Differential effects of Rhodiola rosea on regulatory T cell differentiation and interferon‑γ production in vitro and in vivo

  • Authors:
    • Xi Xu
    • Pingping Li
    • Peng Zhang
    • Ming Chu
    • Hongju Liu
    • Xiaoping Chen
    • Qing Ge
  • View Affiliations

  • Published online on: May 13, 2016     https://doi.org/10.3892/mmr.2016.5278
  • Pages: 529-536
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Rhodiola rosea (R. rosea), a type of adaptogen, has been previously reported to exhibit immunostimulating activity in rodents and in human peripheral blood mononuclear cells (PBMCs) in vitro. To examine the effect of R. rosea on T cells under simulated microgravity, spaceflight analogs of human head‑down bed rest (HDBR) at ‑6˚ and murine hind limb unloading (HU) were used. A decrease in the levels of interferon‑γ (IFN‑γ) and interleukin‑17 (IL‑17) and an increase in regulatory T (Treg) cells were observed in the placebo group following HDBR. The R. rosea treated HBDR group demonstrated further decreased IFN‑γ production, however, R. rosea exhibited no effect on the ratio of circulating Tregs or Treg cell differentiation. By contrast, the treatment of R. rosea on human T cells in vitro did not alter IFN‑γ secretion, however, Treg differentiation was significantly reduced. An R. rosea‑induced upregulation of hypoxia‑inducible factor 1α (HIF‑1α) contributed to the suppression of Treg differentiation in vitro. Differences in the effect of R. rosea in vitro and in vivo were also observed using a mouse model of microgravity. The results of the current study suggest that R. rosea has differential modulatory effects on T cells in vivo and in vitro and care should be taken when evaluating the effects of R. rosea on the immune system.

Introduction

Rhodiola rosea (R. rosea), a type of adaptogen, belongs to the Crassulaceae plant family, of the Sedoideae subfamily and Rhodiola genus. It has been used as traditional medicine in Europe, Asia and Russia for centuries (1). Greater than 20 compounds are present in the R. rosea root, including salidroside (rhodioloside), rosavins and p-tyrosol, which are understood to have important therapeutic activities (2). The commonly described functional activities include, performance enhancement, fatigue reduction, alleviation of depression symptoms, stimulation of the nervous system and prevention of high altitude sickness (3,4). R. rosea was previously demonstrated to have immunostimulatory potential in rodents in vivo, and in human peripheral blood mononuclear cells (PBMCs) in vitro (510). Additionally, in vivo administration of salidroside, the major component of R. rosea, enhanced the proliferation of murine T cells and the production of antibodies and cytokines, including interleukin (IL)-2, IL-4 and interferon-γ (IFN-γ) (5). In vitro administration of the aqueous extract of Rhodiola imbricate rhizome induced increased expression levels of IL-1β in human PBMCs, and of toll-like receptor-4 and granzyme-B in mouse splenocytes (10). Thus, R. rosea may potentially be used to enhance cellular immunity under microgravity conditions. However, it has not been previously reported whether R. rosea has an in vivo immune-modulating effect in humans (7,8). The effects of R. rosea on cytokine production by human T cells and the differentiation of regulatory T cells (Tregs) in vivo and in vitro is currently unknown.

Spaceflight changes the immune system in various ways. These include altered leukocyte distribution, altered serum cytokine levels, reduced functions of natural killer cells, granulocytes and monocytes, reduced leukocyte proliferation following activation, decreased delayed-type hypersensitivity to recall antigens, and latent viral reactivation (1123). A number of studies have investigated strategies to monitor the immune system during spaceflight and to develop countermeasures. To study whether R. rosea may enhance the functions of the immune system during spaceflights, the effect of R. rosea and its main component, salidroside, on human and mouse T cells was examined in vitro and in vivo. Head-down bed rest (HDBR) at −6° was used as a ground-based spaceflight model for the study of human T cells in vivo, and hind limb unloading (HU) was used as the in vivo mouse model.

Materials and methods

Ethical issues

The current study was approved by the Ethics Committee of China Astronaut Research and Training Center. Written consent was obtained from the subjects, who had been informed of the risks and the experimental details.

Subjects

Fifteen male volunteers of age 26.63±4.03, height 171.8±3.0 cm and weight 63.6±6.2 kg (all presented as means ± SEM) were recruited into the present study. All subjects were educated to a junior high level or above. The subjects were healthy and physically fit. Subjects with the following chronic or recent acute illnesses were precluded from the study: Skeletal muscle diseases; organic and functional diseases of psychiatry and neurology; and sleep disorders. No subjects had a history of cancer, hepatitis or any other relevant diseases, including autoimmune disorders. The final selection was based on typical clinical results comprising medical history, physical and psychological examination, complete blood count, blood chemistry analysis and several other tests. Eight subjects were randomly selected for the placebo group and seven for the RR group. The subjects in the RR group received R. rosea 0.5 g per day (prophylactic dose), twice a day from R1 to R7, then 1.0 g per day (therapeutic dose), twice a day from R8 to R45 during HDBR period. The dose of R. rosea was doubled for therapeutic purposes as changes in muscles and bones began to be significant following one week of bed rest. No medication, smoking, alcohol or caffeinated drinks were permitted during the study. Emergency medical surveillance and service were available throughout bed rest protocol.

Head-down bed rest (HDBR) protocol

The entire study included 45 days of HDBR, 10 days of adaptation prior to HDBR and 10 days of recovery following HDBR. During HDBR, the subjects were in a flat, resting, head-down position to −6° from horizontal. Lights were switched off between 22:00 and 06:00, with normal daylight illumination for the rest of the day. All the subjects were housed in an air-conditioned bedroom, maintained at 25±0.5°C with a relative humidity of 60–70%. All dining, washing, urination and defecation activities were carried out in a bedridden state. Changing position around the body axis was permitted.

Human PBMC preparation

Sterile heparinized peripheral blood samples were obtained from 12 healthy volunteers and the 15 test subjects prior to (R-1), during (R15, R30 and R45) and following (R+9) the HDBR protocol at 6:00 a.m. PBMCs were collected by Ficoll-Hypaque density-gradient centrifugation.

Mice

Male C57BL/6 mice (age, 6–8 weeks old) were purchased from Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China), and housed in a specific pathogen-free facility at the Chinese Astronaut Research and Training Center (Beijing, China). The mice were maintained under a 12-h light/dark cycle, 25±2°C and access to food and water ad libitum. All mice were allowed to adapt to the environment for a minimum of 3 days following shipment and prior to the onset of the experiment. The experimental procedures used and the care of the animals were approved by the ethics committee of the Chinese Astronaut Research and Training Center.

Mouse hind limb unloading (HU) model

C57BL/6 mice at 8 weeks of age were randomly assigned to four groups, with 3 mice in each group as follows: Saline group; salidroside group; saline with HU group; and salidroside with HU group. Mice in the HU groups were suspended by their tails at a 30° head-down tilt with no load bearing on their hind limbs, with unlimited access to food and water. Mice without HU were housed individually in standard caging. The mice received salidroside 50 mg/kg/day by intragastric administration for 28 days prior to HU and for 14 days during HU. Immediately following the HU, the mice were sacrificed by cervical dislocation and cells from the spleens were collected for further experimentation.

Reagents

Salidroside was purchased from Sigma-Aldrich (St. Louis, MO, USA). The following monoclonal antibodies were used for staining: Anti-human cluster of differentiation 4-peridinin chlorophyll (CD4-PerCP) Cy5.5 (OKT4; BioLegend, Inc., San Diego, CA, USA); anti-human CD25-phycoerythrin (PE; MEM-181; QuantoBio, Beijing, China); anti-human forkhead box P3-Allophycocyanin (Foxp3-APC; PCH101) and anti-mouse Foxp3-APC (FJK-16s; eBioscience, Inc., San Diego, CA, USA); and anti-human IFN-γ-fluorescein isothiocyanate (FITC; 4S.B3), anti-mouse CD4-FITC (H129.19), anti-mouse CD25-PE (PC61) and anti-mouse IFN-γ-APC (XMG1.2; BD Biosciences, San Jose, CA, USA). The following antibodies and regents were purchased from BD Biosciences were used for cell cultures: Anti-human CD3 (HIT3a); anti-human CD28 (CD28.2); anti-mouse CD3 (145-2C11); anti-mouse CD28 (37.51); and protein transport inhibitor (containing Brefelding A). Recombinant human transforming growth factor (rhTGF)-β1 and rhIL-2 were purchased from R&D Systems China Co., Ltd. (Shanghai, China). The mouse IFN-γ enzyme linked immunosorbent assay (ELISA) kit was purchased from eBioscience.

IFN-γ production

Human PBMCs were stimulated with 1 μg/ml anti-CD3 and 1 μg/ml anti-CD28 for 2 days. Protein transport inhibitor was added 4 hours prior to intracellular IFN-γ staining. Mouse splenocytes were stimulated with 2 μg/ml anti-CD3 and 1 μg/ml anti-CD28 for 2 days. The supernatants were collected and the concentrations of IFN-γ were measured using the mouse IFN-γ ELISA kit.

Helper T cell differentiation

Human PBMCs were stimulated by anti-CD3 and anti-CD28 under induced regulatory T cell (iTreg)-inducing conditions (rhIL-2, 5 ng/ml; and rhTGF-β1, 10 ng/ml) with various doses of salidroside (0, 10, 30, 50 and 100 μg/ml). After 5 days, the cells were collected and stained with CD4 and CD25 antibodies, then intracellular staining of Foxp3 was conducted according to the manufacturer's protocols and the cells were examined by FACSCalibur flow cytometry (BD Biosciences).

Mouse splenocytes were stimulated by anti-CD3 and anti-CD28 under iTreg-inducing condition (rhIL-2, 2 ng/ml; and rhTGF-β1, 1 ng/ml). iTreg-skewing cells were directly stimulated for 3 days prior to intracellular Foxp3 staining.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RNA was extracted from iTreg-skewing PBMCs using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA), according to the manufacturer's protocol, and cDNA was obtained using the FastQuant RT kit (Tiangen Biotech Co., Ltd., Beijing, China). RT-qPCR was performed using SYBR Green Supermix (Bio-Rad Laboratories, Inc., Hercules, CA, USA) on an iCycler Real-Time PCR Detection System (Bio-Rad Laboratories, Inc.), with each sample in triplicate. The primers used for measurement were as follows: Forward, 5′-TTCACCTGAGCCTAATAGTCC-3′ and reverse, 5′-CAAGTCTAAATCTGTGTCCTG-3′ for hypoxia inducible factor-1α (HIF-1α); and forward, 5′-GCACCGTCAAGGCTGAGAAC-3′ and reverse, 5′-TGGTGAAGACGCCAGTGGA-3′ for GAPDH. PCRs were performed for 40 cycles of 95°C for 20 sec, 56°C for 20 sec and 72°C for 20 sec. The quantification was based on ΔΔCq calculations and were normalized to GAPDH as the reference gene (24).

Statistical analysis

Statistical analysis of the results was performed using Prism 5 (GraphPad Software, Inc., La Jolla, CA, USA). The differences between the placebo group and RR group were analyzed by repeated measures analysis of variance (ANOVA), with time and treatment as two factors for repeated measures and were further evaluated using Bonferroni correction. Unpaired or two-tail paired t test was further used to evaluate the significance of the differences. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of R. rosea on human T cells in the HDBR model

To investigate the in vivo effects of R. rosea on human T cells under simulated microgravity, placebo- and RR-treated groups underwent a 45-day HDBR protocol (25). The subjects in the RR group received Rhodiola rosea 0.50 g (prophylactic dose) twice a day from R1 to R7, then 1.0 g (therapeutic dose) twice a day from R8 to R45 during the HDBR period. As presented in Fig. 1A, the percentages of total T cells, CD4+ and CD8+ T cells in the peripheral blood of the placebo group did not change until 9 days following the completion of bed rest (R+9), where the percentages were significantly different to those at day R45 (Fig. 1A) (25). The changes included a significant increase in the total T and CD4+ T cells and a decrease in CD8+ T cells on R+9 compared with R45 (P=0.008, P=0.013 and P=0.013, respectively). Consistently, an ~14% increase in CD4:CD8 ratio was observed at R+9 compared with R45 (P=0.015, Fig. 1A) (25). Compared with the placebo group, the adaptogen RR-treated group exhibited a similar pattern of changes in the T cell subsets (Fig. 1B).

The percentage of circulating Treg cells (CD4+ CD25+ Foxp3+ CD127) in the HDBR PBMCs was observed to be increased at R45 compared with R-1 (P=0.026) and had returned to the baseline levels at R+9 (Fig. 1C) (25). Similar Treg changes were observed in the group treated with RR (Fig. 1C). A late increase in circulatory Tregs was observed, whereas, the percentage of iTregs was increased at R15 compared with R-1, however, there was no change in iTreg levels at R45(Fig. 1C) (25). The pattern of T cell levels in the RR group was not significantly different to the placebo group (Fig. 1C). The differentiation of iTregs at various time points was induced by stimulation of PBMCs with anti-CD3, anti-CD28 and TGF-β1 under 1g conditions for 3 days (26).

The levels of cytokines produced by the activated T cells were examined after stimulation of HDBR PBMCs with anti-CD3 and anti-CD28 under 1g conditions for 2 days. The levels of IFN-γ and IL-17A in the placebo group exhibited a gradual decrease during HDBR, reaching the lowest level at R45 (P=0.05, 0.003 vs. R1, repeated measures ANOVA; 25.0%±26.2% and 53.8%±20.3%, respectively; Fig. 1D) (25). Unlike the findings of previous post-flight and HDBR studies (11,22,27), the current study did not observe a decrease in IL-2 expression in the placebo group (Fig. 1D) (25). No consistent or significant changes were observed in the production of IL-4 by T cells (data not shown). In the RR group, the levels of IFN-γ production by T cell were reduced upon anti-CD3 and anti-CD28 stimulation (R15). The decrease to IFN-γ levels was significantly enhanced in the RR group compared with the placebo group at R15 (P=0.046, repeated measures ANOVA; Fig. 1E) (25). There was no difference in the IL-17A and IL-2 levels between the placebo and the RR groups (Fig. 1E).

Effect of R. rosea on human T cells in vitro

To investigate whether the suppressive effect of R. rosea on T cell function may also be observed in vitro under 1g conditions, normal human PBMCs were treated with salidroside, the main component of R. rosea, and stimulated with anti-CD3 and anti-CD28 monoclonal antibodies. In contrast to the in vivo results obtained from the HDBR experiment, no significant alterations of IFN-γ production was observed between salidroside-treated or untreated PBMCs (Fig. 2A). Notably, a significant decrease in iTreg cell differentiation was observed in the cells treated with salidroside (Fig. 2B). The inhibition of iTreg differentiation by salidroside was dose-dependent, as an increase in salidroside concentration resulted in a further decrease to the iTreg cell percentage, with the lowest percentage of T cells observed following 100 μg/ml salidroside treatment (P=0.019 vs. 0 μg/ml; Fig. 2B). These results suggest that, in contrast to the in vivo effect, salidroside may have a direct and suppressive impact on regulatory T cell differentiation in vitro.

As the signaling pathways downstream of CD28 and the TGF-β1 receptor are crucial to iTreg differentiation, the concentration of anti-CD28 antibody and TGF-β1 were titrated to examine whether salidroside increased the effects of these two pathways. As presented in Fig. 3A and B, the optimal concentration for induction of iTreg differentiation in the absence of salidroside was 1 μg/ml for anti-CD28 antibody and 10 ng/ml for TGF-β1. Compared with cells that received anti-CD28 or TGF-β1 only, salidroside treatment significantly reduced iTreg differentiation (P=0.046 and P=0.016, respectively). Increasing the concentrations of anti-CD28 or TGF-β1 did not abolish the suppressive effect of salidroside. The suppression of Treg differentiation in vitro was not due to the inhibition of T cell survival or proliferation by salidroside treatment (data not shown).

R. rosea, specifically, salidroside, was previously reported to increase HIF-1α expression and its nuclear translocation in cardiomyocytes, fibroblasts, kidney and liver cells (2830). HIF-1α was also previously observed to suppress Treg differentiation by promoting the glycolytic activity of T cells and by binding Foxp3 to promote its proteasomal degradation (31,32). Thus, the current study examined whether the treatment of salidroside alters the expression of HIF-1α in T cells when cultured in regulatory T cell inducing conditions. As presented in Fig. 3C, the HIF-1α mRNA levels in PBMCs were significantly increased by salidroside in vitro, compared with PBS treated cells (P=0.010).

These data demonstrate that salidroside has a significant effect on iTred differentiation, however, the effects are different in vitro and in vivo. Salidroside directly suppressed the differentiation of iTregs in vitro under 1g conditions. However, when R. rosea was administered in vivo during the HDBR model, it did not significantly alter iTreg differentiation, though the actual T cell differentiation assay was also performed in vitro under 1g condition.

Effect of R. rosea on murine T cells in HU model

To investigate whether similar differences are also observed in the murine system, a 14-day HU mouse model was used. Mice received salidroside at 50 mg/kg/day by intragastric administration for 28 days prior to HU and 14 days following HU. The mice were subsequently sacrificed and splenic T cells were cultured under various conditions at 1g. The control groups included saline treatment with and without HU and salidroside treatment without HU. There was no difference in the levels of IFN-γ in T cells following anti-CD3 and anti-CD28 stimulation between the saline with and without HU groups, and in the salidroside without HU group (Fig. 4A). However, the salidroside with HU group exhibited a significant reduction in IFN-γ production by T cells, compared with the saline with HU group (P=0.032), a similar pattern to the results obtained from human HDBR samples (Fig. 4A). The differentiation of iTregs was also similar among saline groups with or without HU, and the salidroside group without HU. However, the salidroside with HU group exhibited a significant increase in the level of iTreg differentiation (Fig. 4B).

Effect of R. rosea on murine T cells in vitro

The direct effect of salidroside on murine T cells in vitro was also investigated. Similar to human PBMCs, mouse splenic T cells treated with salidroside showed no significant difference in the levels of IFN-γ, however, compared with PBS treatment, a significant decrease in iTreg differentiation was observed (P=0.034; Fig. 5A and B).

Discussion

Various studies have previously reported that R. rosea has anti-stress and immunostimulatory activities (310). Thus, the current study investigated whether R. rosea may improve the function of the immune system during spaceflight. R. rosea exhibited differential effects in vitro and in vivo. The administration of R. rosea in vivo decreased the production of IFN-γ by human T cells following simulated microgravity (HDBR). The treatment with R. rosea in vitro, however, did not change the production of IFN-γ by T cells. Similarly, the differentiation of iTregs was not altered in R. rosea-treated human or mouse cells following microgravity simulation, whereas, iTreg differentiation was significantly decreased when R. rosea was directly added into the T cell culture. These differences suggest that R. rosea may have a direct suppressive effect on regulatory T cell differentiation in vitro and may have an indirect impact on regulatory T cell differentiation by the production of Th1 type cytokines under microgravity conditions in vivo. It is possible that the differences in doses and durations of R. rosea treatments in vitro and in vivo may account for the different effects demonstrated. This may be difficult to confirm as T cells cultured in vitro for >40 days may need multiple rounds of T cell receptor-mediated activation and the presence of cytokines to promote cell survival. It is also possible that the different modulatory effects that were observed following R. rosea treatment occurred as a result of the different experimental conditions used for the in vivo microgravity model and the in vitro 1g model. However, this is unlikely as PBMCs derived from humans/mice with or without microgravity were eventually cultured in the same culture conditions as the in vitro experiment (anti-CD3 and anti-CD28 with or without TGF-β1). In addition, mice receiving saline and R. rosea under 1g conditions exhibited similar levels of IFN-γ production and iTreg differentiation (Fig. 3). This further suggests that R. rosea may have differential modulatory functions on T cells directly (in vitro) and indirectly under microgravity (in vivo).

Regarding the direct suppressive effect of R. rosea on iTregs, these data suggest a casual link between R. rosea-promoted HIF-1α transcription in T cells and a reduction in iTreg cell differentiation. Whether R. rosea may alter HIF-1α transcription in vivo is awaiting further investigation.

Collectively, the results of the present study obtained from human and mouse T cells indicate that R. rosea has a direct negative impact on the differentiation of regulatory T cells in vitro. Thus, the increase in Treg differentiation and decrease in IFN-γ production by R. rosea in vivo under microgravity conditions is probably due to the effect of R. rosea on cells other than T cells. Whether they are antigen presenting cells or even cells from neuronal pathways remains unclear. The results of the current study do not support an immunostimulatory effect of R. rosea and suggest that R. rosea may not improve T cell immunity under microgravity in vivo.

Acknowledgments

The current work was supported by grants from the Natural Basic Research Program of China (2011CB711000), the National Natural Science Foundation of China (31270935 and 81471525, Q.G.; 31171144 and 81272177, X.C.), Beijing Natural Science Foundation (5152010, Q.G.) and the Opening Foundation of the State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center (SMFA12K08).

Abbreviations:

HDBR

head-down bed rest

PBMC

peripheral blood mononuclear cell

IFN-γ

interferon-γ

IL

interleukin

Treg

regulatory T cell

iTreg

induced regulatory T cell

R. rosea

Rhodiola rosea

HIF-1α

hypoxia-inducible factor 1α

HU

hind limb unloading

TGF-β1

transforming growth factor β1

References

1 

Ishaque S, Shamseer L, Bukutu C and Vohra S: Rhodiola rosea for physical and mental fatigue: A systematic review. BMC Complement Altern Med. 12:702012. View Article : Google Scholar : PubMed/NCBI

2 

Brekhman II and Dardymov IV: New substances of plant origin which increase nonspecific resistance. Annu Rev Pharmacol. 9:419–430. 1969. View Article : Google Scholar : PubMed/NCBI

3 

Petkov VD, Yonkov D, Mosharoff A, Kambourova T, Alova L, Petkov VV and Todorov I: Effects of alcohol aqueous extract from Rhodiola rosea L. roots on learning and memory. Acta Physiol Pharmacol Bulg. 12:3–16. 1986.PubMed/NCBI

4 

Lee Y, Jung JC, Jang S, Kim J, Ali Z, Khan IA and Oh S: Anti-inflammatory and neuroprotective effects of constituents isolated from Rhodiola rosea. Evid Based Complement Alternat Med. 2013:5140492013. View Article : Google Scholar : PubMed/NCBI

5 

Guan S, He J, Guo W, Wei J, Lu J and Deng X: Adjuvant effects of salidroside from Rhodiola rosea L. on the immune responses to ovalbumin in mice. Immunopharmacol Immunotoxicol. 33:738–743. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Mishra KP, Chanda S, Shukla K and Ganju L: Adjuvant effect of aqueous extract of Rhodiola imbricata rhizome on the immune responses to tetanus toxoid and ovalbumin in rats. Immunopharmacol Immunotoxicol. 32:141–146. 2010. View Article : Google Scholar

7 

Li HX, Sze SC, Tong Y and Ng TB: Production of Th1- and Th2-dependent cytokines induced by the Chinese medicine herb, Rhodiola algida, on human peripheral blood monocytes. J Ethnopharmacol. 123:257–266. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Mishra KP, Padwad YS, Jain M, Karan D, Ganju L and Sawhney RC: Aqueous extract of Rhodiola imbricata rhizome stimulates proinflammatory mediators via phosphorylated IkappaB and transcription factor nuclear factor-kappaB. Immunopharmacol Immunotoxicol. 28:201–212. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Wang H, Ding Y, Zhou J, Sun X and Wang S: The in vitro and in vivo antiviral effects of salidroside from Rhodiola rosea L. against coxsackievirus B3. Phytomedicine. 16:146–155. 2009. View Article : Google Scholar

10 

Mishra KP, Ganju L, Chanda S, Karan D and Sawhney RC: Aqueous extract of Rhodiola imbricata rhizome stimulates Toll-like receptor 4, granzyme-B and Th1 cytokines in vitro. Immunobiology. 214:27–31. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Crucian BE, Stowe RP, Pierson DL and Sams CF: Immune system dysregulation following short- vs long-duration spaceflight. Aviat Space Environ Med. 79:835–843. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Sonnenfeld G and Shearer WT: Immune function during space flight. Nutrition. 8:899–903. 2002. View Article : Google Scholar

13 

Buravkova LB, Rykova MP, Grigorieva V and Antropova EN: Cell interactions in microgravity: Cytotoxic effects of natural killer cells in vitro. J Gravit Physiol. 11:177–180. 2004.

14 

Kaur I, Simons ER, Castro VA, Mark Ott C and Pierson DL: Changes in neutrophil functions in astronauts. Brain Behav Immun. 18:443–450. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Pierson DL, Stowe RP, Phillips TM, Lugg DJ and Mehta SK: Epstein-Barr virus shedding by astronauts during space flight. Brain Behav Immun. 19:235–242. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Crucian BE, Stowe RP, Mehta SK, Yetman DL, Leal MJ, Quiriarte HD, Pierson DL and Sams CF: Immune status, latent viral reactivation, and stress during long-duration head-down bed rest. Aviat Space Environ Med. 80(Suppl): A37–A44. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Kaur I, Simons ER, Castro VA, Ott CM and Pierson DL: Changes in monocyte functions of astronauts. Brain Behav Immun. 19:547–554. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Kaur I, Simons ER, Kapadia AS, Ott CM and Pierson DL: Effect of spaceflight on ability of monocytes to respond to endotoxins of gram-negative bacteria. Clin Vaccine Immunol. 15:1523–1528. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Stowe RP, Sams CF and Pierson DL: Effects of mission duration on neuroimmune responses in astronauts. Aviat Space Environ Med. 74:1281–1284. 2003.PubMed/NCBI

20 

Baqai FP, Gridley DS, Slater JM, Luo-Owen X, Stodieck LS, Ferguson V, Chapes SK and Pecaut MJ: Effects of spaceflight on innate immune function and antioxidant gene expression. J Appl Physiol. 106:1935–1942. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Gridley DS, Slater JM, Luo-Owen X, Rizvi A, Chapes SK, Stodieck LS, Ferguson VL and Pecaut MJ: Spaceflight effects on T lymphocyte distribution, function and gene expression. J Appl Physiol (1985). 106:194–202. 2009. View Article : Google Scholar

22 

Kelsen J, Bartels LE, Dige A, Hvas CL, Frings-Meuthen P, Boehme G, Thomsen MK, Fenger-Grøn M and Dahlerup JF: 21 Days head-down bed rest induces weakening of cell-mediated immunity - Some spaceflight findings confirmed in a ground-based analog. Cytokine. 59:403–409. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Guéguinou N, Huin-Schohn C, Bascove M, Bueb JL, Tschirhart E, Legrand-Frossi C and 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. View Article : Google Scholar

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real time quantitative PCR and the 2 (Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

25 

Xu X, Tan C, Li P, Zhang S, Pang X, Liu H, Li L, Sun X, Zhang Y, Wu H, et al: Changes of cytokines during a spaceflight analog-a 45-day head-down bed rest. PLoS One. 8:e774012013. View Article : Google Scholar

26 

Tauber S, Hauschild S, Paulsen K, Gutewort A, Raig C, Hürlimann E, Biskup J, Philpot C, Lier H, Engelmann F, et al: Signal transduction in primary human T lymphocytes in altered gravity during parabolic flight and clinostat experiments. Cell Physiol Biochem. 35:1034–1051. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Crucian BE, Cubbage ML and Sams CF: Altered cytokine production by specific human peripheral blood cell subsets immediately following space flight. J Interferon Cytokine Res. 20:547–556. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Zhang J, Liu A, Hou R, Zhang J, Jia X, Jiang W and Chen J: Salidroside protects cardiomyocyte against hypoxia-induced death: A HIF-1-alpha-activated and VEGF-mediated pathway. Eur J Pharmacol. 607:6–14. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Zheng KY, Guo AJ, Bi CW, Zhu KY, Chan GK, Fu Q, Xu SL, Zhan JY, Lau DT, Dong TT, et al: The extract of Rhodiolae Crenulatae Radix et Rhizoma induces the accumulation of HIF-1alpha via blocking the degradation pathway in cultured kidney fibroblasts. Planta Med. 77:894–899. 2011. View Article : Google Scholar

30 

Zheng KY, Zhang ZX, Guo AJ, Bi CW, Zhu KY, Xu SL, Zhan JY, Lau DT, Dong TT, Choi RC and Tsim KW: Salidroside stimulates the accumulation of HIF-1α protein resulted in the induction of EPO expression: A signaling via blocking the degradation pathway in kidney and liver cells. Eur J Pharmacol. 679:34–39. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, Bordman Z, Fu J, Kim Y, Yen HR, et al: Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell. 146:772–784. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR and Chi H: HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med. 208:1367–1376. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 14 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu X, Li P, Zhang P, Chu M, Liu H, Chen X and Ge Q: Differential effects of Rhodiola rosea on regulatory T cell differentiation and interferon‑γ production in vitro and in vivo. Mol Med Rep 14: 529-536, 2016
APA
Xu, X., Li, P., Zhang, P., Chu, M., Liu, H., Chen, X., & Ge, Q. (2016). Differential effects of Rhodiola rosea on regulatory T cell differentiation and interferon‑γ production in vitro and in vivo. Molecular Medicine Reports, 14, 529-536. https://doi.org/10.3892/mmr.2016.5278
MLA
Xu, X., Li, P., Zhang, P., Chu, M., Liu, H., Chen, X., Ge, Q."Differential effects of Rhodiola rosea on regulatory T cell differentiation and interferon‑γ production in vitro and in vivo". Molecular Medicine Reports 14.1 (2016): 529-536.
Chicago
Xu, X., Li, P., Zhang, P., Chu, M., Liu, H., Chen, X., Ge, Q."Differential effects of Rhodiola rosea on regulatory T cell differentiation and interferon‑γ production in vitro and in vivo". Molecular Medicine Reports 14, no. 1 (2016): 529-536. https://doi.org/10.3892/mmr.2016.5278