Next Article in Journal
A Retrospective Observational Study Using Administrative Databases to Assess the Risk of Spontaneous Abortions Related to Environmental and Socioeconomic Conditions
Previous Article in Journal
Outcomes of Third-Line Trastuzumab Deruxtecan in a Patient with De Novo Stage 4 HER2-Positive Gastric Adenocarcinoma with Enteroblastic Differentiation: A Case Report
Previous Article in Special Issue
New Radiobiological Principles for the CNS Arising from Space Radiation Research
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Circuits and Biomarkers of the Central Nervous System Relating to Astronaut Performance: Summary Report for a NASA-Sponsored Technical Interchange Meeting

1
NASA Ames Research Center, Moffett Field, CA 94035, USA
2
KBR, Houston, TX 77058, USA
3
Universities Space Research Association (USRA), Moffett Field, CA 94035, USA
4
Department of Physical Therapy & Rehabilitation Science, University of California, San Francisco, CA 94110, USA
5
Department of Neurological Surgery, University of California, San Francisco, CA 94110, USA
6
Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
7
Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20814, USA
8
Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
9
Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92354, USA
10
Integrative Neurochemistry Laboratory, Behavioral Biology Program, McLean Hospital-Harvard Medical School, Belmont, MA 02478, USA
11
NASA Johnson Space Center, Houston, TX 77058, USA
*
Authors to whom correspondence should be addressed.
Life 2023, 13(9), 1852; https://doi.org/10.3390/life13091852
Submission received: 15 June 2023 / Revised: 24 August 2023 / Accepted: 25 August 2023 / Published: 31 August 2023
(This article belongs to the Special Issue Current Challenges in Space Neuroscience)

Abstract

:
Biomarkers, ranging from molecules to behavior, can be used to identify thresholds beyond which performance of mission tasks may be compromised and could potentially trigger the activation of countermeasures. Identification of homologous brain regions and/or neural circuits related to operational performance may allow for translational studies between species. Three discussion groups were directed to use operationally relevant performance tasks as a driver when identifying biomarkers and brain regions or circuits for selected constructs. Here we summarize small-group discussions in tables of circuits and biomarkers categorized by (a) sensorimotor, (b) behavioral medicine and (c) integrated approaches (e.g., physiological responses). In total, hundreds of biomarkers have been identified and are summarized herein by the respective group leads. We hope the meeting proceedings become a rich resource for NASA’s Human Research Program (HRP) and the community of researchers.

1. Introduction

Astronauts on long-duration space missions (e.g., transits to Mars) will experience the combined, potentially synergistic, impacts of simultaneous exposures to spaceflight hazards that affect the central nervous system (CNS) and operationally relevant behavior and performance [1]. While individual spaceflight hazards are often individually well quantified, in long-duration spaceflight, astronauts will experience multiple hazards simultaneously [2,3].
Parcelsus’ famous dictum on dose effects of exposures [4] reinforces the importance of an integrated approach to systematically identify and investigate the relationships of how spaceflight exposures may synergistically interact to pose a risk to the astronauts and the mission. NASA developed the Combined Behavioral Stressors (CBS) project which integrates research topics across three high-impact spaceflight hazard exposures—space radiation, isolation & confinement, and altered gravity—to inform performance outcome limits and permissible exposure limits, and to help identify and establish mitigation strategies. An integrated research approach is focused on identifying biomarker changes associated with exposures to the CBS-associated hazards to identify and develop effective monitoring, and apply countermeasures for mitigating risk to crew health and performance [5]. This is consistent with recent calls for more comprehensive and integrated biomarkers to better identify how different biomarkers can exert different causal effects between and among them [6].
The CBS Integrated Research Plan identifies biomarkers that are linked to in-flight and post-flight decrements in an astronaut’s operational performance resulting from simultaneous exposures to the CBS-relevant spaceflight hazards. In this context, a biomarker is defined as a characteristic that is “objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention” [7].
As sampling of in situ biomarkers in astronauts is not necessarily possible, translational models are useful. To promote the utility of translational models, NASA consistently updates the exposure levels in rodents as they relate to humans; for example, NASA recently adjusted their integrated research platforms involving animal exposures to expected levels of spaceflight radiation related to dose and duration [8]. It is, therefore, essential that biomarkers are useful for bi-directional translation of homologous human and animal measures, which is a cornerstone of the NASA’s CBS project—allowing for the linking of the probability for performance decrements (during and/or after mission) to the level of exposure to a CBS relevant spaceflight hazard, such as radiation exposure.
This paper reviews the results of NASA’s biomarker technical interchange meeting (TIM) that was focused on creating a comprehensive list of constructs, identifying underlying and related brain regions, neural circuits, and biomarkers for inclusion in predictive models to assess and validate changes in future astronaut risk status, as well as to identify changes in operationally relevant brain pathways (e.g., procedural memory) after exposures to varying types and amounts of potentially synergistically acting spaceflight hazards. The overall goals of this biomarker TIM were to (i) identify relevant brain regions, neural circuits, functions, and associated biomarkers, and relate them to operationally relevant performance, and (ii) identify any critical needs for new biomarker knowledge (“gaps”) that can be filled by additional focused and translational animal experiments that include a plausible pathway toward eventual biomarker validation in humans.

2. Meeting Synopsis

Biomarkers—ranging from molecules to behavior—can be used to identify thresholds beyond which performance of mission tasks may be compromised and could potentially trigger the activation of countermeasures. Identification of homologous brain regions and/or neural circuits related to operational performance may allow for translational studies between species. Three discussion groups were asked to use operationally relevant performance tasks as drivers when identifying biomarkers and regions or circuits for the constructs listed in Appendix A. Participants are listed in Appendix B. Here, we summarize the discussions below across the three groups. In total, hundreds of biomarkers have been identified, with references provided mainly in the respective tables for each group. We hope the meeting proceedings become a rich resource for NASA’s Human Research Program (HRP) and the community of researchers.

3. Summaries of Discussions and Recommendations from Each of the Breakout Sessions

3.1. Sensorimotor Influences on Operational Performance (Leads: S. Rosi, M. Shelhamer)

The goal of Group 1 was to create lists of biomarkers and brain regions and/or neural circuits related to operational performance for constructs that are prioritized in HRP’s sensorimotor risk. Group 1 assessed the following 13 key constructs in Table 1: visual function, spatial orientation, vestibular, proprioception, hearing, motion sickness, smell and taste, postural control and balance, locomotion, fine motor control, perception, gaze, and pain. Note that the panel assessed translatability based on the existence of rodent models and did not suggest using non-human primates (NHPs), nor did they identify a construct that should be tested in NHPs.

3.1.1. Summary of Discussions

During discussion of each of the 13 constructs, 10 themes emerged. Although identification of themes was outside the scope of the panel, these themes were applicable to nearly all constructs discussed and, therefore, we define them here:
1. Connections between constructs. Distinctions between the constructs are, in many cases, artificial. Although segregated disciplinary expertise has achieved a great deal in the sensorimotor domain, the different constructs are so closely interconnected that it is hard to discuss them separately in a way that is true to the science and to the operational implications. As an example, vestibular function, gaze control, balance, and locomotion are very closely related, and yet they are often addressed as specific and separable. Another example is perception. Almost all sensorimotor constructs involve perception in some way; vestibular perception—perception of the upright—affects the ability to balance. Perception of upright is influenced by changes that occur in microgravity, which is a vestibular effect. Again, these specific constructs become tightly entangled and it is difficult to separate them in terms of biomarkers and operational relevance.
2. Many spaceflight stressors and sensorimotor effects occur simultaneously with different time courses. Not only do the different constructs interact, they do so with different time courses. The most overt and acute forms of vestibular adaptation (related to space motion sickness) occur over the course of a few days, whereas other vestibular-mediated functions (e.g., the sense of being truly comfortable with the three-dimensional aspects of motion in a weightless environment) develop over several weeks. Some adaptive sensorimotor changes in space occur with similar time courses as those seen in analogous environments on the ground. For example, the changing contributions of vestibular, proprioceptive, and efference copy information during recovery from labyrinthectomy in an animal model [9] have time courses that mimic recovery of motor control during locomotion after spaceflight [10]. Similarly, ground-based studies in animals show that development of efference copy over several weeks mimics the time course of the development of three-dimensional spatial sense in astronauts over the same time period. The similar time courses suggest that these may be aspects of the same underlying process. This might provide translational opportunities from ground-based animal models and may inform a process for preadaptation paradigms for spaceflight.
3. Multi-sensory integration. This is related to the theme of interacting constructs. Most sensorimotor behaviors and perceptions arise from the simultaneous activation of multiple sensory systems. An obvious example is the combination of visual and vestibular information for gaze control (vestibulo-ocular reflex (VOR)). Another is the prevalence of proprioceptive and kinesthetic influences, in addition to vestibular and visual influences, on posture and locomotion.
4. Stress. Spaceflight involves multiple simultaneous stressors—physiological, psychological, and environmental. These have widespread and sometimes unknown influences on sensorimotor function, and likely on the ability to adaptively alter sensorimotor function. The effects of stress on motor learning and on motion sickness are two examples: stress affects motor learning, which alters adaptation, which can change the ability to recover from motion sickness, which can increase stress.
5. Learning. Almost all the individual constructs exhibit adaptive behaviors to spaceflight and these adaptive behaviors may complicate the usefulness of the constructs as biomarkers because the response that is being assessed will change with adaptation to spaceflight. Of course, such adaptation is desirable and should be promoted, but it complicates the use of a biomarker to identify increased risk to astronaut health and performance. This would be especially true in missions of extended duration where the adaptive processes might not be understood. A specific biomarker for learning and adaptation would be desirable.
6. Some constructs might be easily measured but lack relevance. As an example, the angular VOR has been extensively studied and is easy to measure, but little or no evidence exists that it changes significantly due to spaceflight, or that any changes have an operational impact.
7. Neural circuits. Interpretation of neural circuitry is not always straightforward. There is not always a direct analogy between animals (where many circuits have been delineated) and humans; the neural circuitry is different in some cases, and there are also adaptive changes that make the definition of standard circuits difficult. Circuit function is implicitly assessed with behavioral measures, so knowledge of some circuit characteristics such as neurotransmitters and common pathways might aid in the interpretation of behavioral markers.
8. Vestibular Cognition. The relationship between cognition and the vestibular system, and the vestibular effects on cognition, is operationally relevant and directly connects cognition and sensorimotor functions. This connection is seen in many patients with vestibular problems. No specific construct exists for this, and it is difficult to conceive of a specific biomarker.
Overall, the sensorimotor issues of multi-sensory/multi-effector interactions and learning, and their relation to stress, are not yet sufficiently studied, and they likely greatly influence human performance in space. These do not yet lend themselves to direct biomarker identification.

3.1.2. Recommendations

The panel evaluated each specific construct to determine if a good biomarker exists that is operationally relevant for astronauts, and that translates from animal models. The panel also commented on gaps in each construct that would need to be filled to produce an effective biomarker.
1. Visual function is easily measured (acuity, visual fields, etc.), and these measures may help to parse out visual effects from motor effects when there is a functional deficit. Retinal remodeling can be assessed with optical coherence tomography (in flight), and is hence a biomarker. Translatability is clear because many of these aspects can be tested in rodents (e.g., visual acuity in mice and even real-time visual tracking). This is clearly a useful biomarker.
2. Spatial orientation is extremely important. The panel extensively discussed grid cells—the cells in the entorhinal cortex that underlie spatial orientation. The firing of grid cells provides information that can be used to assess spatial orientation as it adapts to alterations in gravity, which is further substantiated as a potential biomarker due to its translational potential as grid cells are present and accessible in rodents. Thus, neural circuits in the hippocampus and medial entorhinal cortex are important.
3. A great deal of information exists on vestibular function in spaceflight. Basic vestibular function is not significantly altered in the microgravity environment of space, although central processing and higher-level derived functions (e.g., spatial orientation, tilt-translation perception) often are. It is, however, important to consider vestibular changes in the context of the integrated spaceflight stressors. So, as noted, the VOR changes little in weightlessness, but it would be useful to assess VOR in the context of other stressors (e.g., radiation, fatigue, etc.); for example, what is the combined impact of multiple stressors? These aspects need to be elucidated, which can be accomplished through rodent studies (e.g., the narrow balance beam as a viable animal assessment). Taken together, vestibular change (e.g., VOR or balance beam performance) is a suitable biomarker.
4. Proprioception was identified as one of the most strongly interconnected constructs, exhibiting significant overlap with several other constructs. Little is known about the effects of (CBS risks) radiation or other stressors on the peripheral nervous system and, consequently, proprioception (this is a gap in knowledge). A rodent model would provide translational opportunities, as proprioception can be measured in that model (e.g., tape removal test, whisker test). Hence, measures of proprioception are suitable biomarkers.
5. Hearing loss is often a factor associated with spaceflight, perhaps due in part to fluid shifts, and hearing assessment in flight may help to parse out the effect of the fluid shift from noise-induced loss. However, the panel noted that these data are not particularly operationally relevant: hearing loss has not been a functional problem. As such, hearing loss is not a priority biomarker.
6. Motion sickness is a known problem that needs to be further assessed because it can have serious operational impacts [11,12], especially when first experiencing a gravity field after extended weightlessness. Motion sickness susceptibility is still unpredictable. This line of work might be revisited with more recent knowledge on learning and adaptation or might be investigated in relation to the impact on specific operational tasks. We do not know how motion sickness induces stress and how stress feeds back to motion sickness and the overall well-being of astronauts. The interaction of motion sickness, sopite, stress, and crew performance has been studied in other contexts. This work should be reviewed; however, it may still be valuable to investigate these effects in the specific context of spaceflight, with its multiple simultaneous stressors and unique demands. Again, there are several overlapping biomarkers. A drawback in this area is translatability, because it is very difficult to measure motion sickness in rodents. This is a useful biomarker, albeit with some uncertainties as to translational aspects.
7. Smell and taste are particularly important for humans as social creatures and are also clearly important in space. These constructs overlap with the well-being and operational performance of astronauts. Smell and olfaction can be markers for neurodegeneration. Loss of olfaction (anosmia) is an early marker in COVID-19 and Alzheimer’s disease, as examples, and is therefore a biomarker for neurodegeneration that can also easily be tested in rodents. This biomarker is rated highly.
8. Posture and balance are important operational issues. They are problematic as biomarkers because, again, their functions cannot be isolated to discrete neural circuits due to the overlap of several circuits for multi-sensory integration and motor control. Rodent models are somewhat problematic because of the difference between neural circuits and functions in organisms with four legs (rodents) relative to two legs (humans).
9. As with posture and balance, locomotion is operationally relevant and important, but good rodent models in spaceflight or microgravity environments are lacking. It might be useful to consider static/dynamic balance control as opposed to posture/locomotion.
10. Fine motor control is difficult to assess because of the large number of confounders. Related factors that can alter fine motor function include changes in proprioception, hand-eye coordination, and others. Although functionally important, it may not be particularly relevant for operational control tasks, and suitable rodent models are lacking. The many confounders alone make this problematic as a discrete biomarker.
11. Perception is in fact a component of almost all the other constructs because it can include spatial orientation, depth perception, vestibular orientation, time perception, and others. Understanding of this construct is important and would address many of the other constructs, but there are many overlaps. Proprioception may be altered and is a critical issue on its own, but it will be most important to address in the context of other stressors. Specific aspects of perception have been noted in spaceflight and can have operational impacts, and so it would also be beneficial to consider perception in this performance context. Nevertheless, parsing out perceptual effects per se remains difficult. Thus, this was not considered to be a good biomarker.
12. The panel did not rate gaze and pain highly as biomarkers. Gaze largely overlaps vestibular function (and has been studied almost as much), so gaze control can be subsumed under vestibular function. Pain per se is not a good biomarker because of confounders between the perception and the sensation of pain. Nociception can depend on sex and other individual factors. Although biomarkers of inflammation exist, these are associated with pain. Hence, pain itself is not a discrete biomarker.
Table 1. Circuits and biomarkers for sensorimotor domains.
Table 1. Circuits and biomarkers for sensorimotor domains.
Key Indicator/ConstructHuman Performance Test (Details about the Actual Test/Assay)Animal Performance Test (Details about the Actual Test/Assay)Caveats/Notes/Related Functional Performance Tasks/Prediction of Behavioral Outcome in HumansBrain RegionHuman/NHP Neural Circuit/PathwaysRodent Neural Circuit/PathwayBiomarkers (Rodents/Humans/NHPs)Gaps/Notes
InaccessibleAccessible (Translatable to Astronauts)
VisualVisual field testingVisual field testing Visual cortex (Occipital lobe of the primary cortex)Retino-geniculate-striate pathway (Conscious vision) Dorsal pathway (spatial location and action): Retina → LGN →V1 → V2 → MT (parietal lobe)
Ventral pathway (characteristics of objects): Retina →LGN →V1 → V2 → V4 (temporal lobe) [13]
Retina-Superior Colliculus-Lateral posterior nucleus-Visual cortex1 pathway [14]Retinal markers-autopsy, superior colliculus pathway—neural circuitry, intracranial pressure in astronauts—lumbar puncture for pressure detection, retinal vasculature imaging—vessel length density and loss of photo receptor cells, role of endothelial structure or vasculature, acceleration of incident of cataract (on cornea, not CNS) and light flashes (post-flight and long-term issue), fluorescent imaging of the retinal vasculature.Imaging: Inflight CT, MRI imaging, ultrasound, OCT, visual field measurements, cataract as predictor Structural changes in eye, nerve, occipital cortex, pretectum, superior colliculus. Vision function test, sampling of tears [15], Intraocular pressure measurement, Saccades [16], Behavioral measures, Live pupil tracking(1) Potential Optical/Eye damage in astronauts—could also be indicator of neurological symptoms.
(2) Any imaging other than ultrasound is difficult to do in space. Difficult to get a gold standard test for intracranial pressure in space.
(3) Possibility of lumbar punctures in astronaut—intracranial pressure.
(4) VR environments for complex sensory integration—Somatosensory component
Spatial Orientation1. Path integration-passive and active
2. Virtual maze perspective taking tests
3. Visual object learning (VOLT)
1. Changes in activity of head direction, grid, place cells
2. Morris water maze
3. Spatial navigation
4. Touch screen cognitive testing [17].
- Test in higher animals: NHP -Spatial navigationHippocampus and parahippocampal regions, cerebellum, brain stem, Retrosplenial cortex (Grid cells, border cells, head direction cells—cortical regions- egocentric and allocentric reference frame) [18]Vestibulospinal pathwayProposed head direction pathway 1: Vestibular nuclei (VN) → Cerebellum → ventral lateral nucleus of thalamus (VLN) → parietal cortex → temporal cortex → hippocampus?
Proposed head direction pathway 2: Vestibular nuclei (VN) → hippocampus [19]
Hippocampal protein lysate: Afg3l1, Tpx2, Neuroligin-3, RB1-inducible coiled-coil 1, Mast3, Kif21a, DnaJ (Hsp40) homolog, SLIT-ROBO Rho GTPase-activating protein 2, Rasgrf1 [20]Structural changes in hippocampus, anterior thalamus, subiculum. Electrodermal activity measured by wrist worn device [21], Optical coherence tomography (OCT), Illusionary experience, somatographic illusion—questionnaire(1) Virtual reality biomarker development for astronauts.
(2) Spatial orientation during g-transitions
(3) Different species have varied responses. Need a model that would be most translatable.
Vestibular1. Drop test/Jump down test
2. VEMP
3. OVAR response (Sensorimotor component after 30 rpm) 4. Time constant or constant rotation
5. ocular counter roll (but noisy)
1. Balance beam test (narrow beam) Righting reflex 2. VEMP (can be done in space and can help distinguish utricular and saccular functions)
3. OVAR response
4. Active vs. Passive motion on vestibular nucleus neurons
5. VSEP (otolith function)
6. Swimming test (for subtle deficits, screening test)
Test in higher animals: NHPThalamus and cortexThalamocortical pathways Anterior vestibulothalamic pathway: Vestibular nuclei (VN) → Nucleus prepositus and supragenual nucleus (NPH/SGN) → Anterior dorsal thalamus (ADN) → Entorhinal cortex → Hippocampus
Posterior vestibulothalamic pathway: Vestibular nuclei (VN) → Ventral posterior lateral nucleus (VPL) → vestibular cortical areas. [9] -Three neuron pathway Vestibulo-ocular reflex: vestibular afferents → vestibular nuclei → Vestibulo-ocular reflex and efferent (vestibular processing)
(1) Vestibular nucleus → Dorsal tegmental nucleus (DTN) → Lateral mammillary nucleus (LMN) → Anterodorsal nucleus (ADN) → Post-subiculum (PS) → Hippocampus
(2) Vestibular nucleus → Pedunculopontine tegmental nucleus (PPTN) → supramammillary nucleus SUM → Medial septum → Hippocampus
(3) Vestibular nucleus → Thalamus → Parietal cortex → Entorhinal/Perirhinal cortices → Hippocampus [22].
Otopetrin1, Alpha 2 adrenergic receptors [23], Glutamate receptor expression [24], c-FOS, vestibular hair cells [25], cerebellar nodulus of adult rats [26,27,28], TEM of synaptic ribbons [29,30,31,32,33]Nausea related—cardiac sensitivity to baroreceptor reflex; raised Heart rate; raised cortisol; reduced dominant power on EGG baseline, questionnaire [34,35], Serum: NSE and S100β [36], Otolin-1 [37]. vibration-induced nystagmus [38](1) Effects of stress on vestibular compensation and adaptation.
(2) Social stress, performance anxiety, other psychological stress—will it impede recovery?
(3) Stress impedes motor learning in mice (Fragile X mice).
Gaze1. Gaze Holding/Gaze stability
2. Eye-head coordination
3. Redirecting gaze
Gaze HoldingTest in higher animals: NHPVisual pathway, Frontal eye fields, vestibular nuclei, cerebellum, oculomotor system, parietal cortex, postcentral gyrus, Entorhinal cortex neuronsHorizontal vestibular-generated eye movement: Horizontal semicircular canal → Vestibular nucleus (Vestibular ganglion) and cerebral cortex inputs (frontal eye field) → Paramedian pontine reticular formation (PPRF or gaze center) → Medial longitudinal fasciculus (MLF) → ipsilateral lateral rectus muscle (eye) and contralateral medial rectus muscle (eye) [39]. Structural changes in cerebellum (conventional and mass-spec imaging), Diplopia, Blurring of vision, vestibulo-ocular reflex. Gaze holding/stability and ability to redirect the gaze with accuracy—integrative Biomarker
Locomotion1. Tandem Walking (=Beam Walking in Animal);
2. Perturbation during walking
3. Navigating obstacle course while walking (eg. Functional Mobility Test)
4. Statistical modeling of actigraphy data
1. Rotarod
2. Beam walking (=tandem walking);
3. Actigraphy in animals;
4. Open field
Test directly in humans when possible.
Animal model tests should be developed:
a. DigiGait 2.0 Analysis with perturbation, belt or surface perturbation (=human perturbation during walking);
b. Dual task test (Catwalk); c. Rodent obstacle course (=FMT)
Mesencephalic locomotor region (MLR) in the midbrain(1) Reticulospinal pathway: Motor cortex → Basal ganglia → Mesencephalic locomotor region → Pons/Medulla (Reticulospinal cells) → Spinal cord/Central pattern generator → Muscle [40].
(2) Vestibulospinal pathway
(1) Reticulospinal pathway (major pathway for initiating locomotion): Motor cortex → Basal ganglia → Thalamus → Mesencephalic locomotor region → Pons/Medulla → Spinal cord/Central pattern generator network → Muscle
(2) Vestibulospinal pathway
(3) Rubriospinal pathway [41]
Behavioral tests. Locomotion and gait as a
biomarker associated with NDs
(1) Can be nested in vestibular, posture, and gait construct (2) Static vs. Dynamic postural control is important
Postural control, Balance1. CDP.
2. Get up From Fall Test
3. Induced stepping (hold and release)
4. Body sway test (non-parallel two-leg model).
5. Engaged leg model of body sway (uneven weight distribution)
(1) Rotarod
(2) Zebrafish Active Posturography (Zap);
(3) Floating Platform Tests–Postural sway–measured by Center of Pressure (COP) Assay (=COP)
Test directly in humans when possible.
Animal model tests should be developed:
(a) Floating Platform Test
(b) Motion Capture Analysis (exists but
advanced version can be developed)
Cerebellum, sensorimotor cortex, vestibular cortex, prefrontal cortexPostural information → Vestibular/Visual/Somatosensory input → Brainstem, cerebellum, thalamus → Temporoparietal cortex (vestibular cortex/posteroparietal cortex) → primary sensory cortex → Supplementary motor area and premotor area (info. integration from hippocampus) → basal ganglia/cerebellum (corticovestibular projections) → Brain stem → Spinal cord (reticulospinal tract) → Muscle [42].Posture-head stabilization: Inner ear vestibular receptors → vestibular nerve → ipsilateral vestibular nuclei in brain stem → vestibulocerebellum/medial vestibulospinal fasciculus → ipsi/contra projections → motor neurons (neck muscle) Locomotion coordination: Inner ear vestibular receptors → vestibular nerve → ipsilateral vestibular nuclei in brain stem → striatum (thalamic relay)/Lateral vestibulospinal fasciculus → ipsilateral projections → locomotor central pattern generator → motor neurons (trunk and leg muscles) [43]. Rodents: Circling, body sway area, the barycenter, the support surface and the weight distribution of the rats when they were moving or stationary [43].(1) Operationally relevant. Need to evaluate before EVA
(2) Animal models not so useful (2 vs. 4 leg)
Motion sickness1. Graybiel scale (comprehensive)
2. Nausea (0 to 10)
3. Eye strain (0–10)
Not reliable in rodent. Ferrets have vomiting response. squirrel monkey and rhesus monkey—difficult to test Brain stem and CerebellumInput (Visual, Vestibular labyrinth, proprioceptive) → vestibular nuclei → cerebellum → brainstem autonomic centers→ vomiting center [44]. Structural changes in inner ear. Increased plasma glucose [45],
Nausea related—cardiac sensitivity to baroreceptor reflex; raised Heart rate; raised cortisol; reduced dominant power on EGG baseline [34,35]
(1) Study the effects of stress, sleep deprivation, head-loading, oscillation vibrations, prolonged fixation, and motion sickness
(2) There are enormous differences in individual susceptibility, with respect to both sensitivity and adaptation/rapid decay of stimulus. So, in long term space missions like to Mars- should we pre-screen the astronauts? But predicting susceptibility is unclear.
(3) How relevant is it to astronaut performance considering it affects only during g transitions (~1% of their time in a 3 year mission).
(4) Sopite syndrome—can affect operational performance—Combined effect.
(5) Translatability -ferret and mouse model, tricky to track
Proprioception1. Force and joint position test;
2. Dysmetria (finger to nose) test +/− eyes closed;
3. Foot sensitivity via pressure algometry (provides objective measure) = Von Frey Fibers;
4. Thesiometry, vibration at different frequency ranges for slow or fast adapting sensors
5. Tendon tap test, tonic vibrations? complementing Hoffman reflexes
1. Von Frey Fibers;
2. Static force von Frey
3. Two-choice mechanosensory assay
4. Cotton swab assay
5. Tail Clip assay
6. Tape response assay
7. Hargreaves assay
8. Randall-Selitto assay
9. Complete Freund’s adjuvant with von Frey
10. Bradykinin with von Frey
11. Two temperature choice assay.
12. Thesiometry testing—withdrawal responses
13. Coupling a Y maze in dark and add tape for tactile responses.
14. Barrel reception system
15. Whisker test coupled with NOR
Animal model tests should be developed:
a. Force and joint position test;
b. No identified animal equivalent of dysmetria
Thalamus, Somatosensory cortex, cerebellum, vestibular cortex, prefrontal cortex, Right putamen, parietal cortex, mouse barrel cortex (homunculus)Dorsal Column pathway:
Proprioceptors → Spinal cord → Nucleus cuneatus (Medulla) → Ventral Posterior lateral nucleus (Thalamus) → primary somatosensory cortex Spinocerebellar pathway (unconscious proprioception): Muscle → Spinal cord → cerebellum
Thalamo-insular pathway [46] Proprioceptive signals from Jaw-closing muscle spindles (JCMSs) → the caudo-ventromedial edge (VPMcvm) of ventral posteromedial thalamic nucleus (VPM) → dorsal part of granular insular cortex rostroventrally adjacent to the rostral most part of the secondary somatosensory cortex (dGIrvs2) Proprioceptive signals → thalamus → cerebral cortexPiezo2 [47], Erg3 transcript levels [48]. Transient receptors which are responsive to camphor, menthol, and capsaicin to stimulate the receptors and check the response.fMRI and Diffusion tensor imaging (DTI): structural differences within the right putamen [49]-not done in orbit(1) Very little data from peripheral nervous system and spinal cord. (2) Need to look at the effects of combined stressors
Fine motor
control
1. Peg board;
2. Fine motor test (Holden iPad);
3. String/rope pull 4. Precision grip post-flight (JL)
1. String pull;
2. Spaghetti eating;
3. Lever manipulation
Animal model tests should be developed:
Peg board
Cerebellum, basal ganglia, motor cortex, thalamus, rubrospinal, sensorimotor cortex, prefrontal cortex, frontal lobeVestibular/Visual input → Brainstem, cerebellum, thalamus → Temporoparietal cortex (vestibular cortex and posterior parietal cortex) → S1 (Primary sensory cortex) → M1 (Primary motor cortex) → Lateral corticospinal tract → Spinal cord → Muscle [42]Visual/Olfactory input → Sensorimotor cortex → Corticospinal tract (Motor and Sensory) → Cervical spinal cord → Sensory and Motor neurons → Muscle [50] Isometric pinch grip force between the thumb and index finger [51](1) Proprioception can be connected to the fine motor control.
(2) Animals have fine motor control, but we need to standardize and develop a model
Perception1. Depth—Egocentric distance
2. Motion illusions—Verbal reports of illusions when
changing modules or looking outside
3. Time—Duration estimates
1. Shape—Novel object recognition
2. Depth—Cognitive Flexibility
3. Time—Navigation and Foraging
4. Visual—Food protection behavior
Test in higher animals: NHPTime perception: Frontal cortex, basal ganglia, parietal cortex, cerebellum, and hippocampus, lateral and medial entorhinal cortex [52]Dorsal stream pathway (where): Retina → Visual cortex (V1, V3) → Middle temporal area (V3A/MT/V5) and Medial superior temporal area → Intra-parietal area → Parieto-occipital area (PO/V6) Ventral stream pathway (what): Retina → Visual cortex (V1) → Visual cortex (V2) → Visual cortex (V4) → Inferior-temporal cortex → Fusiform gyrus (Fusiform face area and occipital face area) [53] Structural changes in somatosensory cortex, Perception as a biomarker?—has many confounding factors(1) Adaptation following flight + return?
(2) Some disagreement regarding the relevance of perception in performing operationally relevant tasks
Pain(1) Back pain (2) Skin sensitivity
(3) Pain modulation while modulating vestibular sensitivity
(4) Joint pain
Crew after one-year long duration mission had significant skin sensitivity for prolonged periodsThalamus, Primary somatosensory cortexPain or Nociception Pathway: Ascending: Nociceptors in Skin → Spinal cord → medulla → midbrain → Thalamus → Primary somatosensory cortex.
Descending: Amygdala → Hypothalamus → PAG → rostral ventromedial medulla → spinal cord → nociceptor [54,55]
Ascending pain pathway: Nociception receptors → spinal cord dorsal horn → parabrachial nucleus (brain stem) → thalamus and amygdala → somatosensory cortex/prefrontal cortex/anterior cingulate [56]Bilateral lesion in mPFC [57]Blood: MFAP3, GNG7, CNTN1, LY9, CCDC144B, and GBP1 [58], sICAM-1 [59], fMRI based brain imaging [60], Autonomic nervous system markers: Pupil reflexes, Electrodermal activity, Peripheral pulsatile component of cardiac cycle, Heart rate, Blood pressure [61]. Blood markers, miRNA markers, inflammatory factors and CCR2 receptor, Pain as biomarker (many confounders).(1) Need to focus on peripheral nervous system and include and utilize blood markers.
(2) Individual pain tolerance is variable
Smell and taste1. University of Pennsylvania Smell/Taste identification Test scratch and smell test1. University of Pennsylvania Smell/Taste identification Test in animals—odor is very important, social interactions, fear conditioning, memory sequences of odor.Smell and Taste has been hypothesized to be modified secondary to fluid shifts causing increase in salt and spice intake leading to dysregulation of body salt compositionGustatory and olfactory cortex, Piriform cortex and homology to hippocampus. Olfactory epithelial, like hippocampus, has continual neurogenesisGustatory pathway: Tongue → solitary nucleus (medulla) → thalamic nucleus (ventral posterior medial nucleus) → gustatory cortex → hippocampus (identification) Olfactory pathway: Olfactory receptors → olfactory bulb → olfactory cortex → hippocampus (odor memory) Olfactory receptors → olfactory bulb → olfactory cortex → thalamus → orbitofrontal cortex (conscious perception of smell)Olfactory pathway: Odor input → olfactory sensory neurons in olfactory epithelium → olfactory bulb → hippocampus → amygdala → learning/behavioral input [62] Smell and hippocampal circuits are similar → can be used to assess broader cognitive dysfunctionOlfactory bulb volume [63]Nasal mucus (smell): Sonic hedgehog levels [64]; Saliva (taste)—Sonic hedgehog [65]
Blood—miRNA panel including mitochondrial stress markers. Smell test: Scratch and sniff test. Smell as a biomarker.
(1) Loss of smell impacts social interaction and can lead to depression. Loss of smell in long term missions can contribute to depression.
(2) Smell can also have a downstream effect. Onset of smell precedes for many years in AD patients.
(3) What about systemic response associated with smell deficits; can we have blood biomarkers for it? Mitochondrial functions are associated with olfactory pathways—can we test mitochondria? can we identify miRNAs associated with olfactory issues?
Hearing1. Otoacoustic emission
2. Auditory evoked potential analysis
1. Otoacoustic emission
2. Auditory evoked potential analysis
Test in higher animals: NHPAuditory cortexAuditory pathway: Ear → cochlea → cochlear nucleus (medulla) → superior olive (medulla) → inferior colliculus (midbrain) → medial geniculate (thalamus) → auditory cortex Lemniscal auditory pathway, olivo-cochlear systemAscending auditory pathway: Ear → Cochlea → Cochlear nucleus → superior olive → inferior colliculus → medial geniculate nucleus (dorsal thalamic nucleus) → auditory cortex [66] Blood: Prestin [67,68], Low frequency hearing loss(1) Need to study combinatorial stressors
(2) Effects of microgravity on hearing/auditory. (3) Largely ignored—as most of behavior test do not rely on hearing ability

3.2. Behavioral Medicine Influences on Operational Performance (Leads: C. Davis, David Dinges)

The goal of Group 2 was to create lists of biomarkers and brain regions and/or neural circuits that are related to operational performance for constructs that are prioritized in the HRP’s Behavioral Medicine (BMed) risk. Group 2 assessed the following key constructs which are summarized below and in Table 2: memory, attention and dual tasking, executive function, working memory, learning and plasticity, social processes, individual behavioral states, arousal and regulatory, emotional regulation, risk taking/tolerance, and stress.

3.2.1. Summary of Discussions

Many of the themes that arose during this panel’s discussion were also discussed by the sensorimotor group (Group 1), including learning and plasticity for assessing an astronaut’s general level of adaptability. The panel also discussed the importance of studying individual differences in these different behaviors, in addition to various modifying factors, such as sex, age, the impact of stress, and immune status. The panel also highlighted the importance of general biomarkers that are not specific to any construct, behavior, or tissue, but could provide a more accurate reflection of overall behavioral health.
Behavior is a biomarker. One major theme that emerged from the discussion was the fact that behavior is an important biomarker. Although biomarkers and brain regions and neural circuits are important for understanding the biological basis of changes in operational performance, the behavior itself needs to be studied as an indicator of changes in operational performance. Variations in behavior, such as increases in variability of response and instability in performance, are often the most sensitive indicators of degradation of operational performance [69,70]. Furthermore, marked inter-individual differences exist in these domains, some of which appear to be phenotypic [70,71]. However, limited knowledge exists regarding the biological basis of these individual differences and how they are modulated by spaceflight stressors. For several constructs, the panel noted specific behavioral changes that should be considered as biomarkers and gave examples of potential neuroimaging modalities that could be used to investigate underlying brain regions and neural circuits. More studies of human behavior in spaceflight are needed. Behavioral tests with greater ethological relevance to animal models would most likely yield better translation of findings to human operational performance. The panel discussed similarities between attention tasks and dual tasking; performance instability, increases in the variability of responding, and increased impulsivity are all behavior markers indicating a problem [70,72,73]. These changes can be subtle, which highlights the importance of knowing the organism’s baseline performance for a task, so that changes to that baseline will then indicate a problem. Finally, behavioral biomarkers can be used to determine when an organism—from rodents to humans—is unable to use new information in the environment to adapt their behavior; these results have been obtained primarily from reversal learning and extinction tasks that are highlighted under General Brain Plasticity below.
Common measurements for studying brain biomarkers. Various neuroimaging modalities were discussed for most of the constructs, and because the panel focused on measures that could be assessed during spaceflight and across species, electroencephalogram (EEG) and event-related potentials were regarded as valuable for identifying markers associated with several constructs, including memory, working memory, attention, dual tasking, and learning and plasticity. The use of whole-brain and region-specific EEGs were both considered useful, with whole-brain EEG being particularly important for learning and plasticity [74,75]. Region-specific EEGs were regarded as most useful when coupled with a behavioral task dependent on that region, such as frontal cortex activity and attention or performance on an adaptive N-back test to assess working memory. Near-infrared spectroscopy (NIRS) and functional NIRS were also regarded as useful for assessing underlying neural targets during task performance during spaceflight.
Magnetic resonance electroencephalography and other frameworks for integrating multiple imaging modalities should also be investigated, such as joint imaging markers from simultaneous magnetic resonance imaging (MRI) and EEG (e.g., temporal volume, cortical thickness) that are associated with cognitive status in healthy individuals, pathophysiological changes in neurodegenerative diseases, and after traumatic brain injury [76,77,78,79,80,81]. The panel contended that these simultaneous recordings could provide a more accurate diagnosis of pathology than either modality alone.
Overlapping markers among constructs. The panel agreed that many biomarkers overlap among the constructs, such as the gastrointestinal (GI) microbiome, immune markers, and the influence of steroid hormones. As such, these markers could be general markers of behavioral health. For translational studies, most of these markers can be measured in animal models and have supporting preclinical evidence to demonstrate their relevance to human CNS function and disease.
  • Immune markers. Several accessible biomarkers are common to various constructs, including inflammatory markers such as Tumor Necrosis Factor alpha (TNF-alpha), Interleukin 6 (IL-6), and Interleukin 8 (IL-8).
  • Oxidative stress markers. The panel considered transthyretin (TTR) as a biomarker of neuronal stress that could be useful for assessing general CNS health, irrespective of a specific BMed construct. Although TTR is possibly inaccessible for spaceflight (e.g., choroid plexus TTR, lumbar puncture for cerebrospinal fluid), recent work suggests serum levels could be indicative of CNS pathology [82].
  • Microbiome. The GI microbiome is connected to the brain through the gut–brain axis and the panel regarded this as an important system to assess potential biomarkers indicative of CNS pathology. Recent research demonstrates a vital role of the GI microbiome in CNS pathology and psychiatric disorders [83,84,85] and the microbiome has important implications for health during long-duration spaceflight [86,87].
Incorporate modifying factors into biomarker studies. The panel discussed additional factors important for spaceflight, and differences in many of the BMed constructs that were not included on the worksheet, such as sex, age, stress, immune status, steroid hormone levels, and prior experiences. The panel noted that any findings regarding the usefulness of the various biomarkers should also include tests of these biomarkers under these additional conditions to determine if the markers were relevant when these other factors are included. For example, a biomarker might be useful for males, but not females, or the menstrual cycle phase could impact the usefulness of the biomarker in females. Studying biomarkers under combined spaceflight factors in analog environments [88] was also viewed as being important to determine the usefulness of these biomarkers, given that individuals might respond differently to various spaceflight factors.
Default mode network (DMN). The panel discussed the importance of the DMN in both normal and pathophysiological processes as it relates to several of the BMed constructs, and they considered DMN to be a marker that might overlap among constructs (e.g., changes in DMN could indicate memory and attention problems, in addition to sensorimotor changes). The DMN is a brain system that is preferentially activated when the brain is at wakeful rest [89,90]. Core regions of the DMN include the medial prefrontal cortex, posterior cingulate cortex, and parts of the precuneus, as well as the hippocampus, retrosplenial cortex, and angular gyrus [91]. Changes in activation of the DMN have been associated with several psychiatric conditions, including post-traumatic stress disorder, Alzheimer’s disease, autism, depression, and chronic pain [92,93,94,95,96]. DMN activation can be modulated by different interventions and physiological processes, including physical activity and exercise, sleeping, resting wakefulness, sleep deprivation [97,98,99], and age [100]. The panel regarded the DMN as an important biomarker of brain function, and given its relationship to other cognitive functions (e.g., attention), they thought it could be useful for understanding changes in operational performance. Because the DMN could be an important marker associated with multiple constructs (e.g., memory, working memory), the panel suggested it could also be an important marker for integration of these constructs and/or how modifying factors influence these constructs (e.g., sleep/wake and sleep deprivation). The DMN seems to be essential to the social understanding of others and could provide a biomarker for spaceflight-associated changes in social cognition and behavior.

3.2.2. Recommendations

The panel evaluated each specific construct to determine if a good biomarker exists that is operationally relevant for astronauts and that translates from animal models. The panel also commented on gaps in each construct that would need to be filled to produce an effective biomarker.
1. Attention. The panel identified several important behavioral markers from attention tests, primarily the psychomotor vigilance test, including increased variability in responses, decreased psychomotor speed, impulsivity, instability in performance, and lapses of attention. Several of these performance measures have been studied on the International Space Station (ISS) and in various analogs of the spaceflight environment [101,102].
2. Dual tasking. This construct overlaps BMed and sensorimotor effects and demonstrates the interconnectedness of numerous constructs relevant to operational performance. Furthermore, dual tasking is argued to be a useful behavioral method for assessing changes in cognitive reserve [103,104,105] during spaceflight and after g-transitions after landing [72,73]. Dual tasking measurements during long-duration spaceflight have identified long-term deficits in visuomotor performance and that cognitive reserve is reduced, possibly due to continued sensorimotor adaptation and stress [72]. Dual tasking measures could be useful behavioral biomarkers of how individuals adapt to the spaceflight environment.
3. Procedural memory. This form of memory [106] was not specifically identified in the two different memory constructs, but the panel felt that it is essential for operational performance and should be mentioned as a subheading under the memory construct.
4. General brain plasticity as an important biomarker of adaptability or lack of adaptability. Operational performance requires a brain that can adapt to stressors under various spaceflight conditions. As such, alterations in brain “adaptability” could be a useful biomarker indicating degradation in operational performance [107]. For example, simple adaptation to repetitive stimuli or general adaptation across multiple tasks (not only task-specific changes) might indicate how the nervous system is faring in a space-like environment (i.e., whether the brain is able to adapt to this new environment, and whether this adaptability is changing over time). This construct is important because it integrates across all measures, can be translated between rodents and humans, and clinical markers of brain damage exist that could be useful biomarkers (e.g., blood brain-derived neurotrophic factor [88]). In addition, learning and plasticity are constructs that have been tested in animal models relevant to astronaut performance (e.g., reversal learning, extinction learning), including after space radiation exposure [108,109].
5. Reversal learning is used extensively in animal models to assess cognitive flexibility and translates well between rodents and humans [110,111]. The panel suggested that reversal learning under stress or under multiple spaceflight stressors could be paired with neuroimaging (e.g., EEG) to identify factors that impair brain adaptability, and to allow translation from rodents to humans.
6. Although social processes were listed as a standalone construct, the panel noted that social interactions are important for the other constructs, and can be affected by the way individuals interact, the way the crew interacts, and how they perceive the interactions of others or the emotional states of others. This is not trivial and is not necessarily easy to assess, but it is integrated into all other constructs. These interactions highlight the need to consider how these individual states impact the group, and the need to determine if there are biomarkers of these interactions, and/or if those interactions then change the individual biomarkers.
7. Inclusion of additional constructs. When the panel took a broad view of the worksheet, they concluded that additional constructs should be added. Although many of these additional constructs were embodied within some of the other constructs, the panel thought they should be discussed as discrete constructs and how they affect operational performance.
Emotion regulation. This includes dysregulation that is subclinical, but not psychiatric disorders such as depression or anxiety, because those are included in the individual behavioral states construct.
Executive function. Assays to measure executive function were included in the attention construct, but executive function, irrespective of attention, is important to operational performance.
Risk taking/tolerance. The Balloon Analog Risk task is included within the astronauts’ Cognition Test Battery test, and the panel thought that risk taking/tolerance should be a discrete construct and not embedded within another construct. Risk taking/tolerance is also important for social interactions and group dynamics [112] and should be examined in animal models under different spaceflight stressors.
Stress. For example, astronauts’ self-reported stress ratings increased during 6-month ISS missions [102,113] and these changes could have important implications for the usefulness of biomarkers throughout the mission.
The panel identified the following gaps in knowledge:
Lack of integrated approach. The panel noted several gaps that could be addressed by first taking an integrated approach to these different constructs. For example, sleep loss or stress will most likely affect all constructs on the list. The constructs are intertwined, and many things can affect them, and for this reason, our group suggested the use of more general biomarkers, instead of construct-specific biomarkers; for example, a “general health” biomarker or a “vulnerability” biomarker that would indicate an individual’s status on some continuum of functioning within the spaceflight environment. What remains unknown is whether the biomarkers that have been identified are informative under all conditions, or if these markers will change as external stressors and internal conditions change.
Importance of stress. The panel noted several modifying factors, but stress emerged as a critical factor that probably deserves its own category on the worksheet.
Lack of sex differences or inclusion of sex. Sex needs to be considered throughout all the constructs. It was not included in any construct and could have important implications for determining what biomarkers are relevant and useful.
Inclusion of microbiome. This appears to be important to brain function, and as such, could affect the majority of the BMed constructs. A better understanding of the specific bacteria, dysbiosis, etc., and how they relate to cognition and the different performance constructs, would be useful for biomarker development.
Lack of measurements for individual differences. The panel noted the importance of inter-individual differences for these constructs and their likelihood of affecting operational performance. All individuals can be trained with the same techniques, but it is not known, nor can we currently predict, how each individual will continue to perform in the spaceflight environment. This is especially true when hazards such as radiation exposure and isolation are combined. Methods are required to measure these differences and to understand how they might impact operational performance.
Additional gaps. These include the need for better technology to quantify biomarkers during spaceflight, and greater understanding of the differences between diurnal humans and nocturnal animal models (e.g., rodents) and how this influences the biomarkers we identify and study.
Table 2. Circuits and biomarkers for behavioral medicine domains.
Table 2. Circuits and biomarkers for behavioral medicine domains.
Key
Indicator/Construct
Human Performance TestAnimal Performance TestCaveats/Notes/Related Functional Performance Tasks/Prediction of Behavioral Outcome in HumansBrain
Region
Human/NHP Neural Circuit/PathwaysRodent Neural Circuit/PathwayBiomarkers (Rodents/Humans/NHPs)Gaps/Notes
InaccessibleAccessible (Translatable to Astronauts)
MemoryMnemonic similarity test (MST) (BPSO)- this test includes Novel object recognition (NOR)1. Object in place
2. Social Recognition
3. Novel Object Recognition
4. Morris Water maze
5. Fear conditioning
6. Temporal Order
7. Mnemonic similarity test (MST) (BPSO)
8. Barnes Maze
-
Needed for recall of training, what you did minutes, hours, days ago
-
Age-related cognitive decline; mild cognitive decline (MCI); neurodegenerative conditions and dementia
-
Post-trauma or prior memory testing administration of glucose to activate hippocampus and contextual learning
Hippocampus and associated regionsExcitatory trisynaptic circuit Direct memory formation: Entorhinal cortex → Dentate gyrus → CA3 → CA1→ Entorhinal cortex V
Indirect episodic memory retrieval:
Entorhinal cortex → Dentate gyrus → CA3 → CA1→ Subiculum → Entorhinal cortex [114,115]
Excitatory trisynaptic circuitCSF: APOE, amyloid. Hippocampus: decreased BDNF, increased GFAP, inflammatory marker, synaptic marker, ArcImaging-CT, fMRI, PET, EEG, MEG, TMS scan for Default mode network activity, mismatch negative amplitude, hippocampal sharp wave ripples (rodents), no contrast fMRI for glymphatic system. Blood: APOE, amyloid, TREM levels, d-cycloserine, neurofilament light chain, BBB breakdown. Behavior -fMRI, EEG and ERPs with behavioral test and stressor. GI microbiome. NIRS/fNIRS(1) Study effects of Stress, immune system?
(2) Study the effect of Combined stressors?
(3) Sex Differences?
(4) Resource constraints for spaceflight mission—development of readily accessible and implementable technology for biomarker quantification
(5) Ethologically relevant animal tests that are relevant to human performance tests
Attention and dual tasking1. Reaction time- PVT 2. Dual Task Test (e.g., cognitive-motor, divided attention):
a. PVT b. Walking with distractors
3. Odd-ball stimulus
1. PVT
2. Attention set-shifting: 3. 5C-CPT 5 choice continuous performance test (selective attention)
  • Used operationally as go/no-go test; operational activities requiring high skill might get most affected;
  • PVT should be considered for performance under pressure with distractions
Prefrontal cortex (lateral PFC) and anterior cingulate cortexSelective attention: Visual cortex → Lateral intraparietal cortex or Middle intraparietal sulcus → prefrontal cortex [116,117]sustained attention (PVT/CPT): pedunculopontine tegmental nucleus (PPTN) → substantia nigra pars compacta (SNc) → striatum and PFC → motor control (cholinergic output) [117]Catecholamine—Noradrenaline, dopamine, mAChR and nAChRImaging: fMRI, PET, EEG scan [118,119], EEG of frontal cortex with behavioral task, pupil diameter, NIRS/fNIRS; Urine: norepinephrine, 3-methoxy-4-hydroxyphenylglycol; Plasma: monoamine oxidase, neuropeptide Y [120], Zinc, ferritin; Saliva: cortisol, Genetic and behavioral biomarkers, inflammation related systemic markers. Behavioral markers—Increase in variability of response, impulsivity, instability in performance, attention lapses, dual tasking (motor control + primary task). ECG heart rate measurement, autonomic measurements, and rest activity cycles with task performance GI microbiome; polysomnography (in sleep) and skin conductance/EDA(1) Correlation between attention, stress, immune dysfunction, and sleep.
(2) Predictive validity of operational performance in astronauts—No data on that. Also need rodent and human analogs.
(3) Access to operational task data and self-monitoring data
(4) Wearable devices for continuous monitoring of heart rate, sleep/wake cycles, rest activity and other autonomic activities without disrupting other crew activities/adding crew time.
(5) Continuous and close tracking of crew behavior.
(6) Note the bias towards response and response strategy of an individual and its dependency towards individuals’ motivation.
Working Memory1. Fractal 2 back
2. Object rotation in space
3. Spatial WM
1. Radial arm water maze-trials to criterion, latency is common across studies, can be modified for each individual animal, can be modified for test-rests 2. modified Barnes maze (operant n-back in rodents lacks stable baseline)
3. NHP: touchscreen, saccades
4. Elevated plus maze and elevated zero maze 5 Forced swim test
6. Light-dark box without elevation
7. Tail suspension test
8. Puzzle box paradigm—adaptive light/dark box with plugging the hole with various substances (mouse)
9. Unconstrained cognitive flexibility—Novel solutions to the problem (Britten test)
- Docking: Egress procedures and EVA-related;—Crew should stop with plans for completion/performance of task with possible catastrophic consequences if not performed correctly—Anxiolytic effects—Anti-depressive effects—Exploratory behavior and measure of anxiety in open areasFronto-parietal brain regions, including the prefrontal, cingulate, and parietal cortices and mediodorsal thalamus (rodent, [121])Prefrontal cortex → Visual componentPFC-hippocampus (dorsal)—visual componentRodents-microglia activation in prefrontal cortex and hippocampus, Afg3l1, Tpx2, Neuroligin-3, RB1-inducible coiled-coil 1, Mast3, Kif21a, DnaJ (Hsp40) homolog, SLIT-ROBO Rho GTPase-activating protein 2, Rasgrf1 [20]Imaging: CT, fMRI, PET, EEG, MEG, TMS scan for default mode network, Neuroimaging with adaptive N-back task, dopaminergic system, whole brain or targeted frontal, parietal, and striatal region Blood: cortisol levels, immune cytokine -chemokine levels (TNFa, IL8, IL-1ra, Tpo, VEGF, CCL2, CCL4, and CXCL5) [122]. Salivary: immune markers. Eye: blink rate for indicator of dopamine sensitivity.
GI microbiome, NIRS/fNIRS
(1) Cross-cutting issue with immune markers?
(2) Integrative approach
Learning and
plasticity
1. Sequence/procedural; 2. Eye-Head/Eye-Head-Hand adaptation tasks—(a) VOR adaptation test (not that relevant-MS) (b) Eye-Head Hand-visuomotor adaptation task
3. Whole body tasks (a) Walking with visuomotor adaptation (b) Split Belt Locomotion Test
4. Mismatch negativity. 5. Gaze control.
6. Reversal learning
1. Odor sequence learning (non-motor)
2. Eye Head and Eye Head Hand adaptation tasks—(a) Nystagmus and compensation following labyrinthectomy
(b) Rodent VOR test
3. Whole body tasks: Ladder rung walk test
4. Mismatch negativity (plasticity + perceptual learning, non-motor component, EEG measure)
5. Barnes maze
6. Extinction learning (Fear extinction).
7. Reversal learning (under stress)
8. Delayed matching to position (DMP)
9. Radial arm maze
- Adaptability is an important trait that will need to be tested with combined stressor because of the need to adapt rapidly after g transitionsPFC, hippocampus (depending on test), cerebellum, striatum (depending on motor component of the test), sensorimotor cortexTrisynaptic pathway, working memory circuitryTrisynaptic pathwayARC, cFos, synaptic markers, BDNF, MMP-9 levels, microstructure of constrained motor connectome and corticospinal tract [123]CT, fMRI, PET, EEG, MEG, TMS scan. EEG of whole brain for plasticity and adaptation with task or repetitive stimuli. Blood- BDNF. GI microbiome. NIRS/fNIRSConvergent tests-adaptable to operational tasks
Social Processes (e.g.,
Socialization,
conflict, communication, bonding)
Socialization: Self-report survey, sociometric badge
Conflict: Self-report survey, journal analysis, observational ratings
Communication: Self-report survey, communication recording analysis, observational ratings
Bonding: Observational ratings
Socialization:
1. Social fear
2. Social approach to a stranger mouse
3. Reciprocal social interactions
4. Conditioned place preference to conspecifics
5. Social recognition
6. Juvenile play
7. Nesting patterns in home cage
Conflict (Aggression)
1. Social Defeat
2. Resident intruder attack
3. Routine observation
4. Isolation-induced fighting
5. Tube test for social dominance
Communication
1. Ultrasonic Vocalizations emitted during social interactions
2. Response to vocalizations form conspecifics
3. Deposition of social olfactory pheromones
Bonding
1. Pair Bonding
2. Observation, Grooming, Inter/Intra-Social Interactions
3. Oxytocin/Vasopressin levels
Social Hierachy
1. Hierarchal testing/Social stability measurements—convergent testing like tube testing
2. Urine marking (sex should be considered)
3. Hotspot testing
Prefrontal cortex, Amygdala, Hypothalamus, striatumAggression: Sensory reception → Prefrontal Cortex → Amygdala → Hypothalamus → Periaqueductal grey (midbrain)/Ventral Tegmental area → Aggressive behavior [124]Social attachment: Olfactory cues → Vomeronasal organ (VNO)/Main olfactory epithelium (MOE) → Accessory olfactory bulb (AOB) → Amygdala → Lateral Septum→ mPFC → Nucleus accumbens Dominance: (1) Olfactory cues → VNO/MOE → AOB → Amygdala (2) Social stimuli → mPFC → Nucleus accumbens/Hypothalamus/Amygdala/Ventral tegmental area./Dorsal raphe nucleus/hippocampus Aggression: Olfactory cues → VNO/MOE → AOB/Main olfactory bulb (MOB) → Amygdala → Hypothalamus/Bed nucleus of the stria terminalis (BNST)/Hippocampus (Hippocampus→ Lateral Septum) [125]TRPc ko mice (loss of aggression) [126], reduced/loss of nNOS (increased aggression and reduced social investigation) [127], Neuroligin-3, PSD95, parvalbumin, bone hormone-osteocalcin. Radiation studies in brain—CCL2, CD206, CD163, PSD-95 in PFC, Dopamine receptor levelsCT, fMRI, PET, EEG scan Blood-Vasopressin and oxytocin levels, 5-HT, nNOS (male mice), testosterone (social regulation), cortisol, progesterone, cortisol to testosterone ratio, cortisol to oxytocin ratio. Imaging- Striatum and reward related brain regions. Psycho variables—heart rate, skin sensitivity. GI microbiome; NIRS/fNIRS; polysomnography (in sleep) and skin conductance/EDA. Behavior-eye gaze and eye tracking(1) Learning effects and sex difference
(2) Behavior of one animal/an astronaut would affect others behavior
(3) Understanding the dynamic social interaction between the crew members, psychological ownership of the space, habitat size to social interaction and any areas that need mitigation.
Individual Behavioral States (e.g., Stress,
Depression, Mood and
Anxiety)
Stress: Visual Analog Scale
Depression: Beck Depression Inventory
Mood: Profile of mood states-short form, Zung self-rating depression scale, Hamilton Rating Scale for Anxiety, Beck Scale for suicide Ideation and Beck Hopelessness Scale, Quality of Life Enjoyment & Satisfaction Questionnaire, Psychological General Well-Being Index, Pittsburgh Sleep Quality Index
Risk Tolerance: balloon analog task
Stress
1. Immobilization
Depression
1. Forced swim test
2. Inescapable shock
3. Low sucrose preference (Anhedonia)
4. Tail suspension
5. Social defeat
6. Leaned helplessness
7. Novelty-Suppressed Feeding
Mood
1. High elevated plus maze
2. High changing reinforcement schedules
3. High open field avoidance
Anxiety
1. Light-dark exploration
2. Vogel conflict test
3. Marble buying
4. Unpredictable chronic mild stress
Risk Tolerance
1. Elevated plus maze (head dips),
2. delayed reward task (impulsivity),
3. Rat gambling task.
4. Predator odor risk taking test
Prefrontal cortex (PFC), subgenual cingulate cortex (Cg25), subcortical hippocampus, nucleus
accumbens, amygdala, ventral tegmental area
5HTergic/NEergic Depression pathway: Locus coeruleus/Dorsal raphe → Amygdala/Hippocampus/Ventral tegmental area/Nucleus accumbens → Prefrontal cortex [128]5HTergic/NEergic Depression pathway: Locus coeruleus/Dorsal raphe → Amygdala/Hippocampus/Ventral tegmental area/Nucleus accumbens → Prefrontal cortex [128]Choroidal plexus: TTR (independent of radiation exposure). CSF: Glutamate, GABA, Acetylcholine, Norepinephrine, Dopamine, Serotonin, Vasopressin, Orexin. Tissue: MAPT, HTT, Presenelin-1, APP (independent of radiation exposure), glial and synaptic dysfunctionfMRI scan Blood: Glutamate, GABA, Acetylcholine, Norepinephrine, Dopamine, Serotonin, Vasopressin, Orexin, cortisol, corticosterone, Immune markers: IL6, B-cells, Cortisol, TNFa, IL4, IL5, IL-10 [122,129,130], CSF—TTR (lumbar puncture) Saliva: Cortisol; NIRS/fNIRS.(1) How individual behavioral state will impact the others in the group (cohesion, behavioral state of the group). This relates to where the crew is in the craft and who interacts with whom, crew member who isolates themselves can be a behavior issue to be detected and dealt with.
Arousal and Regulatory (e.g., sleep, circadian phase)Sleep duration and Architecture: Actigraphy and EEG 1. PVT 2. Visual analog scale towards alertness—assessing sleep quality Circadian phase: Actigraphy (not good biomarker)Sleep duration and Architecture: Actigraphy, Sleep Island, EEG
Circadian phase: Actigraphy (not a good biomarker)
Sleep duration and Architecture: Actigraphy and EEG, PVT, sleep quality Circadian phase: Actigraphy (not good biomarker)Hypothalamus, Brain stem, Spinal cord, Suprachiasmatic nucleusSleep: Retina (light) and metabolic inputs (peptidergic hormones, nutrient signals) → Retinohypothalamic tract and Arcuate nucleus → suprachiasmatic nucleus → ventral sub paraventricular zone → dorsomedial hypothalamus → ventrolateral preoptic nucleus → sleep Wakefulness: Retina (light) and metabolic inputs (peptidergic hormones, nutrient signals) → Retinohypothalamic tract and Arcuate nucleus → suprachiasmatic nucleus → lateral hypothalamic area (melanocyte concentrating hormone/orexin-producing neurons) → wakefulness [131]Circadian rhythm: Retina → Retinohypothalamic tract → Suprachiasmatic nucleus → Paraventricular nucleus → Medial forebrain bundle → Intermediolateral cell column → Superior cervical ganglion → Nervi conarii → Pineal gland (Melanocyte—Melanin secretion) [132]Brain Melatonin levels (not accurate with rodents) nocturnal animals and light cycle and when the test is conducted (light or dark cycle) Sex differenceCT, fMRI, PET, EEG, polysomnography scan 6-sulphatoxymelatonin (aMT6) collected every 2 to 8 h. over 24 to 48 h period, melatonin, Timeless, period 1–3, growth hormone (SOCS) [133]
Actiwatch (sleep quality, duration), Urine: 6-sulphatoxymelatonin (good biomarker); Melatonin in blood and saliva (not accurate), core body temperature (susceptible to masking), GI microbiome, genotype changes—per3 polymorphisms (human), Dqb10602 gene (narcolepsy), Immune markers—IL6; behavioral tests; NIRS/fNIRS
(1) Sex differences
(2) Associations between menstrual cycle phase, sleep need and circadian (major gap!) → actually, not only estrogen, but testosterone cycles too, so should consider both!
(3) Differences between nocturnal and diurnal species! Most rodents are nocturnal, but most behavioral tests on rodents (in general, not sleep specific) are done in light.
(4) New technology for measuring fluid shift and shift of brain in the cranial compartments. Tympanic membrane movement measurement
(5) Sleep duration, quality, and continuity. Need to ensure that sleep is: not disturbed, adequate for operations, at appropriate circadian time, entrained by light, exercise etc. Sleep quality is an orthogonal component to stress and emotional status.
(6) Diet and its contribution
(7) Intersubject variability
Emotional regulation Hippocampus, striatum, PFC Psychology, subclinical—Facial expression, emotional regulation. Regulation of the conflict. Executive functions.

3.3. Integrated Biomarker and Signaling-Pathway Approaches for Understanding Operational Performance (Leads: X.W. Mao, R. I. Desai)

The goal Group 3 was to use a systems-biology approach to generate lists of biomarkers and signaling pathways related to CNS circuitry and operational performance that will be important to monitor in astronauts during spaceflight and after return to Earth. To achieve this goal, the integrated approaches team (a) reviewed and identified a broad array of biomarkers of important mechanisms known from space research (i.e., what is known); this panel discussed research on biomarkers and signaling pathways in animals and humans that could be used to assess the effects of acute or long-duration exposure to spaceflight stressors on operationally relevant performance; (b) considered knowledge from other CNS-health studies that could be repurposed for assessing astronauts (e.g., aging, disorder, disease); and (c) documented open questions and research gaps in the knowledge base that connect genes and biological pathways to brain regions and neural circuits that link to operational performance (i.e., what is not known, needed experiments). Discussions are summarized below and in Table 3 and Table 4. The goal of this integrated approaches team was to provide recommendations regarding the availability, validity, and limitations of biomarkers and signaling pathways to be examined in future research.

3.3.1. Summary of Discussions

It should be emphasized at the outset that the results of this integrated approaches exercise did not reveal any biomarker (or combination thereof) that was uniformly responsive across different regions of the brain to a single or given combination of spaceflight stressors. The panel raised the following distinct, yet overlapping questions:
  • Does the literature provide any useful insight regarding if or how combined exposure to spaceflight stressors might interact to alter (additive, synergize, diminish) biomarkers and signaling pathways involved in CNS function?
  • What experiments need to be performed to inform how these combined stressors interact and affect biomarkers and signaling pathways associated with CNS function?
  • What are the challenges that need to be addressed for data collection and storage?
  • What information do we need for successful biomarker repurposing?
  • What new experiments, analysis, and techniques are needed?
  • What information about biomarkers and signaling pathways is needed to identify and implement effective spaceflight countermeasures that will minimize CNS decrements associated with the long-duration spaceflight beyond Earth’s protective magnetosphere?
Below is a summary of the key issues that were raised by the integrated approaches panel.
  • First and foremost, all group members recognized the need for standardizing certain aspects of the experimental protocol across laboratories; in particular, standardizing (a) factors related to the degree of exposure to a spaceflight stressor (e.g., space radiation (Galactic Cosmic Radiation simulation), dose, dose rate, and energy; isolation/confinement; altered gravitational levels (Mars, lunar or Earth)); (b) the type of animal models used (e.g., age, sex, strain, species; see below) and the time of tissue collection. This approach will permit meaningful comparisons and interpretations of data from different endpoints collected among investigators.
  • The panel overwhelmingly agreed that a paucity of information exists on how CNS-related neurocognitive performance is affected in laboratory animals that have been exposed to space-relevant radiation (e.g., a low-dose (<0.5Gy)/low-dose-rate of simulated galactic cosmic rays) and that such effects have not yet been systematically studied.
  • Although studies using several species (e.g., rats, mice) have provided important information about how spaceflight stressors may affect behavior and cognitive function, extrapolating data from rodents to humans is an imperfect science. Notably, the translational value of larger size animals (e.g., NHPs) used in various research domains, including neurobiological, neurobehavioral, and complex cognitive processes, has been validated and established over many decades. These successes are based on numerous factors including (1) the considerable overlap in the genetic, physiological, pharmacokinetic, neurobiology, and neurobehavioral effects in NHPs and humans; (2) the proven reliability of NHPs as subjects in long-duration (i.e., longitudinal) neurobehavioral and cognitive studies; and (3) the ability to use powerful within-subject designs that are similar to those used in human studies, which permit meaningful conclusions or inferences by evaluating all treatment effects in individuals as well as in groups. Considerations such as these suggest that NHPs are especially well-suited for ground-based study of the acute and long-term neurobehavioral effects induced by spaceflight stressors, either alone or in combination, and for translating effects to astronauts. Thus, there was considerable appreciation in the group that the use of appropriate animal models, especially targeted studies in NHPs to confirm or advance observations in rodents, should be carefully considered by NASA in future work.
  • The panel recognized that an integrated “omics” profiling strategy using technologies such as genomics, proteomics, and metabolomics is desperately needed to further expand understanding of the underlying brain systems/mechanisms that may be affected by exposure to spaceflight stressors. This multimodal approach will be highly beneficial to determine biomarker datasets of differentially expressed genes, proteins, or metabolomic/lipidomic signatures and the pathways that lead to pathological and possible degenerative changes in the brain. An omics-based molecular phenotyping approach for characterizing biosignatures associated with low-dose space radiation, simulated microgravity, and other space environmental stressors will provide a deeper understanding of the underlying mechanisms responsible for brain structure and pathophysiological changes. This approach will also provide critical information about how individual sensitivity (e.g., genetic, epigenetic, previous injury, age, and sex/gender) will influence how spaceflight stressors affect operational performance. However, as stated above, it will be critical for protocols and metadata from experiments in different laboratories to be standardized and processed on a uniform pipeline.
  • A need was identified for longitudinal studies that provide information about changes within the brain (i.e., acute to chronic). This is especially germane for determining if exposure to spaceflight stressors produces short- or long-term neurobiological (or degenerative) adaptations that affect operationally relevant behavioral and neurocognitive performance. A major complication associated with determining how the brain responds to stress insults is the latency between exposure and the expression of injury (e.g., cell loss or dysfunction). Thus, it is essential that longitudinal studies are conducted to meaningfully quantify the development and progression of the CNS injury response.
  • At present, few studies have examined the combined impact of spaceflight stressors on operational performance and/or associated neurobiological changes in the brain. Thus, it is critical that future studies use ground-based animal models that incorporate stressors that are inherent to the spaceflight environment, i.e., space-like radiation exposure and other spaceflight environment stressors including high pCO2, fluid shifts, microgravity, environmental constraints, emotional stress, and circadian misalignment/sleep deprivation. This will permit data to be extrapolated more accurately to estimate potential risks encountered by astronauts during deep space missions. Ground-based studies to examine the impact of combined spaceflight conditions and the underlying mechanism(s) of potential interaction on structural and functional deficits in the brain are very limited.
  • The panel overwhelmingly agreed that significant effort and resources are needed to develop new cutting-edge techniques to identify brain biomarkers that may indicate operationally relevant neurocognitive performance. Novel imaging techniques that provide an early detection of the subtle changes in the brain and identify the target population and biomarkers for intervention are essential. Thus, to improve knowledge about anatomical, physiological, and functional changes to the brain, especially for longitudinal evaluation, an effort is needed to develop advanced computerized tomography scan, functional magnetic resonance imaging (fMRI), positron emission tomography scan, EEG, magnetoencephalography, and transcranial magnetic stimulation scan imaging technologies.
The panel members agreed that a critical need exists to use data better and carefully from flown astronauts to evaluate the actual acute and long-term health risk of the spaceflight environment. Importantly, there was appreciation that human data could be better related to outcomes from animal studies, which may help characterize alterations in circadian rhythm and sleep, immune system, neurotransmitters, neurobiology (i.e., brain structure and function), and vasculature. If used carefully, follow-up analysis of omics, biochemistry, imaging, and a battery of behavior and neurocognitive testing will provide critical human data that may be used to evaluate the actual acute and long-term health risk of the space environment.

3.3.2. Recommendations

Table 3 highlights the major observations and points of discussion that were addressed by the integrated approaches panel. Although it is likely that exposure to combined spaceflight stressors will alter a wide range of biomarkers in different endpoints in animals and humans, ultimately, it is critical that these biomarkers are consistently and reliably linked with changes in operationally relevant behavior and neurocognitive performance. Evidence so far suggests that specific neurocognitive impairments may manifest under evolving mission scenarios (i.e., increased cognitive load) and, therefore, assessing the impact of spaceflight hazards on a wide range of operationally relevant behavioral and neurocognitive tasks is critical. Moreover, the panel suggested that NASA should explore both novel and trained paradigms with increased difficulty of determining the level of impairment. Finally, to promote translation between animal models and humans, parallel behavioral and neurocognitive testing paradigms exist between rodents ↔ NHPs ↔ humans that should be further exploited.
The panel identified the following gaps in knowledge:
  • How can data be integrated across many biology scales for CNS endpoints?
  • How can system biology approaches with new technologies—organ cultures, organs-on-a-chip made from normal human cells, integrated “omics” (genomics, proteomics, metabolomics) and cutting-edge brain imaging techniques—be used to estimate acute CNS risks to astronauts from space environment?
  • How can knowledge of space environment-induced biomarkers/pathways in neuroinflammation, blood–brain barrier function, vasculature, glia activation be integrated towards better understanding of their impact on acute pathophysiological changes in the brain and late neurodegeneration?
  • What is the likelihood of increases in the brain susceptibility to later development of neurological disorders as results of observed changes?
  • What is the relationship between neurochemical biomarkers and operationally relevant performance?
  • What are the temporal and regional differences in neurochemical biomarkers and their influence on operationally relevant performance? What is the right neurochemical balance?
  • What CNS neurotransmitter metabolites can be measured peripherally? Can wearable devices/sensors be used instead of blood?
  • Is personalized nutrition (i.e., B-vitamin supplementation) a viable SANS countermeasure?
  • Do recurring cycles of sleep deprivation affect performance/vestibular/sensorimotor changes, recovery, and biomarkers?
  • What is the role of individual susceptibility—genetic, epigenetic, previous injury, age, and sex/gender—in addressing CNS risk?
Information that is lacking includes astronaut data to monitor the level of DNA damage over time; miRNA signatures as neurodegeneration markers for acute/chronic injury; data from integrated phenotypic studies in models; and omics to identify molecular changes at the synaptic level.

4. Overall Summary and Recommendations

In total, hundreds of biomarkers have been identified and synthesized through this effort. Synthesizing across all three topical groups, the following common responses emerged as general themes:
  • Biomarkers span all levels of data from molecules to behavior.
  • Integrated stressors and integrated effects should be studied, including studies using multi-sensory approaches, for example, combined sleep and radiation exposure.
    o
    Note combined effects of HZE radiation exposure and sleep fragmentation in rodent models show dramatic effects specific to brain regions [109].
    o
    Integrated sensorimotor and cognition effects should be considered for study, e.g., olfaction and vestibular.
  • The responses themselves will have multiple downstream impacts. Treatment may not be successful following a reductionist manner.
  • Modifying factors should be identified and tracked throughout assessment, e.g., cognitive load, stress, circadian aspects, and sex, and their impacts on executive function and attention.
  • Learning and plasticity were highlighted as critical areas to assess during spaceflight to determine the astronaut’s general level of cognitive and sensorimotor adaptability.
  • Biomarkers were recommended not just for immediate predictiveness, but also for long-term predictiveness of damage (late effects that can follow the initial injury by months or longer). As an example, some omics biomarkers may precede pathologies by months.
  • Studying appropriate animal models in parallel with astronauts is extremely valuable for determining applicable constructs/responses, and to better understand the astronaut’s condition.
We hope this effort yields usable knowledge and an effective tool for HRP and the CBS Project to improve monitoring and management of astronaut cognitive and behavioral health.

Author Contributions

All co-authors played a critical role in collecting and disseminating the knowledge in this article. Meeting organizers, discussion leads, participants, and facilitators are itemized in Appendix B. All authors have read and agreed to the published version of the manuscript.

Funding

This technical interchange meeting was funded by NASA Human Research Program, through the Human Factors and Behavioral Performance Element in conjunction with Space Radiation Element and the Human Health Countermeasures Element.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the MTSBI and CBS team members and Facilitators (Appendix B) for supporting all facets of the TIM and thank the Human Health Countermeasures, Space Radiation, and Human Factors & Behavioral Performance Elements of HRP and HRP Chief Scientist’s Office for supporting this TIM. In particular, we thank David Dinges, Gregory Nelson, S. Robin Elgart, Janice Zawaski, and Scott J. Wood for expert consultation and support and Kerry George for critical manuscript review.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-HT5-hydroxytryptamine
5MTHFL-Methylfolate
8-oxo-dG8-Oxo-2′-deoxyguanosine
AchAcetylcholine
AOPAdverse Outcome Pathways
AQP-4Aquaporin-4
ARCAmes Research Center
BBBBlood Brain Barrier
BDNFBrain-derived Neurotrophic Factor
BMedBehavioral Medicine
CBSCentral Nervous System, Behavioral Medicine, and Sensorimotor
CNSCentral Nervous System
COX-2Cyclooxygenase-2
CSFCerebrospinal Fluid
DADopamine
DMNDefault Mode Network
EEGElectroencephalogram
Flt-1Fms Related Receptor Tyrosine Kinase 1
fMRIFunctional Magnetic Resonance Imaging
GFAPGlial Fibrillary Acidic Protein
GIGastrointestinal
HNE4-hydroxynonenal
HRPHuman Research Program
ICAM-1Intercellular Adhesion Molecule 1
IL-15Interleukin-15
IL-4Interleukin-4
ISSInternational Space Station
JSCJohnson Space Center
MAPKMitogen-activated Protein Kinase
MDAMalondialdehyde
MMP-9Matrix Metallopeptidase 9
MMPsMatrix metalloproteinase
MRIMagnetic Resonance Imaging
MTSBIModel Translation & Space Biology Integration
NFKbNuclear Factor kappa B
NHPNon-human Primates
NIRSNear-Infrared Spectroscopy
PIPrincipal Investigator
ROSReactive Oxygen Species
S100bS100 Calcium Binding Protein B
SMSensorimotor
TIMTechnical Interchange Meeting
TNFTumor Necrosis Factor
TREMTriggering Receptor Expressed on Myeloid cells
TRRTransthyretin
UCSFUniversity of California San Francisco
USRAUniversities Space Research Association
USUHSUniformed Services University of the Health Sciences
VCAM-1Vascular Cell Adhesion Molecule 1
VORVestibular-ocular Reflex
YKL-40Chitinase-3-like protein 1
ZO-1Zonula occludens-1

Appendix A. Agenda of Meeting

A NASA translational working group TIM titled Circuits and Biomarkers of the Central Nervous System Relating to Astronaut Performance (Biomarker TIM) was held virtually between 21–25 September 2020, and was supported by the NASA HRP’s Human Factors and Behavioral Performance Element in conjunction with Space Radiation Element and the Human Health Countermeasures Element. The goals of this Biomarker TIM were to (1) identify relevant brain regions, neural circuits, functions, and associated biomarkers that relate to operationally relevant performance and (2) identify any critical needs for new biomarker knowledge (“gaps”) that can be filled by additional focused and translational animal experiments that include a plausible pathway toward eventual biomarker validation in humans.
Deliverables addressing these goals may ultimately inform countermeasure strategies to maintain performance standards and identify performance limits for astronauts. To address the goals, 22 extramural experts from 19 academic institutions and 26 intramural experts from various NASA centers contributed to 15 talks reviewing findings from biomarker research on animals and humans in response to terrestrial and spaceflight stressors, and then participated in virtual thematic breakout sessions to systematically and qualitatively review biomarkers and associated brain circuits for 30 cognitive or behavioral constructs or physiological responses. The topics of the breakout sessions were sensorimotor influences (Group 1), behavioral medicine influences (Group 2), and integrated approaches to understanding operationally relevant performance (Group 3), and respective behavioral constructs listed in Table A1. Before the TIM, a portfolio of documents and scientific literature was shared with participants to frame the workshop and help the participants prepare.
Table A1. List of behavioral constructs for discussion groups.
Table A1. List of behavioral constructs for discussion groups.
SensorimotorBehavioral MedicineIntegrated Approaches:
Physiological Responses
  • Visual
  • Memory
  • Neuroinflammation
  • Spatial Orientation
  • Attention and Dual Tasking
  • Neurotransmitters
  • Vestibular
  • Executive Function
  • One-Carbon Metabolism
  • Proprioception
  • Working Memory
  • Oxidative Stress
  • Hearing
  • Learning and Plasticity
  • Synaptic Plasticity and Neurotrophic Factors
  • Motion Sickness
  • Social Processes
  • Vestibular and Sensorimotor alterations
  • Smell and Taste
  • Individual Behavioral States
  • DNA Damage
  • Postural Control and Balance
  • Arousal and Regulatory
  • Blood Brain Barrier Permeability
  • Locomotion
  • Emotional Regulation
  • Vasculature
  • Fine Motor Control
  • Risk Taking/Tolerance
  • miRNA Regulation
  • Perception
  • Stress
  • Circadian Phase
  • Gaze
  • Neuronal Damage
  • Pain

Appendix B. Organizers & Participants

Lead Organizers
Joshua Alwood, PhD, NASA ARC
Ajitkumar Mulavara, PhD, KBR
Organizer Team
CBS/Johnson Space Center
Jayati Roy Choudhury, PhD, MEI
Kerry George, KBR
Jimmy Zaid, MEI
MTSBI/NASA Ames Research Center
Jared Broddrick, PhD
Egle Cekanaviciute, PhD
Janani Iyer, PhD, USRA
Laura Lewis
Siddhita D. Mhatre, PhD, KBR
April Ronca, PhD
Marianne Sowa, PhD
Participants
Group 1: Sensorimotor Influences on Operational Performance
Leads
Susanna Rosi, PhD, UCSF
Mark Shelhamer, ScD, Johns Hopkins University
Facilitator
Scott J. Wood, PhD, NASA JSC/Azusa Pacific University
Expert Observers
Millard Reschke, PhD, NASA JSC
Meghan Downs, PhD, NASA JSC
Sudhakar Rajulu, PhD, NASA JSC
Jeffrey Somers, PhD, NASA JSC
Science Team
Afshin Beheshti, PhD, KBR/Broad Institute
Kathleen Cullen, PhD, Johns Hopkins University
Sandeep Robert Datta, MD, PhD, Harvard University
Lisa Giocomo, PhD, Stanford University
James Lackner, PhD, Brandeis University
Gregory Nelson, PhD, Loma Linda University
Group 2: Behavioral Medicine Influences on Operational Performance (includes Cognition)
Leads
Catherine Davis-Takács, PhD, USUHS
David Dinges, PhD, University of Pennsylvania
Facilitator
Pete Roma, PhD, KBR
Expert Observers
Gillés Clement, PhD, KBR
Tim Macaulay, PhD, KBR
Sara Whiting, PhD, KBR
Erin Flynn-Evans, PhD, MPH, NASA ARC
Gary Strangman, PhD, Massachusetts General Hospital/Harvard Medical School
Science Team
Amelia Eisch, PhD, University of Pennsylvania
Thomas Jhou, PhD, The Medical University of South Carolina
Rachel Seidler, PhD, University of Florida
Steven Siegel, MD, PhD, University of Southern California
Andy Wyrobek, PhD, Lawrence Berkeley National Laboratory
Group 3: Integrated biomarkers and pathways relating to Operational Performance
Leads
Vivien Mao, MD, Loma Linda University
Rajeev I. Desai, PhD, Harvard Medical School/McLean Hospital
Facilitators
Ajitkumar Mulavara, PhD, KBR
Joshua Alwood, PhD, NASA ARC
Expert Observers
Honglu Wu, PhD, NASA JSC
Lisa Carnell, PhD, NASA Langley Research Center
Satish Mehta, PhD, KBR
Sara Zwart, PhD, KBR
Science Team
Janet Baulch, PhD, University of California, Irvine
Sylvain Costes, PhD, NASA ARC
Brian Crucian, PhD, NASA JSC
Daniel Geschwind, MD, PhD, University of California, Los Angeles
Steven Lockley, PhD, Harvard University
Scott M. Smith, PhD, NASA JSC
Patrick Stover, PhD, Texas A&M University
Donna Wilcock, PhD, University of Kentucky

References

  1. Kiffer, F.; Alexander, T.; Anderson, J.E.; Groves, T.; Wang, J.; Sridharan, V.; Boerma, M.; Allen, A.R. Late Effects of 16O-Particle Radiation on Female Social and Cognitive Behavior and Hippocampal Physiology. Radiat. Res. 2019, 191, 278–294. [Google Scholar] [CrossRef] [PubMed]
  2. NASA; CBS. Integrated Research Plan to Assess the Combined Effects of Space Radiation, Altered Gravity, and Isolation and Confinement on Crew Health and Performance: Problem Statement; NASA: Washington, DC, USA, 2019. [Google Scholar]
  3. Tu, D.; Basner, M.; Smith, M.G.; Williams, E.S.; Ryder, V.E.; Romoser, A.A.; Ecker, A.; Aeschbach, D.; Stahn, A.C.; Jones, C.W.; et al. Dynamic ensemble prediction of cognitive performance in spaceflight. Sci. Rep. 2022, 12, 11032. [Google Scholar] [CrossRef] [PubMed]
  4. Grandjean, P. Paracelsus Revisited: The Dose Concept in a Complex World. Basic. Clin. Pharmacol. Toxicol. 2016, 119, 126–132. [Google Scholar] [CrossRef] [PubMed]
  5. Carnell, L.S. Spaceflight medical countermeasures: A strategic approach for mitigating effects from solar particle events. Int. J. Radiat. Biol. 2021, 97, S125–S131. [Google Scholar] [CrossRef]
  6. Alfano, C.; Farina, L.; Petti, M. Networks as Biomarkers: Uses and Purposes. Genes 2023, 14, 429. [Google Scholar] [CrossRef]
  7. Institute of Medicine. Evaluation of Biomarkers and Surrogate Endpoints in Chronic Disease; Micheel, C.M., Ball, J.R., Eds.; The National Academies Press: Washington, DC, USA, 2010; ISBN 978-0-309-15129-0. [Google Scholar]
  8. Simonsen, L.C.; Slaba, T.C.; Guida, P.; Rusek, A. NASA’s first ground-based Galactic Cosmic Ray Simulator: Enabling a new era in space radiobiology research. PLoS Biol. 2020, 18, e3000669. [Google Scholar] [CrossRef]
  9. Cullen, K.E. Vestibular processing during natural self-motion: Implications for perception and action. Nat. Rev. Neurosci. 2019, 20, 346–363. [Google Scholar] [CrossRef]
  10. Mulavara, A.P.; Peters, B.T.; Miller, C.A.; Kofman, I.S.; Reschke, M.F.; Taylor, L.C.; Lawrence, E.L.; Wood, S.J.; Laurie, S.S.; Lee, S.M.C.; et al. Physiological and Functional Alterations after Spaceflight and Bed Rest. Med. Sci. Sports Exerc. 2018, 50, 1961–1980. [Google Scholar] [CrossRef]
  11. Diamond, S.G.; Markham, C.H. Prediction of space motion sickness susceptibility by disconjugate eye torsion in parabolic flight. Aviat. Space. Environ. Med. 1991, 62, 201–205. [Google Scholar]
  12. Markham, C.H.; Diamond, S.G. A predictive test for space motion sickness. J. Vestib. Res. 1993, 3, 289–295. [Google Scholar] [CrossRef]
  13. Bachatene, L.; Bharmauria, V.; Molotchnikoff, S. Adaptation and Neuronal Network in Visual Cortex. In Visual Cortex—Current Status and Perspectives; InTech: London, UK, 2012. [Google Scholar]
  14. Fang, Q.; Chou, X.L.; Peng, B.; Zhong, W.; Zhang, L.I.; Tao, H.W. A Differential Circuit via Retino-Colliculo-Pulvinar Pathway Enhances Feature Selectivity in Visual Cortex through Surround Suppression. Neuron 2020, 105, 355–369.e6. [Google Scholar] [CrossRef] [PubMed]
  15. Tamhane, M.; Cabrera-Ghayouri, S.; Abelian, G.; Viswanath, V. Review of Biomarkers in Ocular Matrices: Challenges and Opportunities. Pharm. Res. 2019, 36, 40. [Google Scholar] [CrossRef] [PubMed]
  16. Hutton, S.B. Cognitive control of saccadic eye movements. Brain Cogn. 2008, 68, 327–340. [Google Scholar] [CrossRef] [PubMed]
  17. Bussey, T.J.; Padain, T.L.; Skillings, E.A.; Winters, B.D.; Morton, A.J.; Saksida, L.M. The touchscreen cognitive testing method for rodents: How to get the best out of your rat. Learn. Mem. 2008, 15, 516–523. [Google Scholar] [CrossRef]
  18. Vesuna, S.; Kauvar, I.V.; Richman, E.; Gore, F.; Oskotsky, T.; Sava-Segal, C.; Luo, L.; Malenka, R.C.; Henderson, J.M.; Nuyujukian, P.; et al. Deep posteromedial cortical rhythm in dissociation. Nature 2020, 586, 87–94. [Google Scholar] [CrossRef]
  19. Aitken, P.; Zheng, Y.; Smith, P.F. The modulation of hippocampal theta rhythm by the vestibular system. J. Neurophysiol. 2018, 119, 548–562. [Google Scholar] [CrossRef]
  20. Dutta, S.M.; Hadley, M.M.; Peterman, S.; Jewell, J.S.; Duncan, V.D.; Britten, R.A. Quantitative Proteomic Analysis of the Hippocampus of Rats with GCR-Induced Spatial Memory Impairment. Radiat. Res. 2018, 189, 136–145. [Google Scholar] [CrossRef]
  21. Tamura, A.; Iwamoto, T.; Ozaki, H.; Kimura, M.; Tsujimoto, Y.; Wada, Y. Wrist-worn electrodermal activity as a novel neurophysiological biomarker of autonomic symptoms in spatial disorientation. Front. Neurol. 2018, 9, 1056. [Google Scholar] [CrossRef]
  22. Smith, P.F.; Horii, A.; Russell, N.; Bilkey, D.K.; Zheng, Y.; Liu, P.; Kerr, D.S.; Darlington, C.L. The effects of vestibular lesions on hippocampal function in rats. Prog. Neurobiol. 2005, 75, 391–405. [Google Scholar] [CrossRef]
  23. Dumas, R.; Mitton, D.; Laporte, S.; Dubousset, J.; Steib, J.P.; Lavaste, F.; Skalli, W. Explicit calibration method and specific device designed for stereoradiography. J. Biomech. 2003, 36, 827–834. [Google Scholar] [CrossRef]
  24. Uno, Y.; Horii, A.; Uno, A.; Fuse, Y.; Fukushima, M.; Doi, K.; Kubo, T. Quantitative changes in mRNA expression of glutamate receptors in the rat peripheral and central vestibular systems following hypergravity. J. Neurochem. 2002, 81, 1308–1317. [Google Scholar] [CrossRef] [PubMed]
  25. Cohen, B.; Yakushin, S.B.; Holstein, G.R.; Dai, M.; Tomko, D.L.; Badakva, A.M.; Kozlovskaya, I.B. Vestibular Experiments in Space. Adv. Space Biol. Med. 2005, 10, 105–164. [Google Scholar] [CrossRef] [PubMed]
  26. Krasnov, I.; D’iachkova, L. Ultrastructure of the cortex of the cerebellar nodulus in rats after a flight on the biosatellite Kosmos-1514. Kosm. Biol. Aviakosmicheskaia Meditsina 1986, 20, 45–48. Available online: https://pubmed.ncbi.nlm.nih.gov/3784524/ (accessed on 24 August 2023).
  27. Krasnov, I.; Dyachkova, L. The effect of space flight on the ultrastructure of the rat cerebellar and hemisphere cortex. Physiologist 1990, 33 (Suppl. 1), S29–S30. Available online: https://pubmed.ncbi.nlm.nih.gov/2371337/ (accessed on 24 August 2023).
  28. Holstein, G.; Kukielka, E.; Martinelli, G. Anatomical observations of the rat cerebellar nodulus after 24 h of spaceflight. J. Gravitat. Physiol. 1999, 6, P47–P50. [Google Scholar]
  29. Sultemeier, D.R.; Choy, K.R.; Schweizer, F.E.; Hoffman, L.F. Spaceflight-induced synaptic modifications within hair cells of the mammalian utricle. J. Neurophysiol. 2017, 117, 2163–2178. [Google Scholar] [CrossRef] [PubMed]
  30. Ross, M.D. Morphological changes in rat vestibular system following weightlessness. J. Vestib. Res. 1993, 3, 241–251. Available online: http://www.ncbi.nlm.nih.gov/pubmed/7903895 (accessed on 24 August 2023).
  31. Ross, M.D. A spaceflight study of synaptic plasticity in adult rat vestibular maculas. Acta Otolaryngol. Suppl. 1994, 516, 3–14. Available online: http://www.ncbi.nlm.nih.gov/pubmed/7976320 (accessed on 24 August 2023).
  32. Ross, M.D. Changes in ribbon synapses and rough endoplasmic reticulum of rat utricular macular hair cells in weightlessness. Acta Otolaryngol. 2000, 120, 490–499. [Google Scholar] [CrossRef] [PubMed]
  33. Ross, M.D.; Varelas, J. Synaptic ribbon plasticity, ribbon size and potential regulatory mechanisms in utricular and saccular maculae. J. Vestib. Res. 2005, 15, 17–30. Available online: http://www.ncbi.nlm.nih.gov/pubmed/15908737 (accessed on 24 August 2023).
  34. Zhang, L.L.; Wang, J.Q.; Qi, R.R.; Pan, L.L.; Li, M.; Cai, Y.L. Motion Sickness: Current Knowledge and Recent Advance. CNS Neurosci. Ther. 2016, 22, 15–24. [Google Scholar] [CrossRef]
  35. Ng, K.; Chua, Y.; Ban, V.F.; Gresty, M.; Coen, S.; Sanger, G.; Williams, S.; Barker, G.; Andrews, P.; Aziz, Q. Identifying human biomarkers of nausea for refining animal studies on emesis. Gut 2011, 60, A162. [Google Scholar] [CrossRef]
  36. Sohn, J.H.; Kim, C.H.; Lee, S.H.; Kim, J.H.; Lee, J.J. Diagnostic Value of Serum Biomarkers for Differentiating Central and Peripheral Causes of Acute Vertigo. Front. Med. 2020, 7, 84. [Google Scholar] [CrossRef]
  37. Wu, Y.; Han, W.; Yan, W.; Lu, X.; Zhou, M.; Li, L.; Guan, Q.; Fan, Z. Increased Otolin-1 in Serum as a Potential Biomarker for Idiopathic Benign Paroxysmal Positional Vertigo Episodes. Front. Neurol. 2020, 11, 367. [Google Scholar] [CrossRef]
  38. Hamann, K.F. Vibration-Induced Nystagmus: A Biomarker for Vestibular Deficits—A Synopsis. ORL 2017, 79, 112–120. [Google Scholar] [CrossRef]
  39. Osborne, D.; Theodorou, M.; Lee, H.; Ranger, M.; Hedley-Lewis, M.; Shawkat, F.; Harris, C.M.; Self, J.E. Supranuclear eye movements and nystagmus in children: A review of the literature and guide to clinical examination, interpretation of findings and age-appropriate norms. Eye 2019, 33, 261–273. [Google Scholar] [CrossRef] [PubMed]
  40. Ryczko, D.; Dubuc, R. Dopamine and the Brainstem Locomotor Networks: From Lamprey to Human. Front. Neurosci. 2017, 11, 295. [Google Scholar] [CrossRef] [PubMed]
  41. Goulding, M. Circuits controlling vertebrate locomotion: Moving in a new direction. Nat. Rev. Neurosci. 2009, 10, 507–518. [Google Scholar] [CrossRef] [PubMed]
  42. Takakusaki, K. Functional Neuroanatomy for Posture and Gait Control. J. Mov. Disord. 2017, 10, 1–17. [Google Scholar] [CrossRef] [PubMed]
  43. Marouane, E.; Rastoldo, G.; El Mahmoudi, N.; Péricat, D.; Chabbert, C.; Artzner, V.; Tighilet, B. Identification of New Biomarkers of Posturo-Locomotor Instability in a Rodent Model of Vestibular Pathology. Front. Neurol. 2020, 11, 470. [Google Scholar] [CrossRef]
  44. Gordon, C.R.; Shupak, A. Prevention and treatment of motion sickness. Am. Fam. Physician 2014, 90, 41–46. Available online: http://www.ncbi.nlm.nih.gov/pubmed/25077505 (accessed on 24 August 2023).
  45. Mo, F.-F.; Qin, H.-H.; Wang, X.-L.; Shen, Z.-L.; Xu, Z.; Wang, K.-H.; Cai, Y.-L.; Li, M. Acute hyperglycemia is related to gastrointestinal symptoms in motion sickness: An experimental study. Physiol. Behav. 2012, 105, 394–401. [Google Scholar] [CrossRef]
  46. Sato, F.; Uemura, Y.; Kanno, C.; Tsutsumi, Y.; Tomita, A.; Oka, A.; Kato, T.; Uchino, K.; Murakami, J.; Haque, T.; et al. Thalamo-insular pathway conveying orofacial muscle proprioception in the rat. Neuroscience 2017, 365, 158–178. [Google Scholar] [CrossRef]
  47. Ranade, S.S.; Woo, S.-H.; Dubin, A.E.; Moshourab, R.A.; Wetzel, C.; Petrus, M.; Mathur, J.; Bégay, V.; Coste, B.; Mainquist, J.; et al. Piezo2 is the major transducer of mechanical forces for touch sensation in mice. Nature 2014, 516, 121–125. [Google Scholar] [CrossRef]
  48. Oliveira Fernandes, M.; Tourtellotte, W.G. Egr3-dependent muscle spindle stretch receptor intrafusal muscle fiber differentiation and fusimotor innervation homeostasis. J. Neurosci. 2015, 35, 5566–5578. [Google Scholar] [CrossRef]
  49. Goble, D.J.; Coxon, J.P.; Van Impe, A.; Geurts, M.; Van Hecke, W.; Sunaert, S.; Wenderoth, N.; Swinnen, S.P. The neural basis of central proprioceptive processing in older versus younger adults: An important sensory role for right putamen. Hum. Brain Mapp. 2012, 33, 895–908. [Google Scholar] [CrossRef] [PubMed]
  50. Ueno, M.; Nakamura, Y.; Li, J.; Gu, Z.; Niehaus, J.; Maezawa, M.; Crone, S.A.; Goulding, M.; Baccei, M.L.; Yoshida, Y. Corticospinal Circuits from the Sensory and Motor Cortices Differentially Regulate Skilled Movements through Distinct Spinal Interneurons. Cell Rep. 2018, 23, 1286–1300.e7. [Google Scholar] [CrossRef] [PubMed]
  51. Pradhan, S.D.; Brewer, B.R.; Carvell, G.E.; Sparto, P.J.; Delitto, A.; Matsuoka, Y. Assessment of fine motor control in individuals with Parkinson’s disease using force tracking with a secondary cognitive task. J. Neurol. Phys. Ther. 2010, 34, 32–40. [Google Scholar] [CrossRef] [PubMed]
  52. Heys, J.G.; Wu, Z.; Allegra Mascaro, A.L.; Dombeck, D.A. Inactivation of the Medial Entorhinal Cortex Selectively Disrupts Learning of Interval Timing. Cell Rep. 2020, 32, 108163. [Google Scholar] [CrossRef]
  53. Milleret, C.; Bui Quoc, E. Beyond Rehabilitation of Acuity, Ocular Alignment, and Binocularity in Infantile Strabismus. Front. Syst. Neurosci. 2018, 12, 29. [Google Scholar] [CrossRef]
  54. Steeds, C.E. The anatomy and physiology of pain. Surgery 2016, 34, 55–59. [Google Scholar] [CrossRef]
  55. Zjawiony, J.K.; Machado, A.S.; Menegatti, R.; Ghedini, P.C.; Costa, E.A.; Pedrino, G.R.; Lukas, S.E.; Franco, O.L.; Silva, O.N.; Fajemiroye, J.O. Cutting-Edge Search for Safer Opioid Pain Relief: Retrospective Review of Salvinorin A and Its Analogs. Front. Psychiatry 2019, 10, 157. [Google Scholar] [CrossRef]
  56. Graham, B.; Callister, R. Pain. In The Mouse Nervous System; Elsevier: Amsterdam, The Netherlands, 2012; pp. 589–606. [Google Scholar]
  57. Pastoriza, L.N.; Morrow, T.J.; Casey, K.L. Medial frontal cortex lesions selectively attenuate the hot plate response: Possible nocifensive apraxia in the rat. Pain 1996, 64, 11–17. [Google Scholar] [CrossRef]
  58. Niculescu, A.B.; Le-Niculescu, H.; Levey, D.F.; Roseberry, K.; Soe, K.C.; Rogers, J.; Khan, F.; Jones, T.; Judd, S.; McCormick, M.A.; et al. Towards precision medicine for pain: Diagnostic biomarkers and repurposed drugs. Mol. Psychiatry 2019, 24, 501–522. [Google Scholar] [CrossRef]
  59. Luchting, B.; Hinske, L.C.G.; Rachinger-Adam, B.; Celi, L.A.; Kreth, S.; Azad, S.C. Soluble intercellular adhesion molecule-1: A potential biomarker for pain intensity in chronic pain patients. Biomark. Med. 2017, 11, 265–276. [Google Scholar] [CrossRef]
  60. Boissoneault, J.; Sevel, L.; Letzen, J.; Robinson, M.; Staud, R. Biomarkers for Musculoskeletal Pain Conditions: Use of Brain Imaging and Machine Learning. Curr. Rheumatol. Rep. 2017, 19, 5. [Google Scholar] [CrossRef]
  61. Cowen, R.; Stasiowska, M.K.; Laycock, H.; Bantel, C. Assessing pain objectively: The use of physiological markers. Anaesthesia 2015, 70, 828–847. [Google Scholar] [CrossRef]
  62. Sakano, H. Developmental regulation of olfactory circuit formation in mice. Dev. Growth Differ. 2020, 62, 199–213. [Google Scholar] [CrossRef]
  63. Latchney, S.E.; Rivera, P.D.; Mao, X.W.; Ferguson, V.L.; Bateman, T.A.; Stodieck, L.S.; Nelson, G.A.; Eisch, A.J. The effect of spaceflight on mouse olfactory bulb volume, neurogenesis, and cell death indicates the protective effect of novel environment. J. Appl. Physiol. 2014, 116, 1593–1604. [Google Scholar] [CrossRef]
  64. Henkin, R.I.; Hosein, S.; Stateman, W.A.; Knöppel, A.B.; Abdelmeguid, M. Improved smell function with increased nasal mucus sonic hedgehog in hyposmic patients after treatment with oral theophylline. Am. J. Otolaryngol. 2017, 38, 143–147. [Google Scholar] [CrossRef]
  65. Henkin, R.I.; Knöppel, A.B.; Abdelmeguid, M.; Stateman, W.A.; Hosein, S. Sonic hedgehog is present in parotid saliva and is decreased in patients with taste dysfunction. J. Oral. Pathol. Med. 2017, 46, 829–833. [Google Scholar] [CrossRef] [PubMed]
  66. Mueller, T. What is the Thalamus in Zebrafish? Front. Neurosci. 2012, 6, 64. [Google Scholar] [CrossRef] [PubMed]
  67. Sun, C.; Xuan, X.; Zhou, Z.; Yuan, Y.; Xue, F. A Preliminary Report on the Investigation of Prestin as a Biomarker for Idiopathic Sudden Sensorineural Hearing Loss. Ear. Nose. Throat J. 2020, 99, 528–531. [Google Scholar] [CrossRef] [PubMed]
  68. Parham, K.; Dyhrfjeld-Johnsen, J. Outer Hair Cell Molecular Protein, Prestin, as a Serum Biomarker for Hearing Loss: Proof of Concept. Otol. Neurotol. 2016, 37, 1217–1222. [Google Scholar] [CrossRef]
  69. Basner, M.; Moore, T.M.; Hermosillo, E.; Nasrini, J.; Dinges, D.F.; Gur, R.C.; Johannes, B. Cognition Test Battery Performance Is Associated with Simulated 6df Spacecraft Docking Performance. Aerosp. Med. Hum. Perform. 2020, 91, 861–867. [Google Scholar] [CrossRef]
  70. Van Dongen, H.P.; Baynard, M.D.; Maislin, G.; Dinges, D.F. Systematic interindividual differences in neurobehavioral impairment from sleep loss: Evidence of trait-like differential vulnerability. Sleep 2004, 27, 423–433. [Google Scholar] [PubMed]
  71. Tkachenko, O.; Dinges, D.F. Interindividual variability in neurobehavioral response to sleep loss: A comprehensive review. Neurosci. Biobehav. Rev. 2018, 89, 29–48. [Google Scholar] [CrossRef] [PubMed]
  72. Bock, O.; Weigelt, C.; Bloomberg, J.J. Cognitive demand of human sensorimotor performance during an extended space mission: A dual-task study. Aviat. Sp. Env. Med. 2010, 81, 819–824. [Google Scholar] [CrossRef] [PubMed]
  73. Moore, S.T.; Dilda, V.; Morris, T.R.; Yungher, D.A.; MacDougall, H.G.; Wood, S.J. Long-duration spaceflight adversely affects post-landing operator proficiency. Sci. Rep. 2019, 9, 2677. [Google Scholar] [CrossRef]
  74. Featherstone, R.E.; Melnychenko, O.; Siegel, S.J. Mismatch negativity in preclinical models of schizophrenia. Schizophr. Res. 2018, 191, 35–42. [Google Scholar] [CrossRef] [PubMed]
  75. Gonzalez-Castillo, J.; Hoy, C.W.; Handwerker, D.A.; Robinson, M.E.; Buchanan, L.C.; Saad, Z.S.; Bandettini, P.A. Tracking ongoing cognition in individuals using brief, whole-brain functional connectivity patterns. Proc. Natl. Acad. Sci. USA 2015, 112, 8762–8767. [Google Scholar] [CrossRef]
  76. Edlow, B.L.; Chatelle, C.; Spencer, C.A.; Chu, C.J.; Bodien, Y.G.; O’Connor, K.L.; Hirschberg, R.E.; Hochberg, L.R.; Giacino, J.T.; Rosenthal, E.S.; et al. Early detection of consciousness in patients with acute severe traumatic brain injury. Brain 2017, 140, 2399–2414. [Google Scholar] [CrossRef]
  77. Haufe, S.; DeGuzman, P.; Henin, S.; Arcaro, M.; Honey, C.J.; Hasson, U.; Parra, L.C. Elucidating relations between fMRI, ECoG, and EEG through a common natural stimulus. Neuroimage 2018, 179, 79–91. [Google Scholar] [CrossRef]
  78. Itthipuripat, S.; Sprague, T.C.; Serences, J.T. Functional MRI and EEG Index Complementary Attentional Modulations. J. Neurosci. 2019, 39, 6162–6179. [Google Scholar] [CrossRef]
  79. Nguyen, T.; Zhou, T.; Potter, T.; Zou, L.; Zhang, Y. The Cortical Network of Emotion Regulation: Insights From Advanced EEG-fMRI Integration Analysis. IEEE Trans. Med. Imaging 2019, 38, 2423–2433. [Google Scholar] [CrossRef]
  80. Waser, M.; Benke, T.; Dal-Bianco, P.; Garn, H.; Mosbacher, J.A.; Ransmayr, G.; Schmidt, R.; Seiler, S.; Sorensen, H.B.D.; Jennum, P.J. Neuroimaging markers of global cognition in early Alzheimer’s disease: A magnetic resonance imaging-electroencephalography study. Brain Behav. 2019, 9, e01197. [Google Scholar] [CrossRef] [PubMed]
  81. Thatcher, R.; McAlaster, R.; Camacho, M.; Salazar, A.; Biver, C. Biophysical linkage between MRI and EEG amplitude in closed head injury. Neuroimage 1998, 7, 352–367. [Google Scholar] [CrossRef] [PubMed]
  82. Tien, Y.T.; Lee, W.J.; Liao, Y.C.; Wang, W.F.; Jhang, K.M.; Wang, S.J.; Fuh, J.L. Plasma Transthyretin as a Predictor of Amnestic Mild Cognitive Impairment Conversion to Dementia. Sci. Rep. 2019, 9, 18691. [Google Scholar] [CrossRef] [PubMed]
  83. Dabrowski, W.; Siwicka-Gieroba, D.; Kotfis, K.; Zaid, S.; Terpilowska, S.; Robba, C.; Siwicki, A.K. The brain-gut axis—Where are we now and how can we modulate these connections? Curr. Neuropharmacol. 2020, 19, 1164–1177. [Google Scholar] [CrossRef] [PubMed]
  84. Hattori, N.; Yamashiro, Y. The Gut-Brain Axis. Ann. Nutr. Metab. 2021, 77 (Suppl. 2), 1–3. [Google Scholar] [CrossRef]
  85. Sun, M.; Ma, K.; Wen, J.; Wang, G.; Zhang, C.; Li, Q.; Bao, X.; Wang, H. A Review of the Brain-Gut-Microbiome Axis and the Potential Role of Microbiota in Alzheimer’s Disease. J. Alzheimers Dis. 2020, 73, 849–865. [Google Scholar] [CrossRef]
  86. LaPelusa, M.; Donoviel, D.; Branzini, S.E.; Carlson, P.E., Jr.; Culler, S.; Cheema, A.K.; Kaddurah-Daouk, R.; Kelly, D.; de Cremoux, I.; Knight, R.; et al. Microbiome for Mars: Surveying microbiome connections to healthcare with implications for long-duration human spaceflight, virtual workshop, July 13, 2020. Microbiome 2021, 9, 2. [Google Scholar] [CrossRef]
  87. Voorhies, A.A.; Mark Ott, C.; Mehta, S.; Pierson, D.L.; Crucian, B.E.; Feiveson, A.; Oubre, C.M.; Torralba, M.; Moncera, K.; Zhang, Y.; et al. Study of the impact of long-duration space missions at the International Space Station on the astronaut microbiome. Sci. Rep. 2019, 9, 9911. [Google Scholar] [CrossRef]
  88. Stahn, A.C.; Gunga, H.C.; Kohlberg, E.; Gallinat, J.; Dinges, D.F.; Kühn, S. Brain Changes in Response to Long Antarctic Expeditions. N. Engl. J. Med. 2019, 381, 2273–2275. [Google Scholar] [CrossRef]
  89. Raichle, M.E. The brain’s default mode network. Annu. Rev. Neurosci. 2015, 38, 433–447. [Google Scholar] [CrossRef]
  90. Raichle, M.E.; MacLeod, A.M.; Snyder, A.Z.; Powers, W.J.; Gusnard, D.A.; Shulman, G.L. A default mode of brain function. Proc. Natl. Acad. Sci. USA 2001, 98, 676–682. [Google Scholar] [CrossRef]
  91. Buckner, R.L.; Andrews-Hanna, J.R.; Schacter, D.L. The brain’s default network: Anatomy, function, and relevance to disease. Ann. N. Y. Acad. Sci. 2008, 1124, 1–38. [Google Scholar] [CrossRef] [PubMed]
  92. Baliki, M.N.; Geha, P.Y.; Apkarian, A.V.; Chialvo, D.R. Beyond feeling: Chronic pain hurts the brain, disrupting the default-mode network dynamics. J. Neurosci. 2008, 28, 1398–1403. [Google Scholar] [CrossRef] [PubMed]
  93. Broyd, S.J.; Demanuele, C.; Debener, S.; Helps, S.K.; James, C.J.; Sonuga-Barke, E.J. Default-mode brain dysfunction in mental disorders: A systematic review. Neurosci. Biobehav. Rev. 2009, 33, 279–296. [Google Scholar] [CrossRef] [PubMed]
  94. Greicius, M.D.; Srivastava, G.; Reiss, A.L.; Menon, V. Default-mode network activity distinguishes Alzheimer’s disease from healthy aging: Evidence from functional MRI. Proc. Natl. Acad. Sci. USA 2004, 101, 4637–4642. [Google Scholar] [CrossRef]
  95. Kucyi, A.; Moayedi, M.; Weissman-Fogel, I.; Goldberg, M.B.; Freeman, B.V.; Tenenbaum, H.C.; Davis, K.D. Enhanced medial prefrontal-default mode network functional connectivity in chronic pain and its association with pain rumination. J. Neurosci. 2014, 34, 3969–3975. [Google Scholar] [CrossRef]
  96. Whitfield-Gabrieli, S.; Ford, J.M. Default mode network activity and connectivity in psychopathology. Annu. Rev. Clin. Psychol. 2012, 8, 49–76. [Google Scholar] [CrossRef]
  97. Basner, M.; Rao, H.; Goel, N.; Dinges, D.F. Sleep deprivation and neurobehavioral dynamics. Curr. Opin. Neurobiol. 2013, 23, 854–863. [Google Scholar] [CrossRef]
  98. Picchioni, D.; Duyn, J.H.; Horovitz, S.G. Sleep and the functional connectome. Neuroimage 2013, 80, 387–396. [Google Scholar] [CrossRef]
  99. Voss, M.W.; Soto, C.; Yoo, S.; Sodoma, M.; Vivar, C.; van Praag, H. Exercise and Hippocampal Memory Systems. Trends Cogn. Sci. 2019, 23, 318–333. [Google Scholar] [CrossRef]
  100. Sambataro, F.; Murty, V.P.; Callicott, J.H.; Tan, H.Y.; Das, S.; Weinberger, D.R.; Mattay, V.S. Age-related alterations in default mode network: Impact on working memory performance. Neurobiol. Aging 2010, 31, 839–852. [Google Scholar] [CrossRef] [PubMed]
  101. Basner, M.; Nasrini, J.; Hermosillo, E.; McGuire, S.; Dinges, D.F.; Moore, T.M.; Gur, R.C.; Rittweger, J.; Mulder, E.; Wittkowski, M.; et al. Effects of −12° head-down tilt with and without elevated levels of CO2 on cognitive performance: The SPACECOT study. J. Appl. Physiol. 2018, 124, 750–760. [Google Scholar] [CrossRef] [PubMed]
  102. Jones, C.W.; Basner, M.; Mollicone, D.J.; Mott, C.M.; Dinges, D.F. Sleep deficiency in spaceflight is associated with degraded neurobehavioral functions and elevated stress in astronauts on six-month missions aboard the International Space Station. Sleep. 2022, 45, zsac006. [Google Scholar] [CrossRef] [PubMed]
  103. Barulli, D.; Stern, Y. Efficiency, capacity, compensation, maintenance, plasticity: Emerging concepts in cognitive reserve. Trends Cogn. Sci. 2013, 17, 502–509. [Google Scholar] [CrossRef] [PubMed]
  104. Clewett, D.V.; Lee, T.H.; Greening, S.; Ponzio, A.; Margalit, E.; Mather, M. Neuromelanin marks the spot: Identifying a locus coeruleus biomarker of cognitive reserve in healthy aging. Neurobiol. Aging 2016, 37, 117–126. [Google Scholar] [CrossRef]
  105. Stern, Y. An approach to studying the neural correlates of reserve. Brain Imaging Behav. 2017, 11, 410–416. [Google Scholar] [CrossRef]
  106. Gao, Z.; van Beugen, B.J.; De Zeeuw, C.I. Distributed synergistic plasticity and cerebellar learning. Nat. Rev. Neurosci. 2012, 13, 619–635. [Google Scholar] [CrossRef]
  107. McGregor, H.R.; Lee, J.K.; Mulder, E.R.; De Dios, Y.E.; Beltran, N.E.; Kofman, I.S.; Bloomberg, J.J.; Mulavara, A.P.; Seidler, R.D. Brain connectivity and behavioral changes in a spaceflight analog environment with elevated CO(2). Neuroimage 2021, 225, 117450. [Google Scholar] [CrossRef]
  108. Acharya, M.M.; Baulch, J.E.; Klein, P.M.; Baddour, A.A.D.; Apodaca, L.A.; Kramár, E.A.; Alikhani, L.; Garcia, C.; Angulo, M.C.; Batra, R.S.; et al. New Concerns for Neurocognitive Function during Deep Space Exposures to Chronic, Low Dose-Rate, Neutron Radiation. eNeuro 2019, 6, ENEURO.0094-19. [Google Scholar] [CrossRef]
  109. Britten, R.A.; Fesshaye, A.S.; Duncan, V.D.; Wellman, L.L.; Sanford, L.D. Sleep Fragmentation Exacerbates Executive Function Impairments Induced by Low Doses of Si Ions. Radiat. Res. 2020, 194, 116–123. [Google Scholar] [CrossRef]
  110. Izquierdo, A.; Brigman, J.L.; Radke, A.K.; Rudebeck, P.H.; Holmes, A. The neural basis of reversal learning: An updated perspective. Neuroscience 2017, 345, 12–26. [Google Scholar] [CrossRef] [PubMed]
  111. Nithianantharajah, J.; McKechanie, A.G.; Stewart, T.J.; Johnstone, M.; Blackwood, D.H.; St Clair, D.; Grant, S.G.; Bussey, T.J.; Saksida, L.M. Bridging the translational divide: Identical cognitive touchscreen testing in mice and humans carrying mutations in a disease-relevant homologous gene. Sci. Rep. 2015, 5, 14613. [Google Scholar] [CrossRef] [PubMed]
  112. Chaumet, G.; Taillard, J.; Sagaspe, P.; Pagani, M.; Dinges, D.F.; Pavy-Le-Traon, A.; Bareille, M.P.; Rascol, O.; Philip, P. Confinement and sleep deprivation effects on propensity to take risks. Aviat. Sp. Env. Med. 2009, 80, 73–80. [Google Scholar] [CrossRef] [PubMed]
  113. Dinges, D.F.; Basner, M.; Mollicone, D.; Ecker, A.; Jones, C. Reaction Self Test on ISS: 6-Month Missions; University of Pennsylvania: Philadephia, PA, USA, 2016. [Google Scholar]
  114. Deng, W.; Aimone, J.B.; Gage, F.H. New neurons and new memories: How does adult hippocampal neurogenesis affect learning and memory? Nat. Rev. Neurosci. 2010, 11, 339–350. [Google Scholar] [CrossRef]
  115. Roy, D.S.; Kitamura, T.; Okuyama, T.; Ogawa, S.K.; Sun, C.; Obata, Y.; Yoshiki, A.; Tonegawa, S. Distinct Neural Circuits for the Formation and Retrieval of Episodic Memories. Cell 2017, 170, 1000–1012.e19. [Google Scholar] [CrossRef]
  116. Mueller, A.; Hong, D.S.; Shepard, S.; Moore, T. Linking ADHD to the Neural Circuitry of Attention. Trends Cogn. Sci. 2017, 21, 474–488. [Google Scholar] [CrossRef]
  117. Barkley, R.A. Behavioral inhibition, sustained attention, and executive functions: Constructing a unifying theory of ADHD. Psychol. Bull. 1997, 121, 65–94. [Google Scholar] [CrossRef]
  118. Mazaheri, A.; Coffey-Corina, S.; Mangun, G.R.; Bekker, E.M.; Berry, A.S.; Corbett, B.A. Functional disconnection of frontal cortex and visual cortex in attention-deficit/hyperactivity disorder. Biol. Psychiatry 2010, 67, 617–623. [Google Scholar] [CrossRef]
  119. Luck, S.J.; Ford, J.M.; Sarter, M.; Lustig, C. CNTRICS final biomarker selection: Control of attention. Schizophr. Bull. 2012, 38, 53–61. [Google Scholar] [CrossRef]
  120. Faraone, S.V.; Bonvicini, C.; Scassellati, C. Biomarkers in the diagnosis of ADHD--promising directions. Curr. Psychiatry Rep. 2014, 16, 497. [Google Scholar] [CrossRef]
  121. Bolkan, S.S.; Stujenske, J.M.; Parnaudeau, S.; Spellman, T.J.; Rauffenbart, C.; Abbas, A.I.; Harris, A.Z.; Gordon, J.A.; Kellendonk, C. Publisher Correction: Thalamic projections sustain prefrontal activity during working memory maintenance. Nat. Neurosci. 2018, 21, 1138. [Google Scholar] [CrossRef] [PubMed]
  122. Crucian, B.E.; Zwart, S.R.; Mehta, S.; Uchakin, P.; Quiriarte, H.D.; Pierson, D.; Sams, C.F.; Smith, S.M. Plasma cytokine concentrations indicate that in vivo hormonal regulation of immunity is altered during long-duration spaceflight. J. Interf. Cytokine Res. 2014, 34, 778–786. [Google Scholar] [CrossRef] [PubMed]
  123. Carey, L.; Nilsson, M.; Boyd, L. Learning following Brain Injury: Neural Plasticity Markers. Neural Plast. 2019, 2019, 4838159. [Google Scholar] [CrossRef]
  124. Rosell, D.R.; Siever, L.J. The neurobiology of aggression and violence. CNS Spectr. 2015, 20, 254–279. [Google Scholar] [CrossRef] [PubMed]
  125. Ko, J. Neuroanatomical Substrates of Rodent Social Behavior: The Medial Prefrontal Cortex and Its Projection Patterns. Front. Neural Circuits 2017, 11, 41. [Google Scholar] [CrossRef]
  126. Freichel, M.; Vennekens, R.; Olausson, J.; Stolz, S.; Philipp, S.E.; Weissgerber, P.; Flockerzi, V. Functional role of TRPC proteins in native systems: Implications from knockout and knock-down studies. J. Physiol. 2005, 567, 59–66. [Google Scholar] [CrossRef]
  127. Trainor, B.C.; Workman, J.L.; Jessen, R.; Nelson, R.J. Impaired nitric oxide synthase signaling dissociates social investigation and aggression. Behav. Neurosci. 2007, 121, 362–369. [Google Scholar] [CrossRef]
  128. Nestler, E.J.; Barrot, M.; DiLeone, R.J.; Eisch, A.J.; Gold, S.J.; Monteggia, L.M. Neurobiology of depression. Neuron 2002, 34, 13–25. [Google Scholar] [CrossRef]
  129. Crucian, B.; Stowe, R.; Quiriarte, H.; Pierson, D.; Sams, C. Monocyte phenotype and cytokine production profiles are dysregulated by short-duration spaceflight. Aviat. Sp. Environ. Med. 2011, 82, 857–862. [Google Scholar] [CrossRef]
  130. Crucian, B.E.; Choukèr, A.; Simpson, R.J.; Mehta, S.; Marshall, G.; Smith, S.M.; Zwart, S.R.; Heer, M.; Ponomarev, S.; Whitmire, A.; et al. Immune system dysregulation during spaceflight: Potential countermeasures for deep space exploration missions. Front. Immunol. 2018, 9, 1437. [Google Scholar] [CrossRef]
  131. Huang, W.; Ramsey, K.M.; Marcheva, B.; Bass, J. Circadian rhythms, sleep, and metabolism. J. Clin. Investig. 2011, 121, 2133–2141. [Google Scholar] [CrossRef]
  132. Moore, R.Y. Neural control of the pineal gland. Behav. Brain Res. 1996, 73, 125–130. [Google Scholar] [CrossRef]
  133. Shang, X.; Xu, B.; Li, Q.; Zhai, B.; Xu, X.; Zhang, T. Neural oscillations as a bridge between glutamatergic system and emotional behaviors in simulated microgravity-induced mice. Behav. Brain Res. 2017, 317, 286–291. [Google Scholar] [CrossRef]
  134. Janelidze, S.; Mattsson, N.; Stomrud, E.; Lindberg, O.; Palmqvist, S.; Zetterberg, H.; Blennow, K.; Hansson, O. CSF biomarkers of neuroinflammation and cerebrovascular dysfunction in early Alzheimer disease. Neurology 2018, 91, e867–e877. [Google Scholar] [CrossRef] [PubMed]
  135. Raber, J.; Allen, A.R.; Rosi, S.; Sharma, S.; Dayger, C.; Davis, M.J.; Fike, J.R. Effects of (56)Fe radiation on hippocampal function in mice deficient in chemokine receptor 2 (CCR2). Behav. Brain Res. 2013, 246, 69–75. [Google Scholar] [CrossRef] [PubMed]
  136. Aïd, S.; Bosetti, F. Targeting cyclooxygenases-1 and -2 in neuroinflammation: Therapeutic implications. Biochimie 2011, 93, 46–51. [Google Scholar] [CrossRef] [PubMed]
  137. Derecki, N.C.; Cardani, A.N.; Yang, C.H.; Quinnies, K.M.; Crihfield, A.; Lynch, K.R.; Kipnis, J. Regulation of learning and memory by meningeal immunity: A key role for IL-4. J. Exp. Med. 2010, 207, 1067–1080. [Google Scholar] [CrossRef] [PubMed]
  138. Guéguinou, N.; Bojados, M.; Jamon, M.; Derradji, H.; Baatout, S.; Tschirhart, E.; Frippiat, J.-P.; Legrand-Frossi, C. Stress response and humoral immune system alterations related to chronic hypergravity in mice. Psychoneuroendocrinology 2012, 37, 137–147. [Google Scholar] [CrossRef]
  139. Kokhan, V.S.; Matveeva, M.I.; Bazyan, A.S.; Kudrin, V.S.; Mukhametov, A.; Shtemberg, A.S. Combined effects of antiorthostatic suspension and ionizing radiation on the behaviour and neurotransmitters changes in different brain structures of rats. Behav. Brain Res. 2017, 320, 473–483. [Google Scholar] [CrossRef]
  140. Kulikova, E.A.; Kulikov, V.A.; Sinyakova, N.A.; Kulikov, A.V.; Popova, N.K. The effect of long-term hindlimb unloading on the expression of risk neurogenes encoding elements of serotonin-, dopaminergic systems and apoptosis; comparison with the effect of actual spaceflight on mouse brain. Neurosci. Lett. 2017, 640, 88–92. [Google Scholar] [CrossRef]
  141. Wu, X.; Li, D.; Liu, J.; Diao, L.; Ling, S.; Li, Y.; Gao, J.; Fan, Q.; Sun, W.; Li, Q.; et al. Dammarane Sapogenins Ameliorates Neurocognitive Functional Impairment Induced by Simulated Long-Duration Spaceflight. Front. Pharmacol. 2017, 8, 315. [Google Scholar] [CrossRef] [PubMed]
  142. Newman, E.L.; Leonard, M.Z.; Arena, D.T.; de Almeida, R.M.M.; Miczek, K.A. Social defeat stress and escalation of cocaine and alcohol consumption: Focus on CRF. Neurobiol. Stress. 2018, 9, 151–165. [Google Scholar] [CrossRef] [PubMed]
  143. Wise, K.C.; Manna, S.K.; Yamauchi, K.; Ramesh, V.; Wilson, B.L.; Thomas, R.L.; Sarkar, S.; Kulkarni, A.D.; Pellis, N.R.; Ramesh, G.T. Activation of nuclear transcription factor-kappaB in mouse brain induced by a simulated microgravity environment. In Vitro Cell. Dev. Biol. Anim. 2005, 41, 118–123. [Google Scholar] [CrossRef] [PubMed]
  144. Delp, M.D.; Charvat, J.M.; Limoli, C.L.; Globus, R.K.; Ghosh, P. Apollo Lunar Astronauts Show Higher Cardiovascular Disease Mortality: Possible Deep Space Radiation Effects on the Vascular Endothelium. Sci. Rep. 2016, 6, 29901. [Google Scholar] [CrossRef] [PubMed]
  145. Frijhoff, J.; Winyard, P.G.; Zarkovic, N.; Davies, S.S.; Stocker, R.; Cheng, D.; Knight, A.R.; Taylor, E.L.; Oettrich, J.; Ruskovska, T.; et al. Clinical Relevance of Biomarkers of Oxidative Stress. Antioxid. Redox Signal. 2015, 23, 1144–1170. [Google Scholar] [CrossRef] [PubMed]
  146. Ikawa, M.; Okazawa, H.; Nakamoto, Y.; Yoneda, M. PET Imaging for Oxidative Stress in Neurodegenerative Disorders Associated with Mitochondrial Dysfunction. Antioxidants 2020, 9, 861. [Google Scholar] [CrossRef]
  147. Sajdel-Sulkowska, E.M.; Xu, M.; Koibuchi, N. Cerebellar brain-derived neurotrophic factor, nerve growth factor, and neurotrophin-3 expression in male and female rats is differentially affected by hypergravity exposure during discrete developmental periods. Cerebellum 2009, 8, 454–462. [Google Scholar] [CrossRef]
  148. Rudobeck, E.; Bellone, J.A.; Szücs, A.; Bonnick, K.; Mehrotra-Carter, S.; Badaut, J.; Nelson, G.A.; Hartman, R.E.; Vlkolinský, R. Low-dose proton radiation effects in a transgenic mouse model of Alzheimer’s disease—Implications for space travel. PLoS ONE 2017, 12, e0186168. [Google Scholar] [CrossRef]
  149. Simpson, R.H.; Rodda, J.; Reinecke, C.J. Adrenoleukodystrophy in a mother and son. J. Neurol. Neurosurg. Psychiatry 1987, 50, 1165–1172. [Google Scholar] [CrossRef]
  150. Li, W.; Pan, R.; Qi, Z.; Liu, K.J. Current progress in searching for clinically useful biomarkers of blood-brain barrier damage following cerebral ischemia. Brain Circ. 2018, 4, 145–152. [Google Scholar] [CrossRef]
  151. Bellone, J.A.; Gifford, P.S.; Nishiyama, N.C.; Hartman, R.E.; Mao, X.W. Long-term effects of simulated microgravity and/or chronic exposure to low-dose gamma radiation on behavior and blood-brain barrier integrity. NPJ Microgravity 2016, 2, 16019. [Google Scholar] [CrossRef] [PubMed]
  152. Lu, A.T.; Quach, A.; Wilson, J.G.; Reiner, A.P.; Aviv, A.; Raj, K.; Hou, L.; Baccarelli, A.A.; Li, Y.; Stewart, J.D.; et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 2019, 11, 303–327. [Google Scholar] [CrossRef] [PubMed]
  153. Brüll, V.; Burak, C.; Stoffel-Wagner, B.; Wolffram, S.; Nickenig, G.; Müller, C.; Langguth, P.; Alteheld, B.; Fimmers, R.; Stehle, P.; et al. Acute intake of quercetin from onion skin extract does not influence postprandial blood pressure and endothelial function in overweight-to-obese adults with hypertension: A randomized, double-blind, placebo-controlled, crossover trial. Eur. J. Nutr. 2017, 56, 1347–1357. [Google Scholar] [CrossRef] [PubMed]
  154. Zhang, H.; Chen, J.; Wang, H.; Lu, X.; Li, K.; Yang, C.; Wu, F.; Xu, Z.; Nie, H.; Ding, B.; et al. Serum Metabolomics Associating With Circulating MicroRNA Profiles Reveal the Role of miR-383-5p in Rat Hippocampus Under Simulated Microgravity. Front. Physiol. 2020, 11, 939. [Google Scholar] [CrossRef]
Table 3. The major observations and points discussed by the panel.
Table 3. The major observations and points discussed by the panel.
Oxidative Stress
Blood biomarkers: 8-oxo-dG in immune cells, MDA, f2-isoprostane, Nitrotryosine; brain HNE, glutathione, lipid peroxidation, ROS, NFKb, MAPK activation, Xanthine oxidase
  • Oxidative stress-associated mitochondrial dysfunction has been shown in many cells, tissue and organ system, their impacts have to be further investigated.
  • The role of diet in mitigating oxidative stress associated with spaceflight.
  • Epigenetic clock measurements in astronauts and related to time in space or deep space and their association with oxidative stress-induced aging.
  • miRNA signatures and exosomes in identifying oxidative stress biomarkers and as novel biomarkers in brain pathogenesis.
Neurotransmitters
Behavioral biomarkers: mood, depression, anxiety tests
  • Limited to in vitro data that are inconsistent across studies. Only one neurotransmitter examined at a time (e.g., DA, glutamate, 5-HT, or ACh).
  • Human studies with MRI spectroscopy are difficult to do in real-time.
  • Only invasive rodent assays are available.
  • Need studies that associate neurotransmitter changes with changes in lipids/metabolites.
  • Neurotransmitters provide a direct readout of CNS functionality at multiple levels: behavioral, emotional, systemic stress, endocrine, and electrophysiological.
  • Cross-species correlates (chemical changes): rodents– NHP–Humans and should be translated to lipidomic and metabolomic findings.
Neuroinflammation
Blood biomarkers: COX-2, TREM, IL-4, TNF, BDNF, corticosterone; YKL-40, ICAM-1, VCAM-1, IL-15, and Flt-1 in CSF; Behavioral biomarkers: cognitive tests
  • Specificity of blood biomarkers such as cytokines (variability with circadian changes and time of collection).
  • Animal to human correlation (circadian and sleep system differences, rhythm differences, immune differences, white-matter differences, vasculature differences).
  • Applying cell-free DNA and subsequent methylation analysis can give high sensitivity measurement of BBB integrity, cell breakdown and inflammation in the brain.
One-carbon metabolism
Blood biomarkers: folate, Vit. B-12, methylmalonic acid and homocysteine, MMPs; CSF: 5MTHF
  • Difficult to correlate biomarker changes between CSF and plasma
  • Genetic variations in folate-mediated one carbon metabolism predict risk of adverse effects in space flight–mechanisms are unknown
  • Relation to white matter hyperintensity, inflammation, BBB permeability, inflammation, behavioral changes, e.g., Hyperhomocysteinemia, vascular dementia.
Table 4. Circuits and biomarkers for integrated approaches/physiological responses.
Table 4. Circuits and biomarkers for integrated approaches/physiological responses.
Physiological ResponsesRelated Gene Ontology TermsBiomarkers (Human/Rodent)Associated Pathways/Signaling CascadeCNS Health RisksHuman Behavioral MeasureRodent/NHP Behavioral MeasureOpen Questions/Gaps (How to Close?)Notes/Limitations on Biomarkers
InaccessibleAccessible
NeuroinflammationGlial activation, neuron apoptotic process, BBB disruption, endothelial dysfunction, oxidative stressCSF: YKL-40, ICAM-1, VCAM-1, IL-15, and Flt-1 [134], Brain lysates-CCR2 [135], Brain lysate-proteomics, IHC, IL21. CSF-cytokine (accurate for neuroinflammation)Blood: COX-2, cytokines, TREM [136], IL4, TNF, BDNF [137], Corticosterone [138], c-reactive protein, IL-6 and TNFa, glial fibrillary acidic protein (GFAP), IL110, IL4 (variability due to circadian disruption or sleep deprivation), IL21
Imaging: CT, fMRI, PET, EEG, MEG, TMS scan, MRS (myoinositol, glutamine to glutamate ratio), Functional biomarker—HSV1 (viral reactivation)
NFKB signaling, Chemokine signaling, TNF signaling, Calcium signaling, Serotonergic synapse, VEGF signaling, Autophagy, oxidative stressNeurodegenerative disorders, meningitisCognition, Mathematical processing (MTH), Running memory continuous performance test (CPT), Delayed matching-to-sample (MTS), Code substitution (CDS)Spontaneous new home behavior,
Elevated plus maze, light/dark box, WMWM and fear conditioning, contextual fear conditioning, Morris water maze test, pass avoidance performance test, climbing pole test
(1) Longitudinal study of blood biomarker (e.g., cytokines) and correlating with individual’s biological clock (variability across individual of approx. 5 h.), clinical and medical history.
(2) Flight deployable ELISA cytokine panel (3) Microfluidics based system that can be deployed, miniaturized microscope and flow cytometer.
(4) For animal to human study correlation—Tissues can be harvested and animal study should be contextual to the question asked. Humanized mouse model—good for immunological study. (5) Leverage omics data.
(6) Countermeasure development requires living system.
(7) Other animal model—Canine, pig, marmoset—reinventing the wheel?
(1) Threshold?
(2) Challenges for data collection and storage:
(3) Unclear whether plasma will be collected and stored in space, then assessed on Earth, or are we looking for measures that can be done in real time in space? Some of these assays require special equipment and assays.
Importance of storage consistency-Plasma biomarkers are very sensitive to processing and storage conditions, including type of plastic for tubes, tube size and volume of aliquots.
(4) Recommend many small aliquots to maximize potential for number of biomarkers that can be assessed, because freeze-thaw also significant influences measurement. (5) Specificity of blood biomarkers such as cytokines (variability with circadian changes).
(6) Animal to human correlation (circadian and sleep system differences, rhythm differences, immune differences, white-matter differences, vasculature differences).
NeurotransmittersNeurotransmitter release and metabolism, cellular metabolismBrain lysates: Serotonin [139], Dopamine and dopamine regulating enzyme, COMPT [139,140], Ach [139], Norepinephrine, Epinephrine, Glutamate [141], Glutamate receptors (NMDAR2A/2B) [133], Stress hormones-cortisol, oxytocin; Corticotrophin-releasing hormone (CRH); Corticotrophin-releasing factor (CRF) [142]Blood: Serotonin [139], Dopamine and dopamine regulating enzyme, COMPT [139,140], Ach [139], Norepinephrine, Epinephrine, Glutamate, GABA [141], Glutamate receptors (NMDAR2A/2B) [133], Stress hormones-cortisol
Imaging: CT, fMRI, PET, EEG, MEG, TMS scan
Monoamine pathway: mesocorticolimbic; nigrostriatal. Hypothalmic-pituitary-adrenal (HPA) axisMood, Depression, Anxiety, Alzheimer’s, schizophrenia, Parkinson’s, other degenerative conditions; Social stress (Stress leading to social dominance)Mnemonic similarity test (MST) (BPSO)-this test includes Novel object recognition (NOR), learning and motor tasksThigmotaxis, water maze, elevated maze, open field test, passive avoidance(1). What is the relationship between brain neurochemistry and behavior?
(2) Are neurochemical signatures differently impacted in different brain regions to influence behavior and what is the right balance?
(3) What can be measured peripherally?
(4) Which dopamine and serotonin metabolites are best measured peripherally?
(5) Wearable devices/sensors to measure metabolites instead of blood tests
Limitations:
(1) Inconsistent data across studies: one neurotransmitter system examined (e.g., DA, glutamate, or 5-HT): comprehensive assessment needed.
(2) Human studies with MRI spectroscopy are difficult to do in real-time.
(3) Rodents’ assays are invasive measures, lack less invasive techniques
(4) Need studies that associate neurotransmitter changes with changes in lipids and other metabolites
Strengths:
(1) Neurotransmitters provide a direct readout of CNS functionality at multiple levels.
(2) Cross-species correlated (chemical changes) rodents—NHP—Humans. Should be translated to lipidomic and metabolomic findings.
One-carbon metabolismSANS, BBB, endothelial dysfunction, CSF pressure, BioenergeticsBrain: B-vitamin and 1C metabolite profiles, DNA strand breaks; uracil in genomic DNA and mitochondrial DNA (higher sensitivity)Blood: serum and RBC, folate, vitamin B12, methylmalonic acid and homocysteine, MMPs, Met, AdoMet (P. Stover),
Formate, one-carbon nutrients, and their methylation profiling (inputs towards one carbon metabolism pathway).
Imaging: OCT for SANS, MRI for WMH; skin autofluorescence for AGE; Ultrasound Elastography (scleral stiffness), OCT angiography CSF: 5MTHF
Folate and methionine production, Epigenetic methylation, DNA synthesis and repair, Neurotransmitter metabolism,
Trans-sulfuration pathway,
Bioenergetic crisis
SANS, Neurodegenerative disorder (AD), neurodevelopment, DepressionCognition: Standardized Mini-Mental State Examination, simple reaction time (SRT), choice reaction time (CRT), digit vigilance task (DVT)Cognitive tests (Morris water maze)Is personalized nutrition (i.e., B-vitamin supplementation) a viable SANS countermeasure?(1) Correlating biomarker changes between CSF and plasma?
(2) Relation to white matter hyperintensity, inflammation, BBB permeability, inflammation, behavioral changes, e.g., Hyperhomocysteinemia, vascular dementia.
Oxidative stressAutophagy, inflammation, Lipid peroxidation, BioenergeticsTissue: Glutathione, lipid peroxidation, ROS, NFKb, MAPK activation [143], Blood vessel-Xanthine oxidase [144]Blood/Urine: Cytokines levels, HNE, MDA, f2-isoprostane, Nitrotryosine levels [145], 8OHdG; reduced/total glutathione, total antioxidant capacity, superoxide dismutase, glutathione peroxidase, advanced glycation end products (AGEs), glycated albumin, 3-nitrotyrosine, oxidized LDL, miR383 (regulating AQP4), cell-free DNA (genetic and epigenetic changes)
Imaging: CT, fMRI, PET, EEG scan, PET with 62Cu-ATSM [146]
Oxidative phosphorylation, Mitochondrial dysfunction, NFR2-mediated oxidative stress response, Superoxide radicals’ degradation, Neuroinflammation, apoptosis, necrosis, neurovascular impairments, Bioenergetic crisisNeurodegenerative disorders, Cardiovascular disorders, affects multiple organs, Anxiety, Depression, Schizophrenia, Metabolic disorders, SANS.Anxiety and depression related behavioral tests (Visual Analog Scale
Depression: Beck Depression Inventory), psychomotor tests (Tandem Walking, Perturbation during walking, navigating obstacle course while walking (e.g., Functional Mobility Test)), Cognitive tests (Mnemonic similarity test (MST) (BPSO)-this test includes Novel object recognition (NOR), Fractal 2B, object rotation in space)
Anxiety related (Elevated plus maze, hole-board, and open field tests), Psychomotor tests (Rod walking, wire suspension/wire hanging, plank walking, inclined screen, accelerating rotarod), Cognitive tests (Morris water maze)(1) Can diet mitigate oxidative stress associated with space flight?
(2) What are the relationships between ox stress, immune function during flight? (3) miRNA signatures? Antagomir-countermeasure, specificity, applicability? (4) Exosomes?
Mitochondrial dysfunction Plasma: Formate (mito one carbon metabolism) biomarker of mitochondrial function.
Synaptic plasticity/Neurotrophic FactorsRegulation of synaptic plasticity, modulation of chemical synaptic transmission, neurotrophin receptor activityBrain lysates: BDNF,
Neurotrophin-3 [147],
synaptophysin [148], CtBP2, Shank1a [29],
14-3-3 proteins (CSF marker of CNS degeneration), EEG markers, BDNF, c-Fos
Imaging: CT, fMRI, PET, EEG, MEG, TMS scan; Plasma: Neurofilament light (NfL), phospho-tau 181 (pTau181), beta-amyloid 40 and 42, BDNF; CSF: NfL, pTau181, beta-amyloid 40 and 42.Ubiquitin-proteosome, lysophosphatidic acid (LPA), kinases, Calcium signaling (PI3K, PLC gamma), MAPK/ERKNeurodegenerative disorders, schizophrenia1.Sequence/procedural; 2. Eye-Head/Eye-Head-Hand adaptation tasks—
(a) VOR adaptation test
(b) Eye-Head Hand- visuo-motor adaptation task
3. Whole body tasks
(a) Walking with visuomotor adaptation
(b) Split Belt Locomotion Test
4. Mismatch negativity
1. Odor sequence learning (non-motor)
2. Eye Head and Eye Head Hand adaptation tasks:
(a) Nystagmus and compensation following labyrinthectomy (b) Rodent VOR test
3. Whole body tasks (a) Ladder rung walk test
4. Mismatch negativity (plasticity + perceptual learning, non-motor component, EEG measure)
5. Mathematical processing (MTH)
(1) Markers of neurodegeneration are missing. Acute and chronic injury can be tracked longitudinally with plasma NfL.
(2) Lacks integration of phenotypic studies in models and omics.
(3) miRNA signatures are missing.
(4) Identify molecular changes at the synaptic level (5) Relatively unexplored area
Which biomarkers can we repurpose from terrestrial disorders to spaceflight?
There have been huge advances in Alzheimer’s and vascular dementia blood-based biomarkers. While associated with aging, these markers can reflect neuronal and vascular injury and later risk of cognitive problems.
NfL is a marker of neuronal injury that is increased significantly in traumatic brain injury, many forms of dementia, and CTE.
Vestibular/Sensorimotor alterationsVestibular reflex, vestibular hair cell stereocilium organization, vestibular receptor cell stereocilium organizationOtopetrin1, Alpha 2 adrenergic receptors [23], Glutamate receptor expression [24], c-FOS, vestibular hair cells [25], cerebellar nodulus of adult rats [26,27,28], TEM of synaptic ribbons [29,30,31,32,33,149].Nausea related—cardiac sensitivity to baroreceptor reflex; raised Heart rate; raised cortisol; reduced dominant power on EGG baseline, questionnaire [34,35], Circadian measurements
Imaging: CT, fMRI, PET, EEG, MEG, TMS scan
Motion sickness, Dizziness, Loss of Hearing, Postural imbalance, VertigoCognition, Spatial memory, Graybiel scale, CDP, get up From Fall Test, Drop test/Jump down test, VEMP, OVAR responseRotarod, Zebrafish Active Posturography (Zap); Floating Platform Tests–Postural sway–measured by Center of Pressure (COP) Assay (=COP), Righting reflex, VEMP, OVAR response, Active vs. Passive motion on vestibular nucleus neurons, Mid-air righting reflex(1) Robotic simulations
(2) What happens in a more regular schedule?
(3) What are the effects of recurring cycles of sleep deprivation? How do they recover? How does it affect performance? We need biomarkers for that.
(1) Sleep loss and circadian changes affect the sensorimotor and cognitive function.
(2) Caffeine + light − effective countermeasures. (3) Primary task is not affected during sleep loss but the secondary tasks are. This should be considered for effects on operational performance.
DNA damageDNA repair, DNA metabolic process, cellular response to DNA damage stimulusBrain/other tissues: Staining with Anti-8-oxo-dg, 53bp1Blood: DNA lesions via HPLC, 8-oxo dg, micronuclei, double strand DNA breaks, chromosomal aberrations/translocations, one carbon metabolitesCell cycle checkpoint activation, DNA Repair, apoptosis,RadiotherapyCognitive testsOxidative stress and inflammation related cognitive testsMonitor the level of DNA damage over time- need astronaut data(1) Since brain and neurons are not proliferative, DNA damage is might not be relevant in CNS. However, peripheral DNA damage is useful to studying the general diversity and individual differences of responses to radiation (again a surrogate, assuming that the brain will respond the same as the rest of the body).
(2) Use baseline DNA damage as a predictor for responses to irradiation/spaceflight (astronaut panel pre/post flight).
(3) Sleep deprivation exacerbates DNA damage in rats and humans. We cannot train/adapt to sleep deprivation.
Note suggested markers for radiation dosage-bio-dosimetry: FLT3LG, SAA1, C3, VCAM
Blood brain barrier permeabilityInflammation, one carbon metabolismCSF: Albumin [150], Brain IHC—Aquaporin 4 [151], IHC, MMP-9, long-term microglial activation, astrocyte morphology, Endothelial cells, Somatic mosaicismBlood: Occludin, c-Fibronectin, Ubiquitin carboxyl-terminal hydrolase isozyme L1, S100 calcium-binding protein B, Circulating brain microvascular endothelial cells ([150], stroke research), Corticosterone MMP-9, Cell free DNA Imaging: fMRI, PET scan, free water MRI; Epigenetic clock (accelerated aging). Endothelial activation, Systemic inflammation, Kynurenine pathway, Tight junction damage, Oxidative stress, glial activation, MAPK pathway, PKC pathway, degradation of basal lamina and ECM.Inflammation, stroke, Alzheimer’sStress: Visual Analog Scale
Depression: Beck Depression Inventory
Locomotor activity, open field, hole-board, and grip strength tests, anxiety, and depressive behaviors(1) Is BBB function altered in astronauts on ISS (or Artemis) missions?
(2) Study the glymphatic system-removal of solutes from the brain across the BBB.
(3) Need to understand the association of MMP9, occludins, S100, etc. with drainage of BBB. What is the physiological relevance? Glymphatic system is important for sleep as well.
(4) Mutations, mosaicism etc. will affect the endothelial cells and may cause BBB leakiness, leading to physiological effects. (5) Association of sleep with debris clearance. Amyloid clearance from the brain occurs during sleep → relevance to both sleep/circadian and glymphatic system.
(6) Astrocyte morphology—unexplored. Astrocyte expressing AQP4 would be important for glymphatic system.
(7) Epigenetic clock measurements in astronauts and related to time in space etc. Or deep space to look at age acceleration
(8) Development of rodent in vivo imaging technologies for BBB integrity.
(9) Radiation induced senescence and functional readout in brain—glial cells, epithelial cells, somatic mosaicism
(1) Circadian changes in astronauts (avg. sleep 6 h. though allocated 8–9 h) can add more stress.
(2) Epigenetic and aging association [152]. Easily conducted. (3) DNA methylation observed in radiation and inhibition on global level can mitigate hypermethylation related cognitive deficits.
VasculatureBlood vessel development, heart developmentAdhesion molecules (VE-cadherin), tight junction proteins (Claudin 3, 5, 12, Occludin), Zo-1, MMPsBlood: Endothelial function markers (serum nitric oxide, tetra- and dihydrobiopterin (BH4) and (BH2), soluble intercellular adhesion molecule-1 (sICAM-1), soluble vascular cell adhesion molecule-1 (sVCAM-1), endothelin-1, asymmetric dimethylarginine (ADMA), L-arginine, formate, and soluble E-selectin. Imaging: fMRI, PET scan. Noninvasive peripheral arterial tonometry (PAT) technology can be used to assess the reactive hyperemia index (RHI) and the augmentation index [153]; Vascular damage MRI measures: Cerebrovascular reactivity (CVR) (Pre and post flight): Present with CO2 challenge; Free water. Plasma: Placental growth factor (PIGF), IL-8; VEGF-D; CSF: PIGF; IL-8 Adherens junction, Endothelial activation, systemic inflammation, oxidative stress, hypoxiaInflammation, stroke, Alzheimer’sStress: Visual Analog Scale
Depression: Beck Depression Inventory
Locomotor activity, open field, hole-board, and grip strength tests, and depressive behaviors(1) What are the biochemical underpinnings of the thrombotic events seen inflight?
(2) Also missing are chronic vascular injury markers. This biomarker has gained rapid adoption in many fields in the last few years.
(3) Lack of cerebrovascular reactivity MRI data pre and post flight
(4) Lack of 7T MRI for perivascular spaces
(5) How do the biomarkers for vascular cognitive impairment change in astronauts?
(6) Developing computational modeling of vascular changes?
Topological difference in vasculature and its susceptibility towards the various stressors
miRNA regulationTranscriptional regulation Serum: miR-383-5p [154]Transcriptional regulation Cognitive testsCognitive tests
Circadian Phase (sleep, sleepiness, performance impairment, immune function, endocrine function, bone metabolism, reproductive function) Lipidomics, metabolomics, transcriptomics, proteomics Accident, injury (short-term/immediate); cardiometabolic and neurological disorders, compromised immunity (long-term)Cognitive testsCognitive testsCandidates identified; operational validation required(1) Currently blood-borne but development of urinomics, saliva and breath matrices ongoing;
(2) Can predict several days in advance; single vs. multiple samples.
(3) Model organism—consideration of diurnal model over nocturnal. Marmoset? Indian palm squirrels?—restarting and reinventing the wheel?
(4) Consistency in animal models and standardization in measurement.
(5) Primary task is not affected during sleep loss but secondary tasks are. (Considered for operationally relevant performance)
Neuronal and brain Damage MarkersBlood: neurofilament, tau, abeta1-42, common pathology radiation and AD biomarkers (need to be explored) Note suggested markers: NAA/Creatine ratio
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alwood, J.S.; Mulavara, A.P.; Iyer, J.; Mhatre, S.D.; Rosi, S.; Shelhamer, M.; Davis, C.; Jones, C.W.; Mao, X.W.; Desai, R.I.; et al. Circuits and Biomarkers of the Central Nervous System Relating to Astronaut Performance: Summary Report for a NASA-Sponsored Technical Interchange Meeting. Life 2023, 13, 1852. https://doi.org/10.3390/life13091852

AMA Style

Alwood JS, Mulavara AP, Iyer J, Mhatre SD, Rosi S, Shelhamer M, Davis C, Jones CW, Mao XW, Desai RI, et al. Circuits and Biomarkers of the Central Nervous System Relating to Astronaut Performance: Summary Report for a NASA-Sponsored Technical Interchange Meeting. Life. 2023; 13(9):1852. https://doi.org/10.3390/life13091852

Chicago/Turabian Style

Alwood, Joshua S., Ajitkumar P. Mulavara, Janani Iyer, Siddhita D. Mhatre, Susanna Rosi, Mark Shelhamer, Catherine Davis, Christopher W. Jones, Xiao Wen Mao, Rajeev I. Desai, and et al. 2023. "Circuits and Biomarkers of the Central Nervous System Relating to Astronaut Performance: Summary Report for a NASA-Sponsored Technical Interchange Meeting" Life 13, no. 9: 1852. https://doi.org/10.3390/life13091852

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop