Elsevier

Acta Astronautica

Volume 174, September 2020, Pages 32-40
Acta Astronautica

Simulated microgravity disarms human NK-cells and inhibits anti-tumor cytotoxicity in vitro

https://doi.org/10.1016/j.actaastro.2020.03.023Get rights and content

Highlights

  • Natural Killer cell function is impaired after simulated microgravity (SMG).

  • Suppression of function without changes to phenotype.

  • Decreased lysosomal degranulation and cytokine production after exposure to SMG.

  • Leukemia(K562) susceptibility to killing doesn't change after SMG.

Abstract

Long-duration spaceflight impairs natural killer (NK) cell function, which could compromise immune surveillance in exploration class mission crew. To determine if microgravity can impair NK-cell function, we established a rotary cell culture system to expose human peripheral blood mononuclear cells to simulated microgravity (SMG) in vitro. We found that 12 h of SMG suppressed NK-cell cytotoxic activity (NKCA) by ~50% against K562, U266 and 721.221 tumor target cells when returned to the 1G environment. Mass cytometry was used to identify 37 individual markers associated with NK-cell activation, maturation and cytotoxicity, revealing that SMG causes reductions in NK-cell degranulation and effector cytokine production. Extended flow cytometry confirmed that SMG lowered NK cell perforin and granzyme b expression by 25% and 17% respectively, but did not affect the surface expression of various activating (NKG2D, NKp30) and inhibitory (NKG2A, KLRG1) receptors or the ability of NK-cells to conjugate with target cells. Flow cytometry further revealed that SMG impaired NK-cell degranulation (reduced CD107a+ expression) and suppressed TNFα and IFNγ secretion in response to stimulation with K562 target cells. These findings indicate that SMG ‘disarms’ human NK-cells of cytolytic granules and impairs NKCA against a range of tumor target cells in vitro. Exposure to microgravity could be a factor that contributes to impaired NK-cell function during long duration space travel.

Introduction

Space travel presents a myriad of challenges to normal human physiology. Crewmembers on long duration missions, especially those that will go beyond low earth orbit, are faced with challenges presented by microgravity, radiation exposure, high gravitational forces during launch and landing, circadian rhythm disruptions, sleep disturbances and the stressors associated with prolonged periods of isolation and confinement [1]. Immune system dysregulation has been observed in response to both short and long duration spaceflight missions, which could leave crewmembers susceptible to latent viral reactivation and impaired anti-tumor surveillance [[1], [2], [3]]. Shedding of latent herpesviruses such as CMV, EBV and VZV have consistently been observed during spaceflight missions and this has been coupled with alterations in plasma cytokine profiles and impairments in T-cell function [[3], [4], [5], [6]]. NK-cells play an important role in both anti-viral and anti-tumor immune surveillance in the host. While NK cells appear to retain their ability to form complexes and kill tumor target cells while on the International Space Station ISS [23,24], we showed recently that NK cell cytotoxic activity against K562 target cells was diminished when crewmember samples collected during the flight phase of a 6-month mission to the ISS were returned to Earth for analysis [7]. It is not known, however, if these effects are due to discrete spaceflight associated factors (e.g. microgravity, stress, radiation exposure, etc.) or a synergistic effect of such factors.

Several terrestrial analogs have been used in an attempt to discern the precise aspects of the spaceflight environment that affect immunity [8]. It is possible that spaceflight alters immunity largely due to the effects of microgravity. Accepted methods for simulating microgravity (SMG) in vitro are the use of 2D rotating wall vessels (RWV), 3D rotary cell culture systems (RCCS) and random positioning machines. Such methods have shown to increase apoptosis [[9], [10], [11], [12]], reduce expression of early activation markers CD69, CD25 and CD71 in PBMCs [13,14], increase expression of cell cycle arrest protein, p21 [15], and decrease cell motility through type 1 collagen [16,17]. T-cell functionality is also suppressed as measured by reduced activation [14,18,19] and reduced proliferation in response to mitogen stimulation [20]. Only one study, to our knowledge, has examined the effects of SMG on the phenotype and function of human NK-cells [21]. Li et al. reported that ex vivo expanded NK-cells were less capable of killing K562 target cells in vitro following 48 h exposure to SMG using a 2D-RWV, and that this was accompanied by a reduced expression of both activation (NKG2D) and inhibitory (NKG2A) receptors on the cell surface. However, we contend that the use of ex vivo expanded NK-cells that require exogenous cytokines to maintain growth and viability will alter the primary expression of activating and inhibitory receptors on the surface masking the true extent and mechanisms of NK susceptibility to SMG. There is also a need to determine how SMG affects the NK-cell arsenal, specifically the expression and degranulation of the cytotoxic granules perforin and granzyme b, and the secretion of effector cytokines that facilitate NK cell killing [22].

The aim of this study was to determine the effects of SMG on the function and phenotypic characteristics of freshly isolated and non-differentiated human NK-cells. We used a 3D rotary cell culture system (RCCS) to maintain cells in a low-shear, continuous free fall and randomizing gravity vector by rotating high-aspect ratio vessels (HARV) on a horizontal axis [13,[25], [26], [27]]. Our model maintained cell viability (>90%) during 12 h of SMG without the use of cytokines or stimulants to avoid any activating or inhibitory effect on NK cells besides SMG. We report for the first time that SMG impairs the function of freshly isolated human NK-cells by ‘disarming’ them of cytotoxic granules and inhibiting their ability to degranulate and secrete effector cytokines. These findings indicate that microgravity might be partially responsible for the inhibitory effects of space travel on NK-cell function and that appropriate countermeasures might be required to protect immunity during prolonged periods of weightlessness.

Section snippets

Sample material

Peripheral blood samples were collected from healthy volunteers in acid citrate dextrose (ACD) tubes and peripheral blood mononuclear cells (PBMCs) were isolated for immunophenotyping and cytotoxicity experiments. PBMCs were obtained from Gulf Coast Regional Blood Center and processed at M.D. Anderson Cancer Center, Houston, TX for experiments using mass cytometry and flow cytometry. The Institutional Review Boards at the University of Houston and University of Arizona approved the procedures

Pre-exposure to simulated microgravity inhibits NK-cell cytotoxicity

NK-cells within the PBMC fractions pre-exposed to SMG killed ~50% fewer target cells compared to cells in STATIC and RC conditions. This suppression in NKCA was evident against all four cell lines that were measured (Fig. 1b). The RC and STATIC conditions had comparable effects on NKCA, indicating that shear stress as a result of rotation in the absence of SMG does not affect NKCA (F = 31.92; p = 1). After confirming the absence of shear stress effect on function or phenotype in NK cells, we

Discussion

Prolonged exposure to weightlessness is one factor that might contribute to impaired NK-cell function observed in astronauts during long-duration spaceflight missions[7]. Using a rotary cell culture system to simulate microgravity in vitro, our aim was to determine if pre-exposing primary non-differentiated human NK-cells to SMG would alter their phenotype and cytotoxic activity. We have reported here that freshly isolated human NK-cells have a lowered expression of cytolytic granules and an

Conclusion

In conclusion, this is the first study to show that an in vitro cell culture model of microgravity suppresses the function of non-differentiated human NK cells following just 12 h of exposure to SMG. This suppression of NK-cell activity was accompanied by ‘disarming’ of cytolytic granules and also an impaired ability to secrete effector cytokines when stimulated with tumor target cells.

Future studies should determine if SMG alters intracellular ATP levels, RNA transcriptome, and NK-cell

Funding

This work was supported by NASA grants NNX16AB29G, NNX16AG02G and 80NSSC19K1059 to R.J. Simpson, and a National Space Biomedical Research Institute (NSBRI) post-doctoral fellowship (NCC 9-58) to A.B. Bigley.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (42)

  • L.L. Licato et al.

    Multiple interleukin-2 signaling pathways differentially regulated by microgravity

    Immunopharmacology

    (1999)
  • N. Guéguinou et al.

    Could spaceflight-associated immune system weakening preclude the expansion of human presence beyond Earth's orbit?

    J. Leukoc. Biol.

    (2009)
  • B. Crucian et al.

    Immune system dysregulation during spaceflight: clinical risk for exploration-class missions

    J. Leukoc. Biol.

    (2009)
  • C.F. Sams et al.

    Immune status, latent viral reactivation, and stress during long-duration head-down bed rest

    Aviat. Space Environ. Med.

    (2009)
  • S.K. Mehta et al.

    Latent virus reactivation in astronauts on the international space station

    Npj Microgravity

    (2017)
  • A.B. Bigley et al.

    NK-cell function is impaired during long-duration spaceflight

    J. Appl. Physiol.

    (2018)
  • N. Battista et al.

    5-Lipoxygenase-dependent apoptosis of human lymphocytes in the International Space Station: data from the ROALD experiment

    FASEB J.

    (2012)
  • R. Kumari et al.

    Simulated microgravity decreases DNA repair capacity and induces DNA damage in human lymphocytes

    J. Cell. Biochem.

    (2009)
  • M.L. Lewis et al.

    Spaceflight alters microtubules and increases apoptosis in human lymphocytes (Jurkat)

    FASEB J.

    (1998)
  • D. Cooper et al.

    Suppressed PHA activation of T lymphocytes in simulated microgravity is restored by direct activation of protein kinase C

    J. Leukoc. Biol.

    (1998)
  • B.B. Hashemi et al.

    T cell activation responses are differentially regulated during clinorotation and in spaceflight

    FASEB J.

    (1999)
  • Cited by (0)

    View full text