151
|
Marini F, Linke J, Binder H. ideal: an R/Bioconductor package for interactive differential expression analysis. BMC Bioinformatics 2020; 21:565. [PMID: 33297942 PMCID: PMC7724894 DOI: 10.1186/s12859-020-03819-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND RNA sequencing (RNA-seq) is an ever increasingly popular tool for transcriptome profiling. A key point to make the best use of the available data is to provide software tools that are easy to use but still provide flexibility and transparency in the adopted methods. Despite the availability of many packages focused on detecting differential expression, a method to streamline this type of bioinformatics analysis in a comprehensive, accessible, and reproducible way is lacking. RESULTS We developed the ideal software package, which serves as a web application for interactive and reproducible RNA-seq analysis, while producing a wealth of visualizations to facilitate data interpretation. ideal is implemented in R using the Shiny framework, and is fully integrated with the existing core structures of the Bioconductor project. Users can perform the essential steps of the differential expression analysis workflow in an assisted way, and generate a broad spectrum of publication-ready outputs, including diagnostic and summary visualizations in each module, all the way down to functional analysis. ideal also offers the possibility to seamlessly generate a full HTML report for storing and sharing results together with code for reproducibility. CONCLUSION ideal is distributed as an R package in the Bioconductor project ( http://bioconductor.org/packages/ideal/ ), and provides a solution for performing interactive and reproducible analyses of summarized RNA-seq expression data, empowering researchers with many different profiles (life scientists, clinicians, but also experienced bioinformaticians) to make the ideal use of the data at hand.
Collapse
Affiliation(s)
- Federico Marini
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 69, 55131 Mainz, Germany
| | - Jan Linke
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 69, 55131 Mainz, Germany
| | - Harald Binder
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Stefan-Meier-Str. 26, 79104 Freiburg, Germany
| |
Collapse
|
152
|
Th17-related mammary immunity, but not a high systemic Th1 immune response is associated with protection against E. coli mastitis. NPJ Vaccines 2020; 5:108. [PMID: 33298970 PMCID: PMC7686320 DOI: 10.1038/s41541-020-00258-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/23/2020] [Indexed: 11/09/2022] Open
Abstract
Vaccination against bovine mastitis lags behind despite high demand from the dairy industry and margin for efficacy improvement. We previously compared two immunization protocols against E. coli using either only the intramuscular route or a combination of intramuscular and mammary ductal routes, also known as 'prime and pull' strategy. A homologous mammary challenge during the memory phase showed that immunization favorably modified the mastitis course, notably in locally immunized cows in comparison to intramuscular and control adjuvant-only groups. Here, we performed whole-blood profiling through RNA-seq transcriptome and plasma cytokine 15-plex analyses at time points of the E. coli mastitis that showed significant clinical and laboratory differences among the groups. Diminished production of inflammatory cytokines and increased IFNγ were detected in the blood of immunized cows, where a T lymphocyte activation profile was evidenced at 12-h post infection. Acute phase neutropenia was less severe in these cows, and pathways related to neutrophil diapedesis and monocyte activation were also present. Furthermore, three intramammary-immunized cows showing faster healing and shorter mastitis duration had gene profiles that differed from their counterparts, but without any clue for the mastitis susceptibility difference. Inasmuch, when gene expression of CD4 T cells was assessed in mammary tissue, enrichment of IL-17-associated pathways was identified in the quarters of intramammary-immunized cows not only after challenge but also in the control quarters that were not infected. These findings indicate that local immunization mobilizes protective mechanisms that rely on the settlement of type 3 immunity-related CD4 T cells prior to infection.
Collapse
|
153
|
Puccio T, Kunka KS, Zhu B, Xu P, Kitten T. Manganese Depletion Leads to Multisystem Changes in the Transcriptome of the Opportunistic Pathogen Streptococcus sanguinis. Front Microbiol 2020; 11:592615. [PMID: 33250881 PMCID: PMC7674665 DOI: 10.3389/fmicb.2020.592615] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022] Open
Abstract
Streptococcus sanguinis is a primary colonizer of teeth and is typically considered beneficial due to its antagonistic relationship with the cariogenic pathogen Streptococcus mutans. However, S. sanguinis can also act as an opportunistic pathogen should it enter the bloodstream and colonize a damaged heart valve, leading to infective endocarditis. Studies have implicated manganese acquisition as an important virulence determinant in streptococcal endocarditis. A knockout mutant lacking the primary manganese import system in S. sanguinis, SsaACB, is severely attenuated for virulence in an in vivo rabbit model. Manganese is a known cofactor for several important enzymes in S. sanguinis, including superoxide dismutase, SodA, and the aerobic ribonucleotide reductase, NrdEF. To determine the effect of manganese depletion on S. sanguinis, we performed transcriptomic analysis on a ΔssaACB mutant grown in aerobic fermentor conditions after the addition of the metal chelator EDTA. Despite the broad specificity of EDTA, analysis of cellular metal content revealed a decrease in manganese, but not in other metals, that coincided with a drop in growth rate. Subsequent supplementation with manganese, but not iron, zinc, or magnesium, restored growth in the fermentor post-EDTA. Reduced activity of Mn-dependent SodA and NrdEF likely contributed to the decreased growth rate post-EDTA, but did not appear entirely responsible. With the exception of the Dps-like peroxide resistance gene, dpr, manganese depletion did not induce stress response systems. By comparing the transcriptome of ΔssaACB cells pre- and post-EDTA, we determined that manganese deprivation led to altered expression of diverse systems. Manganese depletion also led to an apparent induction of carbon catabolite repression in a glucose-independent manner. The combined results suggest that manganese limitation produces effects in S. sanguinis that are diverse and complex, with no single protein or system appearing entirely responsible for the observed growth rate decrease. This study provides further evidence for the importance of this trace element in streptococcal biology. Future studies will focus on determining mechanisms for regulation, as the multitude of changes observed in this study indicate that multiple regulators may respond to manganese levels.
Collapse
Affiliation(s)
| | | | | | | | - Todd Kitten
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
154
|
Newhouse DJ, Vernasco BJ. Developing a transcriptomic framework for testing testosterone-mediated handicap hypotheses. Gen Comp Endocrinol 2020; 298:113577. [PMID: 32739436 DOI: 10.1016/j.ygcen.2020.113577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 12/17/2022]
Abstract
Sexually selected traits are hypothesized to be honest signals of individual quality due to the costs associated with their maintenance, development, and/or production. Testosterone, a sex steroid associated with the development and/or production of sexually selected traits, has been proposed to enforce the honesty of sexually selected traits via its immunosuppressive effects (i.e., the Immunocompetence Handicap Hypothesis) and/or by influencing an individual's exposure/susceptibility to oxidative stress (i.e., the Oxidation Handicap Hypothesis). Previous work testing these hypotheses has primarily focused on physiological measurements of immunity or oxidative stress, but little is known about the molecular pathways by which testosterone could influence immunity and/or oxidative stress pathways. To further understand the transcriptomic consequences of experimentally elevated testosterone in the context of handicap hypotheses, we used previously published RNA-seq data from studies that measured the transcriptome of individuals treated with either a testosterone-filled or an empty (i.e., control) implant. Two studies encompassing three species of bird and three tissue types fit our selection criteria and we reanalyzed the data using weighted gene co-expression network analysis. Testosterone-treated individuals exhibited signatures of immunosuppression and our results describe the molecular pathways underlying this effect. We also provide some evidence to suggest that the transcriptomic signature of immunosuppression is evolutionarily conserved between the three species. While our results provide no evidence to suggest testosterone mediates handicaps via pathways associated with oxidative stress, they do support the hypothesis that testosterone enforces the honesty of sexually-selected traits by influencing an individual's immunocompetence. Overall, this study develops a framework for testing testosterone-mediated handicap hypotheses and provides guidelines for future integrative and comparative studies focused on the proximate mechanisms mediating sexually selected traits.
Collapse
Affiliation(s)
- Daniel J Newhouse
- Department of Biology, East Carolina University, Greenville, NC, USA.
| | - Ben J Vernasco
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
155
|
Swaegers J, Spanier KI, Stoks R. Genetic compensation rather than genetic assimilation drives the evolution of plasticity in response to mild warming across latitudes in a damselfly. Mol Ecol 2020; 29:4823-4834. [PMID: 33031581 DOI: 10.1111/mec.15676] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022]
Abstract
Global warming is causing plastic and evolutionary changes in the phenotypes of ectotherms. Yet, we have limited knowledge on how the interplay between plasticity and evolution shapes thermal responses and underlying gene expression patterns. We assessed thermal reaction norm patterns across the transcriptome and identified associated molecular pathways in northern and southern populations of the damselfly Ischnura elegans. Larvae were reared in a common garden experiment at the mean summer water temperatures experienced at the northern (20°C) and southern (24°C) latitudes. This allowed a space-for-time substitution where the current gene expression levels at 24°C in southern larvae are a proxy for the expected responses of northern larvae under gradual thermal evolution to the predicted 4°C warming. Most differentially expressed genes showed fixed differences across temperatures between latitudes, suggesting that thermal genetic adaptation will mainly evolve through changes in constitutive gene expression. Northern populations also frequently showed plastic responses in gene expression to mild warming, while southern populations were much less responsive to temperature. Thermal responsive genes in northern populations showed to a large extent a pattern of genetic compensation, namely gene expression that was induced at 24°C in northern populations remained at a lower constant level in southern populations, and were associated with metabolic and translation pathways. There was instead little evidence for genetic assimilation of an initial plastic response to mild warming. Our data therefore suggest that genetic compensation rather than genetic assimilation may drive the evolution of plasticity in response to mild warming in this damselfly species.
Collapse
Affiliation(s)
- Janne Swaegers
- Laboratory of Evolutionary Stress Ecology and Ecotoxicology, University of Leuven, Leuven, Belgium
| | - Katina I Spanier
- Laboratory of Computational Biology, University of Leuven, Leuven, Belgium.,Laboratory of Aquatic Ecology, Evolution and Conservation, University of Leuven, Leuven, Belgium
| | - Robby Stoks
- Laboratory of Evolutionary Stress Ecology and Ecotoxicology, University of Leuven, Leuven, Belgium
| |
Collapse
|
156
|
Hauptmann J, Johann L, Marini F, Kitic M, Colombo E, Mufazalov IA, Krueger M, Karram K, Moos S, Wanke F, Kurschus FC, Klein M, Cardoso S, Strauß J, Bolisetty S, Lühder F, Schwaninger M, Binder H, Bechman I, Bopp T, Agarwal A, Soares MP, Regen T, Waisman A. Interleukin-1 promotes autoimmune neuroinflammation by suppressing endothelial heme oxygenase-1 at the blood-brain barrier. Acta Neuropathol 2020; 140:549-567. [PMID: 32651669 PMCID: PMC7498485 DOI: 10.1007/s00401-020-02187-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/05/2020] [Accepted: 06/26/2020] [Indexed: 12/18/2022]
Abstract
The proinflammatory cytokine interleukin 1 (IL-1) is crucially involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Herein, we studied the role of IL-1 signaling in blood-brain barrier (BBB) endothelial cells (ECs), astrocytes and microglia for EAE development, using mice with the conditional deletion of its signaling receptor IL-1R1. We found that IL-1 signaling in microglia and astrocytes is redundant for the development of EAE, whereas the IL-1R1 deletion in BBB-ECs markedly ameliorated disease severity. IL-1 signaling in BBB-ECs upregulated the expression of the adhesion molecules Vcam-1, Icam-1 and the chemokine receptor Darc, all of which have been previously shown to promote CNS-specific inflammation. In contrast, IL-1R1 signaling suppressed the expression of the stress-responsive heme catabolizing enzyme heme oxygenase-1 (HO-1) in BBB-ECs, promoting disease progression via a mechanism associated with deregulated expression of the IL-1-responsive genes Vcam1, Icam1 and Ackr1 (Darc). Mechanistically, our data emphasize a functional crosstalk of BBB-EC IL-1 signaling and HO-1, controlling the transcription of downstream proinflammatory genes promoting the pathogenesis of autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Judith Hauptmann
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lisa Johann
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Federico Marini
- Center of Thrombosis and Hemostasis Mainz (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Maja Kitic
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisa Colombo
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ilgiz A Mufazalov
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Martin Krueger
- Anatomical Institute, University of Leipzig, Leipzig, Germany
| | - Khalad Karram
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sonja Moos
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Dermatology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Florian Wanke
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area Roche Innovation Center, Basel, Switzerland
| | - Florian C Kurschus
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Dermatology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Judith Strauß
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Subhashini Bolisetty
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Harald Binder
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Ingo Bechman
- Anatomical Institute, University of Leipzig, Leipzig, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anupam Agarwal
- Nephrology Research and Training Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
157
|
Canales J, Uribe F, Henríquez-Valencia C, Lovazzano C, Medina J, Vidal EA. Transcriptomic analysis at organ and time scale reveals gene regulatory networks controlling the sulfate starvation response of Solanum lycopersicum. BMC PLANT BIOLOGY 2020; 20:385. [PMID: 32831040 PMCID: PMC7444261 DOI: 10.1186/s12870-020-02590-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/10/2020] [Indexed: 05/17/2023]
Abstract
BACKGROUND Sulfur is a major component of biological molecules and thus an essential element for plants. Deficiency of sulfate, the main source of sulfur in soils, negatively influences plant growth and crop yield. The effect of sulfate deficiency on plants has been well characterized at the physiological, transcriptomic and metabolomic levels in Arabidopsis thaliana and a limited number of crop plants. However, we still lack a thorough understanding of the molecular mechanisms and regulatory networks underlying sulfate deficiency in most plants. In this work we analyzed the impact of sulfate starvation on the transcriptome of tomato plants to identify regulatory networks and key transcriptional regulators at a temporal and organ scale. RESULTS Sulfate starvation reduces the growth of roots and leaves which is accompanied by major changes in the organ transcriptome, with the response being temporally earlier in roots than leaves. Comparative analysis showed that a major part of the Arabidopsis and tomato transcriptomic response to sulfate starvation is conserved between these plants and allowed for the identification of processes specifically regulated in tomato at the transcript level, including the control of internal phosphate levels. Integrative gene network analysis uncovered key transcription factors controlling the temporal expression of genes involved in sulfate assimilation, as well as cell cycle, cell division and photosynthesis during sulfate starvation in tomato roots and leaves. Interestingly, one of these transcription factors presents a high identity with SULFUR LIMITATION1, a central component of the sulfate starvation response in Arabidopsis. CONCLUSIONS Together, our results provide the first comprehensive catalog of sulfate-responsive genes in tomato, as well as novel regulatory targets for future functional analyses in tomato and other crops.
Collapse
Affiliation(s)
- Javier Canales
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile.
- Millennium Institute for Integrative Biology (iBio), Santiago, Chile.
| | - Felipe Uribe
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
- Millennium Institute for Integrative Biology (iBio), Santiago, Chile
| | - Carlos Henríquez-Valencia
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
- Millennium Institute for Integrative Biology (iBio), Santiago, Chile
| | - Carlos Lovazzano
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
- Millennium Institute for Integrative Biology (iBio), Santiago, Chile
| | - Joaquín Medina
- Centro de Biotecnología y Genómica de Plantas (CBGP), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Elena A Vidal
- Millennium Institute for Integrative Biology (iBio), Santiago, Chile.
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
| |
Collapse
|
158
|
Roewe J, Stavrides G, Strueve M, Sharma A, Marini F, Mann A, Smith SA, Kaya Z, Strobl B, Mueller M, Reinhardt C, Morrissey JH, Bosmann M. Bacterial polyphosphates interfere with the innate host defense to infection. Nat Commun 2020; 11:4035. [PMID: 32788578 PMCID: PMC7423913 DOI: 10.1038/s41467-020-17639-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Polyphosphates are linear polymers and ubiquitous metabolites. Bacterial polyphosphates are long chains of hundreds of phosphate units. Here, we report that mouse survival of peritoneal Escherichia coli sepsis is compromised by long-chain polyphosphates, and improves with bacterial polyphosphatekinase deficiency or neutralization using recombinant exopolyphosphatase. Polyphosphate activities are chain-length dependent, impair pathogen clearance, antagonize phagocyte recruitment, diminish phagocytosis and decrease production of iNOS and cytokines. Macrophages bind and internalize polyphosphates, in which their effects are independent of P2Y1 and RAGE receptors. The M1 polarization driven by E. coli derived LPS is misdirected by polyphosphates in favor of an M2 resembling phenotype. Long-chain polyphosphates modulate the expression of more than 1800 LPS/TLR4-regulated genes in macrophages. This interference includes suppression of hundreds of type I interferon-regulated genes due to lower interferon production and responsiveness, blunted STAT1 phosphorylation and reduced MHCII expression. In conclusion, prokaryotic polyphosphates disturb multiple macrophage functions for evading host immunity.
Collapse
Affiliation(s)
- Julian Roewe
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, 55131, Mainz, Germany
| | - Georgios Stavrides
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, 55131, Mainz, Germany
| | - Marcel Strueve
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, 55131, Mainz, Germany
| | - Arjun Sharma
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, 55131, Mainz, Germany
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Federico Marini
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, 55131, Mainz, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center Mainz, 55131, Mainz, Germany
| | - Amrit Mann
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, 55131, Mainz, Germany
| | - Stephanie A Smith
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, 48109-1085, USA
| | - Ziya Kaya
- Department of Medicine III, University of Heidelberg, 69120, Heidelberg, Germany
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, Department of Biomedical Science, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Mathias Mueller
- Institute of Animal Breeding and Genetics, Department of Biomedical Science, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, 55131, Mainz, Germany
| | - James H Morrissey
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, 48109-1085, USA
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, 55131, Mainz, Germany.
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
159
|
Machado FB, Moharana KC, Almeida-Silva F, Gazara RK, Pedrosa-Silva F, Coelho FS, Grativol C, Venancio TM. Systematic analysis of 1298 RNA-Seq samples and construction of a comprehensive soybean (Glycine max) expression atlas. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2020; 103:1894-1909. [PMID: 32445587 DOI: 10.1111/tpj.14850] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/15/2020] [Accepted: 05/06/2020] [Indexed: 05/23/2023]
Abstract
Soybean (Glycine max [L.] Merr.) is a major crop in animal feed and human nutrition, mainly for its rich protein and oil contents. The remarkable rise in soybean transcriptome studies over the past 5 years generated an enormous amount of RNA-seq data, encompassing various tissues, developmental conditions and genotypes. In this study, we have collected data from 1298 publicly available soybean transcriptome samples, processed the raw sequencing reads and mapped them to the soybean reference genome in a systematic fashion. We found that 94% of the annotated genes (52 737/56 044) had detectable expression in at least one sample. Unsupervised clustering revealed three major groups, comprising samples from aerial, underground and seed/seed-related parts. We found 452 genes with uniform and constant expression levels, supporting their roles as housekeeping genes. On the other hand, 1349 genes showed heavily biased expression patterns towards particular tissues. A transcript-level analysis revealed that 95% (70 963 of 74 490) of the assembled transcripts have intron chains exactly matching those from known transcripts, whereas 3256 assembled transcripts represent potentially novel splicing isoforms. The dataset compiled here constitute a new resource for the community, which can be downloaded or accessed through a user-friendly web interface at http://venanciogroup.uenf.br/resources/. This comprehensive transcriptome atlas will likely accelerate research on soybean genetics and genomics.
Collapse
Affiliation(s)
- Fabricio B Machado
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| | - Kanhu C Moharana
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| | - Fabricio Almeida-Silva
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| | - Rajesh K Gazara
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| | - Francisnei Pedrosa-Silva
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| | - Fernanda S Coelho
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| | - Clícia Grativol
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| | - Thiago M Venancio
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| |
Collapse
|
160
|
Organ of Corti size is governed by Yap/Tead-mediated progenitor self-renewal. Proc Natl Acad Sci U S A 2020; 117:13552-13561. [PMID: 32482884 DOI: 10.1073/pnas.2000175117] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Precise control of organ growth and patterning is executed through a balanced regulation of progenitor self-renewal and differentiation. In the auditory sensory epithelium-the organ of Corti-progenitor cells exit the cell cycle in a coordinated wave between E12.5 and E14.5 before the initiation of sensory receptor cell differentiation, making it a unique system for studying the molecular mechanisms controlling the switch between proliferation and differentiation. Here we identify the Yap/Tead complex as a key regulator of the self-renewal gene network in organ of Corti progenitor cells. We show that Tead transcription factors bind directly to the putative regulatory elements of many stemness- and cell cycle-related genes. We also show that the Tead coactivator protein, Yap, is degraded specifically in the Sox2-positive domain of the cochlear duct, resulting in down-regulation of Tead gene targets. Further, conditional loss of the Yap gene in the inner ear results in the formation of significantly smaller auditory and vestibular sensory epithelia, while conditional overexpression of a constitutively active version of Yap, Yap5SA, is sufficient to prevent cell cycle exit and to prolong sensory tissue growth. We also show that viral gene delivery of Yap5SA in the postnatal inner ear sensory epithelia in vivo drives cell cycle reentry after hair cell loss. Taken together, these data highlight the key role of the Yap/Tead transcription factor complex in maintaining inner ear progenitors during development, and suggest new strategies to induce sensory cell regeneration.
Collapse
|
161
|
Sadler E, Ryals MM, May LA, Martin D, Welsh N, Boger ET, Morell RJ, Hertzano R, Cunningham LL. Cell-Specific Transcriptional Responses to Heat Shock in the Mouse Utricle Epithelium. Front Cell Neurosci 2020; 14:123. [PMID: 32528249 PMCID: PMC7247426 DOI: 10.3389/fncel.2020.00123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/15/2020] [Indexed: 12/21/2022] Open
Abstract
Sensory epithelia of the inner ear contain mechanosensory hair cells (HCs) and glia-like supporting cells (SCs), both of which are required for hearing and balance functions. Each of these cell types has unique responses to ototoxic and cytoprotective stimuli. Non-lethal heat stress in the mammalian utricle induces heat shock proteins (HSPs) and protects against ototoxic drug-induced hair cell death. Induction of HSPs in the utricle demonstrates cell-type specificity at the protein level, with HSP70 induction occurring primarily in SCs, while HSP32 (also known as heme oxygenase 1, HMOX1) is induced primarily in resident macrophages. Neither of these HSPs are robustly induced in HCs, suggesting that HCs may have little capacity for induction of stress-induced protective responses. To determine the transcriptional responses to heat shock of these different cell types, we performed cell-type-specific transcriptional profiling using the RiboTag method, which allows for immunoprecipitation (IP) of actively translating mRNAs from specific cell types. RNA-Seq differential gene expression analyses demonstrated that the RiboTag method identified known cell type-specific markers as well as new markers for HCs and SCs. Gene expression differences suggest that HCs and SCs exhibit differential transcriptional heat shock responses. The chaperonin family member Cct8 was significantly enriched only in heat-shocked HCs, while Hspa1l (HSP70 family), and Hspb1 and Cryab (HSP27 and HSP20 families, respectively) were enriched only in SCs. Together our data indicate that HCs exhibit a limited but unique heat shock response, and SCs exhibit a broader and more robust transcriptional response to protective heat stress.
Collapse
Affiliation(s)
- Erica Sadler
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Matthew M Ryals
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States.,Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lindsey A May
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Daniel Martin
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States.,Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Nora Welsh
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Erich T Boger
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Robert J Morell
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| | - Ronna Hertzano
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.,Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Lisa L Cunningham
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
162
|
Hirata AHDL, Rocha LADJ, da Silva VA, de Almeida RJ, Bacigalupo LDS, Varela P, Martins L, Pesquero JB, Dellê H, Camacho CP. Circulating RNA Transcriptome of Pregnant Women with TSH Just Above the Trimester-Specific Reference and its Correlation with the Hypertensive Phenotype. Sci Rep 2020; 10:6439. [PMID: 32296081 PMCID: PMC7160149 DOI: 10.1038/s41598-020-63040-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 03/19/2020] [Indexed: 11/09/2022] Open
Abstract
During gestation, a woman’s body undergoes physiological changes that alter thyroid function. Pregnant women with hypothyroidism may exhibit gestational complications, including hypertension and preeclampsia. We investigated differentially expressed genes (DEGs) in circulating RNAs from pregnant women with TSH levels just above the normal range to determine the impact of a mild elevation of TSH in pregnancy. We selected three women with healthy thyroid pregnancy (HTP), three pregnant women with gestational hypothyroidism (GHT), and three nonpregnant women (NPG) to construct transcriptome libraries. We also compared our results with data from the GEO dataset and DisGeNET. We identified 1500 DEG in GHT and 1656 DEG in HTP. From GEO dataset, we recognized 453 DEGs in trimester-specific plasma RNA, 1263 DEGs in placental tissues from healthy women, 1031 DEGs from preeclamptic uteroplacental tissues and 1657 DEGs from placental tissues from severely preeclamptic women. In this scenario, 12.26% and 12.86% genes were shared between these datasets in GHT and HTP, respectively. We stablished 62 genes in GHT DEGs related to hypertensive phenotype hallmarks. In conclusion, even in women with a mild TSH increment, we were able to detect some DEGs that could be associated with a hypertensive phenotype.
Collapse
Affiliation(s)
- Andréa Harumy de Lima Hirata
- Molecular Innovation and Biotechnology Laboratory, Postgraduate Program in Medicine, Universidade Nove de Julho (Uninove), São Paulo, SP, Brazil
| | - Luiz Antônio de Jesus Rocha
- Molecular Innovation and Biotechnology Laboratory, Postgraduate Program in Medicine, Universidade Nove de Julho (Uninove), São Paulo, SP, Brazil
| | - Valdelena Alessandra da Silva
- Molecular Innovation and Biotechnology Laboratory, Postgraduate Program in Medicine, Universidade Nove de Julho (Uninove), São Paulo, SP, Brazil
| | - Robson José de Almeida
- Molecular Innovation and Biotechnology Laboratory, Postgraduate Program in Medicine, Universidade Nove de Julho (Uninove), São Paulo, SP, Brazil
| | - Lucas Dos Santos Bacigalupo
- Molecular Innovation and Biotechnology Laboratory, Postgraduate Program in Medicine, Universidade Nove de Julho (Uninove), São Paulo, SP, Brazil.,Department of Obstetrics and Gynecology, Conjunto Hospitalar do Mandaqui, São Paulo, SP, Brazil
| | - Patrícia Varela
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Leonardo Martins
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - João Bosco Pesquero
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Humberto Dellê
- Molecular Innovation and Biotechnology Laboratory, Postgraduate Program in Medicine, Universidade Nove de Julho (Uninove), São Paulo, SP, Brazil
| | - Cleber Pinto Camacho
- Molecular Innovation and Biotechnology Laboratory, Postgraduate Program in Medicine, Universidade Nove de Julho (Uninove), São Paulo, SP, Brazil.
| |
Collapse
|
163
|
Dogra P, Rancan C, Ma W, Toth M, Senda T, Carpenter DJ, Kubota M, Matsumoto R, Thapa P, Szabo PA, Li Poon MM, Li J, Arakawa-Hoyt J, Shen Y, Fong L, Lanier LL, Farber DL. Tissue Determinants of Human NK Cell Development, Function, and Residence. Cell 2020; 180:749-763.e13. [PMID: 32059780 DOI: 10.1016/j.cell.2020.01.022] [Citation(s) in RCA: 283] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/09/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022]
Abstract
Immune responses in diverse tissue sites are critical for protective immunity and homeostasis. Here, we investigate how tissue localization regulates the development and function of human natural killer (NK) cells, innate lymphocytes important for anti-viral and tumor immunity. Integrating high-dimensional analysis of NK cells from blood, lymphoid organs, and mucosal tissue sites from 60 individuals, we identify tissue-specific patterns of NK cell subset distribution, maturation, and function maintained across age and between individuals. Mature and terminally differentiated NK cells with enhanced effector function predominate in blood, bone marrow, spleen, and lungs and exhibit shared transcriptional programs across sites. By contrast, precursor and immature NK cells with reduced effector capacity populate lymph nodes and intestines and exhibit tissue-resident signatures and site-specific adaptations. Together, our results reveal anatomic control of NK cell development and maintenance as tissue-resident populations, whereas mature, terminally differentiated subsets mediate immunosurveillance through diverse peripheral sites. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Pranay Dogra
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Chiara Rancan
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Wenji Ma
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Marta Toth
- Department of Immunology, Faculty of Medicine, University of Debrecen and Doctoral School of Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Takashi Senda
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Dustin J Carpenter
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Masaru Kubota
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Rei Matsumoto
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Puspa Thapa
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Peter A Szabo
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Maya Meimei Li Poon
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Jacky Li
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Janice Arakawa-Hoyt
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lewis L Lanier
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
164
|
Lückel C, Picard F, Raifer H, Campos Carrascosa L, Guralnik A, Zhang Y, Klein M, Bittner S, Steffen F, Moos S, Marini F, Gloury R, Kurschus FC, Chao YY, Bertrams W, Sexl V, Schmeck B, Bonetti L, Grusdat M, Lohoff M, Zielinski CE, Zipp F, Kallies A, Brenner D, Berger M, Bopp T, Tackenberg B, Huber M. IL-17 + CD8 + T cell suppression by dimethyl fumarate associates with clinical response in multiple sclerosis. Nat Commun 2019; 10:5722. [PMID: 31844089 PMCID: PMC6915776 DOI: 10.1038/s41467-019-13731-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/21/2019] [Indexed: 12/20/2022] Open
Abstract
IL-17-producing CD8+ (Tc17) cells are enriched in active lesions of patients with multiple sclerosis (MS), suggesting a role in the pathogenesis of autoimmunity. Here we show that amelioration of MS by dimethyl fumarate (DMF), a mechanistically elusive drug, associates with suppression of Tc17 cells. DMF treatment results in reduced frequency of Tc17, contrary to Th17 cells, and in a decreased ratio of the regulators RORC-to-TBX21, along with a shift towards cytotoxic T lymphocyte gene expression signature in CD8+ T cells from MS patients. Mechanistically, DMF potentiates the PI3K-AKT-FOXO1-T-BET pathway, thereby limiting IL-17 and RORγt expression as well as STAT5-signaling in a glutathione-dependent manner. This results in chromatin remodeling at the Il17 locus. Consequently, T-BET-deficiency in mice or inhibition of PI3K-AKT, STAT5 or reactive oxygen species prevents DMF-mediated Tc17 suppression. Overall, our data disclose a DMF-AKT-T-BET driven immune modulation and suggest putative therapy targets in MS and beyond. Dimethyl fumarate (DMF) is a therapy for multiple sclerosis (MS) with undetermined mechanism of action. Here the authors find that clinical response to DMF associates with decrease in IL-17-producing CD8+ T cells (Tc17), delineate molecular pathways involved, and show that DMF suppresses Tc17 pathogenicity in a mouse model of MS.
Collapse
Affiliation(s)
- Christina Lückel
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, 35043, Marburg, Germany.,Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Felix Picard
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, 35043, Marburg, Germany
| | - Hartmann Raifer
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, 35043, Marburg, Germany.,Core-Facility Flow Cytometry, University of Marburg, 35043, Marburg, Germany
| | - Lucia Campos Carrascosa
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, 35043, Marburg, Germany.,Laboratory of Gastroentrology and Hepatology, Erasmus MC University Medical Center, 3015 CE, Rotterdam, Netherlands
| | - Anna Guralnik
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, 35043, Marburg, Germany
| | - Yajuan Zhang
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, 35043, Marburg, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology at the University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Falk Steffen
- Department of Neurology at the University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Sonja Moos
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Federico Marini
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Renee Gloury
- The Peter Doherty Institute for Infection and Immunity, Dept. of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, 3000, Australia.,The Walter and Eliza Hall Institute of Medical Research, 1 G Royal Parade, Parkville, VIC, 3052, Australia
| | - Florian C Kurschus
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany.,Department of Dermatology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Ying-Yin Chao
- Center for Translational Cancer Research TranslaTUM, Technical University of Munich, 81675, Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| | - Wilhelm Bertrams
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), 35043, Marburg, Germany
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), 35043, Marburg, Germany.,Dept. of Medicine, Pulmonary and Critical Care Medicine, University Medical Center Giessen and Marburg, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), 35043, Marburg, Germany
| | - Lynn Bonetti
- Dept. of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, L-4354, Luxembourg
| | - Melanie Grusdat
- Dept. of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, L-4354, Luxembourg
| | - Michael Lohoff
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, 35043, Marburg, Germany
| | - Christina E Zielinski
- Center for Translational Cancer Research TranslaTUM, Technical University of Munich, 81675, Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| | - Frauke Zipp
- Department of Neurology at the University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity, Dept. of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, 3000, Australia.,The Walter and Eliza Hall Institute of Medical Research, 1 G Royal Parade, Parkville, VIC, 3052, Australia
| | - Dirk Brenner
- Dept. of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, L-4354, Luxembourg.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg.,Odense Research Center for Anaphylaxis, Dept. of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, DK-5000, Denmark
| | - Michael Berger
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, 9112001, Israel
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Björn Tackenberg
- Center of Neuroimmunology, Dept. of Neurology, University of Marburg, 35043, Marburg, Germany
| | - Magdalena Huber
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, 35043, Marburg, Germany.
| |
Collapse
|
165
|
Alonso DP, Campos M, Troca H, Kunii R, Tripet F, Ribolla PEM. Gene expression profile of Aedes aegypti females in courtship and mating. Sci Rep 2019; 9:15492. [PMID: 31664152 PMCID: PMC6820793 DOI: 10.1038/s41598-019-52268-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 10/15/2019] [Indexed: 11/24/2022] Open
Abstract
Aedes aegypti is the most synanthropic and anthropophilic mosquito of Culicidae. This species always cohabits with humans and is extremely opportunistic. Vector dispersal is directly related to the ability of the females on successfully finding a mate in a generally patchy urban scenario. In the present work, we investigate transcriptional changes in Ae. aegypti females during the courtship process and after mating. We observe a substantial alteration in gene expression triggered just upon contact with Ae. aegypti males, which in turn was not fully correlated to the changes triggered by the contact. After analysing shared significant differentially regulated genes between conspecific contact and insemination, the major part of the observed transcriptomic change triggered by contact is reversed after mating, indicating an intermediary situation between naive and mating conditions that we hypothesize to be crucial for mating success. Upon contact, several chemosensory related genes are repressed, especially odorant binding proteins. Most of these genes return to higher expression rates after mating. None of these genes are significantly regulated by the encounter of a different species, Aedes albopictus. The results presented here might be applied to an innovative control approach focusing on the semiochemical systems of mosquitoes in an effort to disrupt undesirable host–insect interaction to reduce the risk of pathogen transmission to humans.
Collapse
Affiliation(s)
- Diego Peres Alonso
- Sao Paulo State University - UNESP, Biotechnology Institute and Bioscience Institute, Botucatu, 18618-689, Brazil
| | - Melina Campos
- Sao Paulo State University - UNESP, Biotechnology Institute and Bioscience Institute, Botucatu, 18618-689, Brazil
| | - Heitor Troca
- Sao Paulo State University - UNESP, Biotechnology Institute and Bioscience Institute, Botucatu, 18618-689, Brazil
| | - Rafael Kunii
- Sao Paulo State University - UNESP, Biotechnology Institute and Bioscience Institute, Botucatu, 18618-689, Brazil
| | - Frédéric Tripet
- Keele University, Centre for Applied Entomology and Parasitology, Keele, ST5 5BG, UK
| | | |
Collapse
|