODS A murine model of eccentric contraction (EC)-induced injury of the tibialis anterior muscle was used to profile the cellular and molecular changes induced by ketorolac tromethamine administered 47 hr post injury. RESULTS NSAID administration inhibited several important muscle regeneration processes and down-regulated multiple cytoprotective proteins known to inhibit the intrinsic pathway of programmed cell death. These activities were associated with increased caspase activity in injured muscles but were independent of any NSAID effect on macrophage influx or phenotype switching. CONCLUSIONS These findings provide new molecular evidence supporting the notion that NSAIDs have a direct negative influence on muscle repair after acute strain injury in mice and thus add to renewed concern about the safety and benefits of NSAIDS in both children and adults, in those with progressive loss of muscle mass such as the elderly or patients with cancer or AIDS, and those at risk of secondary infection after trauma or surgery.
Collapse
Affiliation(s)
- Amy E. Bryant
- U.S. Department of Veterans Affairs, Office of Research and Development, Boise, ID, United States of America
- University of Washington School of Medicine, Seattle, WA, United States of America
| | - Michael J. Aldape
- U.S. Department of Veterans Affairs, Office of Research and Development, Boise, ID, United States of America
- Northwest Nazarene University, Nampa, ID, United States of America
| | - Clifford R. Bayer
- U.S. Department of Veterans Affairs, Office of Research and Development, Boise, ID, United States of America
| | - Eva J. Katahira
- U.S. Department of Veterans Affairs, Office of Research and Development, Boise, ID, United States of America
| | - Laura Bond
- Boise State University, Boise, ID, United States of America
| | - Carrie D. Nicora
- Pacific Northwest National Laboratory, Richland, WA, United States of America
| | - Thomas L. Fillmore
- Pacific Northwest National Laboratory, Richland, WA, United States of America
| | | | - Thomas O. Metz
- Pacific Northwest National Laboratory, Richland, WA, United States of America
| | | | - Dennis L. Stevens
- U.S. Department of Veterans Affairs, Office of Research and Development, Boise, ID, United States of America
- University of Washington School of Medicine, Seattle, WA, United States of America
| |
Collapse
|
48
|
Burnum-Johnson KE, Kyle JE, Eisfeld AJ, Casey CP, Stratton KG, Gonzalez JF, Habyarimana F, Negretti NM, Sims AC, Chauhan S, Thackray LB, Halfmann PJ, Walters KB, Kim YM, Zink EM, Nicora CD, Weitz KK, Webb-Robertson BJM, Nakayasu ES, Ahmer B, Konkel ME, Motin V, Baric RS, Diamond MS, Kawaoka Y, Waters KM, Smith RD, Metz TO. MPLEx: a method for simultaneous pathogen inactivation and extraction of samples for multi-omics profiling. Analyst 2017; 142:442-448. [PMID: 28091625 PMCID: PMC5283721 DOI: 10.1039/c6an02486f] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The continued emergence and spread of infectious agents is of great concern, and systems biology approaches to infectious disease research can advance our understanding of host-pathogen relationships and facilitate the development of new therapies and vaccines. Molecular characterization of infectious samples outside of appropriate biosafety containment can take place only subsequent to pathogen inactivation. Herein, we describe a modified Folch extraction using chloroform/methanol that facilitates the molecular characterization of infectious samples by enabling simultaneous pathogen inactivation and extraction of proteins, metabolites, and lipids for subsequent mass spectrometry-based multi-omics measurements. This single-sample metabolite, protein and lipid extraction (MPLEx) method resulted in complete inactivation of clinically important bacterial and viral pathogens with exposed lipid membranes, including Yersinia pestis, Salmonella Typhimurium, and Campylobacter jejuni in pure culture, and Yersinia pestis, Campylobacter jejuni, and West Nile, MERS-CoV, Ebola, and influenza H7N9 viruses in infection studies. In addition, >99% inactivation, which increased with solvent exposure time, was also observed for pathogens without exposed lipid membranes including community-associated methicillin-resistant Staphylococcus aureus, Clostridium difficile spores and vegetative cells, and adenovirus type 5. The overall pipeline of inactivation and subsequent proteomic, metabolomic, and lipidomic analyses was evaluated using a human epithelial lung cell line infected with wild-type and mutant influenza H7N9 viruses, thereby demonstrating that MPLEx yields biomaterial of sufficient quality for subsequent multi-omics analyses. Based on these experimental results, we believe that MPLEx will facilitate systems biology studies of infectious samples by enabling simultaneous pathogen inactivation and multi-omics measurements from a single specimen with high success for pathogens with exposed lipid membranes.
Collapse
Affiliation(s)
| | - Jennifer E Kyle
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Amie J Eisfeld
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Cameron P Casey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Kelly G Stratton
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Juan F Gonzalez
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Fabien Habyarimana
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Nicholas M Negretti
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Amy C Sims
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sadhana Chauhan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Larissa B Thackray
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Kevin B Walters
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Young-Mo Kim
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Erika M Zink
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Carrie D Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Bobbie-Jo M Webb-Robertson
- Computational and Statistical Analytics Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Brian Ahmer
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Michael E Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Vladimir Motin
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Katrina M Waters
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
49
|
Khadempour L, Burnum-Johnson KE, Baker ES, Nicora CD, Webb-Robertson BJM, White RA, Monroe ME, Huang EL, Smith RD, Currie CR. The fungal cultivar of leaf-cutter ants produces specific enzymes in response to different plant substrates. Mol Ecol 2016; 25:5795-5805. [PMID: 27696597 DOI: 10.1111/mec.13872] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/17/2016] [Accepted: 09/19/2016] [Indexed: 01/20/2023]
Abstract
Herbivores use symbiotic microbes to help derive energy and nutrients from plant material. Leaf-cutter ants are a paradigmatic example, cultivating their mutualistic fungus Leucoagaricus gongylophorus on plant biomass that workers forage from a diverse collection of plant species. Here, we investigate the metabolic flexibility of the ants' fungal cultivar for utilizing different plant biomass. Using feeding experiments and a novel approach in metaproteomics, we examine the enzymatic response of L. gongylophorus to leaves, flowers, oats or a mixture of all three. Across all treatments, our analysis identified and quantified 1766 different fungal proteins, including 161 putative biomass-degrading enzymes. We found significant differences in the protein profiles in the fungus gardens of subcolonies fed different plant substrates. When provided with leaves or flowers, which contain the majority of their energy as recalcitrant plant polymers, the fungus gardens produced more proteins predicted to break down cellulose: endoglucanase, exoglucanase and β-glucosidase. Further, the complete metaproteomes for the leaves and flowers treatments were very similar, while the mixed substrate treatment closely resembled the treatment with oats alone. This indicates that when provided a mixture of plant substrates, fungus gardens preferentially break down the simpler, more digestible substrates. This flexible, substrate-specific enzymatic response of the fungal cultivar allows leaf-cutter ants to derive energy from a wide range of substrates, which likely contributes to their ability to be dominant generalist herbivores.
Collapse
Affiliation(s)
- Lily Khadempour
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, 53706, USA.,Department of Zoology, University of Wisconsin-Madison, Madison, WI, 53706, USA.,Department of Energy Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Erin S Baker
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Carrie D Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | | | - Richard A White
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Matthew E Monroe
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Eric L Huang
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Cameron R Currie
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, 53706, USA.,Department of Energy Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
50
|
Integrating Transcriptomic and Proteomic Data Using Predictive Regulatory Network Models of Host Response to Pathogens. PLoS Comput Biol 2016; 12:e1005013. [PMID: 27403523 PMCID: PMC4942116 DOI: 10.1371/journal.pcbi.1005013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 06/06/2016] [Indexed: 12/17/2022] Open
Abstract
Mammalian host response to pathogenic infections is controlled by a complex regulatory network connecting regulatory proteins such as transcription factors and signaling proteins to target genes. An important challenge in infectious disease research is to understand molecular similarities and differences in mammalian host response to diverse sets of pathogens. Recently, systems biology studies have produced rich collections of omic profiles measuring host response to infectious agents such as influenza viruses at multiple levels. To gain a comprehensive understanding of the regulatory network driving host response to multiple infectious agents, we integrated host transcriptomes and proteomes using a network-based approach. Our approach combines expression-based regulatory network inference, structured-sparsity based regression, and network information flow to infer putative physical regulatory programs for expression modules. We applied our approach to identify regulatory networks, modules and subnetworks that drive host response to multiple influenza infections. The inferred regulatory network and modules are significantly enriched for known pathways of immune response and implicate apoptosis, splicing, and interferon signaling processes in the differential response of viral infections of different pathogenicities. We used the learned network to prioritize regulators and study virus and time-point specific networks. RNAi-based knockdown of predicted regulators had significant impact on viral replication and include several previously unknown regulators. Taken together, our integrated analysis identified novel module level patterns that capture strain and pathogenicity-specific patterns of expression and helped identify important regulators of host response to influenza infection. An important challenge in infectious disease research is to understand how the human immune system responds to different types of pathogenic infections. An important component of mounting proper response is the transcriptional regulatory network that specifies the context-specific gene expression program in the host cell. However, our understanding of this regulatory network and how it drives context-specific transcriptional programs is incomplete. To address this gap, we performed a network-based analysis of host response to influenza viruses that integrated high-throughput mRNA- and protein measurements and protein-protein interaction networks to identify virus and pathogenicity-specific modules and their upstream physical regulatory programs. We inferred regulatory networks for human cell line and mouse host systems, which recapitulated several known regulators and pathways of the immune response and viral life cycle. We used the networks to study time point and strain-specific subnetworks and to prioritize important regulators of host response. We predicted several novel regulators, both at the mRNA and protein levels, and experimentally verified their role in the virus life cycle based on their ability to significantly impact viral replication.
Collapse
|