Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Immune-enhancing effects of anionic macromolecules extracted from Codium fragile on cyclophosphamide-treated mice

  • Chaiwat Monmai,

    Roles Formal analysis, Methodology, Writing – original draft

    Affiliation Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon, Korea

  • SangGuan You,

    Roles Conceptualization, Project administration, Writing – review & editing

    Affiliation Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon, Korea

  • Woo Jung Park

    Roles Project administration, Supervision, Writing – review & editing

    pwj0505@gwnu.ac.kr

    Affiliation Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon, Korea

Abstract

Immune-regulation and homeostasis are critical in cancer therapy and immunomodulatory biomaterials have been used to decrease side effects of immunosuppressant drugs. Anionic macromolecules (CFAMs) were isolated from the seaweed Codium fragile, and its immune-enhancing biological activities were examined in CY-induced immunosuppressed mice. CFAMs improved the splenic lymphocyte proliferation, NK cell activity, and spleen index. The expression of immune-associated genes was highly upregulated in splenic lymphocytes, and gene expression was differently regulated according to mitogens such as T-cell (Con A) and B-cell (LPS) mitogens. Additionally, CFAMs boosted the proliferation, NO production, and phagocytosis of peritoneal macrophages. CFAMs also considerably stimulated immune-associated gene expression in peritoneal macrophages. Moreover, our results showed CFAMs mediated its immune-enhancing effects via the MAPK pathway. These suggested CFAMs can be used as a potent immunomodulatory material under immune-suppressive condition. Furthermore, CFAMs may also be used as a bio-functional and pharmaceutical material for improving human health and immunity.

Introduction

Immunomodulation, consisting of immune-stimulation and immunosuppression, is a sophisticated mechanism regulating the pathophysiology and etiology of various diseases affecting the immune system [1]. Immuno-regulation and homeostasis are crucial factors affecting cellular processes [2]. An immunomodulatory material can be used as an immune-stimulator to decrease the side effects of immunosuppressant drugs [3]. From ancient times, natural products with immune-regulatory properties have been considered to be important. They also act as important nutrients for improving human health and preventing chronic diseases and physiological problems, such as allergies, infections, and autoimmune diseases. Beta-glucan, a polysaccharide, is useful for treating respiratory tract infections [4]. Hemicellulose is known to possess immune-regulatory, antibacterial, and antitumor biological activities [5]. Chitin also enhances innate immunity and modulates inflammation in occupational allergies in humans [6]. Moreover, carbohydrates are known to be associated with the regulation of gut microbiota, which affect host immunity and health [7].

Most anticancer chemotherapeutic drugs, such as cyclophosphamide (CY), gemcitabine, and 5-fluorouracil, kill cancer cells; however, they also destroy normal cells, including immune cells, during the cytokine storm [8]. CY is a cancer prodrug and its therapeutic effectiveness depends on its metabolism by cytochrome P450 (CYP). It is the most widely used alkylating agent in chemotherapy for treating several cancers [9]. It acts on immune cells (such as T and B cells) which are xenoreactive, and thus, proliferate only after antigen stimulation [10]. The antitumor effect of CY is proportional to the amount of CY administered, and often leads to immunosuppression and cytotoxic effects [11]. Biomaterials that reduce the side effects of anticancer drugs are important for improving the quality of life of cancer patients [12].

Codium fragile is a popular, edible green alga belonging to the family Codiaceae It is widely distributed along the shores of East Asia, Oceania, and Northern Europe. C. fragile has also been used as an oriental medicine for treatment of diseases, such as enterobiosis, dropsy, and dysuria [13]. C. fragile extracts have been demonstrated to possess thrombolytic, anticoagulant, antiplatelet [14], anti-inflammatory [15], and immune-stimulatory activities on murine macrophage cells via the activation of NF-κB and MAPK pathways [16].

Recently, the crude anionic macromolecules from C. fragile (CFAMs) were extracted and their immunomodulatory activities was demonstrated on macrophages and NK cells in addition to chemical compositional analysis [1618]. However, no previous study reports their immune-enhancing regulation activity using in vivo immune-suppressed animal physiological system. We hypothesize that CFAMs may have immune-enhancing activity in immune organs and cells such as spleen, NK cells, and peritoneal macrophages under in vivo immunosuppressed animal physiological condition. Herein, the present study induced immunosuppressed condition of in vivo mice using CY and investigated the immune-enhancing effects through oral administration of CFAMs on CY-induced immunosuppressed mice as well as analyzed the immune signaling pathways involved in mediating those effects.

Materials and methods

Animals

Six-week-old inbred male BALB/c mice weighing 23 g were obtained from Central Lab. Animal Inc. (South Korea). These animals were kept in pathogen-free, environmentally controlled rooms maintained at 22 ± 2°C temperature and a 12-h dark–light cycle, for at least a week before the start of the experiment. They were fed on standard laboratory diet and water. All experimental procedures were approved by the Gangneung-Wonju National University committee for animal experiments (Approval number: GWNU-2016-31).

Isolation of the polysaccharides

Extraction and purification of crude anionic macromolecules from C. fragile (CFAMs) were performed as did previously [16] and these CFAMs were used in this study. Briefly, CFAMs were extracted from the milled sample of C. fragile using EtOH and distilled water, following centrifugation, filtration, and evaporation after removal of proteins [19].

Induction of immunosuppression in mice

Mice were randomly divided into seven groups (n = 5), after acclimatizing them for one week. One group was designated as the control group (normal group) and was administrated saline orally. The other groups were orally administrated saline (saline group) supplemented with varying concentrations of CFAMs (50, 100, 250, and 500 mg/kg BW) or with 100 mg/kg BW of commercial ginseng syrup (ginseng group). All groups received the respective treatment once per day for 10 consecutive days. At day 4–6 post-administration, mice (except those in the normal group) were injected intraperitoneally once a day with CY (80 mg/kg BW; Sigma–Aldrich, USA), and all the mice were sacrificed 24 h after completion of the treatment regimen.

Preparation of peritoneal macrophages and splenocytes

Peritoneal macrophages were prepared using the Ray and Dittel method [20]. Five milliliters of ice-cold phosphate buffered saline (PBS, supplemented with 3% FCS) was injected into the peritoneal cavity of each mouse, and subsequently the macrophages were collected. After collection, the cell suspension was centrifuged and cell pellet were resuspended in the RPMI-1640 medium or PBS for cell counting.

Splenocytes were isolated from the spleen of BALB/c mice. The spleen was weighed and collected in ice-cold PBS. After treating the spleen with 1 × RBC Lysis Buffer (eBioscience, USA), the lysate cells were centrifuged at 400 × g for 10 min and washed using PBS. The splenocytes were resuspended in RPMI-1640 growth medium supplemented with 10% fetal bovine serum, streptomycin (100 μg/mL), and penicillin (100 IU/mL). Spleen index was evaluated using the following formula:

Determination of peritoneal macrophage proliferation and nitric oxide production

In order to determine the proliferation of macrophages, the EZ-Cytox Cell Viability Assay kit (Daeil Labservice, Korea) was used, as described previously [21]. Macrophage proliferation ratio (%) was analyzed using the following formula:

The immune-enhancing activity was measured based on nitric oxide (NO) production by macrophages. The cells were cultured with or without LPS, and NO concentration was measured using the Griess reagent (Sigma–Aldrich, USA) [22].

Phagocytosis of peritoneal macrophages

Phagocytosis of peritoneal macrophages was determined using the neutral red uptake method, as described previously [23]. Macrophages were seeded in triplicates in microplates and grown overnight in complete RPMI-1640 medium at 37°C in a humidified atmosphere of 5% CO2. Phagocytosis of macrophages was measured using a microplate reader [absorbance (A), 540 nm]. Phagocytosis ratio was calculated using the following formula

Proliferation of splenic lymphocytes

Isolated splenic lymphocytes were induced using a T-cell (Con A, 5 μg/mL) or B-cell (LPS, 10 μg/mL) mitogen. Cells in the negative control group were incubated in RPMI at 37°C in a humidified atmosphere of 5% CO2. After culturing for 48 h, cell proliferation was evaluated using the EZ-Cytox Cell Viability Assay kit.

Cytotoxicity assays for measuring the natural killer cell activity of splenocytes

Natural killer cell activity of splenocytes (effector cells) was analyzed using YAC-1 cells (target cells) [24] and lactate dehydrogenase (LDH) solution (Promega Co., USA) [25, 26]. Splenocyte suspensions were added to the YAC-1 cells to obtain an effector-to-target cell ratio of 50:1. After 4 h incubation, the culture supernatants were mixed with a LDH solution. Absorbance was measured at 490 nm and the percentage of NK cell cytotoxicity was evaluated using the following formula: Cytotoxicity (%) = [(Experimental release-Spontaneous release)/(Maximum release-Spontaneous release)] × 100.

RNA isolation and reverse transcription

Total RNA was prepared from the LPS-stimulated peritoneal macrophages and mitogen-stimulated (Con A and LPS) splenic lymphocytes using the Tri reagent® (Molecular Research Center, Inc., USA). After isolation, RNA was dissolved in nuclease-free water and its concentration was analyzed using a nanophotometer (Implen, Germany). cDNA was synthesized from the extracted RNA using the High Capacity cDNA Reverse Transcription kit (Applied biosystems, USA), according to the manufacturer’s instructions.

Analysis of immune gene expression using real-time PCR

Quantification of immune gene expression in peritoneal macrophages and splenocytes was performed using the QuantStudio™ 7 FlexReal-Time PCR System (ThermoFisher scientific, USA) and SYBR® Premix Ex Taq™ II (Takara Bio Inc., Japan). Relative gene expression was calculated using the 2-ΔΔCT method [27] and β-Actin as the reference gene (Table 1).

thumbnail
Table 1. Oligonucleotide primers used in real-time PCR for evaluating immune gene expression.

https://doi.org/10.1371/journal.pone.0211570.t001

Western blotting for analyzing the NF-κB and MAPK signaling pathways

RAW264.7 cells were cultured in different concentrations of CFP (1.875, 3.75, 7.5, or 15 μg/mL) or 1 μg/mL of LPS. Using RIPA buffer (Tech & Innovation, China), the cells were lysed and protein was extracted. Protein concentration was measured using the Pierce™ BCA Protein Assay kit (Thermo Scientific, USA). After resolving the proteins using SDS-polyacrylamide gel electrophoresis (SDS-PAGE), they were transferred on to a polyvinylidene fluoride (PVDF) membrane. Western blotting was performed as described previously [28]. using antibodies specific for p-NF-κB p65 (R&D systems), p-IκBα (Cell Signaling Technology), p-p38 (R&D systems), p-ERK1/2 (R&D systems), p-JNK (R&D systems), and α-tubulin (Abcam). Target proteins were detected using the Pierce® ECL Plus Western Blotting Substrate (Thermo Scientific, USA). Imaging was performed using the ChemiDoc XRS+ imaging system (Bio-Rad) and protein band intensities were analyzed using the ImageLab software (version 4.1, Bio-Rad).

Effect of NF-κB and MAPK activation inhibitors on TNF-α expression

In order to evaluate the effects of NF-κB and MAPK activation inhibitors (such as ERK, JNK, and p38) on TNF-α expression, RAW264.7 cells were used. RAW264.7 cells were treated with 100 nM of NF-κB activation inhibitor (Merck, 481406), and 20 μM of ERK activation inhibitors (Merck, 328006), JNK (Merck, 420119), and p38 (Merck, 559389) [29]. After that, these cells were stimulated with varying concentrations of CFP (1.875, 3.75, 7.5, and 15 μg/mL) or 1 μg/mL of LPS. After extraction of total RNA, TNF-α expression was analyzed using the QuantStudio™ 7 FlexReal-Time PCR System, as described above.

Statistical analysis

Statistical analysis was performed using the Statistix 8.1 software. Data were analyzed using one-way ANOVA followed by comparison with the saline or ginseng group. Differences between groups were considered to be statistically significant at P < 0.05 and P < 0.01.

Results

Effect of CFAMs treatment on spleen index

Fig 1A and 1B show the spleen size and spleen index of CY-induced mice treated with four different concentrations of CFAMs, respectively. Analysis was performed on the day of sacrifice. Compared to the normal group, the CY-treated group showed a marked reduction in the spleen index and spleen size. Compared to the CY group, spleen index of the group treated with 250 or 500 mg/kg BW CFAMs was significantly different; however, no significant differences in the body weight were observed between these groups (S1 Fig). Further, significant differences were not observed in the spleen size and spleen index of the group treated with 50 or 100 mg/kg BW CFAMs and the CY group.

thumbnail
Fig 1. Effects of various concentrations of Codium fragile extracts.

The spleen was collected from each group of mice (A). The spleen weight was divided with body weight for calculating the spleen index (B). The splenocytes were isolated and were stimulated with mitogens for splenic lymphocyte proliferation assay (C). The splenocytes were co-cultured with YAC-1 cells for splenic NK cells cytotoxic activity (D). Data were observed to be significantly different compared to the saline group (*, P < 0.01) and ginseng group (#, P < 0.05).

https://doi.org/10.1371/journal.pone.0211570.g001

Effect of CFAMs treatment on splenic lymphocyte proliferation

Splenocytes from each group were incubated with two mitogens, namely Con A and LPS. Con A and LPS stimulate the T and B cells in splenocytes, respectively. Fig 1C shows that compared to the normal group, the proliferative response of splenic lymphocytes to both the T- and B-cell mitogens decreased markedly in the CY-treated group. However, compared to the CY-treated group, the CFAMs-treated group showed enhanced T and B cell proliferation in a dose-dependent manner. Further, the proliferation in the CFAMs-treated group was similar to that observed in the red ginseng-treated group, and significantly higher than that observed in the normal group.

Effect of CFAMs on NK cell activity of splenocytes

In order to evaluate the NK cell activity of splenocytes, the splenocytes were co-cultured with YAC-1 cells. Cytotoxicity was determined using the LDH assay. Fig 1D shows that CY treatment suppressed splenic NK cell activity. However, CFAMs treatment gradually stimulated the NK cell activity of splenocytes. Notably, compared to the red ginseng-treated and normal groups, higher splenic NK cell activity was observed in the group treated with a high dose of CFAMs.

Effect of CFAM on immune-associated gene expression in splenic lymphocytes

By analyzing the expression of immune-associated genes (including those for cytokines) using qRT-PCR, the immuno-enhancing effects of CFAMs were elucidated. Fig 2 shows that compared to the normal group, the CY-treated group showed reduced expression of immune-associated genes in the spleen lymphocytes. However, treatment with varying concentrations of CFAMs enhanced immune-associated gene expression in splenic lymphocytes, which responded to the T- and B-cell mitogens. After CFAMs treatment, genes such as IL-4, IL-10, TNF-α, IFN-γ, TLR-4, and COX-2 in splenic lymphocytes were highly upregulated and responded more to the B cell mitogen (LPS) than to the T cell mitogen (Con A). In contrast, the expression of IL-1β, IL-2, and IL-6 was considerably higher in the Con A-stimulated cells than in the LPS-stimulated cells.

thumbnail
Fig 2. Relative expression (fold-change) of immune genes in mitogen-stimulated splenic lymphocytes.

Splenic lymphocytes were stimulated with the mitogens (5 μg/mL of Con A or 10 μg/mL of LPS). After 48 h stimulation, total RNA was extracted from the stimulated cells. The cytokine expression was analyzed using the QuantStudio™ 7 FlexReal-Time PCR System. (A) IL-1β, (B) IL-2, (C) IL-4, (D) IL-6, (E) IL-10, (F) TNF-α, (G) IFN-γ, (H) TLR-4, and (I) COX-2 genes. Significant differences were observed at P < 0.05 compared with the saline (*) and ginseng (#) group.

https://doi.org/10.1371/journal.pone.0211570.g002

Effect of CFAMs treatment on peritoneal macrophage proliferation and nitric oxide production

In order to investigate the effect of CFAMs on macrophage proliferation and nitric oxide production, peritoneal macrophages were isolated from each group of mice and stimulated using LPS. As shown in Fig 3A and 3B, compared to the CY-treated mice, the CFAMs-treated mice displayed a significant and dose-dependent increase in peritoneal macrophage proliferation and nitric oxide (NO) production. At high doses (250–500 mg/kg BW), CFAMs restored peritoneal macrophage proliferation and nitric oxide production. These restored levels were similar to those observed for the non-treated mice (normal group), and similar or higher than those observed for the ginseng-treated group.

thumbnail
Fig 3. Effects of different concentrations of CFAMs on peritoneal macrophage.

Peritoneal macrophages were isolated from each group of mice in the sacrifice day. The cells were stimulated with or without 1 μg/mL of LPS. LPS was used to investigate the immune response of macrophages isolated from each group. After 24h stimulated, the stimulated-cells were used for the peritoneal macrophage proliferation assay (A), and the cultured medium was used for the nitric oxide production assay (B). Phagocytosis of peritoneal macrophages was determined using the neutral red uptake (C). Significant differences were observed at P < 0.01 compared with the saline group (*) and at P < 0.05 compared with the ginseng group (#).

https://doi.org/10.1371/journal.pone.0211570.g003

Effect of CFAMs on peritoneal macrophage phagocytosis

The neutral red uptake method has been used for the evaluation of phagocytosis by mouse peritoneal macrophages. Fig 3C shows that peritoneal macrophage phagocytosis was significantly lower in the CY group than in the normal group. Compared to the CY-treated group, peritoneal macrophage phagocytosis rates were remarkably and dose-dependently higher in the CFAMs-treated group. High doses of CFAMs also induced the recovery of peritoneal macrophage phagocytosis.

Effect of CFAMs treatment on immune-associated gene expression in peritoneal macrophages

Immune-associated gene expression in peritoneal macrophages, in addition to that observed in splenic lymphocytes, was analyzed in order to investigate the effect of CFAMs treatment on the activation of peritoneal macrophages from the CY-treated mice. Fig 4 shows that the expression of iNOS, IL-6, TNF-α, COX-2, and IFN-γ was remarkably downregulated in the CY-treated mice. Compared to the CY-treated group, the CFP-treated group showed a dose-dependent and significantly higher expression of these immune-associated genes. Furthermore, peritoneal macrophages treated with 100, 250, and 500 mg/kg BW CFAMs showed similar or higher gene expression than the red ginseng-treated group (positive control).

thumbnail
Fig 4. Relative expression (fold-change) of immune genes in LPS-stimulated peritoneal macrophages.

Peritoneal macrophages were stimulated with 1 μg/mL of LPS. After 24 h stimulation, total RNA was extracted from the stimulated cells. The cytokine expression was analyzed using the QuantStudio™ 7 FlexReal-Time PCR System. (A) iNOS, (B) IL-6, (C) TNF-α, (D) COX-2, and (E) IFN-γ. Significant differences were observed at P < 0.05 compared to the saline (*) and ginseng (#) groups.

https://doi.org/10.1371/journal.pone.0211570.g004

Effect of CFAMs treatment on NF-κB and MAPK signaling pathways

To analyze the mechanism by which CFAMs regulated immune responses in physiological systems, RAW264.7 cells were grown in the presence of varying concentrations of CFAMs. As shown in Fig 5, compared to the negative control (RPMI), CFAMs treatment increased phospholyration of IκBα and NF-κB-p65 in a dose-dependent manner. In addition, phospholyration of ERK, JNK, and p38, which are the key biomarkers for the mitogen-activated protein kinase (MAPK) pathway, was also investigated. It was observed that CFAMs increased the phosphorylation of these proteins in a dose-dependent manner.

thumbnail
Fig 5. Effects of CFAMs on the proteins associated with NF-κB and MAPK pathways in RAW264.7 cells.

Cells were stimulated with various concentrations of CFAMs or 1 μg/mL of LPS for 24 h. Total protein was extracted from each treatment and was separated on SDS-PAGE. The separated protein was transferred on to PVDF membrane. The membrane was incubated with the specific antibodies and was detected using the Pierce® ECL Plus Western Blotting Substrate and were imaged using the ChemiDoc XRS+ imaging system. (A) Western blots showing protein bands, (B) Relative band intensity.

https://doi.org/10.1371/journal.pone.0211570.g005

Effect of CFAMs treatment on NF-κB and MAPK activation inhibited-RAW264.7 cells

In order to investigate the effects of CFAMs on cellular signaling for immune-regulation, TNF-α expression was examined in NF-κB and MAPK activation inhibited-RAW264.7 cells (Fig 6). Compared to the RPMI group (negative control), the CFAMs-treated group showed increased expression of TNF-α in the NF-κB and MAPK activation inhibited-macrophages in a dose-dependent manner. Notably, in the NF-κB and ERK inhibited-RAW264.7 cells treated with high doses of CFAMs (for NF-κB inhibited cells, CFAMs = 7.5 and 15 μg/mL; for ERK inhibited cells, CFAMs = 15 μg/mL), TNF-α expression was restored and similar to or higher than that observed for the RPMI group (negative control).

thumbnail
Fig 6. Relative expression (fold-change) of TNF-α in CFAM-treated NF-κB and MAPK activation inhibited-RAW264.7 cells.

RAW264.7 cells were treated with the f NF-κB activation inhibitor and MAPKs activation inhibitors. After that, these cells were stimulated with various concentrations of CFP or 1 μg/mL of LPS. After total RNA extraction, TNF-α expression was analyzed using the QuantStudio™ 7 FlexReal-Time PCR System. Significant differences were observed at P < 0.01 compared to the control group (*).

https://doi.org/10.1371/journal.pone.0211570.g006

Discussions

In humans, immune-regulation is important for protection against several diseases, including tumorigenesis [30]. CY, a well-known chemotherapeutic drug used for tumor treatment, induces adverse effects on the immune system and causes immunosuppression, which can be life threatening [31]. Therefore, many natural compounds having immune-modulation activities have been reported recently. These compounds alleviate the deleterious effects of chemotherapeutic drugs [1, 3032]. Particularly, polysaccharides with low toxic side-effects have been used as adjuvant medications or immunomodulators for strengthening host defense responses [33, 34]. In the present study, we determined the efficacy of polysaccharides isolated from C. fragile in enhancing immunological functions in immunosuppressed BALB/c mice.

The spleen is an important lymphoid organ consisting of immune cells, such as monocytes, macrophages, B and T lymphocytes, for mediating immune responses [35]. Spleen-associated factors, such as spleen index and NK cell activities, reflect the immune status of an individual [36]. Especially, lymphocyte proliferation is the most direct indicator of the state of host immunity [37]. T and B lymphocytes are known to react with stimulation activated by antigens or mitogens, which is a common type of non-specific immune regulation [12]. Proliferation of T and B lymphocytes is induced by Con A and LPS, respectively [38]. Fig 1 shows that CFAM accelerated the recovery of spleen size and spleen index in the CY-induced immunosuppressed mice. CFAM also promoted T- and B-cell proliferation and NK cell activities in splenocytes in a dose-dependent manner. Previous studies reported that CY could inhibit both humoral and cellular immune responses [39, 40]. Notably, CFAM-treated mice did not show any mortality. Additionally, compared to the normal group, their body weights also did not change significantly (S1 Fig) [30, 41, 42]. These results indicated that under immunosuppressive conditions, CFAMs treatment establishes immune homeostasis in physiological systems.

Macrophages are immune cells which play important roles in combating infection and removing tumor cells in hosts. They are also closely associated with immune-modulation in cancer therapy [43]. Phagocytes, including macrophages, are important for the regulation of innate immune responses. Phagocytosis is the mechanism by which the phagocytes ingest and kill microorganisms or cancer cells [32]. Increased production of NO by macrophages protects host cells from infection [44]. Macrophages also play an important role as antigen-presenting cells and coordinate with T lymphocytes for regulation of adaptive immune responses [32]. Fig 3 shows that in immunosuppressed mice, CY impaired peritoneal macrophage proliferation and NO production; however, CFAMs enhanced proliferation and NO production of peritoneal macrophages in a dose-dependent manner. Furthermore, CFAMs considerably increased phagocytosis by peritoneal macrophages in a dose-dependent manner, suggesting that CFAMs could enhance non-specific immune functions in CY-treated immunosuppressed mice to improve immune homeostasis.

Several subtypes of T cells, including T helper (Th) cells, cytotoxic T cells, suppressor T cells, effector T cells, are present. Particularly, the type 1 (Th1) and type 2 (Th2) T helper cells play different roles under different immune conditions. Th1 cells are activated during cell-mediated immune responses; however, Th2 cells are stimulated during humoral or allergic responses [45]. Th1 and Th2 cells produce specific cytokines to directly and indirectly control specific immune responses. IL-2, IFN-γ, and TNF-α are regulated by Th1 cells, whereas IL-4, IL-6, and IL-10 are modulated by Th2 cells [46]. Our results showed that cytokine expression in the CFAMs-treated group was higher than that in the CY-treated group. This suggested that CFAMs, unlike in CY-treated group, stimulated the secretion of Th1 and Th2 cytokines to restore immunosuppression and control the dynamic balance between the Th1 and Th2 cells in mice [47].

To regulate immune responses, intermediates of the NF-κB signaling pathway coordinate with inflammatory intermediates, including iNOS, COX-2, and the pro-inflammatory cytokines [48]. In addition, the MAPK signaling pathway involving ERK1/2, JNK, and p38 regulates cellular differentiation and growth by modulating immune responses during stress [49]. MAPK, along with NF-κB, regulates inflammatory cytokine production and related processes [50]. Our results showed that CFAMs increased the phosphorylation of IκBα, NF-κB-p65, ERK, JNK, and p38. Among these, phosphorylation of JNK was the highest. Moreover, it was shown that TNF-α expression, widely used as an experimental model for studying immunomodulatory activities of polysaccharides [36], was affected in the NF-κB and MAPK inhibited cells. In these cells, the expression of TNF-α was observed to be considerably low. These data suggested that CFAMs activated macrophages via the MAPK pathway and involved the phosphorylation of JNK.

Conclusions

The present study demonstrated that CFAMs, anionic macromolecules from C. fragile, enhanced the immune responses in CY-treated immunosuppressed mice via peritoneal macrophage proliferation, NO production and phagocytosis by peritoneal macrophages, enhancing macrophage gene expression, increasing splenic lymphocyte proliferation and NK cell activity, spleen index, and splenic lymphocyte gene expression. Additionally, it was shown using macrophages and NF-κB- and MAPK-inhibited cells that CFAMs stimulated macrophages via the MAPK pathway. Consequently, our results might help in elucidating the immune-enhancing mechanisms of CFAMs under immune-suppressive conditions in cancer treatment. In conclusion, CFAMs may be used as a potent biofunctional and pharmaceutical material for enhancing immunity in humans.

Supporting information

S1 Fig. Effects of different concentrations of CFAMs on body weight of mice.

https://doi.org/10.1371/journal.pone.0211570.s001

(TIF)

References

  1. 1. Raj S, Gothandam KM. Immunomodulatory activity of methanolic extract of Amorphophallus commutatus var. wayanadensis under normal and cyclophosphamide induced immunosuppressive conditions in mice models. Food Chem Toxicol. 2015;81: 151–159. pmid:25916916
  2. 2. Walls J, Sinclair L, Finlay D. Nutrient sensing, signal transduction and immune responses. Semin Immunol. 2016;28(5): 396–407. pmid:27681670
  3. 3. Prendergast GC, Jaffee EM. Cancer immunologists and cancer biologists: why we didn't talk then but need to now. Cancer Res. 2007;67(8): 3500–3504. pmid:17413003
  4. 4. Jesenak M, Urbancikova I, Banovcin P. Respiratory tract infections and the role of biologically active polysaccharides in their management and prevention. Nutrients. 2017;9(7). pmid:28726737
  5. 5. Liu X, Lin Q, Yan Y, Peng F, Sun R, Ren J. Hemicellulose from plant biomass in medical and pharmaceutical application: A critical review. Curr Med Chem. 2017. pmid:28685685
  6. 6. Patel S, Goyal A. Chitin and chitinase: Role in pathogenicity, allergenicity and health. Int J Biol Macromol. 2017;97: 331–338. pmid:28093332
  7. 7. Daien CI, Pinget GV, Tan JK, Macia L. Detrimental impact of microbiota-accessible carbohydrate-deprived diet on gut and immune homeostasis: An overview. Front Immunol. 2017;8: 548–554. pmid:28553291
  8. 8. Iida Y, Harashima N, Motoshima T, Komohara Y, Eto M, Harada M. Contrasting effects of cyclophosphamide on anti-CTL-associated protein 4 blockade therapy in two mouse tumor models. Cancer Sci. 2017;108(10): 1974–1984. pmid:28787548
  9. 9. Pass GJ, Carrie D, Boylan M, Lorimore S, Wright E, Houston B, et al. Role of hepatic cytochrome p450s in the pharmacokinetics and toxicity of cyclophosphamide: studies with the hepatic cytochrome p450 reductase null mouse. Cancer Res. 2005;65(10): 4211–4217. pmid:15899812
  10. 10. Umesue M, Mayumi H, Nishimura Y, Kong YY, Omoto K, Murakami Y, et al. Donor-specific prolongation of rat skin graft survival induced by rat-donor cells and cyclophosphamide under coadministration of monoclonal antibodies against T cell receptor alpha beta and natural killer cells in mice. Transplantation. 1996;61(1): 116–124. pmid:8560549
  11. 11. Singh KP, Gupta RK, Shau H, Ray PK. Effect of ASTA-Z 7575 (INN Maphosphamide) on human lymphokine-activated killer cell induction. Immunopharmacol Immunotoxicol. 1993;15(5): 525–538. pmid:8301017
  12. 12. Wang JX, Tong X, Li PB, Cao H, Su WW. Immuno-enhancement effects of Shenqi Fuzheng injection on cyclophosphamide-induced immunosuppression in Balb/c mice. J Ethnopharmacol. 2012;139(3): 788–795. pmid:22212503
  13. 13. Lee JB, Ohta Y, Hayashi K, Hayashi T. Immunostimulating effects of a sulfated galactan from Codium fragile. Carbohydr Res. 2010;345(10): 1452–1454. pmid:20362278
  14. 14. Choi JH, Sapkota K, Park SE, Kim S, Kim SJ. Thrombolytic, anticoagulant and antiplatelet activities of codiase, a bi-functional fibrinolytic enzyme from Codium fragile. Biochimie. 2013;95(6): 1266–1277. pmid:23402909
  15. 15. Kang CH, Choi YH, Park SY, Kim GY. Anti-inflammatory effects of methanol extract of Codium fragile in lipopolysaccharide-stimulated RAW264.7 cells. J Med Food. 2012;15(1): 44–50. pmid:22082064
  16. 16. Tabarsa M, Karnjanapratum S, Cho M, Kim JK, You SG. Molecular characteristics and biological activities of anionic macromolecules from Codium fragile. Int J Biol Macromol. 2013;59: 1–12. pmid:23597705
  17. 17. Surayot U, You SG. Structural effects of sulfated polysaccharides from Codium fragile on NK cell activation and cytotoxicity. Int J Biol Macromol. 2017;98: 117–124. pmid:28130139
  18. 18. Tabarsa M, Park GM, Shin IS, Lee E, Kim JK, You SG. Structure-activity relationships of sulfated glycoproteins from Codium fragile on nitric oxide releasing capacity from RAW264.7 Cells. Mar Biotechnol (NY). 2015;17(3): 266–276. pmid:25627693
  19. 19. Sevag MG, Lackman DB, Smolens J. The isolation of the components of streptococcal nucleoproteins in serologically active form. J Biol Chem. 1938;124: 425–436.
  20. 20. Ray A, Dittel BN. Isolation of mouse peritoneal cavity cells. J Vis Exp. 2010(35): e1488–1490. pmid:20110936
  21. 21. Kim JK, Cho ML, Karnjanapratum S, Shin IS, You SG. In vitro and in vivo immunomodulatory activity of sulfated polysaccharides from Enteromorpha prolifera. Int J Biol Macromol. 2011;49(5): 1051–1058. pmid:21907732
  22. 22. Cao RA, Surayot U, You S. Structural characterization of immunostimulating protein-sulfated fucan complex extracted from the body wall of a sea cucumber, Stichopus japonicus. Int J Biol Macromol. 2017;99: 539–548. pmid:28279766
  23. 23. Weeks BA, Keisler AS, Myrvik QN, Warinner JE. Differential uptake of neutral red by macrophages from three species of estuarine fish. Dev Comp Immunol. 1987;11(1): 117–124. pmid:3595937
  24. 24. Cho CW, Han CJ, Rhee YK, Lee YC, Shin KS, Shin JS, et al. Cheonggukjang polysaccharides enhance immune activities and prevent cyclophosphamide-induced immunosuppression. Int J Biol Macromol. 2015;72: 519–525. pmid:25236609
  25. 25. Park HR, Lee HS, Cho SY, Kim YS, Shin KS. Anti-metastatic effect of polysaccharide isolated from Colocasia esculenta is exerted through immunostimulation. Int J Mol Med. 2013;31(2): 361–368. pmid:23292184
  26. 26. Sarangi I, Ghosh D, Bhutia SK, Mallick SK, Maiti TK. Anti-tumor and immunomodulating effects of Pleurotus ostreatus mycelia-derived proteoglycans. Int J Immunopharmacol. 2006;6(8): 1287–1297. pmid:16782541
  27. 27. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4): 402–408. pmid:11846609
  28. 28. Narayanan BA, Narayanan NK, Simi B, Reddy BS. Modulation of inducible nitric oxide synthase and related proinflammatory genes by the omega-3 fatty acid docosahexaenoic acid in human colon cancer cells. Cancer Res. 2003;63(5): 972–979. pmid:12615711
  29. 29. Bhattacharyya S, Ratajczak CK, Vogt SK, Kelley C, Colonna M, Schreiber RD, et al. TAK1 targeting by glucocorticoids determines JNK and IkappaB regulation in Toll-like receptor-stimulated macrophages. Blood. 2010;115(10): 1921–1931. pmid:20065289
  30. 30. Yu ZD, Kong ML, Zhang PY, Sun QJ, Chen KH. Immune-enhancing activity of extracellular polysaccharides isolated from Rhizopus nigricans. Carbohydr Polym. 2016;148: 318–325. pmid:27185145
  31. 31. Wang H, Wang MY, Chen J, Tang Y, Dou J, Yu J, et al. A polysaccharide from Strongylocentrotus nudus eggs protects against myelosuppression and immunosuppression in cyclophosphamide-treated mice. Int J Immunopharmacol. 2011;11(11): 1946–1953. pmid:21723424
  32. 32. Chen JR, Yang ZQ, Hu TJ, Yan ZT, Niu TX, Wang L, et al. Immunomodulatory activity in vitro and in vivo of polysaccharide from Potentilla anserina. Fitoterapia. 2010;81(8): 1117–1124. pmid:20624446
  33. 33. Sun YX. Structure and biological activities of the polysaccharides from the leaves, roots and fruits of Panax ginseng C.A. Meyer: An overview. Carbohydr Polym. 2011;85(3): 490–499.
  34. 34. Tzianabos AO. Polysaccharide immunomodulators as therapeutic agents: structural aspects and biologic function. Clin Microbiol Rev. 2000;13(4): 523–533. pmid:11023954
  35. 35. Lori A, Perrotta M, Lembo G, Carnevale D. The spleen: A hub connecting nervous and immune systems in cardiovascular and metabolic diseases. Int J Mol Sci. 2017;18(6): 1216–1227. pmid:28590409
  36. 36. Chen X, Nie W, Fan S, Zhang J, Wang Y, Lu J, et al. A polysaccharide from Sargassum fusiforme protects against immunosuppression in cyclophosphamide-treated mice. Carbohydr Polym. 2012;90(2): 1114–1119. pmid:22840047
  37. 37. Tu J, Sun H-X, Ye Y-P. Immunomodulatory and antitumor activity of triterpenoid fractions from the rhizomes of Astilbe chinensis. J Ethnopharmacol. 2008;119(2): 266–271. pmid:18692123
  38. 38. Quakyi EK, Frasch CE, Buller N, Tsai CM. Immunization with meningococcal outer-membrane protein vesicles containing lipooligosaccharide protects mice against lethal experimental group B Neisseria meningitidis infection and septic shock. J Infect Dis. 1999;180(3): 747–754. pmid:10438363
  39. 39. Bear HD. Tumor-specific suppressor T-cells which inhibit the in vitro generation of cytolytic T-cells from immune and early tumor-bearing host spleens. Cancer Res. 1986;46(4 Pt 1): 1805–1812.
  40. 40. Hoover SK, Barrett SK, Turk TM, Lee TC, Bear HD. Cyclophosphamide and abrogation of tumor-induced suppressor T cell activity. Cancer Immunol Immunother. 1990;31(2): 121–127. pmid:2138931
  41. 41. Ren Z, He CH, Fan YH, Guo LW, Si HM, Wang YW, et al. Immuno-enhancement effects of ethanol extract from Cyrtomium macrophyllum (Makino) Tagawa on cyclophosphamide-induced immunosuppression in BALB/c mice. J Ethnopharmacol. 2014;155(1): 769–775. pmid:24960181
  42. 42. Zheng Y, Zong ZM, Chen SL, Chen AH, Wei XY. Ameliorative effect of Trametes orientalis polysaccharide against immunosuppression and oxidative stress in cyclophosphamide-treated mice. Int J Biol Macromol. 2017;95: 1216–1222. pmid:27825995
  43. 43. Manosroi A, Saraphanchotiwitthaya A, Manosroi J. Immunomodulatory activities of Clausena excavata Burm. f. wood extracts. J Ethnopharmacol. 2003;89(1): 155–160. pmid:14522448
  44. 44. Rahat MA, Hemmerlein B. Macrophage-tumor cell interactions regulate the function of nitric oxide. Front Physiol. 2013;4: 144–159. pmid:23785333
  45. 45. Constant SL, Bottomly K. Induction of Th1 and Th2 CD4+ T cell responses: the alternative approaches. Annu Rev Immunol. 1997;15: 297–322. pmid:9143690
  46. 46. Mosmann TR, Coffman RL. Heterogeneity of cytokine secretion patterns and functions of helper T cells. Adv Immunol. 1989;46: 111–147. pmid:2528896
  47. 47. Lee EJ, Ko E, Lee J, Rho S, Ko S, Shin MK, et al. Ginsenoside Rg1 enhances CD4(+) T-cell activities and modulates Th1/Th2 differentiation. Int J Immunopharmacol. 2004;4(2): 235–244. pmid:14996415
  48. 48. Tak PP, Firestein GS. NF-kappaB: a key role in inflammatory diseases. J Clin Invest. 2001;107(1): 7–11. pmid:11134171
  49. 49. Kim JB, Han AR, Park EY, Kim JY, Cho W, Lee J, et al. Inhibition of LPS-induced iNOS, COX-2 and cytokines expression by poncirin through the NF-kappaB inactivation in RAW264.7 macrophage cells. Biol Pharm Bull. 2007;30(12): 2345–2351. pmid:18057724
  50. 50. Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 2011;75(1): 50–83. pmid:21372320