Skip to main content
Journal of Applied Physiology logoLink to Journal of Applied Physiology
. 2018 Nov 29;126(2):469–476. doi: 10.1152/japplphysiol.00789.2018

B cell homeostasis is maintained during long-duration spaceflight

Guillaume Spielmann 1,, Nadia Agha 2, Hawley Kunz 2,3, Richard J Simpson 2,4,5,6, Brian Crucian 7, Satish Mehta 7, Mitzi Laughlin 2,8, John Campbell 1,9
PMCID: PMC6397409  PMID: 30496712

Abstract

Long-duration spaceflights reportedly induce immune dysregulation, which is considered a risk to astronaut safety and mission success. Recent studies have examined the impact of spaceflight on markers of adaptive and innate immunity, but no study, to date, has comprehensively evaluated humoral immunity and serological markers of B cell function. The aim of this study was to characterize changes in B cell numbers and phenotypes, along with plasma Igs and polyclonal free light chains (FLCs)—near-“real-time” biomarkers of Ig synthesis—in response to an ~6-mo mission to the International Space Station (ISS). Whole-blood samples were collected before flight, during flight (“Early flight,” “Mid-flight,” and “Late flight”), immediately upon return, and during a recovery period (R + 18, R + 30/R + 33, and R + 60/R + 66) from 23 ISS crew members. B Cell counts and phenotypes were measured throughout the duration of the mission, along with total plasma Ig and FLC levels. There was no effect of spaceflight on the number and proportion of the different B cell subsets. There was no difference in kappa FLC between preflight samples and either in-flight or recovery samples (P > 0.05), and only a marginal reduction was observed in lambda FLC levels upon return to Earth (P < 0.05). Furthermore, IgG and IgM remained unchanged during and after spaceflight compared with preflight values (P > 0.05). Of note, plasma IgA concentrations were elevated in-flight compared with baseline and recovery values (P < 0.05). These results indicate that B cell homeostasis is maintained during long-duration spaceflight, advocating for potential in-flight vaccination as viable countermeasures against viral reactivation during exploration-class missions.

Keywords: B cell homeostasis, free light chains, immunoglobulins, long-duration spaceflight

INTRODUCTION

Long-duration spaceflights are associated with alterations to both innate and adaptive immunity (11, 24), including impaired cytokine response to antigenic stimuli (8, 49), natural killer (NK) cell cytotoxicity (46), neutrophil and monocyte oxidative burst capacity (30, 31), and lymphocyte distribution and proliferative capacity (40). As exposure to pathogens (45) and rate of latent viral reactivations (35, 36) are known to increase in space, profound immune dysregulation would likely have great clinical and operational significance during perennial missions. Logistical constraints unique to spaceflight have compelled the majority of space immunology research to be collected using short-duration missions or by comparing pre- to postflight measures of immune function. Whereas these studies provided valuable insights on the immune status of astronauts upon return on Earth, the magnitude of immune alterations during spaceflight is less clear. Furthermore, few studies have attempted to characterize comprehensively B cell and plasma cell homeostasis during orbital missions.

Effective humoral immunity is of fundamental importance to ensure adequate destruction of extracellular pathogens and the control of intracellular viral infections. As such, humoral immunity relies on the induction of different effector functions to antibody production, and any alteration in B cell production, ability to differentiate, and Ig output from plasma cells could lead to profound immune dysregulation and potentially endanger crew safety. The use of ground-based analogs to spaceflight, including hind-limb unloading, has highlighted significant reductions in B cell progenitor cells in the bone marrow of mice that led to reduced B lymphopoiesis (32). Furthermore, short-duration spaceflight missions have been shown to impact negatively cell phenotypes in the bone marrow (41) and splenic B cell counts in mice (21), whereas simulated microgravity reduces the hematopoietic stem cell proliferation rate (42). Although these changes appear to promote reductions in Ig output in animals following spaceflight (33), without affecting the breadth of the Ig repertoire (58), they may not translate to humans, as preflight Ig levels remain unchanged in cosmonauts following 6 mo in the International Space Station (ISS) (46). With the suggestion that exploration crew members receive in-flight vaccination against certain latent herpesviruses, to maintain optimal protection throughout the duration of an ~3-yr mission (13), optimal B cell homeostasis is likely to be of paramount importance to preserve vaccine response and crew member health. Unfortunately, however, those studies failed to measure in-flight changes in B cell phenotypes and Ig concentrations.

A limitation of the measurement of intact, circulating Igs, such as total plasma IgG, IgA, and IgM concentrations, is their slow clearance via cellular catabolism that confers them a relatively long biological half-life (1–3 wk) (16). As this limits assessment of shorter-term changes to Ig production, plasma free light chains (FLCs), with a short half-life of 2–6 h (16), are conventionally used for the diagnosis, prognostication, and monitoring of plasma cell dyscrasias (43, 44). In this context, plasma Ig FLCs are considered to be a sensitive barometer of plasma cell activation and immune competency (28, 39). Ig FLCs are produced by activated plasma cells during antibody synthesis at a rate influenced by the magnitude of immune activation (28, 57). Elevated FLC levels are indicative of inflammation and have been associated with metabolic disorders and type 2 diabetes (4, 27), chronic low-grade inflammation (6), myeloma (53), and mortality in the general population (17), whereas low levels identify immune suppression (28). Consequently, plasma Ig FLCs are likely the ideal candidates to detect early immune suppression in astronauts, and the characterization of the effects of long-duration spaceflight on plasma Ig FLC is of paramount importance.

The successful implementation of exploration-class missions to Mars or other near-Earth objects requires a better understanding of the impact of long-duration spaceflight on the immune system to evaluate the risks of crew adverse health events associated with immune dysregulations. The aim of this study was to assess the impact of long-duration spaceflight on B cell and plasma cell homeostasis by comprehensively analyzing changes in B cell number, phenotype, and soluble markers of humoral function during a 6-mo mission in the ISS.

MATERIALS AND METHODS

Subjects and Study Design

Data for this study were collected from two independent [National Aeronautics and Space Administration (NASA)]-funded studies: The “Integrated Immune” study, conducted at NASA–Johnson Space Center (JSC; B.C.) and the “Salivary Markers” study conducted at the University of Houston (R.J.S.). A total of 23 ISS crew members (15 from Integrated Immune and eight from Salivary Markers), ranging in age from 37 to 57 yr old (three women, age 47.1 ± 5.6 yr) were enrolled in this study. The ISS crew was affiliated with NASA, European Space Agency, Japanese Aerospace Exploration Agency, or Canadian Space Agency and participated in a 6-mo mission to the ISS. Data were collected over 18 separate ISS missions. Additionally, six ground-based controls (one woman, age 33.0 ± 7.1 yr) were enrolled in this study, as part of Salivary Markers, to ensure assay validity. The Institutional Review Board at Louisiana State University, the University of Houston, and NASA–JSC approved the study, and informed consent was obtained from all subjects.

Sample Collection

For both studies, plasma samples were collected in lithium heparin tubes, and whole-blood samples were collected in acid-citrate dextrose (ACD) tubes for cellular phenotyping during the Salivary Markers study (Vacutainer; Becton Dickinson, Franklin Lakes, NJ), before, during, and after the 6-mo mission in the ISS.

Specifically, plasma samples from the Integrated Immune study were collected at 180 and 45 days before launch (L − 180 and L − 45), 10 days after launch [“Early”; flight day (FD)-10], “Mid” (FD-90), last day in space [“Late”; FD-180/return (R) − 1], upon return on Earth (R + 0), and R + 30 from 15 ISS crew members. Plasma samples collected during the Salivary Markers study were drawn at L − 180, L − 60, FD-10, FD-90, FD-180/R − 1, R + 0, R + 18, R + 33, and R + 66 from eight astronauts and six ground-based controls. Blood samples used for B cell phenotyping were collected during Salivary Markers on eight ISS crew members in a Vacutainer, supplemented with an acidified glucose nutrient solution (ACD) to maintain optimal cellular viability during sample return on Earth. Technical constraints did not allow for timely return of whole blood on Earth at FD-10, and consequently, no B cell phenotype data were measured at that time point.

A temperature-controlled environment (temperature range: 6–24°C) was maintained during the 24–36 h of sample storage/transport to the laboratories at NASA–JSC and the University of Houston. Upon arrival to the respective laboratories, the plasma samples were centrifugally separated from whole blood and stored at −80°C, and B cell phenotype was characterized using whole-blood samples from the ACD tubes. Cryopreserved plasma samples were transported to the ImmunoEnergetics Laboratory at Louisiana State University, and freshly isolated B cell phenotypes were characterized on a BD Accuri C6 flow cytometer (BD Biosciences, San Jose, CA) at the University of Houston. Lymphocyte counts were determined using a BC-3200 auto hematology analyzer (Mindray, Mahwah, NJ).

Urine and saliva samples were collected during the Integrated Immune study to determine cytomegalovirus (CMV), Epstein-Barr virus (EBV), and varicella zoster virus (VZV) viral load in astronauts throughout the mission; results are reported in Mehta et al. (35, 36). In brief, aliquots of urine were sampled from a 24-h urine pool at each of the aforementioned time points, and fasting saliva samples were collected using sterile Salivette cotton rolls (Sardstedt, Newton, NC) immediately upon awakening. The urine samples were frozen until return to Earth, whereas the Salivette was stored in stability buffer (0.5% SDS, 10 mM Tris-Cl, and 1 mM EDTA) at room temperature for up to 2 wk before return to Earth for subsequent analysis (37). Viral DNA was extracted and quantified by Mehta et al. (38) at NASA–JSC by PCR, as described previously.

B Cell Phenotyping

B Cells were labeled with directly conjugated MAb and analyzed using four-color flow cytometry, as previously described (54). Briefly, aliquots of 50 µl whole blood were incubated with 5 µl each MAb for 30 min at room temperature in the dark. The following MAb were used to stain the cells: anti-CD20-FITC Clone #LT20 and anti-CD43-phycoerythrin (PE) Clone #84-3C1 (eBioscience, San Diego, CA); anti-IgD-PE Clone #IA6-2 and anti-IgM-PE Clone #G20-127 (BD PharMingen, San Diego, CA); anti-IgG-PE Clone #H2 (SouthernBiotech, Birmingham, AL); and anti-CD27-peridinin-chlorophyll Clone #0323, anti-CD25-allophycocyanin (APC) Clone #CD25-4E3, anti-CD5-APC Clone #L17F12, and anti-CD38-APC Clone #HIT2 (eBioscience). With the use of these MAb, CD20+ B cells were classified into immature (CD20+/CD43/CD27/IgD), naive/transitional (CD20+/CD43/CD27/IgD+), and memory B cells (CD20+/CD43+/CD27+/CD38); plasmablasts/plasma cells (CD20+/CD43+/CD27+/CD38hi); B1 cells (CD20+/CD43+/CD27+/CD5); regulatory B cells (CD20+/CD43+/CD27/CD5+); IgG+ memory B cells (CD20+/CD27+/IgG+); and IgM+ B cells (CD20+/CD27+/IgM+; Table 1). Following incubation, erythrocytes were lysed by increased osmotic pressure, induced by the addition of 500 µl red blood cell lysis buffer (eBioscience) for 20 min at room temperature. Samples were then washed twice with PBS and resuspended in 250 µl PBS to be analyzed on a BD Accuri C6 flow cytometer. Flow cytometry analysis was conducted on the BD Accuri proprietary flow cytometry analysis software. Total cell numbers of each B cell subset were determined by the multiplication of the percentages of cells expressing each marker of interest by the total lymphocyte count.

Table 1.

Phenotypic characterization of the different B cell subsets

Cell Type Phenotype Reference
B Cells CD20+ 31a
Immature B cells CD20+/CD43/CD27/IgD 28a, 47a, 59a
Naïve/transitional B cells CD20+/CD43/CD27/IgD+ 28a, 47a, 59a
IgM+ B cells CD20+/CD27+/IgM+ 4a
Memory B cells CD20+/CD43+/CD27+/CD38 42a
IgG+ memory B cells CD20+/CD27+/IgG+ 4a
B1 cells CD20+/CD43+/CD27+/CD5 22a
Regulatory B cells CD20+/CD43+/CD27/CD5+ 34a
Plasmablasts/plasma cells CD20+/CD43+/CD27+/CD38hi 42a

Ig Analyses

Plasma samples from the 23 crew members (Integrated Immune, n = 15; Salivary Markers, n = 8) were thawed, and a total volume of 100 μl was analyzed for kappa (κ) and lambda (λ) FLCs using commercially available ELISAs (Seralite; Abingdon Health, Oxford, UK), using previously published methods (7). Briefly, diluted plasma samples were incubated in 96-well plates, precoated with either anti-κ or anti-λ FLC MAb at room temperature for 60 min. Following initial incubation, the wells were washed four consecutive times and incubated with horseradish peroxidase-labeled anti-κ or anti-λ detection antibody for 30 min at room temperature. After another washing step, the presence of κ or λ FLC in the plasma samples was detected using a colorimetric reaction and read on a SpectraMax i3× plate reader (Molecular Devices, San Jose, CA). The color intensity was directly correlated with the κ and λ FLC concentration in the samples. Plasma cystatin C was measured with a commercially available ELISA (R&D Systems, Minneapolis, MN) and used to calculate estimated glomerular filtration rate (eGFR) throughout the missions, based on an established algorithm (47). This estimate of renal function was used to account for changes in renal clearance of FLC in response to spaceflight.

Total IgA, IgM, and IgG were measured in a total of 150 μl thawed plasma sample from all astronauts and corresponding controls using commercially available ELISA kits (eBioscience).

Statistical Analysis

A longitudinal, repeated-measures design was used to determine the effects of long-term exposure to microgravity on proportions and numbers of the different B cell subsets, κ and λ FLCs, and Ig levels. Linear mixed models were used to evaluate potential differences in the main and interaction effects of time (L − 180, L − 45 to 60, early flight, mid-flight, late flight, R + 0, R + 1, R + 18, R + 30 to 33, and R + 66) after controlling for a potential change in eGFR. When a significant time effect was observed, post hoc tests were performed with Bonferroni correction. Data are presented as means ± SE. All statistical analyses were performed using SPSS version 24 (IBM, Armonk, NY), and significance was set at P < 0.05.

RESULTS

Effect of Long-Duration Spaceflight on Circulating B Cell Subsets

Cell counts for the different B cell subsets isolated from crew members throughout the Salivary Markers study are presented in Table 2. There was no change in the percentages of total B cells within the lymphocyte population or in the total number of B cells during the 6-mo mission (FB cell frequency = 1.603, pB cell frequency = 0.142; FB cell count = 0.248, pB cell count = 0.972), where F is the F-statistic, and p is the P value. Furthermore, long-duration spaceflight was not associated with any statistically significant change in plasma cells, immature, naïve/transitional, memory, and regulatory B cells; and B1 cells (P > 0.05) in crew members.

Table 2.

Changes in the numbers (cells/100 µl) of B lymphocyte lineage cells in 6-mo ISS crew members (n = 8)

Sample Time Point
L − 180 L − 60 FD-90/Mid-Flight R − 1/Late Flight R + 0 R + 18 R + 33 R + 66 Main Effect of Time F-Statistic (P value)
Total B cells, cells/100 µl
ISS crew members (n = 8) 12,128 ± 4,080 15,316 ± 3,036 21,892 ± 12,747 24,920 ± 14,955 19,865 ± 13,955 14,279 ± 6,157 13,831 ± 3,272 11,856 ± 4,376 0.248 (0.972)
% Lymphocytes
ISS crew members (n = 8) 10 ± 4 12 ± 3 13 ± 4 11 ± 4 13 ± 7 11 ± 5 11 ± 4 9 ± 4 1.603 (0.142)
Immature B cells
ISS crew members (n = 8) 482 ± 271 625 ± 568 2,618 ± 2,936 2,508 ± 3,785 2,322 ± 3,534 1,022 ± 1,233 866 ± 768 602 ± 645 0.665 (0.702)
Naïve/transitional B cells
ISS crew members (n = 8) 7,091 ± 3,751 9,787 ± 4,250 11,083 ± 5,605 12,896 ± 6,237 11,196 ± 7,842 7,855 ± 3,706 8,045 ± 3,292 7,044 ± 3,447 0.626 (0.733)
IgM+ B cells
ISS crew members (n = 8) 53 ± 155 11 ± 13 16 ± 22 29 ± 45 34 ± 46 6 ± 9 10 ± 11 11 ± 11 0.702 (0.670)
Memory B cells
ISS crew members (n = 8) 179 ± 158 208 ± 133 327 ± 243 409 ± 310 381 ± 327 202 ± 141 394 ± 477 196 ± 122 1.878 (0.080)
IgG+ memory B cells
ISS crew members (n = 8) 74 ± 177 38 ± 29 67 ± 63 41 ± 46 42 ± 43 57 ± 95 61 ± 73 28 ± 21 1.004 (0.432)
B1 cells
ISS crew members (n = 8) 123 ± 117 211 ± 256 225 ± 177 283 ± 357 242 ± 216 211 ± 171 167 ± 221 306 ± 383 0.932 (0.485)
Regulatory B cells
ISS crew members (n = 8) 129 ± 104 177 ± 64 288 ± 240 269 ± 151 312 ± 494 152 ± 51 252 ± 206 202 ± 124 0.943 (0.476)
Plasmablasts/plasma cells
ISS crew members (n = 8) 104 ± 86 178 ± 155 169 ± 102 197 ± 177 185 ± 202 161 ± 157 99 ± 93 173 ± 233 0.225 (0.979)

Average values are presented ± SD. FD-90, 90 days after launch (flight day); ISS, International Space Station; L − 180/L − 60, 180 and 60 days before launch; R − 1, last day in space before return (R).

Long-Duration Spaceflight and Blood Igs

Plasma intact Ig concentration during a 6-mo mission in the ISS.

There was no change in plasma IgG and IgM concentrations in astronauts throughout the mission (P > 0.05). The impact of long-duration spaceflight on total plasma Ig concentrations is presented in Table 3. Astronauts exhibited an increase in plasma IgA during flight compared with baseline values (L − 60/45; F = 7.077; P < 0.001). Upon return on Earth, plasma IgA concentrations decreased from in-flight levels and were back to preflight values (L − 60/L − 45) during recovery (R + 30; P = 0.047). All changes withstood adjustment for latent viral reactivation status and DNA load, along with eGFR.

Table 3.

Changes in plasma IgA, IgM, and IgG in astronauts before, during, and following 6 mo in the ISS (n = 23)

L − 180 L − 60/L − 45 Early Flight FD-90/Mid-Flight R − 1/Late Flight R + 0 R + 18 R + 30/R + 33 R + 66
IgA (mg/dl) ± SD Crew members (n = 23) 131.51 ± 63.73 112.73 ± 49.64 126.60 ± 58.05* 136.38 ± 68.12* 140.58 ± 75.08* 126.64 ± 59.56 103.14 ± 32.67 112.46 ± 55.11 101.73 ± 27.51
IgG (mg/dl) ± SD Crew members (n = 23) 1,218.41 ± 231.66 1,190.41 ± 253.88 1,181.47 ± 310.80 1,235.41 ± 277.07 1,249.98 ± 298.99 1,184.38 ± 209.64 1,359.33 ± 304.05 1,226.67 ± 399.88 1,267.50 ± 255.92
IgM (mg/dl) ± SD Crew members (n = 23) 373.51 ± 546.67 408.84 ± 530.97 476.08 ± 665.62 422.57 ± 476.36 410.57 ± 491.51 346.95 ± 360.77 651.04 ± 751.93 295.78 ± 268.07 471.20 ± 324.15

Average values are presented ± SD. FD-90, 90 days after launch (flight day); ISS, International Space Station; L − 180/L − 60/L− 45, 180 and 60/45 days before launch; R − 1, last day in space before return (R).

*

P < 0.05, significant differences from baseline preflight values (L − 180 and L − 60/L − 45).

Plasma FLC concentration during a 6-mo mission in the ISS.

The effects of long-duration spaceflight on κ and λ FLCs, along with the ratio of κ/λ and total FLCs, are presented in Fig. 1. There was no effect of spaceflight on plasma κ FLC (P > 0.05), and only a minor decrease in the concentration of plasma λ FLC was observed immediately upon return on Earth (R + 0) in crew members compared with in-flight plasma λ FLC concentrations (Early: P = 0.03; Mid: P = 0.005; Late/R − 1: P = 0.012). The preferential reduction in plasma λ FLC at landing without any change in plasma κ FLC concentration led to a minor decrease in the κ/λ ratio at the mid-flight time point compared with baseline L − 60/L − 45 and return R + 0 and R + 30 κ/λ ratio (pL − 45 = 0.029; pR + 0 = 0.037; pR + 18 = 0.053 ; pR + 33 = 0.037). As plasma FLC levels can be impacted by altered production from plasma cells and/or impaired clearance from renal metabolism, cystatin C was measured to calculate eGFR (1) and account for variation in renal function during spaceflight. There was no change in kidney function during flight (P > 0.05); however, postflight eGFR values (R + 30) were significantly lower than preflight values (L − 60/L − 45; P = 0.015). Subtle changes in plasma FLC withstood adjustment for eGFR.

Fig. 1.

Fig. 1.

Changes in kappa (A) and lambda (B) free light chains (FLC) in 6-mo International Space Station astronauts (n = 23). The ratio of kappa/lambda and total FLC are presented in C and D, respectively. Mean values are presented ± SE. Significant differences between immediately postflight values (R + 0; *P < 0.05) and baseline values (L − 45; aP < 0.05). L, days before launch; Early, 10 days after launch; Mid, 90 days after launch; Late, last day in space; R, days after return on Earth. Horizontal lines represent where the significant differences are located.

In related analyses, we explored whether latent viral reactivation was associated with FLC levels in blood. CMV, EBV, and VZV reactivation status and viral DNA load from a subset of samples (Integrated Immune study) were included in the model as covariates. The rate and magnitude of latent viral reactivation observed in the Integrated Immune cohort are described elsewhere (51). In brief, 47% (7/15) of astronauts exhibited CMV reactivation at any point during the mission, 73% (11/15) of astronauts exhibited EBV reactivation at any point during the mission, and 60% (9/15) of astronauts exhibited VZV reactivation at any point during the mission. There was no difference in plasma κ and λ FLC concentrations in astronauts who exhibited CMV and EBV reactivation at any point during the mission (P > 0.05), and there was no association between CMV and EBV DNA load and plasma FLC at any time point (P > 0.05). When VZV DNA load was included in the model, the greater plasma λ FLC concentration observed in-flight compared with postflight values was associated with the magnitude of VZV reactivation (FVZV DNA load = 4.937; pVZV DNA load = 0.029).

DISCUSSION

The rapid progress made in aerospace technologies over the past 50 yr has dramatically increased the scope and duration of manned space-exploration missions. In addition to the novel technological challenges, safe implementation of exploration-class missions requires a profound understanding of the impact that prolonged exposure to space environments has on astronauts’ biology and health. In particular, numerous studies have raised concerns about the potential clinical risks associated with reported immune impairments observed during spaceflight (10, 11, 14, 50), such as reduced T cell (25, 26, 34) and NK cell functions (46), sustained production of proinflammatory cytokines (15, 35), altered neutrophil and monocyte microbicidal activity (30, 31), diminished antimicrobial protein concentration, and increased rate and magnitude of latent viral reactivation (36, 51). However, no study, to date, has attempted to characterize comprehensively the impact of long-duration spaceflight on humoral immunity in humans. The goal of this study was to identify changes in a broad range of phenotypically distinct B cell subsets, along with secreted Igs and FLCs in astronauts who completed 6 mo on the ISS. We found no change in the number of total B cells in response to spaceflight; however, there was a trend for increased memory B cells during spaceflight compared with baseline values. Contrary to supposition, only marginal changes were observed in soluble biomarkers of B cell homeostasis during spaceflight. Indeed, we found no effect of spaceflight on plasma κ FLC, IgG, and IgM levels and only modest changes to plasma λ FLC concentrations. Interestingly, long-duration spaceflight increased plasma IgA levels, which with the consideration of the correlation between mucosal and plasma IgA (55), may represent alterations in mucosal immunity.

Spaceflight has been shown to alter human and animal leukocyte distribution (12, 48), but the majority of studies was focused on T cells and NK cells (22, 46) and was limited to either short-duration missions (8–15 days) (23, 52) or to comparisons between pre- and postflight. Our data are in accordance with previous findings showing that total B cell counts and proportion within the lymphocyte compartment are not affected by long-duration spaceflight (46). However, the more advanced and detailed phenotypic analysis performed in this study highlighted for the first time that spaceflight does not have a detrimental impact on the phenotypic composition of the B cell compartment. Indeed, memory B cell counts changed modestly in-flight, whereas the number of naive/transitional and regulatory B cells remained unchanged during the mission. Animal studies have shown that short-duration spaceflight (41) and ground-based analogs to spaceflight, such as hind-limb unloading (32), had deleterious impacts on immune cell phenotypes in the bone marrow and led to a reduction in de novo generation of B cells (32) and total splenic B cell numbers (21). As both bone marrow progenitor cells (18) and B cells are known to respond to acute stressors, such as intense exercise (54), it can be hypothesized that the changes in B cell lymphopoiesis, observed in animals immediately after short-duration spaceflight, may be due to a variety of factors other than microgravity, including landing-associated stressors, rather than exposure to the space exposome.

Effective humoral responses rely on B cell activation, differentiation, and antibody output. Although there was no effect of spaceflight on the number of circulating plasma cells in astronauts, it is interesting to note that they had a greater amount of B1 cells than ground-based controls (data not shown). As B1 cells have recently been characterized as preplasmablasts (9), a greater circulating number could be associated with the increased capability to produce antibodies following activation. Conflicting data exist on the effect of spaceflight on Ig outputs, with some studies showing a reduction in in vitro IgG production in response to altered gravity (19), whereas others showed no change from baseline in IgG production after a 10- to 15-day flight (52, 56) and no alteration in the B cell repertoire of unimmunized animals (58). Our data are in partial agreement with the current literature, as we found no difference in total plasma IgG and IgM at any point in the mission, even after controlling for latent viral reactivation and DNA load. Interestingly, however, astronauts exhibited an increase in total plasma IgA during flight. Elevated plasma IgA has been observed in the urodele amphibian Pleurodeles waltl, exposed to microgravity for 6 mo (5), but also in rodents and human subjects under chronic psychological and physiological stress (20, 60), similar to those experienced by the astronauts on board the ISS. Furthermore, although the in-flight increases in plasma IgA concentrations were statistically significant compared with baseline values, astronauts’ plasma IgA levels did not exceed the normal clinical range observed on Earth (59). Consequently, the fluctuations in plasma IgA observed in this study are likely attributable to long-duration exposure to spaceflight-associated psychological and operative stressors rather than to alterations in B cell function.

The immune impairments associated with prolonged exposure to myriad stressors specific to the spaceflight environment are known to result in increased latent CMV, EBV, and VZV reactivation (35, 36, 51). However, no study, to date, has attempted to determine whether potential alterations in B cell activation and function could play a role in latent viral reactivations in space. Whereas our data did not show any biologically relevant change in total plasma Ig concentrations in response to spaceflight, it could be argued that the relatively long half-life of intact plasma Ig (?21 days) curtails their sensitivity at detecting changes in B cell function (16). As such, we sought to determine changes in κ and λ Ig FLCs, a more sensitive barometer of immune competency with short half-life (κ: 2–4 h; λ: 3–6 h), due to rapid renal clearance (16). Plasma κ FLC levels remained unchanged during the missions; however, there was a slight decrease in the concentration of plasma λ FLC immediately upon return on Earth (R + 0) in crew members compared with in-flight plasma λ FLC concentrations. Whereas this reduction in λ FLC at R + 0 compared with in-flight values reached statistical significance, no crew member reached clinically significant low levels of λ FLC (29). As there was no change in kidney function in-flight, this preferential reduction in plasma λ FLC is not due to increased renal clearance. However, whereas there was no effect of CMV and EBV reactivation on λ FLC concentrations, the magnitude of VZV reactivation during flight was associated with the greater levels of λ FLC observed at the same time points. This discrepancy, in response, between plasma κ and λ FLC could be explained by the difference in size between both light chains. Indeed, although κ FLCs are produced in greater quantities than λ FLCs during antibody synthesis (57), the larger size of the dimeric λ FLCs impedes their clearance by the kidney (16). Our results suggest that the B cell ability to produce FLCs and consequently, Igs in response to a viral or vaccine challenge in space remains intact. It should, however, be noted that whereas we measured the quantity of plasma κ and λ FLCs and total Igs in crew members, we did not characterize the quality of these antibodies. Animal studies have indeed shown that whereas similar quantities of antibodies could be produced in response to antigenic challenge in space (3), the quality of the produced antibodies remained inferior to those produced on Earth (2). Consequently, although the present study shows that B cell homeostasis appears to be preserved during long-duration spaceflight, it could be hypothesized that suboptimal immune responses could still be observed upon antigenic challenge in space.

One limitation of our study lies in its retrospective nature. These results were obtained from samples collected during two previously completed NASA studies, and all plasma Ig and FLC analyses were performed on previously archived samples. As such, we were unable to measure the antibody response to a specific vaccine but rather, measured B cell homeostasis during long-duration spaceflight. Furthermore, the changes in B cell number and phenotype during spaceflight were only characterized on the Salivary Markers cohort (n = 8). Technical constraints unique to spaceflight research prevented the immediate analysis of the isolated B cell populations. To mitigate this limitation, we performed validation work in our laboratory before this study and found that B cell number and phenotype remained unaltered for 48 h when collected from a Vacutainer supplemented with an acidified glucose nutrient solution. Finally, we also collected blood samples from ground-based controls to parallel samples collected on the ISS, thus controlling for any potential ex vivo aging of the blood samples. Unfortunately however, as the ground-based control subjects were recruited to ensure assay validity, rather than to serve as paired controls with each astronaut, they were not age or sex matched with the different crew members. Future studies should attempt to recruit populations of ground-based controls that closely match the astronaut population to reduce further the effects of various confounding factors (age, sex, fitness level, etc.) on the observed changes in immune function during spaceflight.

In conclusion, this is the first study to show comprehensively that long-duration spaceflight in human astronauts has no—or very limited—effect on B cell number, phenotype, and antibody output. These important results suggest that plasma immune competency is maintained in microgravity and that future in-flight vaccine-based countermeasures are likely to be efficient at further protecting astronauts from immune dysregulation and symptomatic latent viral reactivations during prolonged exploration-class missions.

GRANTS

Funding for this study was provided by grants from NASA (Omnibus Grant NNX17AB16G to G. Spielmann and Grants SMO 015 to B. Crucian and NNX12AB48G to R. J. Simpson).

DISCLOSURES

J. Campbell reports minor shares in Abingdon Health, the company that produces one of the assay (FLC ELISA Kits) used in this project.

AUTHOR CONTRIBUTIONS

G.S. and J.C. conceived and designed research; G.S., N.A., H.K., R.J.S., B.C., S.M., M.L., and J.C. performed experiments; G.S., N.A., H.K., R.J.S., S.M., M.L., and J.C. analyzed data; G.S., N.A., H.K., and J.C. interpreted results of experiments; G.S. prepared figures; G.S. and B.C. drafted manuscript; G.S., N.A., H.K., R.J.S., B.C., S.M., M.L., and J.C. edited and revised manuscript; G.S., N.A., H.K., R.J.S., B.C., S.M., M.L., and J.C. approved final version of manuscript.

REFERENCES

  • 1.Aulakh NK, Bansal E, Bose A, Aulakh GS, Aulakh BS, Singh MR. Can cystatin C become an easy and reliable tool for anesthesiologists to calculate glomerular filtration rate? J Anaesthesiol Clin Pharmacol 31: 44–48, 2015. doi: 10.4103/0970-9185.150538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bascove M, Guéguinou N, Schaerlinger B, Gauquelin-Koch G, Frippiat JP. Decrease in antibody somatic hypermutation frequency under extreme, extended spaceflight conditions. FASEB J 25: 2947–2955, 2011. doi: 10.1096/fj.11-185215. [DOI] [PubMed] [Google Scholar]
  • 3.Bascove M, Huin-Schohn C, Guéguinou N, Tschirhart E, Frippiat JP. Spaceflight-associated changes in immunoglobulin VH gene expression in the amphibian Pleurodeles waltl. FASEB J 23: 1607–1615, 2009. doi: 10.1096/fj.08-121327. [DOI] [PubMed] [Google Scholar]
  • 4.Bellary S, Faint JM, Assi LK, Hutchison CA, Harding SJ, Raymond NT, Barnett AH. Elevated serum free light chains predict cardiovascular events in type 2 diabetes. Diabetes Care 37: 2028–2030, 2014. doi: 10.2337/dc13-2227. [DOI] [PubMed] [Google Scholar]
  • 4a.Berkowska MA, Driessen GJ, Bikos V, Grosserichter-Wagener C, Stamatopoulos K, Cerutti A, He B, Biermann K, Lange JF, van der Burg M, van Dongen JJ, van Zelm MC. Human memory B cells originate from three distinct germinal center-dependent and -independent maturation pathways. Blood 118: 2150–2158, 2011. doi: 10.1182/blood-2011-04-345579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Boxio R, Dournon C, Frippiat JP. Effects of a long-term spaceflight on immunoglobulin heavy chains of the urodele amphibian Pleurodeles waltl. J Appl Physiol (1985) 98: 905–910, 2005. doi: 10.1152/japplphysiol.00957.2004. [DOI] [PubMed] [Google Scholar]
  • 6.Brebner JA, Stockley RA. Polyclonal free light chains: a biomarker of inflammatory disease or treatment target? F1000 Med Rep 5: 4, 2013. doi: 10.3410/M5-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Campbell J, Heaney J, Gleeson M, He C, Killer S, Svendson S, Taylor I, Phillips AC, Drayson M. Salivary Immunoglobulin Free Light Chains: Reference Ranges in Younger and Older Adults and Responses to Exercise. Vienna, Austria: International Society of Exercise Immunology, 2015. [PubMed] [Google Scholar]
  • 8.Chapes SK, Morrison DR, Guikema JA, Lewis ML, Spooner BS. Production and action of cytokines in space. Adv Space Res 14: 5–9, 1994. doi: 10.1016/0273-1177(94)90380-8. [DOI] [PubMed] [Google Scholar]
  • 9.Covens K, Verbinnen B, Geukens N, Meyts I, Schuit F, Van Lommel L, Jacquemin M, Bossuyt X. Characterization of proposed human B-1 cells reveals pre-plasmablast phenotype. Blood 121: 5176–5183, 2013. doi: 10.1182/blood-2012-12-471953. [DOI] [PubMed] [Google Scholar]
  • 10.Crucian B, Johnston S, Mehta S, Stowe R, Uchakin P, Quiriarte H, Pierson D, Laudenslager ML, Sams C. A case of persistent skin rash and rhinitis with immune system dysregulation onboard the International Space Station. J Allergy Clin Immunol Pract 4: 759–762.e8, 2016. doi: 10.1016/j.jaip.2015.12.021. [DOI] [PubMed] [Google Scholar]
  • 11.Crucian B, Sams C. Immune system dysregulation during spaceflight: clinical risk for exploration-class missions. J Leukoc Biol 86: 1017–1018, 2009. doi: 10.1189/jlb.0709500. [DOI] [PubMed] [Google Scholar]
  • 12.Crucian B, Stowe RP, Mehta S, Quiriarte H, Pierson D, Sams C. Alterations in adaptive immunity persist during long-duration spaceflight. NPJ Microgravity 1: 15013, 2015. doi: 10.1038/npjmgrav.2015.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Crucian BE, Choukèr A, Simpson RJ, Mehta S, Marshall G, Smith SM, Zwart SR, Heer M, Ponomarev S, Whitmire A, Frippiat JP, Douglas GL, Lorenzi H, Buchheim JI, Makedonas G, Ginsburg GS, Ott CM, Pierson DL, Krieger SS, Baecker N, Sams C. Immune system dysregulation during spaceflight: potential countermeasures for deep space exploration missions. Front Immunol 9: 1437, 2018. doi: 10.3389/fimmu.2018.01437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Crucian BE, Stowe RP, Pierson DL, Sams CF. Immune system dysregulation following short- vs long-duration spaceflight. Aviat Space Environ Med 79: 835–843, 2008. doi: 10.3357/ASEM.2276.2008. [DOI] [PubMed] [Google Scholar]
  • 15.Crucian BE, Zwart SR, Mehta S, Uchakin P, Quiriarte HD, Pierson D, Sams CF, Smith SM. Plasma cytokine concentrations indicate that in vivo hormonal regulation of immunity is altered during long-duration spaceflight. J Interferon Cytokine Res 34: 778–786, 2014. doi: 10.1089/jir.2013.0129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Davids MS, Murali MR, Kuter DJ. Serum free light chain analysis. Am J Hematol 85: 787–790, 2010. doi: 10.1002/ajh.21815. [DOI] [PubMed] [Google Scholar]
  • 17.Dispenzieri A, Katzmann JA, Kyle RA, Larson DR, Therneau TM, Colby CL, Clark RJ, Mead GP, Kumar S, Melton LJ III, Rajkumar SV. Use of nonclonal serum immunoglobulin free light chains to predict overall survival in the general population. Mayo Clin Proc 87: 517–523, 2012. doi: 10.1016/j.mayocp.2012.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Emmons R, Niemiro GM, Owolabi O, De Lisio M. Acute exercise mobilizes hematopoietic stem and progenitor cells and alters the mesenchymal stromal cell secretome. J Appl Physiol (1985) 120: 624–632, 2016. doi: 10.1152/japplphysiol.00925.2015. [DOI] [PubMed] [Google Scholar]
  • 19.Fitzgerald W, Chen S, Walz C, Zimmerberg J, Margolis L, Grivel JC. Immune suppression of human lymphoid tissues and cells in rotating suspension culture and onboard the International Space Station. In Vitro Cell Dev Biol Anim 45: 622–632, 2009. doi: 10.1007/s11626-009-9225-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Gaignier F, Legrand-Frossi C, Stragier E, Mathiot J, Merlin JL, Cohen-Salmon C, Lanfumey L, Frippiat JP. A model of chronic exposure to unpredictable mild socio-environmental stressors replicates some spaceflight-induced immunological changes. Front Physiol 9: 514, 2018. doi: 10.3389/fphys.2018.00514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gridley DS, Pecaut MJ. Changes in the distribution and function of leukocytes after whole-body iron ion irradiation. J Radiat Res (Tokyo) 57: 477–491, 2016. doi: 10.1093/jrr/rrw051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gridley DS, Slater JM, Luo-Owen X, Rizvi A, Chapes SK, Stodieck LS, Ferguson VL, Pecaut MJ. Spaceflight effects on T lymphocyte distribution, function and gene expression. J Appl Physiol (1985) 106: 194–202, 2009. doi: 10.1152/japplphysiol.91126.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22a.Griffin DO, Holodick NE, Rothstein TL. Human B1 cells in umbilical cord and adult peripheral blood express the novel phenotype CD20+ CD27+ CD43+ CD70−. J Exp Med 208: 67–80, 2011. [Errata in J Exp Med 208: 409, 2011; J Exp Med 208: 871, 2011; J Exp Med 208: 67, 2011.] 10.1084/jem.20101499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Grove DS, Pishak SA, Mastro AM. The effect of a 10-day space flight on the function, phenotype, and adhesion molecule expression of splenocytes and lymph node lymphocytes. Exp Cell Res 219: 102–109, 1995. doi: 10.1006/excr.1995.1210. [DOI] [PubMed] [Google Scholar]
  • 24.Guéguinou N, Huin-Schohn C, Bascove M, Bueb JL, Tschirhart E, Legrand-Frossi C, Frippiat JP. Could spaceflight-associated immune system weakening preclude the expansion of human presence beyond Earth’s orbit? J Leukoc Biol 86: 1027–1038, 2009. doi: 10.1189/jlb.0309167. [DOI] [PubMed] [Google Scholar]
  • 25.Hauschild S, Tauber S, Lauber B, Thiel CS, Layer LE, Ullrich O. T Cell regulation in microgravity—the current knowledge from in vitro experiments conducted in space, parabolic flights and ground-based facilities. Acta Astronaut 104: 365–377, 2014. doi: 10.1016/j.actaastro.2014.05.019. [DOI] [Google Scholar]
  • 26.Hughes-Fulford M, Chang TT, Martinez EM, Li CF. Spaceflight alters expression of microRNA during T-cell activation. FASEB J 29: 4893–4900, 2015. doi: 10.1096/fj.15-277392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hutchison CA, Cockwell P, Harding S, Mead GP, Bradwell AR, Barnett AH. Quantitative assessment of serum and urinary polyclonal free light chains in patients with type II diabetes: an early marker of diabetic kidney disease? Expert Opin Ther Targets 12: 667–676, 2008. doi: 10.1517/14728222.12.6.667. [DOI] [PubMed] [Google Scholar]
  • 28.Hutchison CA, Landgren O. Polyclonal immunoglobulin free light chains as a potential biomarker of immune stimulation and inflammation. Clin Chem 57: 1387–1389, 2011. doi: 10.1373/clinchem.2011.169433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28a.Inui M, Hirota S, Hirano K, Fujii H, Sugahara-Tobinai A, Ishii T, Harigae H, Takai T. Human CD43+ B cells are closely related not only to memory B cells phenotypically but also to plasmablasts developmentally in healthy individuals. Int Immunol 27: 345–355, 2015. doi: 10.1093/intimm/dxv009. [DOI] [PubMed] [Google Scholar]
  • 29.Katzmann JA, Clark RJ, Abraham RS, Bryant S, Lymp JF, Bradwell AR, Kyle RA. Serum reference intervals and diagnostic ranges for free kappa and free lambda immunoglobulin light chains: relative sensitivity for detection of monoclonal light chains. Clin Chem 48: 1437–1444, 2002. [PubMed] [Google Scholar]
  • 30.Kaur I, Simons ER, Castro VA, Mark Ott C, Pierson DL. Changes in neutrophil functions in astronauts. Brain Behav Immun 18: 443–450, 2004. doi: 10.1016/j.bbi.2003.10.005. [DOI] [PubMed] [Google Scholar]
  • 31.Kaur I, Simons ER, Castro VA, Ott CM, Pierson DL. Changes in monocyte functions of astronauts. Brain Behav Immun 19: 547–554, 2005. doi: 10.1016/j.bbi.2004.12.006. [DOI] [PubMed] [Google Scholar]
  • 31a.Leandro MJ. B-Cell subpopulations in humans and their differential susceptibility to depletion with anti-CD20 monoclonal antibodies. Arthritis Res Ther 15, Suppl 1: S3, 2013. doi: 10.1186/ar3908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lescale C, Schenten V, Djeghloul D, Bennabi M, Gaignier F, Vandamme K, Strazielle C, Kuzniak I, Petite H, Dosquet C, Frippiat JP, Goodhardt M. Hind limb unloading, a model of spaceflight conditions, leads to decreased B lymphopoiesis similar to aging. FASEB J 29: 455–463, 2015. doi: 10.1096/fj.14-259770. [DOI] [PubMed] [Google Scholar]
  • 33.Lesnyak AT, Sonnenfeld G, Rykova MP, Meshkov DO, Mastro A, Konstantinova I. Immune changes in test animals during spaceflight. J Leukoc Biol 54: 214–226, 1993. doi: 10.1002/jlb.54.3.214. [DOI] [PubMed] [Google Scholar]
  • 34.Martinez EM, Yoshida MC, Candelario TL, Hughes-Fulford M. Spaceflight and simulated microgravity cause a significant reduction of key gene expression in early T-cell activation. Am J Physiol Regul Integr Comp Physiol 308: R480–R488, 2015. doi: 10.1152/ajpregu.00449.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34a.Mauri C, Menon M. The expanding family of regulatory B cells. Int Immunol 27: 479–486, 2015. doi: 10.1093/intimm/dxv038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mehta SK, Crucian BE, Stowe RP, Simpson RJ, Ott CM, Sams CF, Pierson DL. Reactivation of latent viruses is associated with increased plasma cytokines in astronauts. Cytokine 61: 205–209, 2013. doi: 10.1016/j.cyto.2012.09.019. [DOI] [PubMed] [Google Scholar]
  • 36.Mehta SK, Laudenslager ML, Stowe RP, Crucian BE, Feiveson AH, Sams CF, Pierson DL. Latent virus reactivation in astronauts on the international space station. NPJ Microgravity 3: 11, 2017. doi: 10.1038/s41526-017-0015-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Mehta SK, Laudenslager ML, Stowe RP, Crucian BE, Sams CF, Pierson DL. Multiple latent viruses reactivate in astronauts during Space Shuttle missions. Brain Behav Immun 41: 210–217, 2014. doi: 10.1016/j.bbi.2014.05.014. [DOI] [PubMed] [Google Scholar]
  • 38.Mehta SK, Stowe RP, Feiveson AH, Tyring SK, Pierson DL. Reactivation and shedding of cytomegalovirus in astronauts during spaceflight. J Infect Dis 182: 1761–1764, 2000. doi: 10.1086/317624. [DOI] [PubMed] [Google Scholar]
  • 39.Nakano T, Matsui M, Inoue I, Awata T, Katayama S, Murakoshi T. Free immunoglobulin light chain: its biology and implications in diseases. Clin Chim Acta 412: 843–849, 2011. doi: 10.1016/j.cca.2011.03.007. [DOI] [PubMed] [Google Scholar]
  • 40.Nash PV, Konstantinova IV, Fuchs BB, Rakhmilevich AL, Lesnyak AT, Mastro AM. Effect of spaceflight on lymphocyte proliferation and interleukin-2 production. J Appl Physiol (1985) 73, Suppl: 186S–190S, 1992. doi: 10.1152/jappl.1992.73.2.S186. [DOI] [PubMed] [Google Scholar]
  • 41.Ortega MT, Pecaut MJ, Gridley DS, Stodieck LS, Ferguson V, Chapes SK. Shifts in bone marrow cell phenotypes caused by spaceflight. J Appl Physiol (1985) 106: 548–555, 2009. doi: 10.1152/japplphysiol.91138.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Plett PA, Frankovitz SM, Abonour R, Orschell-Traycoff CM. Proliferation of human hematopoietic bone marrow cells in simulated microgravity. In Vitro Cell Dev Biol Anim 37: 73–78, 2001. doi:. [DOI] [PubMed] [Google Scholar]
  • 42a.Quách TD, Rodríguez-Zhurbenko N, Hopkins TJ, Guo X, Hernández AM, Li W, Rothstein TL. Distinctions among circulating antibody-secreting cell populations, including B-1 cells, in human adult peripheral blood. J Immunol 196: 1060–1069, 2016. doi: 10.4049/jimmunol.1501843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rao M, Lamont JL, Chan J, Concannon TW, Comenzo R, Ratichek SJ, Avendano EE. Serum free light chain analysis for the diagnosis, management, and prognosis of plasma cell dyscrasias: future research needs: identification of future research needs from comparative effectiveness review No. 73 (Online). In: AHRQ Future Research Needs Papers. Rockville, MD: Agency for Healthcare Research and Quality (US), 2012. [PubMed] [Google Scholar]
  • 44.Rao M, Yu WW, Chan J, Patel K, Comenzo R, Lamont JL, Ip S, Lau J. Serum Free Light Chain Analysis for the Diagnosis. Rockville, MD: Management, and Prognosis of Plasma Cell Dyscrasias, 2012. [PubMed] [Google Scholar]
  • 45.Rosenzweig JA, Abogunde O, Thomas K, Lawal A, Nguyen YU, Sodipe A, Jejelowo O. Spaceflight and modeled microgravity effects on microbial growth and virulence. Appl Microbiol Biotechnol 85: 885–891, 2010. doi: 10.1007/s00253-009-2237-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rykova MP, Antropova EN, Larina I, Morukov BV. Humoral and cellular immunity in cosmonauts after the ISS missions. Acta Astronaut 63: 697–705, 2008. doi: 10.1016/j.actaastro.2008.03.016. [DOI] [Google Scholar]
  • 47.Shlipak MG, Coresh J, Gansevoort RT. Cystatin C versus creatinine in determining risk based on kidney function. N Engl J Med 369: 932–943, 2013. doi: 10.1056/NEJMoa1214234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47a.Sims GP, Ettinger R, Shirota Y, Yarboro CH, Illei GG, Lipsky PE. Identification and characterization of circulating human transitional B cells. Blood 105: 4390–4398, 2005. doi: 10.1182/blood-2004-11-4284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Sonnenfeld G. Animal models for the study of the effects of spaceflight on the immune system. Adv Space Res 32: 1473–1476, 2003. doi: 10.1016/S0273-1177(03)90383-8. [DOI] [PubMed] [Google Scholar]
  • 49.Sonnenfeld G. Effect of space flight on cytokine production. Acta Astronaut 33: 143–147, 1994. doi: 10.1016/0094-5765(94)90119-8. [DOI] [PubMed] [Google Scholar]
  • 50.Sonnenfeld G, Shearer WT. Immune function during space flight. Nutrition 18: 899–903, 2002. doi: 10.1016/S0899-9007(02)00903-6. [DOI] [PubMed] [Google Scholar]
  • 51.Spielmann G, Laughlin MS, Kunz H, Crucian BE, Quiriarte HD, Mehta SK, Pierson DL, Simpson RJ. Latent viral reactivation is associated with changes in plasma antimicrobial protein concentrations during long-duration spaceflight. Acta Astronaut 146: 111–116, 2018. doi: 10.1016/j.actaastro.2018.02.039. [DOI] [Google Scholar]
  • 52.Stowe RP, Sams CF, Mehta SK, Kaur I, Jones ML, Feeback DL, Pierson DL. Leukocyte subsets and neutrophil function after short-term spaceflight. J Leukoc Biol 65: 179–186, 1999. doi: 10.1002/jlb.65.2.179. [DOI] [PubMed] [Google Scholar]
  • 53.Tosi P, Tomassetti S, Merli A, Polli V. Serum free light-chain assay for the detection and monitoring of multiple myeloma and related conditions. Ther Adv Hematol 4: 37–41, 2013. doi: 10.1177/2040620712466863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Turner JE, Spielmann G, Wadley AJ, Aldred S, Simpson RJ, Campbell JP. Exercise-induced B cell mobilisation: preliminary evidence for an influx of immature cells into the bloodstream. Physiol Behav 164: 376–382, 2016. doi: 10.1016/j.physbeh.2016.06.023. [DOI] [PubMed] [Google Scholar]
  • 55.van Ravenhorst MB, den Hartog G, van der Klis FRM, van Rooijen DM, Sanders EAM, Berbers GAM. Induction of salivary antibody levels in Dutch adolescents after immunization with monovalent meningococcal serogroup C or quadrivalent meningococcal serogroup A, C, W and Y conjugate vaccine. PLoS One 13: e0191261, 2018. doi: 10.1371/journal.pone.0191261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Voss EW., Jr Prolonged weightlessness and humoral immunity. Science 225: 214–215, 1984. doi: 10.1126/science.6729476. [DOI] [PubMed] [Google Scholar]
  • 57.Waldmann TA, Strober W, Mogielnicki RP. The renal handling of low molecular weight proteins. II. Disorders of serum protein catabolism in patients with tubular proteinuria, the nephrotic syndrome, or uremia. J Clin Invest 51: 2162–2174, 1972. doi: 10.1172/JCI107023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ward C, Rettig TA, Hlavacek S, Bye BA, Pecaut MJ, Chapes SK. Effects of spaceflight on the immunoglobulin repertoire of unimmunized C57BL/6 mice. Life Sci Space Res (Amst) 16: 63–75, 2018. doi: 10.1016/j.lssr.2017.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Webster AD. Laboratory investigations of primary deficiencies of the lymphoid system. Clin Allergy Immunol 5: 447–467, 1985. [Google Scholar]
  • 59a.Wei C, Jung J, Sanz I. OMIP-003: phenotypic analysis of human memory B cells. Cytometry A 79A: 894–896, 2011. doi: 10.1002/cyto.a.21112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yadav AP, Mishra KP, Ganju L, Singh SB. Wintering in Antarctica: impact on immune response of Indian expeditioners. Neuroimmunomodulation 19: 327–333, 2012. doi: 10.1159/000339512. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Applied Physiology are provided here courtesy of American Physiological Society

RESOURCES