Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.150
Filter
Add more filters

Publication year range
1.
Cell ; 183(6): 1457-1461, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33242423

ABSTRACT

As of 2020, fewer than 600 individuals have left Earth to experience work and life in space. That number will grow as government-funded and commercial space programs move forward in countries around the globe. There are however major questions about how humans respond to spaceflight at every level, from the whole body to individual organs to specific cells to molecular pathways. Preparing for a future where longer-duration spaceflights are anticipated and people can begin to contemplate space tourism, researchers are studying astronauts to understand how the human system is affected by and adapts to space. Lara Szewczak got a window on this world, speaking with retired astronaut Scott Kelly about his late-blooming interest in science, what he's learned through the NASA Twins Study, and why space vacations might not be for everyone. They were joined by Chris Mason, a lead investigator looking at the 'omics of spaceflight. Excerpts from this conversation are presented below, and the full conversation is available with the article online.


Subject(s)
Astronauts , History, 20th Century , History, 21st Century , Humans , Space Flight
2.
Cell ; 183(5): 1162-1184, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33242416

ABSTRACT

Research on astronaut health and model organisms have revealed six features of spaceflight biology that guide our current understanding of fundamental molecular changes that occur during space travel. The features include oxidative stress, DNA damage, mitochondrial dysregulation, epigenetic changes (including gene regulation), telomere length alterations, and microbiome shifts. Here we review the known hazards of human spaceflight, how spaceflight affects living systems through these six fundamental features, and the associated health risks of space exploration. We also discuss the essential issues related to the health and safety of astronauts involved in future missions, especially planned long-duration and Martian missions.


Subject(s)
Extraterrestrial Environment , Space Flight , Astronauts , Health , Humans , Microbiota , Risk Factors
3.
Cell ; 183(5): 1185-1201.e20, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33242417

ABSTRACT

Spaceflight is known to impose changes on human physiology with unknown molecular etiologies. To reveal these causes, we used a multi-omics, systems biology analytical approach using biomedical profiles from fifty-nine astronauts and data from NASA's GeneLab derived from hundreds of samples flown in space to determine transcriptomic, proteomic, metabolomic, and epigenetic responses to spaceflight. Overall pathway analyses on the multi-omics datasets showed significant enrichment for mitochondrial processes, as well as innate immunity, chronic inflammation, cell cycle, circadian rhythm, and olfactory functions. Importantly, NASA's Twin Study provided a platform to confirm several of our principal findings. Evidence of altered mitochondrial function and DNA damage was also found in the urine and blood metabolic data compiled from the astronaut cohort and NASA Twin Study data, indicating mitochondrial stress as a consistent phenotype of spaceflight.


Subject(s)
Genomics , Mitochondria/pathology , Space Flight , Stress, Physiological , Animals , Circadian Rhythm , Extracellular Matrix/metabolism , Humans , Immunity, Innate , Lipid Metabolism , Metabolic Flux Analysis , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscles/immunology , Organ Specificity , Smell/physiology
4.
Cell ; 179(5): 1003-1009, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31730844

ABSTRACT

Astronauts and cancer patients are subject to similar multisystem physiological toxicities. Over the past sixty years, NASA developed a state-of-the-art countermeasures program (CMP) to characterize and mitigate the physiological consequences of spaceflight. Here, we propose a NASA-modeled CMP to elucidate and abrogate physiological toxicities in patients with cancer.


Subject(s)
Neoplasms/pathology , United States National Aeronautics and Space Administration , Astronauts , Humans , Medical Countermeasures , Space Flight , United States
5.
Nature ; 632(8027): 995-1008, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38862027

ABSTRACT

The recent acceleration of commercial, private and multi-national spaceflight has created an unprecedented level of activity in low Earth orbit, concomitant with the largest-ever number of crewed missions entering space and preparations for exploration-class (lasting longer than one year) missions. Such rapid advancement into space from many new companies, countries and space-related entities has enabled a 'second space age'. This era is also poised to leverage, for the first time, modern tools and methods of molecular biology and precision medicine, thus enabling precision aerospace medicine for the crews. The applications of these biomedical technologies and algorithms are diverse, and encompass multi-omic, single-cell and spatial biology tools to investigate human and microbial responses to spaceflight. Additionally, they extend to the development of new imaging techniques, real-time cognitive assessments, physiological monitoring and personalized risk profiles tailored for astronauts. Furthermore, these technologies enable advancements in pharmacogenomics, as well as the identification of novel spaceflight biomarkers and the development of corresponding countermeasures. In this Perspective, we highlight some of the recent biomedical research from the National Aeronautics and Space Administration, Japan Aerospace Exploration Agency, European Space Agency and other space agencies, and detail the entrance of the commercial spaceflight sector (including SpaceX, Blue Origin, Axiom and Sierra Space) into aerospace medicine and space biology, the first aerospace medicine biobank, and various upcoming missions that will utilize these tools to ensure a permanent human presence beyond low Earth orbit, venturing out to other planets and moons.


Subject(s)
Aerospace Medicine , Astronauts , Multiomics , Space Flight , Humans , Aerospace Medicine/methods , Aerospace Medicine/trends , Biological Specimen Banks , Biomarkers/metabolism , Biomarkers/analysis , Cognition , Internationality , Monitoring, Physiologic/methods , Monitoring, Physiologic/trends , Multiomics/methods , Multiomics/trends , Pharmacogenetics/methods , Pharmacogenetics/trends , Precision Medicine/methods , Precision Medicine/trends , Space Flight/methods , Space Flight/trends
6.
Nature ; 632(8027): 1155-1164, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38862026

ABSTRACT

Human spaceflight has historically been managed by government agencies, such as in the NASA Twins Study1, but new commercial spaceflight opportunities have opened spaceflight to a broader population. In 2021, the SpaceX Inspiration4 mission launched the first all-civilian crew to low Earth orbit, which included the youngest American astronaut (aged 29), new in-flight experimental technologies (handheld ultrasound imaging, smartwatch wearables and immune profiling), ocular alignment measurements and new protocols for in-depth, multi-omic molecular and cellular profiling. Here we report the primary findings from the 3-day spaceflight mission, which induced a broad range of physiological and stress responses, neurovestibular changes indexed by ocular misalignment, and altered neurocognitive functioning, some of which match those of long-term spaceflight2, but almost all of which did not differ from baseline (pre-flight) after return to Earth. Overall, these preliminary civilian spaceflight data suggest that short-duration missions do not pose a significant health risk, and moreover present a rich opportunity to measure the earliest phases of adaptation to spaceflight in the human body at anatomical, cellular, physiological and cognitive levels. Finally, these methods and results lay the foundation for an open, rapidly expanding biomedical database for astronauts3, which can inform countermeasure development for both private and government-sponsored space missions.


Subject(s)
Adaptation, Physiological , Astronauts , Space Flight , Adult , Female , Humans , Male , Cognition/physiology , Stress, Physiological/physiology , Time Factors , Weightlessness/adverse effects , Monitoring, Physiologic , Multiomics , Adaptation, Physiological/physiology , Databases as Topic
7.
Nature ; 634(8032): 48-52, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39294379

ABSTRACT

Space radiation is a notable hazard for long-duration human spaceflight1. Associated risks include cancer, cataracts, degenerative diseases2 and tissue reactions from large, acute exposures3. Space radiation originates from diverse sources, including galactic cosmic rays4, trapped-particle (Van Allen) belts5 and solar-particle events6. Previous radiation data are from the International Space Station and the Space Shuttle in low-Earth orbit protected by heavy shielding and Earth's magnetic field7,8 and lightly shielded interplanetary robotic probes such as Mars Science Laboratory and Lunar Reconnaissance Orbiter9,10. Limited data from the Apollo missions11-13 and ground measurements with substantial caveats are also available14. Here we report radiation measurements from the heavily shielded Orion spacecraft on the uncrewed Artemis I lunar mission. At differing shielding locations inside the vehicle, a fourfold difference in dose rates was observed during proton-belt passes that are similar to large, reference solar-particle events. Interplanetary cosmic-ray dose equivalent rates in Orion were as much as 60% lower than previous observations9. Furthermore, a change in orientation of the spacecraft during the proton-belt transit resulted in a reduction of radiation dose rates of around 50%. These measurements validate the Orion for future crewed exploration and inform future human spaceflight mission design.


Subject(s)
Cosmic Radiation , Moon , Radiation Dosage , Radiation Protection , Space Flight , Spacecraft , Cosmic Radiation/adverse effects , Humans , Radiation Protection/methods , Radiation Monitoring/methods , Astronauts , Protons/adverse effects
8.
Nature ; 632(8027): 1145-1154, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38862028

ABSTRACT

Spaceflight induces molecular, cellular and physiological shifts in astronauts and poses myriad biomedical challenges to the human body, which are becoming increasingly relevant as more humans venture into space1-6. Yet current frameworks for aerospace medicine are nascent and lag far behind advancements in precision medicine on Earth, underscoring the need for rapid development of space medicine databases, tools and protocols. Here we present the Space Omics and Medical Atlas (SOMA), an integrated data and sample repository for clinical, cellular and multi-omic research profiles from a diverse range of missions, including the NASA Twins Study7, JAXA CFE study8,9, SpaceX Inspiration4 crew10-12, Axiom and Polaris. The SOMA resource represents a more than tenfold increase in publicly available human space omics data, with matched samples available from the Cornell Aerospace Medicine Biobank. The Atlas includes extensive molecular and physiological profiles encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics and microbiome datasets, which reveal some consistent features across missions, including cytokine shifts, telomere elongation and gene expression changes, as well as mission-specific molecular responses and links to orthologous, tissue-specific mouse datasets. Leveraging the datasets, tools and resources in SOMA can help to accelerate precision aerospace medicine, bringing needed health monitoring, risk mitigation and countermeasure data for upcoming lunar, Mars and exploration-class missions.


Subject(s)
Aerospace Medicine , Astronauts , Biological Specimen Banks , Databases, Factual , Internationality , Space Flight , Animals , Female , Humans , Male , Mice , Aerospace Medicine/methods , Atlases as Topic , Cytokines/metabolism , Datasets as Topic , Epigenomics , Gene Expression Profiling , Genomics , Metabolomics , Microbiota/genetics , Multiomics , Organ Specificity , Precision Medicine/trends , Proteomics , Space Flight/statistics & numerical data , Telomere/metabolism , Twins
9.
Proc Natl Acad Sci U S A ; 121(40): e2404644121, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39312653

ABSTRACT

With current plans for manned missions to Mars and beyond, the need to better understand, prevent, and counteract the harmful effects of long-duration spaceflight on the body is becoming increasingly important. In this study, an automated heart-on-a-chip platform was flown to the International Space Station on a 1-mo mission during which contractile cardiac function was monitored in real-time. Upon return to Earth, engineered human heart tissues (EHTs) were further analyzed with ultrastructural imaging and RNA sequencing to investigate the impact of prolonged microgravity on cardiomyocyte function and health. Spaceflight EHTs exhibited significantly reduced twitch forces, increased incidences of arrhythmias, and increased signs of sarcomere disruption and mitochondrial damage. Transcriptomic analyses showed an up-regulation of genes and pathways associated with metabolic disorders, heart failure, oxidative stress, and inflammation, while genes related to contractility and calcium signaling showed significant down-regulation. Finally, in silico modeling revealed a potential link between oxidative stress and mitochondrial dysfunction that corresponded with RNA sequencing results. This represents an in vitro model to faithfully reproduce the adverse effects of spaceflight on three-dimensional (3D)-engineered heart tissue.


Subject(s)
Myocardial Contraction , Myocytes, Cardiac , Space Flight , Space Flight/methods , Humans , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Lab-On-A-Chip Devices , Weightlessness/adverse effects , Oxidative Stress , Mitochondria/metabolism , Mitochondria, Heart/metabolism
10.
Physiol Rev ; 99(1): 807-851, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30540225

ABSTRACT

This review presents lower body negative pressure (LBNP) as a unique tool to investigate the physiology of integrated systemic compensatory responses to altered hemodynamic patterns during conditions of central hypovolemia in humans. An early review published in Physiological Reviews over 40 yr ago (Wolthuis et al. Physiol Rev 54: 566-595, 1974) focused on the use of LBNP as a tool to study effects of central hypovolemia, while more than a decade ago a review appeared that focused on LBNP as a model of hemorrhagic shock (Cooke et al. J Appl Physiol (1985) 96: 1249-1261, 2004). Since then there has been a great deal of new research that has applied LBNP to investigate complex physiological responses to a variety of challenges including orthostasis, hemorrhage, and other important stressors seen in humans such as microgravity encountered during spaceflight. The LBNP stimulus has provided novel insights into the physiology underlying areas such as intolerance to reduced central blood volume, sex differences concerning blood pressure regulation, autonomic dysfunctions, adaptations to exercise training, and effects of space flight. Furthermore, approaching cardiovascular assessment using prediction models for orthostatic capacity in healthy populations, derived from LBNP tolerance protocols, has provided important insights into the mechanisms of orthostatic hypotension and central hypovolemia, especially in some patient populations as well as in healthy subjects. This review also presents a concise discussion of mathematical modeling regarding compensatory responses induced by LBNP. Given the diverse applications of LBNP, it is to be expected that new and innovative applications of LBNP will be developed to explore the complex physiological mechanisms that underline health and disease.


Subject(s)
Blood Pressure/physiology , Hemodynamics/physiology , Hypotension, Orthostatic/physiopathology , Hypovolemia/physiopathology , Lower Body Negative Pressure , Animals , Humans , Space Flight
11.
Clin Microbiol Rev ; 37(3): e0016322, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39136453

ABSTRACT

SUMMARYThe human microbiota encompasses the diverse communities of microorganisms that reside in, on, and around various parts of the human body, such as the skin, nasal passages, and gastrointestinal tract. Although research is ongoing, it is well established that the microbiota exert a substantial influence on the body through the production and modification of metabolites and small molecules. Disruptions in the composition of the microbiota-dysbiosis-have also been linked to various negative health outcomes. As humans embark upon longer-duration space missions, it is important to understand how the conditions of space travel impact the microbiota and, consequently, astronaut health. This article will first characterize the main taxa of the human gut microbiota and their associated metabolites, before discussing potential dysbiosis and negative health consequences. It will also detail the microbial changes observed in astronauts during spaceflight, focusing on gut microbiota composition and pathogenic virulence and survival. Analysis will then turn to how astronaut health may be protected from adverse microbial changes via diet, exercise, and antibiotics before concluding with a discussion of the microbiota of spacecraft and microbial culturing methods in space. The implications of this review are critical, particularly with NASA's ongoing implementation of the Moon to Mars Architecture, which will include weeks or months of living in space and new habitats.


Subject(s)
Astronauts , Dysbiosis , Space Flight , Humans , Dysbiosis/microbiology , Microbiota/physiology , Gastrointestinal Microbiome/physiology
12.
Physiol Rev ; 98(1): 59-87, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29167331

ABSTRACT

Visual impairment intracranial pressure (VIIP) syndrome is considered an unexplained major risk for future long-duration spaceflight. NASA recently redefined this syndrome as Spaceflight-Associated Neuro-ocular Syndrome (SANS). Evidence thus reviewed supports that chronic, mildly elevated intracranial pressure (ICP) in space (as opposed to more variable ICP with posture and activity on Earth) is largely accounted for by loss of hydrostatic pressures and altered hemodynamics in the intracranial circulation and the cerebrospinal fluid system. In space, an elevated pressure gradient across the lamina cribrosa, caused by a chronic but mildly elevated ICP, likely elicits adaptations of multiple structures and fluid systems in the eye which manifest themselves as the VIIP syndrome. A chronic mismatch between ICP and intraocular pressure (IOP) in space may acclimate the optic nerve head, lamina cribrosa, and optic nerve subarachnoid space to a condition that is maladaptive to Earth, all contributing to the pathogenesis of space VIIP syndrome. Relevant findings help to evaluate whether artificial gravity is an appropriate countermeasure to prevent this seemingly adverse effect of long-duration spaceflight.


Subject(s)
Intracranial Hypertension/physiopathology , Intracranial Pressure/physiology , Intraocular Pressure/physiology , Posture/physiology , Space Flight , Vision Disorders/physiopathology , Animals , Humans , Intracranial Hypertension/etiology , Intracranial Hypertension/pathology , Vision Disorders/etiology , Vision Disorders/pathology
13.
FASEB J ; 38(14): e23831, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39037540

ABSTRACT

Depression is a significant concern among astronauts, yet the molecular mechanisms underlying spaceflight-induced depression remain poorly understood. MicroRNAs (miRNAs) have emerged as potential regulators of neuropsychiatric disorders, including depression, but their specific role in space-induced depression remains unexplored. This study aimed to elucidate the involvement of candidate miRNAs (miR-455-3p, miR-206-3p, miR-132-3p, miR-16-5p, miR-124-3p, and miR-145-3p) and their interaction with differentially expressed genes (DEGs) in the neurobiology of spaceflight-induced depressive behavior. Using a simulated space environmental model (SCSE) for 21 days, depressive behavior was induced in rats, and candidate miRNA expressions and DEGs in the cortex region were analyzed through qRT-PCR and HPLC, respectively. Results showed that SCSE-exposed rats exhibited depressive behaviors, including anhedonia, increased immobility, and anxiousness compared to controls. Further analysis revealed increased hydrogen peroxide levels and decreased superoxide dismutase levels in the SCSE group, indicating abnormal oxidative stress in the cerebral cortex. Moreover, miRNA analysis demonstrated significant upregulation of miR-455-3p, miR-206-3p, miR-132-3p, and miR-16-5p expression. Among the DEGs identified, the in silico analysis highlighted their involvement in crucial pathways such as glutamatergic signaling, GABA synaptic pathway, and calcium signaling, implicating their role in spaceflight-induced depression. Protein-protein interaction analysis identified hub genes, including DLG4, DLG3, GRIN1, GRIN2B, GRIN2A, SYNGAP1, DLGAP1, GRIK2, and GRIN3A, impacting neuronal dysfunction functions in the cortex region of SCSE depressive rats. DLG4 emerged as a core gene regulated by miR-455-3p and miR-206-3p. Overall, this study underscores the potential of miRNAs as biomarkers for mood disorders and neurological abnormalities associated with spaceflight, advancing health sciences, and space health care.


Subject(s)
Depression , MicroRNAs , Space Flight , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Male , Depression/metabolism , Depression/etiology , Depression/genetics , Rats, Sprague-Dawley , Disease Models, Animal
14.
Nature ; 568(7750): 55-60, 2019 04.
Article in English | MEDLINE | ID: mdl-30890786

ABSTRACT

NASA'S Origins, Spectral Interpretation, Resource Identification and Security-Regolith Explorer (OSIRIS-REx) spacecraft recently arrived at the near-Earth asteroid (101955) Bennu, a primitive body that represents the objects that may have brought prebiotic molecules and volatiles such as water to Earth1. Bennu is a low-albedo B-type asteroid2 that has been linked to organic-rich hydrated carbonaceous chondrites3. Such meteorites are altered by ejection from their parent body and contaminated by atmospheric entry and terrestrial microbes. Therefore, the primary mission objective is to return a sample of Bennu to Earth that is pristine-that is, not affected by these processes4. The OSIRIS-REx spacecraft carries a sophisticated suite of instruments to characterize Bennu's global properties, support the selection of a sampling site and document that site at a sub-centimetre scale5-11. Here we consider early OSIRIS-REx observations of Bennu to understand how the asteroid's properties compare to pre-encounter expectations and to assess the prospects for sample return. The bulk composition of Bennu appears to be hydrated and volatile-rich, as expected. However, in contrast to pre-encounter modelling of Bennu's thermal inertia12 and radar polarization ratios13-which indicated a generally smooth surface covered by centimetre-scale particles-resolved imaging reveals an unexpected surficial diversity. The albedo, texture, particle size and roughness are beyond the spacecraft design specifications. On the basis of our pre-encounter knowledge, we developed a sampling strategy to target 50-metre-diameter patches of loose regolith with grain sizes smaller than two centimetres4. We observe only a small number of apparently hazard-free regions, of the order of 5 to 20 metres in extent, the sampling of which poses a substantial challenge to mission success.


Subject(s)
Extraterrestrial Environment/chemistry , Minor Planets , Space Flight , Exobiology , Origin of Life , Space Flight/instrumentation , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL