1
|
Vinken M, Grimm D, Baatout S, Baselet B, Beheshti A, Braun M, Carstens AC, Casaletto JA, Cools B, Costes SV, De Meulemeester P, Doruk B, Eyal S, Ferreira MJS, Miranda S, Hahn C, Helvacıoğlu Akyüz S, Herbert S, Krepkiy D, Lichterfeld Y, Liemersdorf C, Krüger M, Marchal S, Ritz J, Schmakeit T, Stenuit H, Tabury K, Trittel T, Wehland M, Zhang YS, Putt KS, Zhang ZY, Tagle DA. Taking the 3Rs to a higher level: replacement and reduction of animal testing in life sciences in space research. Biotechnol Adv 2025; 81:108574. [PMID: 40180136 PMCID: PMC12048243 DOI: 10.1016/j.biotechadv.2025.108574] [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: 01/02/2025] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
Human settlements on the Moon, crewed missions to Mars and space tourism will become a reality in the next few decades. Human presence in space, especially for extended periods of time, will therefore steeply increase. However, despite more than 60 years of spaceflight, the mechanisms underlying the effects of the space environment on human physiology are still not fully understood. Animals, ranging in complexity from flies to monkeys, have played a pioneering role in understanding the (patho)physiological outcome of critical environmental factors in space, in particular altered gravity and cosmic radiation. The use of animals in biomedical research is increasingly being criticized because of ethical reasons and limited human relevance. Driven by the 3Rs concept, calling for replacement, reduction and refinement of animal experimentation, major efforts have been focused in the past decades on the development of alternative methods that fully bypass animal testing or so-called new approach methodologies. These new approach methodologies range from simple monolayer cultures of individual primary or stem cells all up to bioprinted 3D organoids and microfluidic chips that recapitulate the complex cellular architecture of organs. Other approaches applied in life sciences in space research contribute to the reduction of animal experimentation. These include methods to mimic space conditions on Earth, such as microgravity and radiation simulators, as well as tools to support the processing, analysis or application of testing results obtained in life sciences in space research, including systems biology, live-cell, high-content and real-time analysis, high-throughput analysis, artificial intelligence and digital twins. The present paper provides an in-depth overview of such methods to replace or reduce animal testing in life sciences in space research.
Collapse
Affiliation(s)
- Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Sarah Baatout
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium; Department of Molecular Biotechnology, Gent University, Gent, Belgium
| | - Bjorn Baselet
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium
| | - Afshin Beheshti
- Center of Space Biomedicine, McGowan Institute for Regenerative Medicine, and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Markus Braun
- German Space Agency, German Aerospace Center, Bonn, Germany
| | | | - James A Casaletto
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Ben Cools
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium; Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium
| | - Sylvain V Costes
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA; Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Phoebe De Meulemeester
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bartu Doruk
- Space Applications Services NV/SA, Sint-Stevens-Woluwe, Belgium; Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Sara Eyal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Silvana Miranda
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium; Department of Molecular Biotechnology, Gent University, Gent, Belgium
| | - Christiane Hahn
- European Space Agency, Human and Robotic Exploration Programmes, Human Exploration Science team, Noordwijk, the Netherlands
| | - Sinem Helvacıoğlu Akyüz
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stefan Herbert
- Space Systems, Airbus Defence and Space, Immenstaad am Bodensee, Germany
| | - Dmitriy Krepkiy
- Office of Special Initiatives, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Yannick Lichterfeld
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Christian Liemersdorf
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany
| | - Shannon Marchal
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany
| | - Jette Ritz
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Theresa Schmakeit
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Hilde Stenuit
- Space Applications Services NV/SA, Sint-Stevens-Woluwe, Belgium
| | - Kevin Tabury
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium
| | - Torsten Trittel
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany; Department of Engineering, Brandenburg University of Applied Sciences, Brandenburg an der Havel, Germany
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany
| | - Yu Shrike Zhang
- Division of Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Karson S Putt
- Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Zhong-Yin Zhang
- Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA; Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Danilo A Tagle
- Office of Special Initiatives, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Gebre SG, Scott RT, Saravia-Butler AM, Lopez DK, Sanders LM, Costes SV. NASA open science data repository: open science for life in space. Nucleic Acids Res 2025; 53:D1697-D1710. [PMID: 39558178 PMCID: PMC11701653 DOI: 10.1093/nar/gkae1116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024] Open
Abstract
Space biology and health data are critical for the success of deep space missions and sustainable human presence off-world. At the core of effectively managing biomedical risks is the commitment to open science principles, which ensure that data are findable, accessible, interoperable, reusable, reproducible and maximally open. The 2021 integration of the Ames Life Sciences Data Archive with GeneLab to establish the NASA Open Science Data Repository significantly enhanced access to a wide range of life sciences, biomedical-clinical and mission telemetry data alongside existing 'omics data from GeneLab. This paper describes the new database, its architecture and new data streams supporting diverse data types and enhancing data submission, retrieval and analysis. Features include the biological data management environment for improved data submission, a new user interface, controlled data access, an enhanced API and comprehensive public visualization tools for environmental telemetry, radiation dosimetry data and 'omics analyses. By fostering global collaboration through its analysis working groups and training programs, the open science data repository promotes widespread engagement in space biology, ensuring transparency and inclusivity in research. It supports the global scientific community in advancing our understanding of spaceflight's impact on biological systems, ensuring humans will thrive in future deep space missions.
Collapse
Affiliation(s)
- Samrawit G Gebre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Ryan T Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | - Danielle K Lopez
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Lauren M Sanders
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| |
Collapse
|
3
|
Francés-Herrero E, Lorenzo-Rebenaque L, Casto-Rebollo C, Vicente JS, Sebastian-Leon P, Bueno-Fernandez C, Rodríguez-Eguren A, Gómez-Álvarez M, Faus A, Diaz-Gimeno P, Marco-Jiménez F, Cervelló I. Oviductal extracellular matrix hydrogels enhance in vitro culture of rabbit embryos and reduce deficiencies during assisted reproductive technologies. Sci Rep 2024; 14:27579. [PMID: 39528559 PMCID: PMC11554825 DOI: 10.1038/s41598-024-77583-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
In vitro embryo culture often falls short of mimicking the physiological dynamism occurring in the reproductive tract, prompting developmental plasticity in mammalian embryos with consequential genotypic and phenotypic deviations. Recent research highlights the potential of biological derivatives in in vitro culture to mitigate these effects, being the extracellular matrix (ECM) one of the most important components in retaining structural and biological signals derived from the native source tissue. Current bioengineering techniques could provide ECM-based biomaterials mimicking the native environment and offering optimal embryonic development. Rabbit oviducts (n = 24) were decellularized and solubilized to create tissue-specific ECM (OviECM) hydrogels. Following physicochemical characterization, these hydrogels were applied as coatings for the in vitro culture of two-cell embryos over 48 h, along with embryos cultured under In vitro control conditions (n = 218/group), which were subsequently transferred to recipient females. A subset of embryos was recovered on day 6 for transcriptomic analysis (n = 75-80/group), while the remaining embryos were used to assess implantation and birth rates. Rabbit weights were monitored over 20 weeks post-delivery, with blood tests conducted at weeks 8 and 20. Bayesian inference methods were used for statistical analysis. Differences were considered relevant if P ≥ 0.8 (80%). No differences in embryo development and morphology were detected between the OviECM coating and In vitro control conditions. However, embryos cultured on these coatings exhibited upregulation of pathways involved in antigen presentation and immune system activation, as well as, increased cellular response to external stimulus and intracellular protein transport. The implantation and live birth rates were significantly higher in the coating group than in the In vitro control group (30.8% vs. 26.1% and 21.2% vs. 18.1%, respectively). During the first 20 weeks of life, the animals from the coating group showed higher weights than the In vitro control group P0 > 0.8. The animals of both experimental groups showed normal blood parameters. Implementation of OviECM coatings allows for improving in vitro conditions and decreases postnatal phenotypic deviations after assisted reproductive technology (ART). This study could initiate a new embryo culture techniques era to guarantee that ART is utilized in the most efficient and safest possible practice.
Collapse
Affiliation(s)
- Emilio Francés-Herrero
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, Universitat de València, 46010, Valencia, Valencia, Spain
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026, Valencia, Valencia, Spain
| | - Laura Lorenzo-Rebenaque
- Institute for Animal Science and Technology (ICTA), Universitat Politècnica de València, 46022, Valencia, Valencia, Spain
| | - Cristina Casto-Rebollo
- Institute for Animal Science and Technology (ICTA), Universitat Politècnica de València, 46022, Valencia, Valencia, Spain
| | - José Salvador Vicente
- Institute for Animal Science and Technology (ICTA), Universitat Politècnica de València, 46022, Valencia, Valencia, Spain
| | - Patricia Sebastian-Leon
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026, Valencia, Valencia, Spain
| | - Clara Bueno-Fernandez
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, Universitat de València, 46010, Valencia, Valencia, Spain
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026, Valencia, Valencia, Spain
| | - Adolfo Rodríguez-Eguren
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026, Valencia, Valencia, Spain
| | - María Gómez-Álvarez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026, Valencia, Valencia, Spain
| | - Amparo Faus
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026, Valencia, Valencia, Spain
| | - Patricia Diaz-Gimeno
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026, Valencia, Valencia, Spain
| | - Francisco Marco-Jiménez
- Institute for Animal Science and Technology (ICTA), Universitat Politècnica de València, 46022, Valencia, Valencia, Spain
| | - Irene Cervelló
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026, Valencia, Valencia, Spain.
| |
Collapse
|
4
|
Casco A, Ohashi M, Johannsen E. Epstein-Barr virus induces host shutoff extensively via BGLF5-independent mechanisms. Cell Rep 2024; 43:114743. [PMID: 39298313 PMCID: PMC11549945 DOI: 10.1016/j.celrep.2024.114743] [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: 03/21/2024] [Revised: 07/08/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Epstein-Barr virus (EBV) is a ubiquitous oncogenic virus associated with multiple cancers and autoimmune diseases. Unlike most herpesviruses, EBV reactivation from latency occurs asymptomatically, allowing it to spread efficiently to other hosts. However, available models are limited by the inefficient and asynchronous reactivation from latency into lytic replication. To address this problem, we develop a dual-fluorescent lytic reporter (DFLR) EBV that specifically labels cells in the early and late stages of replication. Using lymphoblastoid cell lines transformed by DFLR EBV as a model for EBV reactivation in B cells, we observe extensive reprogramming of the host cell transcriptome during lytic cycle progression. This includes widespread shutoff of host gene expression and disruption of mRNA processing. Unexpectedly, host shutoff remains extensive even in cells infected with DFLR EBV deleted for the BGLF5 nuclease. These findings implicate BGLF5-independent mechanisms as the primary drivers of host transcriptome remodeling during EBV lytic replication.
Collapse
Affiliation(s)
- Alejandro Casco
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA
| | - Makoto Ohashi
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA
| | - Eric Johannsen
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA; Department of Medicine, Division of Infectious Diseases, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
5
|
Busoms S, da Silva AC, Escolà G, Abdilzadeh R, Curran E, Bollmann-Giolai A, Bray S, Wilson M, Poschenrieder C, Yant L. Local cryptic diversity in salinity adaptation mechanisms in the wild outcrossing Brassica fruticulosa. Proc Natl Acad Sci U S A 2024; 121:e2407821121. [PMID: 39316046 PMCID: PMC11459175 DOI: 10.1073/pnas.2407821121] [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: 04/18/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
It is normally supposed that populations of the same species should evolve shared mechanisms of adaptation to common stressors due to evolutionary constraint. Here, we describe a system of within-species local adaptation to coastal habitats, Brassica fruticulosa, and detail surprising strategic variability in adaptive responses to high salinity. These different adaptive responses in neighboring populations are evidenced by transcriptomes, diverse physiological outputs, and distinct genomic selective landscapes. In response to high salinity Northern Catalonian populations restrict root-to-shoot Na+ transport, favoring K+ uptake. Contrastingly, Central Catalonian populations accumulate Na+ in leaves and compensate for the osmotic imbalance with compatible solutes such as proline. Despite contrasting responses, both metapopulations were salinity tolerant relative to all inland accessions. To characterize the genomic basis of these divergent adaptive strategies in an otherwise non-saline-tolerant species, we generate a long-read-based genome and population sequencing of 18 populations (nine inland, nine coastal) across the B. fruticulosa species range. Results of genomic and transcriptomic approaches support the physiological observations of distinct underlying mechanisms of adaptation to high salinity and reveal potential genetic targets of these two very recently evolved salinity adaptations. We therefore provide a model of within-species salinity adaptation and reveal cryptic variation in neighboring plant populations in the mechanisms of adaptation to an important natural stressor highly relevant to agriculture.
Collapse
Affiliation(s)
- Silvia Busoms
- Department of Plant Physiology, Universitat Autònoma de Barcelona, Barcelona08193, Spain
| | - Ana C. da Silva
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, NottinghamNG7 2RD, United Kingdom
| | - Glòria Escolà
- Department of Plant Physiology, Universitat Autònoma de Barcelona, Barcelona08193, Spain
| | - Raziyeh Abdilzadeh
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, NottinghamNG7 2RD, United Kingdom
| | - Emma Curran
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, NottinghamNG7 2RD, United Kingdom
| | - Anita Bollmann-Giolai
- Department of Cell and Developmental Biology, John Innes Centre, Norwich Research Park, NorwichNR4 7UH, United Kingdom
| | - Sian Bray
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, NottinghamNG7 2RD, United Kingdom
| | - Michael Wilson
- School of Computer Sciences, Faculty of Science, University of Nottingham, NottinghamNG7 2RD, United Kingdom
| | | | - Levi Yant
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, NottinghamNG7 2RD, United Kingdom
- Department of Botany, Faculty of Science, Charles University, Prague128 01, Czech Republic
| |
Collapse
|
6
|
Tabaro F, Boulard M. 3t-seq: automatic gene expression analysis of single-copy genes, transposable elements, and tRNAs from RNA-seq data. Brief Bioinform 2024; 25:bbae467. [PMID: 39322626 PMCID: PMC11424182 DOI: 10.1093/bib/bbae467] [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: 11/15/2023] [Revised: 06/16/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
RNA sequencing is the gold-standard method to quantify transcriptomic changes between two conditions. The overwhelming majority of data analysis methods available are focused on polyadenylated RNA transcribed from single-copy genes and overlook transcripts from repeated sequences such as transposable elements (TEs). These self-autonomous genetic elements are increasingly studied, and specialized tools designed to handle multimapping sequencing reads are available. Transfer RNAs are transcribed by RNA polymerase III and are essential for protein translation. There is a need for integrated software that is able to analyze multiple types of RNA. Here, we present 3t-seq, a Snakemake pipeline for integrated differential expression analysis of transcripts from single-copy genes, TEs, and tRNA. 3t-seq produces an accessible report and easy-to-use results for downstream analysis starting from raw sequencing data and performing quality control, genome mapping, gene expression quantification, and statistical testing. It implements three methods to quantify TEs expression and one for tRNA genes. It provides an easy-to-configure method to manage software dependencies that lets the user focus on results. 3t-seq is released under MIT license and is available at https://github.com/boulardlab/3t-seq.
Collapse
Affiliation(s)
- Francesco Tabaro
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ercole Ramarini 32, Monterotondo 00015, Italy
| | - Matthieu Boulard
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ercole Ramarini 32, Monterotondo 00015, Italy
| |
Collapse
|
7
|
Abdelfattah F, Schulz H, Wehland M, Corydon TJ, Sahana J, Kraus A, Krüger M, González-Torres LF, Cortés-Sánchez JL, Wise PM, Mushunuri A, Hemmersbach R, Liemersdorf C, Infanger M, Grimm D. Omics Studies of Specialized Cells and Stem Cells under Microgravity Conditions. Int J Mol Sci 2024; 25:10014. [PMID: 39337501 PMCID: PMC11431953 DOI: 10.3390/ijms251810014] [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: 08/20/2024] [Revised: 09/06/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
The primary objective of omics in space with focus on the human organism is to characterize and quantify biological factors that alter structure, morphology, function, and dynamics of human cells exposed to microgravity. This review discusses exciting data regarding genomics, transcriptomics, epigenomics, metabolomics, and proteomics of human cells and individuals in space, as well as cells cultured under simulated microgravity. The NASA Twins Study significantly heightened interest in applying omics technologies and bioinformatics in space and terrestrial environments. Here, we present the available publications in this field with a focus on specialized cells and stem cells exposed to real and simulated microgravity conditions. We summarize current knowledge of the following topics: (i) omics studies on stem cells, (ii) omics studies on benign specialized different cell types of the human organism, (iii) discussing the advantages of this knowledge for space commercialization and exploration, and (iv) summarizing the emerging opportunities for translational regenerative medicine for space travelers and human patients on Earth.
Collapse
Affiliation(s)
- Fatima Abdelfattah
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
- Department of Ophthalmology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Jayashree Sahana
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
| | - Armin Kraus
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Luis Fernando González-Torres
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - José Luis Cortés-Sánchez
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Petra M. Wise
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
| | - Ashwini Mushunuri
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Ruth Hemmersbach
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (R.H.); (C.L.)
| | - Christian Liemersdorf
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (R.H.); (C.L.)
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
| |
Collapse
|
8
|
Gonzalez E, Lee MD, Tierney BT, Lipieta N, Flores P, Mishra M, Beckett L, Finkelstein A, Mo A, Walton P, Karouia F, Barker R, Jansen RJ, Green SJ, Weging S, Kelliher J, Singh NK, Bezdan D, Galazska J, Brereton NJB. Spaceflight alters host-gut microbiota interactions. NPJ Biofilms Microbiomes 2024; 10:71. [PMID: 39209868 PMCID: PMC11362537 DOI: 10.1038/s41522-024-00545-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The ISS rodent habitat has provided crucial insights into the impact of spaceflight on mammals, inducing symptoms characteristic of liver disease, insulin resistance, osteopenia, and myopathy. Although these physiological responses can involve the microbiome on Earth, host-microbiota interactions during spaceflight are still being elucidated. We explore murine gut microbiota and host gene expression in the colon and liver after 29 and 56 days of spaceflight using multiomics. Metagenomics revealed significant changes in 44 microbiome species, including relative reductions in bile acid and butyrate metabolising bacteria like Extibacter muris and Dysosmobacter welbionis. Functional prediction indicate over-representation of fatty acid and bile acid metabolism, extracellular matrix interactions, and antibiotic resistance genes. Host gene expression described corresponding changes to bile acid and energy metabolism, and immune suppression. These changes imply that interactions at the host-gut microbiome interface contribute to spaceflight pathology and that these interactions might critically influence human health and long-duration spaceflight feasibility.
Collapse
Affiliation(s)
- E Gonzalez
- Microbiome Unit, Canadian Centre for Computational Genomics, Department of Human Genetics, McGill University, Montréal, Canada
- Centre for Microbiome Research, McGill University, Montréal, Canada
| | - M D Lee
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - B T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - N Lipieta
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - P Flores
- BioServe Space Technologies, University of Colorado Boulder, Boulder, CO, USA
| | - M Mishra
- Grossman School of Medicine, New York University, New York, USA
| | - L Beckett
- University of Nottingham, Nottingham, NG7 2RD, UK
| | - A Finkelstein
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - A Mo
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - P Walton
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - F Karouia
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Centre for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - R Barker
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Yuri GmbH, Wiesentalstr. 40, 88074, Meckenbeuren, Germany
- University of Wisconsin-Madison, Madison, WI, USA
| | - R J Jansen
- Department of Public Health, North Dakota State University, Fargo, ND, USA
- Genomics, Phenomics, and Bioinformatics Program, North Dakota State University, Fargo, ND, USA
| | - S J Green
- Genomics and Microbiome Core Facility, Rush University Medical Centre, 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - S Weging
- Institute of Computer Science, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - J Kelliher
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - N K Singh
- Department of Industrial Relations, Division of Occupational Safety and Health, Oakland, USA
| | - D Bezdan
- University of Wisconsin-Madison, Madison, WI, USA
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - J Galazska
- Space Biosciences Research Branch, NASA Ames Research Centre, Moffett Field, CA, USA
| | - N J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
9
|
Vicente JS, Valdés-Hernández J, Marco-Jiménez F. Transcriptomic Signatures of the Foetal Liver and Late Prenatal Development in Vitrified Rabbit Embryos. Vet Sci 2024; 11:347. [PMID: 39195801 PMCID: PMC11360234 DOI: 10.3390/vetsci11080347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Assisted reproduction technologies (ARTs) are generally considered safe; however, emerging evidence highlights the need to evaluate potential risks in adulthood to improve safety further. ART procedures like rederivation of embryos by vitrification differ from natural conditions, causing significant disparities between in vitro and in vivo embryos, affecting foetal physiology and postnatal life. This study aims to investigate whether hepatic transcriptome and metabolome changes observed postnatally are already present in foetal livers at the end of gestation. This study compared fresh and vitrified rabbit embryos, finding differences between foetuses obtained by the transfer of fresh and vitrified embryos at 24 days of gestation. Rederived embryos had reduced foetal and liver weights and crown-rump length. However, the offspring of vitrified embryos tended to be born with higher weight, showing compensatory growth in the final week of gestation (59.2 vs. 49.8 g). RNA-Seq analysis revealed 43 differentially expressed genes (DEGs) in the foetal liver of vitrified embryos compared to the fresh group. Notably, downregulated genes included BRAT1, CYP4A7, CYP2B4, RPL23, RPL22L1, PPILAL1, A1BG, IFGGC1, LRRC57, DIPP2, UGT2B14, IRGM1, NUTF2, MPST, and PPP1R1B, while upregulated genes included ACOT8, ERICH3, UBXN2A, METTL9, ALDH3A2, DERPC-like, NR5A2-like, AP-1, COG8, INHBE, and PLA2G4C. Overall, a functional annotation of these DEGs indicated an involvement in lipid metabolism and the stress and inflammatory process or immune response. Thus, our results suggest that vitrification and embryo transfer manipulation induce an adaptive response that can be observed in the liver during the last week of gestation.
Collapse
Affiliation(s)
| | | | - Francisco Marco-Jiménez
- Instituto de Ciencia y Tecnología Animal, Universitat Politècnica de València, 46022 Valencia, Spain; (J.S.V.); (J.V.-H.)
| |
Collapse
|
10
|
Cope H, Elsborg J, Demharter S, McDonald JT, Wernecke C, Parthasarathy H, Unadkat H, Chatrathi M, Claudio J, Reinsch S, Avci P, Zwart SR, Smith SM, Heer M, Muratani M, Meydan C, Overbey E, Kim J, Chin CR, Park J, Schisler JC, Mason CE, Szewczyk NJ, Willis CRG, Salam A, Beheshti A. Transcriptomics analysis reveals molecular alterations underpinning spaceflight dermatology. COMMUNICATIONS MEDICINE 2024; 4:106. [PMID: 38862781 PMCID: PMC11166967 DOI: 10.1038/s43856-024-00532-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/23/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Spaceflight poses a unique set of challenges to humans and the hostile spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. METHODS To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on space flown murine transcriptomic datasets focused on the skin, biochemical profiles of 50 NASA astronauts and human transcriptomic datasets generated from blood and hair samples of JAXA astronauts, as well as blood samples obtained from the NASA Twins Study, and skin and blood samples from the first civilian commercial mission, Inspiration4. RESULTS Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation are identified as potential drivers for skin health risks during spaceflight. Additionally, a machine learning model is utilized to determine gene pairings associated with spaceflight response in the skin. While we identified spaceflight-induced dysregulation, such as alterations in genes associated with skin barrier function and collagen formation, our results also highlight the remarkable ability for organisms to re-adapt back to Earth via post-flight re-tuning of gene expression. CONCLUSION Our findings can guide future research on developing countermeasures for mitigating spaceflight-associated skin damage.
Collapse
Affiliation(s)
- Henry Cope
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
| | - Jonas Elsborg
- Department of Energy Conversion and Storage, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
- Abzu, Copenhagen, 2150, Denmark
| | | | - J Tyson McDonald
- Department of Radiation Medicine, School of Medicine, Georgetown University, Washington D.C., WA, 20057, USA
| | - Chiara Wernecke
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Department of Aerospace and Geodesy, TUM School of Engineering and Design, Technical University of Munich, Munich, Germany
| | - Hari Parthasarathy
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- College of Engineering and Haas School of Business, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Hriday Unadkat
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- School of Engineering and Applied Science, Princeton University, Princeton, NJ, 08540, USA
| | - Mira Chatrathi
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- College of Letters and Science, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jennifer Claudio
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA
| | - Sigrid Reinsch
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA
| | - Pinar Avci
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 80337, Munich, Germany
| | - Sara R Zwart
- University of Texas Medical Branch, Galveston, TX, USA
| | - Scott M Smith
- Biomedical Research and Environmental Sciences Division, Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, 77058, USA
| | - Martina Heer
- IU International University of Applied Sciences, Erfurt and University of Bonn, Bonn, Germany
| | - Masafumi Muratani
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, 305-8575, Japan
- Department of Genome Biology, Institute of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Cem Meydan
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Eliah Overbey
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Jangkeun Kim
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Christopher R Chin
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Jiwoon Park
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, USA
| | - Jonathan C Schisler
- McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher E Mason
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, USA
| | - Nathaniel J Szewczyk
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Craig R G Willis
- School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Amr Salam
- St John's Institute of Dermatology, King's College London, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Afshin Beheshti
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
11
|
Grigorev K, Nelson TM, Overbey EG, Houerbi N, Kim J, Najjar D, Damle N, Afshin EE, Ryon KA, Thierry-Mieg J, Thierry-Mieg D, Melnick AM, Mateus J, Mason CE. Direct RNA sequencing of astronaut blood reveals spaceflight-associated m6A increases and hematopoietic transcriptional responses. Nat Commun 2024; 15:4950. [PMID: 38862496 PMCID: PMC11166648 DOI: 10.1038/s41467-024-48929-3] [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: 03/29/2023] [Accepted: 05/17/2024] [Indexed: 06/13/2024] Open
Abstract
The advent of civilian spaceflight challenges scientists to precisely describe the effects of spaceflight on human physiology, particularly at the molecular and cellular level. Newer, nanopore-based sequencing technologies can quantitatively map changes in chemical structure and expression at single molecule resolution across entire isoforms. We perform long-read, direct RNA nanopore sequencing, as well as Ultima high-coverage RNA-sequencing, of whole blood sampled longitudinally from four SpaceX Inspiration4 astronauts at seven timepoints, spanning pre-flight, day of return, and post-flight recovery. We report key genetic pathways, including changes in erythrocyte regulation, stress induction, and immune changes affected by spaceflight. We also present the first m6A methylation profiles for a human space mission, suggesting a significant spike in m6A levels immediately post-flight. These data and results represent the first longitudinal long-read RNA profiles and RNA modification maps for each gene for astronauts, improving our understanding of the human transcriptome's dynamic response to spaceflight.
Collapse
Affiliation(s)
- Kirill Grigorev
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Theodore M Nelson
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Center for STEM, University of Austin, Austin, TX, USA
- BioAstra, Inc, New York, NY, USA
| | - Nadia Houerbi
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Evan E Afshin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jean Thierry-Mieg
- National Center for Biotechnology Information (NCBI), National Library of Medicine, NIH, Bethesda, MD, 20894, USA
| | - Danielle Thierry-Mieg
- National Center for Biotechnology Information (NCBI), National Library of Medicine, NIH, Bethesda, MD, 20894, USA
| | - Ari M Melnick
- Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jaime Mateus
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
- WorldQuant Initiative for Quantitative Prediction, New York, NY, USA.
| |
Collapse
|
12
|
Ilangovan H, Kothiyal P, Hoadley KA, Elgart R, Eley G, Eslami P. Harmonizing heterogeneous transcriptomics datasets for machine learning-based analysis to identify spaceflown murine liver-specific changes. NPJ Microgravity 2024; 10:61. [PMID: 38862523 PMCID: PMC11167036 DOI: 10.1038/s41526-024-00379-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 03/08/2024] [Indexed: 06/13/2024] Open
Abstract
NASA has employed high-throughput molecular assays to identify sub-cellular changes impacting human physiology during spaceflight. Machine learning (ML) methods hold the promise to improve our ability to identify important signals within highly dimensional molecular data. However, the inherent limitation of study subject numbers within a spaceflight mission minimizes the utility of ML approaches. To overcome the sample power limitations, data from multiple spaceflight missions must be aggregated while appropriately addressing intra- and inter-study variabilities. Here we describe an approach to log transform, scale and normalize data from six heterogeneous, mouse liver-derived transcriptomics datasets (ntotal = 137) which enabled ML-methods to classify spaceflown vs. ground control animals (AUC ≥ 0.87) while mitigating the variability from mission-of-origin. Concordance was found between liver-specific biological processes identified from harmonized ML-based analysis and study-by-study classical omics analysis. This work demonstrates the feasibility of applying ML methods on integrated, heterogeneous datasets of small sample size.
Collapse
Affiliation(s)
- Hari Ilangovan
- Science Applications International Corporation (SAIC), Reston, VA, 20190, USA.
| | | | - Katherine A Hoadley
- Department of Genetics, Computational Medicine Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Greg Eley
- Scimentis LLC, Statham, GA, 30666, USA
| | - Parastou Eslami
- Universal Artificial Intelligence Inc, Boston, MA, 02130, USA
| |
Collapse
|
13
|
Dixit AR, Meyers AD, Richardson B, Richards JT, Richards SE, Neelam S, Levine HG, Cameron MJ, Zhang Y. Simulated galactic cosmic ray exposure activates dose-dependent DNA repair response and down regulates glucosinolate pathways in arabidopsis seedlings. FRONTIERS IN PLANT SCIENCE 2023; 14:1284529. [PMID: 38162303 PMCID: PMC10757676 DOI: 10.3389/fpls.2023.1284529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/30/2023] [Indexed: 01/03/2024]
Abstract
Outside the protection of Earth's magnetic field, organisms are constantly exposed to space radiation consisting of energetic protons and other heavier charged particles. With the goal of crewed Mars exploration, the production of fresh food during long duration space missions is critical for meeting astronauts' nutritional and psychological needs. However, the biological effects of space radiation on plants have not been sufficiently investigated and characterized. To that end, 10-day-old Arabidopsis seedlings were exposed to simulated Galactic Cosmic Rays (GCR) and assessed for transcriptomic changes. The simulated GCR irradiation was carried out in the NASA Space Radiation Laboratory (NSRL) at Brookhaven National Lab (BNL). The exposures were conducted acutely for two dose points at 40 cGy or 80 cGy, with sequential delivery of proton, helium, oxygen, silicon, and iron ions. Control and irradiated seedlings were then harvested and preserved in RNAlater at 3 hrs post irradiation. Total RNA was isolated for transcriptomic analyses using RNAseq. The data revealed that the transcriptomic responses were dose-dependent, with significant upregulation of DNA repair pathways and downregulation of glucosinolate biosynthetic pathways. Glucosinolates are important for plant pathogen defense and for the taste of a plant, which are both relevant to growing plants for spaceflight. These findings fill in knowledge gaps of how plants respond to radiation in beyond-Earth environments.
Collapse
Affiliation(s)
- Anirudha R. Dixit
- AETOS Systems Inc., LASSO II Contract, Huntsville, AL, United States
| | - Alexander D. Meyers
- NASA Postdoctoral Program, John F. Kennedy Space Center, Merritt Island, FL, United States
| | | | | | | | - Srujana Neelam
- NASA Postdoctoral Program, John F. Kennedy Space Center, Merritt Island, FL, United States
| | - Howard G. Levine
- NASA John F. Kennedy Space Center, Kennedy Space Center, FL, United States
| | - Mark J. Cameron
- Case Western Reserve University, Cleveland, OH, United States
| | - Ye Zhang
- NASA John F. Kennedy Space Center, Kennedy Space Center, FL, United States
| |
Collapse
|
14
|
Li K, Desai R, Scott RT, Steele JR, Machado M, Demharter S, Hoarfrost A, Braun JL, Fajardo VA, Sanders LM, Costes SV. Explainable machine learning identifies multi-omics signatures of muscle response to spaceflight in mice. NPJ Microgravity 2023; 9:90. [PMID: 38092777 PMCID: PMC10719374 DOI: 10.1038/s41526-023-00337-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
The adverse effects of microgravity exposure on mammalian physiology during spaceflight necessitate a deep understanding of the underlying mechanisms to develop effective countermeasures. One such concern is muscle atrophy, which is partly attributed to the dysregulation of calcium levels due to abnormalities in SERCA pump functioning. To identify potential biomarkers for this condition, multi-omics data and physiological data available on the NASA Open Science Data Repository (osdr.nasa.gov) were used, and machine learning methods were employed. Specifically, we used multi-omics (transcriptomic, proteomic, and DNA methylation) data and calcium reuptake data collected from C57BL/6 J mouse soleus and tibialis anterior tissues during several 30+ day-long missions on the international space station. The QLattice symbolic regression algorithm was introduced to generate highly explainable models that predict either experimental conditions or calcium reuptake levels based on multi-omics features. The list of candidate models established by QLattice was used to identify key features contributing to the predictive capability of these models, with Acyp1 and Rps7 proteins found to be the most predictive biomarkers related to the resilience of the tibialis anterior muscle in space. These findings could serve as targets for future interventions aiming to reduce the extent of muscle atrophy during space travel.
Collapse
Affiliation(s)
- Kevin Li
- KBR, Moffett Field, CA, USA
- NASA Space Life Sciences Training Program, Moffett Field, CA, USA
| | - Riya Desai
- College of Letters and Science, University of California at Davis, Davis, CA, USA
| | - Ryan T Scott
- KBR, Moffett Field, CA, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Joel Ricky Steele
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Blue Marble Space, Seattle, WA, USA
| | | | | | | | - Jessica L Braun
- Department of Kinesiology, Centre for Bone and Muscle Health, Brock University, St. Catharines, Canada
| | - Val A Fajardo
- Department of Kinesiology, Centre for Bone and Muscle Health, Brock University, St. Catharines, Canada
| | - Lauren M Sanders
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
- Blue Marble Space, Seattle, WA, USA.
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
| |
Collapse
|
15
|
Manzano A, Weging S, Bezdan D, Borg J, Cahill T, Carnero-Diaz E, Cope H, Deane CS, Etheridge T, Giacomello S, Hardiman G, Leys N, Madrigal P, Mastroleo F, Medina FJ, Mieczkowski J, Fernandez-Rojo MA, Siew K, Szewczyk NJ, Walsh SB, da Silveira WA, Herranz R. Enhancing European capabilities for application of multi-omics studies in biology and biomedicine space research. iScience 2023; 26:107289. [PMID: 37636054 PMCID: PMC10448007 DOI: 10.1016/j.isci.2023.107289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023] Open
Abstract
Following on from the NASA twins' study, there has been a tremendous interest in the use of omics techniques in spaceflight. Individual space agencies, NASA's GeneLab, JAXA's ibSLS, and the ESA-funded Space Omics Topical Team and the International Standards for Space Omics Processing (ISSOP) groups have established several initiatives to support this growth. Here, we present recommendations from the Space Omics Topical Team to promote standard application of space omics in Europe. We focus on four main themes: i) continued participation in and coordination with international omics endeavors, ii) strengthening of the European space omics infrastructure including workforce and facilities, iii) capitalizing on the emerging opportunities in the commercial space sector, and iv) capitalizing on the emerging opportunities in human subjects research.
Collapse
Affiliation(s)
- Aránzazu Manzano
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain
| | | | - Daniela Bezdan
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, Germany
- Yuri GmbH, Meckenbeuren, Germany
| | - Joseph Borg
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, 2080 Msida MSD, Malta
| | - Thomas Cahill
- Faculty of Medicine, Health and Life Sciences, School of Biological Sciences, Institute for Global Food Security, Queen’s University Belfast, Belfast BT7 1NN, UK
| | - Eugénie Carnero-Diaz
- Institut Systématique, Evolution, Biodiversité (ISYEB), Muséum National d’Histoire Naturelle, Sorbonne Université, CNRS, EPHE, UA, 75005 Paris, France
| | - Henry Cope
- School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Colleen S. Deane
- Human Development & Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Timothy Etheridge
- Department of Sport and Health Science, College of Life and Environmental Sciences, University of Exeter, Exeter EX1 2LU, UK
| | - Stefania Giacomello
- Science for Life Laboratory, KTH Royal Institute of Technology, 17165 Stockholm, Sweden
| | - Gary Hardiman
- Faculty of Medicine, Health and Life Sciences, School of Biological Sciences, Institute for Global Food Security, Queen’s University Belfast, Belfast BT7 1NN, UK
| | - Natalie Leys
- Microbiology Unit, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium
| | - Pedro Madrigal
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Hinxton CB10 1SD, UK
| | - Felice Mastroleo
- Microbiology Unit, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium
| | - F. Javier Medina
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain
| | - Jakub Mieczkowski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
| | - Manuel A. Fernandez-Rojo
- Hepatic Regenerative Medicine Lab, Madrid Institute for Advanced Studies in Food, 28049 Madrid, Spain
| | - Keith Siew
- University College London, London NW3 2PF, UK
| | - Nathaniel J. Szewczyk
- School of Medicine, University of Nottingham, Derby DE22 3DT, UK
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | | | - Willian A. da Silveira
- Department of Biological Sciences, School of Health, Science and Wellbeing, Staffordshire University, Stoke-on-Trent ST4 2DF, UK
- International Space University, 67400 Illkirch-Graffenstaden, France
| | - Raúl Herranz
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain
| |
Collapse
|
16
|
Hachem AA, Filkins LM, Kidane YH, Raj P, Tareen NG, Arana CA, Muthukrishnan G, Copley LA. Staphylococcus aureus isolates from children with clinically differentiated osteomyelitis exhibit distinct transcriptomic signatures. PLoS One 2023; 18:e0288758. [PMID: 37561761 PMCID: PMC10414669 DOI: 10.1371/journal.pone.0288758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/04/2023] [Indexed: 08/12/2023] Open
Abstract
There is substantial genomic heterogeneity among Staphylococcus aureus isolates of children with acute hematogenous osteomyelitis (AHO) but transcriptional behavior of clinically differentiated strains has not been previously described. This study evaluates transcriptional activity of S. aureus isolates of children with AHO that may regulate metabolism, biosynthesis, or virulence during bacterial growth and pathogenesis. In vitro growth kinetics were compared between three S. aureus clinical isolates from children with AHO who had mild, moderate, and severe illness. Total RNA sequencing was performed for each isolate at six separate time points throughout the logarithmic phase of growth. The NASA RNA-Sequencing Consensus Pipeline was used to identify differentially expressed genes allowing for 54 comparisons between the three isolates during growth. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathways were used to evaluate transcriptional variation in metabolism, biosynthesis pathways and virulence potential of the isolates. The S. aureus isolates demonstrated differing growth kinetics under standardized conditions with the mild isolate having higher optical densities with earlier and higher peak rates of growth than that of the other isolates (p<0.001). Enrichment pathway analysis established distinct transcriptional signatures according to both sampling time and clinical severity. Moderate and severe isolates demonstrated pathways of bacterial invasion, S. aureus infection, quorum sensing and two component systems. In comparison, the mild strain favored biosynthesis and metabolism. These findings suggest that transcriptional regulation during the growth of S. aureus may impact the pathogenetic mechanisms involved in the progression of severity of illness in childhood osteomyelitis. The clinical isolates studied demonstrated a tradeoff between growth and virulence. Further investigation is needed to evaluate these transcriptional pathways in an animal model or during active clinical infections of children with AHO.
Collapse
Affiliation(s)
- Ahmad A. Hachem
- Department of Pediatrics, University of Florida College of Medicine –Jacksonville, Jacksonville, FL, United States of America
| | - Laura M. Filkins
- Department of Microbiology, University of Texas Southwestern, Children’s Health System of Texas, Dallas, TX, United States of America
| | - Yared H. Kidane
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, United States of America
| | - Prithvi Raj
- Microbiome Research Laboratory, University of Texas Southwestern, Dallas, TX, United States of America
| | - Naureen G. Tareen
- Department of Pediatric Orthopaedic Surgery, Children’s Health System of Texas, Dallas, TX, United States of America
| | - Carlos A. Arana
- Genomics Core, University of Texas Southwestern, Dallas, TX, United States of America
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Lawson A. Copley
- Department of Pediatric Orthopaedic Surgery, Children’s Health System of Texas, Dallas, TX, United States of America
- Department of Pediatric Orthopaedic Surgery, University of Texas Southwestern, Dallas, TX, United States of America
| |
Collapse
|
17
|
Bisutti V, Mach N, Giannuzzi D, Vanzin A, Capra E, Negrini R, Gelain ME, Cecchinato A, Ajmone-Marsan P, Pegolo S. Transcriptome-wide mapping of milk somatic cells upon subclinical mastitis infection in dairy cattle. J Anim Sci Biotechnol 2023; 14:93. [PMID: 37403140 DOI: 10.1186/s40104-023-00890-9] [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: 01/17/2023] [Accepted: 05/07/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Subclinical intramammary infection (IMI) represents a significant problem in maintaining dairy cows' health. Disease severity and extent depend on the interaction between the causative agent, environment, and host. To investigate the molecular mechanisms behind the host immune response, we used RNA-Seq for the milk somatic cells (SC) transcriptome profiling in healthy cows (n = 9), and cows naturally affected by subclinical IMI from Prototheca spp. (n = 11) and Streptococcus agalactiae (S. agalactiae; n = 11). Data Integration Analysis for Biomarker discovery using Latent Components (DIABLO) was used to integrate transcriptomic data and host phenotypic traits related to milk composition, SC composition, and udder health to identify hub variables for subclinical IMI detection. RESULTS A total of 1,682 and 2,427 differentially expressed genes (DEGs) were identified when comparing Prototheca spp. and S. agalactiae to healthy animals, respectively. Pathogen-specific pathway analyses evidenced that Prototheca's infection upregulated antigen processing and lymphocyte proliferation pathways while S. agalactiae induced a reduction of energy-related pathways like the tricarboxylic acid cycle, and carbohydrate and lipid metabolism. The integrative analysis of commonly shared DEGs between the two pathogens (n = 681) referred to the core-mastitis response genes, and phenotypic data evidenced a strong covariation between those genes and the flow cytometry immune cells (r2 = 0.72), followed by the udder health (r2 = 0.64) and milk quality parameters (r2 = 0.64). Variables with r ≥ 0.90 were used to build a network in which the top 20 hub variables were identified with the Cytoscape cytohubba plug-in. The genes in common between DIABLO and cytohubba (n = 10) were submitted to a ROC analysis which showed they had excellent predictive performances in terms of discriminating healthy and mastitis-affected animals (sensitivity > 0.89, specificity > 0.81, accuracy > 0.87, and precision > 0.69). Among these genes, CIITA could play a key role in regulating the animals' response to subclinical IMI. CONCLUSIONS Despite some differences in the enriched pathways, the two mastitis-causing pathogens seemed to induce a shared host immune-transcriptomic response. The hub variables identified with the integrative approach might be included in screening and diagnostic tools for subclinical IMI detection.
Collapse
Affiliation(s)
- Vittoria Bisutti
- DAFNAE, University of Padova, Viale Dell'Università 16, Legnaro, PD, 35020, Italy.
| | - Núria Mach
- IHAP, Université de Toulouse, INRAE, ENVT, 23 Chemin Des Capelles, Toulouse, 31300, France
| | - Diana Giannuzzi
- DAFNAE, University of Padova, Viale Dell'Università 16, Legnaro, PD, 35020, Italy
| | - Alice Vanzin
- DAFNAE, University of Padova, Viale Dell'Università 16, Legnaro, PD, 35020, Italy
| | - Emanuele Capra
- IBBA, National Research Council, Via Einstein, Lodi, 26900, Italy
| | - Riccardo Negrini
- DIANA, Università Cattolica del Sacro Cuore, Via E. Parmense 84, Piacenza, 29122, Italy
| | - Maria Elena Gelain
- BCA, University of Padova, Viale Dell'Università 16, Legnaro, PD, 35020, Italy
| | - Alessio Cecchinato
- DAFNAE, University of Padova, Viale Dell'Università 16, Legnaro, PD, 35020, Italy
| | - Paolo Ajmone-Marsan
- DIANA, Università Cattolica del Sacro Cuore, Via E. Parmense 84, Piacenza, 29122, Italy
| | - Sara Pegolo
- DAFNAE, University of Padova, Viale Dell'Università 16, Legnaro, PD, 35020, Italy
| |
Collapse
|
18
|
Cope H, Elsborg J, Demharter S, Mcdonald JT, Wernecke C, Parthasarathy H, Unadkat H, Chatrathi M, Claudio J, Reinsch S, Zwart S, Smith S, Heer M, Muratani M, Meydan C, Overbey E, Kim J, Park J, Schisler J, Mason C, Szewczyk N, Willis C, Salam A, Beheshti A. More than a Feeling: Dermatological Changes Impacted by Spaceflight. RESEARCH SQUARE 2023:rs.3.rs-2367727. [PMID: 36798347 PMCID: PMC9934743 DOI: 10.21203/rs.3.rs-2367727/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Spaceflight poses a unique set of challenges to humans and the hostile Spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on spaceflown murine transcriptomic datasets focused on the skin, biomedical profiles from fifty NASA astronauts, and confirmation via transcriptomic data from JAXA astronauts, the NASA Twins Study, and the first civilian commercial mission, Inspiration4. Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation were determined to be involved with skin health risks during Spaceflight. Additionally, a machine learning model was utilized to determine key genes driving Spaceflight response in the skin. These results can be used for determining potential countermeasures to mitigate Spaceflight damage to the skin.
Collapse
|
19
|
Costa-Silva J, Domingues DS, Menotti D, Hungria M, Lopes FM. Temporal progress of gene expression analysis with RNA-Seq data: A review on the relationship between computational methods. Comput Struct Biotechnol J 2022; 21:86-98. [PMID: 36514333 PMCID: PMC9730150 DOI: 10.1016/j.csbj.2022.11.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Analysis of differential gene expression from RNA-seq data has become a standard for several research areas. The steps for the computational analysis include many data types and file formats, and a wide variety of computational tools that can be applied alone or together as pipelines. This paper presents a review of the differential expression analysis pipeline, addressing its steps and the respective objectives, the principal methods available in each step, and their properties, therefore introducing an organized overview to this context. This review aims to address mainly the aspects involved in the differentially expressed gene (DEG) analysis from RNA sequencing data (RNA-seq), considering the computational methods. In addition, a timeline of the computational methods for DEG is shown and discussed, and the relationships existing between the most important computational tools are presented by an interaction network. A discussion on the challenges and gaps in DEG analysis is also highlighted in this review. This paper will serve as a tutorial for new entrants into the field and help established users update their analysis pipelines.
Collapse
Affiliation(s)
- Juliana Costa-Silva
- Department of Informatics – Federal University of Paraná, Rua Coronel Francisco Heráclito dos Santos, 100, 81531-990 Curitiba, Paraná, Brazil
| | - Douglas S. Domingues
- Department of Genetics, “Luiz de Queiroz” College of Agriculture, University of São Paulo, Av. Pádua Dias, 11, 13418-900 Piracicaba, São Paulo, Brazil
| | - David Menotti
- Department of Informatics – Federal University of Paraná, Rua Coronel Francisco Heráclito dos Santos, 100, 81531-990 Curitiba, Paraná, Brazil
| | - Mariangela Hungria
- Department of Soil Biotecnology - Embrapa Soybean, Cx. Postal 231, 86000-970 Londrina, Paraná, Brazil
| | - Fabrício Martins Lopes
- Department of Computer Science, Universidade Tecnológica Federal do Paraná – UTFPR, Av. Alberto Carazzai, 1640, 86300-000, Cornélio Procópio, Paraná, Brazil
| |
Collapse
|
20
|
Overbey EG, Das S, Cope H, Madrigal P, Andrusivova Z, Frapard S, Klotz R, Bezdan D, Gupta A, Scott RT, Park J, Chirko D, Galazka JM, Costes SV, Mason CE, Herranz R, Szewczyk NJ, Borg J, Giacomello S. Challenges and considerations for single-cell and spatially resolved transcriptomics sample collection during spaceflight. CELL REPORTS METHODS 2022; 2:100325. [PMID: 36452864 PMCID: PMC9701605 DOI: 10.1016/j.crmeth.2022.100325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) and spatially resolved transcriptomics (SRT) have experienced rapid development in recent years. The findings of spaceflight-based scRNA-seq and SRT investigations are likely to improve our understanding of life in space and our comprehension of gene expression in various cell systems and tissue dynamics. However, compared to their Earth-based counterparts, gene expression experiments conducted in spaceflight have not experienced the same pace of development. Out of the hundreds of spaceflight gene expression datasets available, only a few used scRNA-seq and SRT. In this perspective piece, we explore the growing importance of scRNA-seq and SRT in space biology and discuss the challenges and considerations relevant to robust experimental design to enable growth of these methods in the field.
Collapse
Affiliation(s)
- Eliah G. Overbey
- Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, New York, NY, USA
| | - Saswati Das
- Department of Biochemistry, Atal Bihari Vajpayee Institute of Medical Sciences & Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Henry Cope
- School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Pedro Madrigal
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Genome Campus, Hinxton, UK
| | - Zaneta Andrusivova
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Solène Frapard
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Rebecca Klotz
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Daniela Bezdan
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, German
- yuri GmbH, Meckenbeuren, Germany
| | | | - Ryan T. Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | | | - Jonathan M. Galazka
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Sylvain V. Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Christopher E. Mason
- Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, New York, NY, USA
- The WorldQuant Initiative for Quantitative Prediction, New York, NY, USA
| | - Raul Herranz
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid 28040, Spain
| | - Nathaniel J. Szewczyk
- School of Medicine, University of Nottingham, Derby DE22 3DT, UK
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Joseph Borg
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
21
|
Kothiyal P, Eley G, Ilangovan H, Hoadley KA, Elgart SR, Mao XW, Eslami P. A multi-omics longitudinal study of the murine retinal response to chronic low-dose irradiation and simulated microgravity. Sci Rep 2022; 12:16825. [PMID: 36207342 PMCID: PMC9547011 DOI: 10.1038/s41598-022-19360-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
The space environment includes unique hazards like radiation and microgravity which can adversely affect biological systems. We assessed a multi-omics NASA GeneLab dataset where mice were hindlimb unloaded and/or gamma irradiated for 21 days followed by retinal analysis at 7 days, 1 month or 4 months post-exposure. We compared time-matched epigenomic and transcriptomic retinal profiles resulting in a total of 4178 differentially methylated loci or regions, and 457 differentially expressed genes. Highest correlation in methylation difference was seen across different conditions at the same time point. Nucleotide metabolism biological processes were enriched in all groups with activation at 1 month and suppression at 7 days and 4 months. Genes and processes related to Notch and Wnt signaling showed alterations 4 months post-exposure. A total of 23 genes showed significant changes in methylation and expression compared to unexposed controls, including genes involved in retinal function and inflammatory response. This multi-omics analysis interrogates the epigenomic and transcriptomic impacts of radiation and hindlimb unloading on the retina in isolation and in combination and highlights important molecular mechanisms at different post-exposure stages.
Collapse
Affiliation(s)
| | - Greg Eley
- Scimentis LLC, Statham, GA, 30666, USA
| | - Hari Ilangovan
- Science Applications International Corporation (SAIC), Reston, VA, 20190, USA
| | - Katherine A Hoadley
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Xiao W Mao
- Basic Sciences, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Parastou Eslami
- Universal Artificial Intelligence Inc., Boston, MA, 02130, USA
| |
Collapse
|
22
|
Mhatre SD, Iyer J, Petereit J, Dolling-Boreham RM, Tyryshkina A, Paul AM, Gilbert R, Jensen M, Woolsey RJ, Anand S, Sowa MB, Quilici DR, Costes SV, Girirajan S, Bhattacharya S. Artificial gravity partially protects space-induced neurological deficits in Drosophila melanogaster. Cell Rep 2022; 40:111279. [PMID: 36070701 PMCID: PMC10503492 DOI: 10.1016/j.celrep.2022.111279] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 03/16/2022] [Accepted: 08/05/2022] [Indexed: 02/03/2023] Open
Abstract
Spaceflight poses risks to the central nervous system (CNS), and understanding neurological responses is important for future missions. We report CNS changes in Drosophila aboard the International Space Station in response to spaceflight microgravity (SFμg) and artificially simulated Earth gravity (SF1g) via inflight centrifugation as a countermeasure. While inflight behavioral analyses of SFμg exhibit increased activity, postflight analysis displays significant climbing defects, highlighting the sensitivity of behavior to altered gravity. Multi-omics analysis shows alterations in metabolic, oxidative stress and synaptic transmission pathways in both SFμg and SF1g; however, neurological changes immediately postflight, including neuronal loss, glial cell count alterations, oxidative damage, and apoptosis, are seen only in SFμg. Additionally, progressive neuronal loss and a glial phenotype in SF1g and SFμg brains, with pronounced phenotypes in SFμg, are seen upon acclimation to Earth conditions. Overall, our results indicate that artificial gravity partially protects the CNS from the adverse effects of spaceflight.
Collapse
Affiliation(s)
- Siddhita D Mhatre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA; COSMIAC Research Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Janani Iyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA; Universities Space Research Association, Mountain View, CA 94043, USA
| | - Juli Petereit
- Nevada Bioinformatics Center, University of Nevada, Reno, NV 89557, USA
| | - Roberta M Dolling-Boreham
- Department of Electrical and Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada; Blue Marble Space Institute of Science, Seattle, WA 94035, USA
| | - Anastasia Tyryshkina
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Amber M Paul
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Universities Space Research Association, Mountain View, CA 94043, USA; Blue Marble Space Institute of Science, Seattle, WA 94035, USA; NASA Postdoctoral Program, Universities Space Research Association, NASA Ames Research Center, Moffett Field, CA 94035, USA; Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL 32114, USA
| | - Rachel Gilbert
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; NASA Postdoctoral Program, Universities Space Research Association, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Matthew Jensen
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | | | - Sulekha Anand
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| | - Marianne B Sowa
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - David R Quilici
- Nevada Proteomics Center, University of Nevada, Reno, NV 89557, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Santhosh Girirajan
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Sharmila Bhattacharya
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Biological and Physical Sciences Division, NASA Headquarters, Washington DC 20024, USA.
| |
Collapse
|
23
|
Manzano A, Carnero-Diaz E, Herranz R, Medina FJ. Recent transcriptomic studies to elucidate the plant adaptive response to spaceflight and to simulated space environments. iScience 2022; 25:104687. [PMID: 35856037 PMCID: PMC9287483 DOI: 10.1016/j.isci.2022.104687] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Discovering the adaptation mechanisms of plants to the space environment is essential for supporting human space exploration. Transcriptomic analyses allow the identification of adaptation response pathways by detecting changes in gene expression at the global genome level caused by the main factors of the space environment, namely altered gravity and cosmic radiation. This article reviews transcriptomic studies carried out from plants grown in spaceflights and in different ground-based microgravity simulators. Despite differences in plant growth conditions, these studies have shown that cell wall remodeling, oxidative stress, defense response, and photosynthesis are common altered processes in plants grown under spaceflight conditions. European scientists have significantly contributed to the acquisition of this knowledge, e.g., by showing the role of red light in the adaptation response of plants (EMCS experiments) and the mechanisms of cellular response and adaptation mostly affecting cell cycle regulation, using cell cultures in microgravity simulators. Cell wall, photosynthesis, and stress response are key in plant adaptation to space DNA methylation and alternative splicing are among the involved molecular mechanisms Light is an essential factor for plant development, even more in the space environment EMCS and simulation cell culture experiments are the main European contributions
Collapse
Affiliation(s)
- Aránzazu Manzano
- PCNPμG Lab (Plant Cell Nucleolus, Proliferation and Microgravity), Centro de Investigaciones Biológicas Margarita Salas - CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Eugénie Carnero-Diaz
- Institut Systématique, Evolution, Biodiversité (ISYEB), Muséum National d'Histoire Naturelle, Sorbonne Université, CNRS, EPHE, UA, Paris, 75005, France
| | - Raúl Herranz
- PCNPμG Lab (Plant Cell Nucleolus, Proliferation and Microgravity), Centro de Investigaciones Biológicas Margarita Salas - CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - F Javier Medina
- PCNPμG Lab (Plant Cell Nucleolus, Proliferation and Microgravity), Centro de Investigaciones Biológicas Margarita Salas - CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| |
Collapse
|
24
|
Cope H, Willis CR, MacKay MJ, Rutter LA, Toh LS, Williams PM, Herranz R, Borg J, Bezdan D, Giacomello S, Muratani M, Mason CE, Etheridge T, Szewczyk NJ. Routine omics collection is a golden opportunity for European human research in space and analog environments. PATTERNS 2022; 3:100550. [PMID: 36277820 PMCID: PMC9583032 DOI: 10.1016/j.patter.2022.100550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
25
|
McDonald JT, Enguita FJ, Taylor D, Griffin RJ, Priebe W, Emmett MR, Sajadi MM, Harris AD, Clement J, Dybas JM, Aykin-Burns N, Guarnieri JW, Singh LN, Grabham P, Baylin SB, Yousey A, Pearson AN, Corry PM, Saravia-Butler A, Aunins TR, Sharma S, Nagpal P, Meydan C, Foox J, Mozsary C, Cerqueira B, Zaksas V, Singh U, Wurtele ES, Costes SV, Davanzo GG, Galeano D, Paccanaro A, Meinig SL, Hagan RS, Bowman NM, Wolfgang MC, Altinok S, Sapoval N, Treangen TJ, Moraes-Vieira PM, Vanderburg C, Wallace DC, Schisler JC, Mason CE, Chatterjee A, Meller R, Beheshti A. Role of miR-2392 in driving SARS-CoV-2 infection. Cell Rep 2021; 37:109839. [PMID: 34624208 PMCID: PMC8481092 DOI: 10.1016/j.celrep.2021.109839] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/13/2021] [Accepted: 09/24/2021] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs involved in post-transcriptional gene regulation that have a major impact on many diseases and provide an exciting avenue toward antiviral therapeutics. From patient transcriptomic data, we determined that a circulating miRNA, miR-2392, is directly involved with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) machinery during host infection. Specifically, we show that miR-2392 is key in driving downstream suppression of mitochondrial gene expression, increasing inflammation, glycolysis, and hypoxia, as well as promoting many symptoms associated with coronavirus disease 2019 (COVID-19) infection. We demonstrate that miR-2392 is present in the blood and urine of patients positive for COVID-19 but is not present in patients negative for COVID-19. These findings indicate the potential for developing a minimally invasive COVID-19 detection method. Lastly, using in vitro human and in vivo hamster models, we design a miRNA-based antiviral therapeutic that targets miR-2392, significantly reduces SARS-CoV-2 viability in hamsters, and may potentially inhibit a COVID-19 disease state in humans.
Collapse
Affiliation(s)
- J Tyson McDonald
- COVID-19 International Research Team; Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Francisco J Enguita
- COVID-19 International Research Team; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Deanne Taylor
- COVID-19 International Research Team; The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert J Griffin
- COVID-19 International Research Team; University of Arkansas for Medical Sciences, Little Rock, AK 72211, USA
| | - Waldemar Priebe
- COVID-19 International Research Team; University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mark R Emmett
- COVID-19 International Research Team; University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | - Anthony D Harris
- University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jean Clement
- University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph M Dybas
- COVID-19 International Research Team; The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Joseph W Guarnieri
- COVID-19 International Research Team; The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Larry N Singh
- COVID-19 International Research Team; The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Peter Grabham
- COVID-19 International Research Team; Columbia University, New York, NY 10032, USA
| | - Stephen B Baylin
- COVID-19 International Research Team; Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Aliza Yousey
- COVID-19 International Research Team; Morehouse School of Medicine, Atlanta, GA 30310, USA
| | | | - Peter M Corry
- COVID-19 International Research Team; University of Arkansas for Medical Sciences, Little Rock, AK 72211, USA
| | - Amanda Saravia-Butler
- COVID-19 International Research Team; Logyx LLC, Mountain View, CA 94043, USA; NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | - Sadhana Sharma
- University of Colorado Boulder, Boulder, CO 80303, USA; Sachi Bioworks Inc., Boulder, CO 80301, USA
| | - Prashant Nagpal
- Sachi Bioworks Inc., Boulder, CO 80301, USA; Antimicrobial Regeneration Consortium, Boulder Labs, Boulder, CO 80301, USA; Quantum Biology Inc., Boulder, CO 80301, USA
| | - Cem Meydan
- Weill Cornell Medicine, New York, NY 10065, USA
| | | | | | - Bianca Cerqueira
- COVID-19 International Research Team; KBR Space & Science, San Antonio, TX 78235, USA; United States Air Force School of Aerospace Medicine, Lackland AFB, San Antonio, TX 78236, USA
| | - Viktorija Zaksas
- COVID-19 International Research Team; University of Chicago, Chicago, IL 60615, USA
| | - Urminder Singh
- COVID-19 International Research Team; Iowa State University, Ames, IA 50011, USA
| | - Eve Syrkin Wurtele
- COVID-19 International Research Team; Iowa State University, Ames, IA 50011, USA
| | | | | | - Diego Galeano
- COVID-19 International Research Team; Fundação Getulio Vargas, Rio de Janeiro, Brazil; National University of Asuncion, San Lorenzo, Central, Paraguay
| | - Alberto Paccanaro
- COVID-19 International Research Team; Fundação Getulio Vargas, Rio de Janeiro, Brazil; University of London, Egham Hill, Egham, UK
| | - Suzanne L Meinig
- University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Robert S Hagan
- University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie M Bowman
- University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Selin Altinok
- University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | - Douglas C Wallace
- COVID-19 International Research Team; The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan C Schisler
- COVID-19 International Research Team; University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christopher E Mason
- COVID-19 International Research Team; Weill Cornell Medicine, New York, NY 10065, USA; New York Genome Center, New York, NY, USA
| | - Anushree Chatterjee
- COVID-19 International Research Team; University of Colorado Boulder, Boulder, CO 80303, USA; Sachi Bioworks Inc., Boulder, CO 80301, USA; Antimicrobial Regeneration Consortium, Boulder Labs, Boulder, CO 80301, USA
| | - Robert Meller
- COVID-19 International Research Team; Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Afshin Beheshti
- COVID-19 International Research Team; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| |
Collapse
|
26
|
McDonald JT, Enguita FJ, Taylor D, Griffin RJ, Priebe W, Emmett MR, Sajadi MM, Harris AD, Clement J, Dybas JM, Aykin-Burns N, Guarnieri JW, Singh LN, Grabham P, Baylin SB, Yousey A, Pearson AN, Corry PM, Saravia-Butler A, Aunins TR, Sharma S, Nagpal P, Meydan C, Foox J, Mozsary C, Cerqueira B, Zaksas V, Singh U, Wurtele ES, Costes SV, Davanzo GG, Galeano D, Paccanaro A, Meinig SL, Hagan RS, Bowman NM, Wolfgang MC, Altinok S, Sapoval N, Treangen TJ, Moraes-Vieira PM, Vanderburg C, Wallace DC, Schisler J, Mason CE, Chatterjee A, Meller R, Beheshti A. The Great Deceiver: miR-2392's Hidden Role in Driving SARS-CoV-2 Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33948587 DOI: 10.1101/2021.04.23.441024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs involved in post-transcriptional gene regulation that have a major impact on many diseases and provides an exciting avenue towards antiviral therapeutics. From patient transcriptomic data, we have discovered a circulating miRNA, miR-2392, that is directly involved with SARS-CoV-2 machinery during host infection. Specifically, we show that miR-2392 is key in driving downstream suppression of mitochondrial gene expression, increasing inflammation, glycolysis, and hypoxia as well as promoting many symptoms associated with COVID-19 infection. We demonstrate miR-2392 is present in the blood and urine of COVID-19 positive patients, but not detected in COVID-19 negative patients. These findings indicate the potential for developing a novel, minimally invasive, COVID-19 detection method. Lastly, using in vitro human and in vivo hamster models, we have developed a novel miRNA-based antiviral therapeutic that targets miR-2392, significantly reduces SARS-CoV-2 viability in hamsters and may potentially inhibit a COVID-19 disease state in humans.
Collapse
|
27
|
Animal models, open science, & space-omics. Lab Anim (NY) 2021; 50:143-145. [PMID: 34021293 DOI: 10.1038/s41684-021-00776-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
28
|
Rad-Bio-App: a discovery environment for biologists to explore spaceflight-related radiation exposures. NPJ Microgravity 2021; 7:15. [PMID: 33976230 PMCID: PMC8113475 DOI: 10.1038/s41526-021-00143-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
In addition to microgravity, spaceflight simultaneously exposes biology to a suite of other stimuli. For example, in space, organisms experience ionizing radiation environments that significantly differ in both quality and quantity from those normally experienced on Earth. However, data on radiation exposure during space missions is often complex to access and to understand, limiting progress towards defining how radiation affects organisms against the unique background of spaceflight. To help address this challenge, we have developed the Rad-Bio-App. This web-accessible database imports radiation metadata from experiments archived in NASA’s GeneLab data repository, and then allows the user to explore these experiments both in the context of their radiation exposure and through their other metadata and results. Rad-Bio-App provides an easy-to-use, graphically-driven environment to enable both radiation biologists and non-specialist researchers to visualize, and understand the impact of ionizing radiation on various biological systems in the context of spaceflight.
Collapse
|