Scientific Research for Immunity-Boosting Power

Immunity-BOOSTING POWER

Immunity-Booster is a unique combo of immune-stimulating mushrooms with Gac fruit extract, intended for poor, weak, and fragile immunity. It significantly provides immunomodulatory properties and protects cellular integrity, strengthening the immune response against various pathological and hazardous agents. Moreover, Immunity-Booster by FUNGAC Essentials Inc. suppresses mild illnesses, stimulates immune cells production, and increases the body's resistance against infectious agents. Also, it modulates gut microbiota, improves gut immunity, and reduces the risk of various gastrointestinal diseases. Thus, regular consumption of Immunity-Booster increase immunity, protects vital body organs, and enhances the quality of life. The scientifically proven immune-stimulatory mushrooms are processes in a well-certified facility following GMP guidelines. Note that Immunity-Booster by FUNGAC Essentials Inc. is a non-GMO, gluten-free, and organic formulation, effective for both men and women. Additionally, the final formulation is packed with Black Pepper Extract for maximum absorption and protection in the gastrointestinal tract.

Features of Immunity-Booster Power

  • Natural Ingredients Formulation
  • Non-GMO and Gluten Free
  • No Risk of Side Effects
  • Boosts Immune Response
  • Protect Cellular Integrity
  • Combat Pathological Attack
  • Promote Quality of Life
  • Support Longevity
  • Effective for both Men and Women
  • Quality Mushrooms at Cheap Price

How does Immunity-Booster Power Works?

Adaptogenic mushrooms combined with Gac fruit extract stimulate immune cells production and level up the immune response against pathological conditions. Moreover, it provides antioxidant and anti-inflammatory properties that relieve oxidative stress and block pro-inflammatory cytokines production. As a result, Immunity-Booster prevents organ damage, removes toxins, and boosts innate and adaptive immunity. Additionally, it increases the natural antioxidant capability of the body by regulating the expression of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px).

Health Benefits of Immune-Booster Power 

Strengthen Immune System

Immunity-Booster delivers potent antioxidant and anti-inflammatory mushrooms that boost and strengthen the immune system. It significantly potentiates immune cells production, including macrophages, T-Lymphocytes, natural killer cells, and other defensive cells, in the body. As a result, Immunity-Booster by FUNGAC Essentials Inc. prepares the body for combat against infectious diseases and other environmental toxins. 

Prevent Medical Conditions

The presence of Immuno-stimulatory mushrooms combined with Gac Fruit increases the body's resistance against pathological agents and decreases the risk of infectious diseases. Moreover, it supports the body to effectively fight against various Bacterial and Viral strains, including Herpes simplex virus types (HSV-1 and HSV-2), B. Subtilis, S. Aureus, E. coli, P. Aeruginosa, and Vesicular stomatitis virus (VSV). Additionally, the combo mushroom formula improves liver and gastrointestinal diseases such as cirrhosis, ulcer, lesions, and abnormal enzymes elevation.

Enhance Quality of Life and Longevity

Regular consumption of Gac fruit extract and adaptogenic mushrooms preserve cellular integrity, promote vital organs functions and prevent various medical conditions. As a result, Immunity-Booster enhance the quality of life, vitality and extends longevity. Besides, it supports cardiovascular functions, improves gastrointestinal health, aid cellular immunity, and provides enormous health benefits. 

Note, please read the Immune-Booster Power label for suggested dose and precautionary measures.

References (Medical Research Studies)

 


 

Copy Data about Immunity Booster Composition

Support Immune system:

Studies also reported the antioxidant and antimicrobial activity of Reishi. It helps infectious diseases caused by different strains of Bacteria and Viruses, including Herpes simplex virus types (HSV-1 and HSV-2), B. Subtilis, S. Aureus, E. coli, P. Aeruginosa, and Vesicular stomatitis virus (VSV). Also, it shows a positive effect on the liver via ameliorating cirrhosis and preventing an elevated serum level of Alkaline phosphatase (ALP), Aspartate, and Alanine transaminases. Plus, provide gastric healing activity and speed up the ulcer or gastrointestinal lesion healing. [1, 4, 12, 13]

  1. Wachtel-Galor S, Yuen J, Buswell JA, et al. Ganoderma lucidum (Lingzhi or Reishi): A Medicinal Mushroom. In: Benzie IFF, Wachtel-Galor S, editors. Herbal Medicine: Biomolecular and Clinical Aspects. 2nd edition. Boca Raton (FL): CRC Press/Taylor & Francis; 2011. Chapter 9. Available from: https://www.ncbi.nlm.nih.gov/books/NBK92757/
  2. Li Z, Liu J, Zhao Y. Possible mechanism underlying the antiherpetic activity of a proteoglycan isolated from the mycelia of Ganoderma lucidum in vitro. J Biochem Mol Biol. 2005;38(1):34–40.
  3. Keypour S, Riahi H, Moradali M. F, Rafati H. Investigation of the antibacterial activity of a chloroformextract of Lingzhi or Reishi medicinal mushroom, Ganoderma lucidum (W. Curt.: Fr.) P. Karst. (Aphyllophoromycetideae). Int J Med Mushrooms. 2008;10(4):345–9.
  4. Lin J. M, Lin C. C, Chen M. F, Ujiie T, Takada A. Radical scavenger and antihepatotoxic activity of Ganoderma formosanum, Ganodermalucidum and Ganoderma neo-japonicum. J Ethnopharmacol. 1995;47:33–41.

Strengthen Immune System

AM extract modulates the immune response against various pathological agents, including viruses, bacteria, and other germs. It boosts the immunity level and prevents the risk of different medical conditions. Medical studies reported that AM stimulates and increases immune cells' production, including T-lymphocytes, Natural Killer cells, and improved the level of certain antibodies such as IgG, IgA, and IgM. Thus, it strengthens both innate and adaptive immunity and supports immune responses against illnesses. Studies reported that AM root extracts could be used as immunological adjuncts in certain conditions for better immune response.

Antioxidant Activity

AM roots extract shows potent antioxidant activity and supports vital organ functions. It neutralizes free radicals and removes toxins from the body. Thus, it preserves cellular integrity and decreases the risk of medical conditions. Moreover, AM extract regulates the expression of natural antioxidant enzymes in the body, including superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Also, it removes reactive oxygen species from mitochondria and protects against oxidative damage. Thus, it improves cellular integrity and promotes cellular energy production.

Antiinflammatory Activity:

AM inhibits the production and secretion of various pro-inflammatory cytokines and decreases the risk of inflammation. Studies showed that it suppresses the production of leukotrienes, tumor necrosis factor-alpha, and nitric oxide. Plus, it also inhibits the cyclooxygenase and other enzymatic pathways responsible for inflammatory response. Thus, it relieves inflammation and decreases the pathogenesis of various underlying inflammatory conditions such as diabetes.

 

Reishi

Agents that enhance the functioning of the host immune system could be expected to enhance health in terms of improved resistance and, thus, removal of malignant or premalignant cells. Many G. lucidum products on the market are labeled or promoted as immunomodulating agents.

There is considerable evidence to support the immunostimulating activities of G. lucidum via induction of cytokines and enhancement of immunological effector (Wang et al. 1997Zhu and Lin 2006). Different components from G. lucidum were proved to enhance the proliferation and maturation of T and B lymphocytes, splenic mononuclear cells, NK cells, and dendritic cells in culture in vitro and in animal studies in vivo (Bao et al. 2001Cao and Lin 2002Zhu, Chen, and Lin 2007Ma et al. 2008). In normal BALB/c mice, a polysaccharide-rich extract of G. lucidum promoted the proliferation of splenocytes and enhanced the activities of macrophages and NK cells, which resulted in the increase of IL-6 and IFN-γ (Chang et al. 2009). Although a commercial G. lucidum extract did not stimulate proliferation of lymphocytes, it activated the gene expression of IL-1β, IL-6, IL-10, and tumor necrosis factor (TNF)-α (Mao et al. 1999). A polysaccharide fraction (F3) was shown to enhance both adaptive and innate immunities by triggering the production of cytokines IL-1, IL-6, IL-12, IFN-γ, TNF-α, and colony stimulating factors (CSFs) from mouse splenocytes (Chen et al. 2004). It was reported also that TNF-α and IL-6 production were stimulated in human and murine macrophages by G. lucidum mycelia (Kuo et al. 2006). This effect might be due to increased synthesis of nitric oxide (NO) induced by β-D-glucan (Ohno et al. 1998). These polysaccharides were also found to be highly suppressive to tumor cell proliferation in vivo while enhancing the host’s immune response (Ooi and Liu 2000).

Wang et al. (1997) found that a polysaccharide-enriched fraction from G. lucidum activated cultured macrophages and T lymphocytes in vitro, which led to an increase of IL-1β, TNF-α, and IL-6 in the culture medium. In another study (Zhang and Lin 1999), incubation of macrophages and T lymphocytes with a polysaccharide resulted in an increase in TNF-α and INF-γ levels in the culture medium. This “conditioned” culture medium was found to inhibit cell growth and induce apoptosis in sarcoma 180 and HL-60 cells (Zhang and Lin 1999). Furthermore, serum-incorporated treatment with a polysaccharide peptide fraction from G. lucidum markedly inhibited the proliferation of human lung carcinoma (PG) cells, whereas the pure fraction by itself did not induce similar effects (Cao and Lin 2004). In addition to polysaccharides, a lanostane triterpenoid, ganoderic acid Me, inhibited tumor growth and metastasis of Lewis lung carcinoma in “T helper 1 responder” C57BL/6 mice by enhancing immune function in terms of IL-2 and IFN-γ expression and NK cell activity (Wang et al. 2007). Zhu and Lin (2006) used cytokine-induced killer (CIK) cells to investigate the interaction between GL-PSs and cytokines, which mediated cell proliferation and antitumor activity. The cytotoxicity of CIK cells was correlated well with the expression of perforin and granzyme B induced by IL-2 and anti-CD3. Results indicated that GL-PSs enhance IL-2 and TNF-α production as well as protein and messenger ribonucleic acid (mRNA) expression of granzyme B and perforin in CIK cells culture, and thus decrease the doses of IL-2 and anti-CD3 without affecting the killing effects on NK-resistant mouse P815 mastocytoma cells and NK-sensitive mouse YAC-1 lymphoma cells (Zhu and Lin 2006).

https://www.ncbi.nlm.nih.gov/books/NBK92757/

Consumption of antioxidant-rich plants may help prevent cancer and other chronic diseases (Collins 2005Benzie and Wachtel-Galor 2009). Antioxidants protect cellular components from oxidative damage, which is likely to decrease risk of mutations and carcinogenesis and also protect immune cells, allowing them to maintain immune surveillance and response. Various components of G. lucidum, in particular polysaccharides and triterpenoids, show antioxidant activity in vitro (Lee et al. 2001Mau, Lin, and Chen 2002Shi et al. 2002Wachtel-Galor, Choi, and Benzie 2005Yuen and Gohel 2008Saltarelli et al. 2009Wu and Wang 2009). As shown in Figure 9.4, antioxidants from lingzhi were found to be absorbed quickly after ingestion, resulting in an increase in the plasma total antioxidant activity of human subjects (Figure 9.4Wachtel-Galor, Szeto et al. 2004).

The goal of research in the treatment of viral and bacterial infections is the discovery of agents that specifically inhibit viral and bacterial multiplication without affecting normal cells. The undesired side effects of antibiotics and antivirals and the appearance of resistant and mutant strains make the development of new agents an urgent requirement. This has led researchers to investigate the antibacterial and antiviral activity of medicinal plants and fungi (Wasser and Weis 1999Zhong and Xiao 2009). Isolation of various water- and methanol-soluble, high-molecular-weight PBPs from G. lucidum showed inhibitory effects on herpes simplex virus type 1 (HSV-1), herpes simplex virus type 2 (HSV-2), and vesicular stomatitis virus (VSV) New Jersey strain in a tissue culture system. Using the plaque reduction method, a significant inhibitory effect was seen at doses that showed no cytotoxicity (Eo et al. 1999Oh et al. 2000). In addition, there was a marked synergistic effect when PBP from G. lucidum was used in tissue culture in conjunction with antiherpetic agents, acyclovir or vidarabine, and with IFN-α (Kim et al. 2000Oh et al. 2000). Similar results were shown in HSV-1 and HSV-2 with a GLPG isolated from the mycelia of G. lucidum (Liu et al. 2004; Li, Liu, and Zhao 2005). The cells were treated before, during, and after infection, and viral titer in the supernatant of cell culture 48 hours postinfection was determined. The antiviral effects of the GLPG were more remarkable before viral treatment than after treatment. Although the mechanism was not defined, the authors concluded that GLPG inhibits viral replication by interfering with early events of viral adsorption (Li, Liu, and Zhao 2005).

For evaluating the antibacterial effects of the mushroom, several in vitro and in vivo animal studies using G. lucidum were performed. Mice injected with G. lucidum extract (2 mg/mouse) 1 day prior to injection with Escherichia coli showed markedly improved survival rates (>80% compared to 33% in controls; Ohno et al. 1998). In an in vitro study that used the disk assay (Keypour et al. 2008), a chloroform extract of G. lucidum was investigated for its antibacterial effect on gram-positive bacteria (Bacillus subtilis, Staphylococcus aureus, Enterococcus faecalis) and gram-negative bacteria (E. coli, Pseudomonas aeruginosa). Results showed that the extract had growth-inhibitory effects on two of the gram-positive bacteria with a minimal inhibitory concentration (MIC) of 8 mg/mL for S. aureus and B. subtilis. In another in vitro study, the direct antimicrobial effect of a G. lucidum water extract was examined against 15 species of bacteria alone and in combination with 4 kinds of antibiotics (Yoon et al. 1994). G. lucidum was found to be more effective than antibiotics against E. coli, Micrococcus luteus, S. aureus, B. cereus, Proteus vulgaris, and Salmonella typhi, but less effective against other species tested. The antimicrobial combination of G. lucidum with four commonly used antibiotics (Yoon et al. 1994) resulted in an additive or synergistic effect in most, but not all, instances, with apparent antagonism against cefazolin and ampicillin effects on P. vulgaris.

To date, the antimicrobial components of the tested crude extracts have not been identified, although antimicrobial polysaccharides have been identified in other fungi and plant terpenes have been reported to have antimicrobial activity (Wasser and Weis 1999Zhong and Xiao 2009). In addition, the bioavailability of putative antimicrobial components of G. lucidum has not been established. Nonetheless, G. lucidum offers a potentially effective therapy. There is also the implication that combination therapy may be more safe and cost effective, as lower amounts of cytotoxic antiviral and antibacterial drugs could be used with a concomitant decrease in the risk of side effects. However, this needs further investigation in terms of in vitro studies and well-designed clinical trials.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4684115/

https://pubmed.ncbi.nlm.nih.gov/16169168/

https://pubmed.ncbi.nlm.nih.gov/30317947/

Ganoderma (Lingzhi) has been used for a long time in China to prevent and treat various diseases. Accumulated studies have demonstrated that the Ganoderma modulates immune function both in vivo and in vitro. The immunomodulating effects of Ganoderma were extensive, including promoting the innate immune function, humoral immunity, and cellular immunity. In particular, G. lucidum polysaccharides may affect immune cells and immune-related cells including B and T lymphocytes, dendritic cells, macrophages, and natural killer cells, with the promotion of immune organ growth, cytokine release, and other immune regulatory functions. Furthermore, cellular and molecular immunomodulatory mechanisms, possible receptors involved, and triggered signaling pathways have also been summarized. However, whole animal experiments are still needed to further establish the mechanism of the immunomodulating effects by Ganoderma. Importantly, evidence-based clinical trials are also needed.

https://pubmed.ncbi.nlm.nih.gov/31777013/

https://pubmed.ncbi.nlm.nih.gov/29953399/

Chaga

It has been reported that oral and intraperitoneal administration of endo-polysaccharides extracted from I. obliquus inhibits the in vivo growth of melanoma cells in mice via humoral immunity of the host defense system (Kim et al., 20052006). Our results showed that oral administration was effective for inhibition of melanoma solid tumor. In particular, pre-oral administration of PFIO was more rapid and effective for the inhibition of melanoma solid tumor than post-oral administration. These results suggested that inhibition of melanoma solid tumor occurred through upregulation and activation of the intestinal immune system. β-glucans, a major component of polysaccharides, is taken up by intestinal macrophages, after which it is transported to lymph nodes, spleen and bone marrow; accordingly, it can be concluded that oral administration of polysaccharides in as effective as parenteral administration for protecting against pathogen infections (Rop et al., 2009Volman et al., 2008). Therefore, our results elucidated that the anti-tumor effects of PFIO may have been exerted through activation of several immune response systems (Youn et al., 2009).

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3932689/

Inonotus obliquus extracts were found to inhibit hepatitis C virus14 and human immunodeficiency virus15 and demonstrated strong antioxidant and immunostimulatory activities in vitro.16,17 At the same time, animal studies revealed that aqueous extracts of I. obliquus exhibited anti-inflammatory effects in experimental colitis18-21 and promoted lipid metabolism.22 The mushroom has the ability to increase peroxisome proliferator-activated receptors γ transcriptional activities, which are expected to be therapeutic targets for dyslipidemia and type 2 diabetes.23

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6142110/

The Chaga mushroom (Inonotus obliquus) is claimed to have beneficial properties for human health, such as anti-bacterial, anti-allergic, anti-inflammatory and antioxidant activities. The antioxidant effects of the mushroom may be partly explained by protection of cell components against free radicals. We evaluated the effect of aqueous Chaga mushroom extracts for their potential for protecting against oxidative damage to DNA in human lymphocytes. Cells were pretreated with various concentrations (10, 50, 100 and 500 microg/mL) of the extract for 1 h at 37 degrees C. Cells were then treated with 100 microM of H2O2 for 5 min as an oxidative stress. Evaluation of oxidative damage was performed using single-cell gel electrophoresis for DNA fragmentation (Comet assay). Using image analysis, the degree of DNA damage was evaluated as the DNA tail moment. Cells pretreated with Chaga extract showed over 40% reduction in DNA fragmentation compared with the positive control (100 micromol H2O2 treatment). Thus, Chaga mushroom treatment affords cellular protection against endogenous DNA damage produced by H2O2.

https://pubmed.ncbi.nlm.nih.gov/15630179/

https://pubmed.ncbi.nlm.nih.gov/22484608/

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7868817/

In this study, we investigated the immunostimulating activity of polysaccharides isolated from fruiting body of Inonotus obliquus (PFIO). Additionally, the signaling pathway of PFIO-mediated macrophage activation was investigated in RAW264.7 macrophage cells. We found that PFIO was capable of promoting NO/ROS production, TNF-α secretion and phagocytic uptake in macrophages, as well as cell proliferation, comitogenic effect and IFN-γ/IL-4 secretion in mouse splenocytes. PFIO was able to induce the phosphorylation of three MAPKs as well as the nuclear translocation of NF-κB, resulting in activation of RAW264.7 macrophages. PFIO also induced the inhibition of TNF-α secretion by anti-TLR2 mAb, consequently, PFIO might be involved in TNF-α secretion via the TLR2 receptor. In addition, our results showed that oral administration of PFIO suppressed in vivo growth of melanoma tumor in tumorbearing mice. In conclusion, our experiments presented that PFIO effectively promotes macrophage activation through the MAPK and NF-κB signaling pathways, suggesting that PFIO may potentially regulate the immune response.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6706056/

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3932689/

Turkey Tail

Both aqueous and solid fractions of TvM triggered robust induction of CD69 on lymphocytes and monocytes, whereas FS only triggered minor induction of CD69, and IS had no activating effect. The aqueous extract of TvM had stronger activating effects than the solid fraction. In contrast, the solid fraction of IS triggered a reduction in CD69, below levels on untreated cells.

Both aqueous and solid fractions of FS triggered large and dose-dependent increases in immune-activating pro-inflammatory cytokines (IL-2, IL-6), anti-inflammatory cytokines Interleukin-1 receptor antagonist (IL-1ra) and Interleukin-10 (IL-10), anti-viral cytokines interferon-gamma (IFN-γ) and Macrophage Inflammatory Protein-alpha (MIP-1α), as well as Granulocyte-Colony Stimulating Factor (G-CSF) and Interleukin-8 (IL-8). TvM triggered more modest cytokine increases. The aqueous extract of IS showed no effects, whereas the solid fraction showed modest effects on induction of cytokines and growth factors.

The results demonstrated that the immune-activating bioactivity of a mycelial-based medicinal mushroom preparation is a combination of the mycelium itself (including insoluble beta-glucans, and also water-soluble components), and the highly bioactive, metabolically fermented substrate, not present in the initial substrate.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6889544/

A recent phase 1, dose-escalation, clinical trial of turkey tail evaluated dosing safety and immune function in women with breast cancer.47 Turkey tail extract was well-tolerated and was immunomodulatory at higher doses (6 g or 9 g) by increasing CD8+ T cells and CD19+ B cells. The researchers also found that the radiation-induced decline in NK cells was improved by a 6-gram dosing per day of turkey tail.

Agaricus has also been tested by Ohno et al in a phase I clinical study of safety with participants in cancer remission.53 At all doses—1.8, 3.6, and 5.4 g/d for 6 months, Agaricus was well-tolerated, with a 12% rate of adverse events that were digestive in nature, such as nausea. While Agaricus was deemed safe, the study did not follow immune outcomes for the enrolled patients.

Gao et al studied the use of reishi polysaccharides in late-stage cancer patients and late-stage, lung cancer patients.17,18 In participants with late-state lung cancer treated with 5.4 g/d of a proprietary reishi extract (Ganopoly), IL-2, IL-6, and IFN-γ increased. Great variability in patients’ responses occurred, with some participants having a very significant increase while others had minimal changes. This finding suggests that subgroups of patients may respond more favorably to reishi, although the mechanisms of such a difference have not been studied at this time. When Ganopoly was studied in late-stage cancer patients, it was found that a dose of 5.4 g/d increased IL-2, IL-6, and IFN-γ and decreased TNF-α and IL-1. This dosage also increased NK cells (CD56+ cells) and NK activity.

The immune-stimulating impact that mushrooms can exert on NK cells, macrophages, and T cells can also provide a protective effect against chemotherapeutic myelosuppression, one of the most serious deleterious effects of chemotherapy. Because severe myelosuppression neutropenia often truncates treatment and requires hospitalization before full therapeutic effects can be achieved, reducing myelosuppression would allow for better response to chemotherapy.54,55 One promising study examined the effect of the MD-fraction from the maitake mushroom on cisplatin-induced myelosuppression in a mouse model. Mice given 8 mg/kg/d while treated with cisplatin did not experience a decrease in NK cells, DCs, and macrophages.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4684115/

https://www.ncbi.nlm.nih.gov/books/NBK424937/

Since the earliest reports of clinical benefits, other investigators have sought to define the mechanism of PSK’s beneficial action. One group hypothesized that T-cell dysfunction, including apoptosis of peripheral blood T cells, commonly occurs in patients receiving chemotherapy.[12] They postulated that reversal of T-cell dysfunction induced by chemotherapy could reduce the adverse effects or enhance the antitumor effect. PSK is reported to enhance natural killer (NK) cell and T-cell activities by upregulation of interleukin-2 or interferon-gamma. Twenty patients with curatively resected stage III gastric cancer were randomly assigned to receive adjuvant therapy with the second-generation dihydropyrimidine dehydrogenase–inhibitory oral fluoropyrimidine S-1 alone (n = 10) or S-1 plus PSK (n = 10). At 5 weeks after adjuvant therapy, T-cell apoptosis was significantly higher in the S-1–alone group than in the S-1–plus-PSK group, leading the authors to conclude that PSK could partially prevent the T-cell apoptosis induced by S-1.

Another group of investigators studied the effect of PSK added to tegafur/uracil (UFT) chemotherapy compared with that of UFT alone.[13Baseline immune parameters were comparable in the two groups. However, CD57-positive T cells decreased more significantly after surgery for patients treated with PSK than for those in the control group (P = .0486). These investigators had previously noted that a high CD57-positive cell count was an indicator of poor prognosis in patients with advanced gastric cancer, leading them to suggest that PSK may improve overall survival (OS) partly by inhibiting CD57-positive T cells.

Noting that hosts become immunocompromised at the time of tumor progression and that decreased expression of major histocompatibility complex (MHC) class I by the tumor is one mechanism that allows it to evade destruction by cytotoxic T lymphocytes, investigators conducted a retrospective study to evaluate the expression of MHC class I by immunohistochemical staining in the primary lesions of patients with stage II or stage III gastric cancer.[14] They analyzed data from 349 patients who had undergone adjuvant therapy (after curative resection) between 1995 and 2008; 225 patients received adjuvant chemotherapy with an oral fluoropyrimidine alone, while 124 patients received adjuvant chemotherapy plus PSK 3 g/d. Although this was not a randomized trial, baseline characteristics of the patients were well matched. The mean duration of follow-up was 49 months. Three-year recurrence-free survival (RFS) rates were the same for both groups (60% for the PSK group and 62% for the chemotherapy-only group). For MHC expression–negative cases, the 3-year RFS rates were 65% for the PSK group and 50% for the chemotherapy-only group; the difference was not considered significant. For 82 MHC expression–negative patients with lymph node status of pN2 or greater, the RFS rates were 65% for the PSK group and 34% for the chemotherapy-only group—a significant difference with no P value offered. The authors concluded that PSK adjuvant immunotherapy may be effective in MHC class I–negative patients with advanced lymph node metastasis of pN2 or greater.

While the mechanism of action for PSK in general and in colorectal cancer specifically is not clearly defined, the potential activity of PSK as an immunomodulatory adjunct to chemoradiation therapy in rectal cancer has been studied.[15] Thirty patients with stage II or III rectal cancer who were treated with S-1 and external-beam radiation therapy were randomly assigned to receive either the standard regimen or standard regimen plus PSK. A number of cellular and humoral immune parameters were tested. An increase in peripheral blood NK cells after therapy was observed in the PSK-treated group compared with the control group. Immunosuppressive acidic protein (IAP) levels have been reported to be elevated in cancer patients and correlated to cancer progression and prognosis. In the study, a more-marked decrease in IAP level was observed in patients treated with PSK than in those treated in the control group. In addition, cytotoxic T cells increased in the peritumoral mucosa and normal mucosa within the radiation field in the PSK-treated group. The authors of the study concluded that PSK treatment may promote local tissue immunity within the radiation field.

One review included preclinical studies conducted in lung cancer models using either PSK or other T. versicolor preparations.[16] Data from the 15 preclinical studies supported the anticancer effects of PSK by way of immunomodulation and potentiation of immune surveillance. In animal models, direct antitumor effects resulted in reduced tumor growth and metastases.

https://www.ncbi.nlm.nih.gov/books/NBK401261/

https://pubmed.ncbi.nlm.nih.gov/10702635/

 

 

Agaricus blazei

One important group within the immunocompetent leucocytes is the phagocytic cells, which include monocytes, monocyte-derived macrophages and polymorphonuclear neutrophils (PMN). They all bind, internalize and eradicate invading micro-organisms. These cells use their own primitive, non-specific recognition systems, which allow them to bind a multitude of microbial products, and elicit the so-called innate immune responses. In effect, the cells act as the first line of defence against infection. Natural killer (NK) cells and NK T cells also belong to the innate immune system and are, together with macrophages, in the first line of defence against tumours [4]. AbM is rich in biological response modulators, such as proteoglucans [5, 6] and b-glucans [7], which are potent stimulators of macrophages [8–10], PMN [11] and NK cells [12]. These substances are main structural components in the cell wall of yeast and fungi, but are also found in some plants such as barley. The effects are mediated via the lectin-binding site for b-glucan in complement receptor 3 (CR3) (CD11b ⁄ 18) [13–15], Toll-like receptor 2 (TLR2) [16] and dectin-1 [17]. Stimulation of these receptors results in the release of proinflammatory cytokines [18], nitric oxide and hydrogen peroxide [19, 20], lysosomal enzyme [21] and activation of arachidonic acid metabolism [22]. b-1,3-glucans also induce activation of another component of innate immunity, the alternative complement pathway [23]. As to AbM, several studies have investigated its stimulating profile in more detail. In activated macrophages, AbM has been shown to induce secretion of nitric oxide, the proinflammatory cytokines TNF-a and IL-8 [24], as well as the Th1 cytokine IL-12 [25]. A dose-dependent in vitro production of proinflammatory cytokines, including IL-1b and IL-6, has been confirmed in AbM-stimulated human monocytes and umbilical vein endothelial cells [26]; however, neither anti-inflammatory cytokine IL-10 nor IL-12 synthesis was observed in this study. On the other hand, an AbM proteoglycan that stimulated mouse dendritic cell maturation also increased IL-12 production [27]. Gene expression microarray analysis of promonocytic THP-1 cells revealed upregulation of genes for chemokine ligands CXCL1-3, TLR2, dectin-1 and the IL-23a subunit of the IL-12 family, in addition to genes for IL-1b, IL-8 and cyclo-oxygenase 2 (Prostaglandin-endoperoxide synthase 2), whereas the IL-10 and IL-12 genes were not upregulated [28]. As to in vivo effects, increased levels of cytokines MIP2 (murine equivalent of IL-8) and TNF-a have been observed in mice receiving AbM extract [29]. Examination of immunomodulatory effects of AbM in mice have revealed increased numbers of antibody-producing spleen cells [30], elevated serum IgG levels and T-cell number in spleen, as well as increased phagocytic capacity of PMN [31], which is induced by proinflammatory cytokines [32, 33]. On the other hand, IL-12- and IFN-cmediated NK cell activity by AbM has been documented both in vitro and in vivo [34]. The latter contrasts somehow with the previous finding of AbM-suppressed PBMC production of IFN-c, IL-2 and IL-4 [35]. Interestingly, also the gene for regulator of G-protein signalling (RGS1), which is important for the G-protein-linked rhodopsin-like chemoattractant receptors for IL-8 (CD128), complement anaphylatoxin C5a (CD88), bacterial formyl peptide (fMLF) and leukotriene B4 [36], was selectively upregulated by an AbM extract in the promonocytic THP-1 cells [28]. The receptor for interferons a and b (IFNAR1) was also upregulated in peripheral blood. Among the extracts, only one (A) had a significant protective effect as demonstrated by reduced bacteraemia and increased survival rate (P < 0.05) (Fig. 2). None of the control mice given PBS survived day 5, whereas 38% (3 ⁄ 8) of mice given extract A lived on day 6, and another two mice were killed due to illness on day 8 (Fig. 2B). One of the other extracts (D) showed a similar, but statistically non-significant, trend for both bacteraemia and survival. The most active product was an aqueous, highly purified extract, which contained 82% AbM, 15% Hericium erinaceum (Yamabushitake) and 3% Grifola frondosa (Maitake). Although the latter species is another known medicinal mushroom with immunomodulating effects, recent examination of NF-jB activation via TLR2 has revealed that the main stimulatory effect of the extract A on monocytes is contained in the AbM fraction [49]. Note that, as extracts B–E were pure AbM extracts according to the producers, one must compare 80% of extract A with 100% of each of the others. The difference in anti-infectivity between the AbM extracts may be due to the presence of additional biological components, such as extracts from other mushrooms, with possible synergistic effects [50]. Cultivation methods may, however, be another reason for biological differences. It is well known that mushrooms and moulds can change their phenotype (e.g. colour and sporulation), and their production of secondary metabolites [such as mycotoxins and microbial volatile organic compounds (MVOC)] depending on growth medium and conditions. For example, mycotoxins are produced under suboptimal growth conditions of moulds [50], and MVOC durin. Other Japanese groups have shown that fat-soluble ergosterol, as well as another antiangiogenetic substance from AbM, reduced tumour growth and metastasis in sarcoma- and lung carcinoma-bearing mice [58, 76]. One interesting study showed myeloma tumour suppression by intake of an AbM extract in myeloma bearing mice, and that the tumour vanished in mice given a combination of AbM extract and marine phospholipids [77]. These results suggest improved uptake through the gut mucosa of active substances in AbM, such as b-glucans, by encapsulation in phospholipids. Recently, a toxicity study in rats that ingested AbM extract over 2 years, found no carcinogenicity or other adverse health effects of AbM [78]. Rather, the study demonstrated significantly lower mortality among the male rats on AbM treatment, which was suggested to be due to a lower tumour incidence in this group. Table 2 shows a summary of reported AbM-induced changes in vitro and in vivo related to cancer. Microarray examination of periph.

https://sci-hub.se/10.1111/j.1365-3083.2008.02156.x

https://pubmed.ncbi.nlm.nih.gov/18782264/

Mice treated with A. blazei aqueous extract or fraction C, that shows antioxidant activity, displayed lower parasitaemia, increased survival, reduced weight loss and protection against the development of CM. The administration of A. blazei resulted in reduced levels of TNF, IL-1β and IL-6 production when compared to untreated P. berghei-infected mice. Agaricus blazei (aqueous extract or fraction C) treated infected mice displayed reduction of brain lesions. Although chloroquine treatment reduced parasitaemia, there was increased production of proinflammatory cytokines and damage in the CNS not observed with A. blazei treatment. Moreover, the in vitro pretreatment of infected erythrocytes followed by in vivo infection resulted in lower parasitaemia, increased survival, and little evidence of clinical signs of disease.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4531523/

Agaricus blazei, or some of its fractions, stimulates the immune response, including TNF and IL-8 production by macrophages [30] and stimulates IL-1β and IL-6 in human monocytes and endothelial cells [6]. Studies in healthy individuals fed A. blazei demonstrated significant reduction in cytokine levels including TNF, IL-1β, IL-2, IL-6, and IL-17 in human blood [31]. Blood cells obtained from patients, with inflammatory bowel disease treated with A. blazei had reduced levels of IL-1β, IL-6, IL-8, MCP-1 and G-CSF when stimulated with LPS in vitro [14]. Johnson et al. [31] observed in human healthy volunteers, after oral A. blazei, elevated levels of, IFN-γ, IL-2, IL-4, IL-10, IL-12 and IL-17 in blood/serum.

An aqueous extract of A. blazei reduced the parasite infectivity, load and viability [18] in murine macrophages infected with different species of Leishmania. Additionally, Leishmania amazonensis-infected mice treated with aqueous extract of A. blazei displayed a reduction of the lesions size, a reduction of parasite load in the spleen and lymph nodes, an elevation of IFN-γ and decreased IL-4 and IL-10 in the spleen and in lymphoid nodules, respectively [19]. A. blazei has also been used in the prophylaxis and treatment of Leishmania chagasi infection [20].

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2249742/

We found that an AbM-based extract (AndoSan, http://www.immunopharma.net/), also containing the medicinal Basidiomycetes mushrooms Hericium erinaceum (15%) and Grifola frondosa (3%), given orally increased survival from bacterial sepsis in mice inoculated i.p. a day afterward with pneumococci (Figure 2) [14] or fecal bacteria [15]. The mixed mushroom extract also protected against IgE-mediated allergy in a mouse model when given p.o. either before or after ovalbumin s.c. sensitization of the animals (Figure 3) [16]. In supernatants of cultured spleen cells from the sacrificed AbM-treated mice, there was an increased T-helper cell 1 response relative to the allergy-inducing Th2 response. The observation fits with the reduced specific serum IgE levels in these animals and shows that also adaptive immunity is engaged by the mushroom. Since the original Th1/Th2 dichotomy [17] says that the antitumor and anti-infection Th1 response is inversely related to the Th2 response, the spleen cell finding above also helps explain the concomitant antiallergic, antitumor, and antiinfection effects of AbM. Moreover, this agrees with the very interesting report finding that AbM extract ameliorated a skewed Th1/Th2 balance both in asthma-induced and in tumor-bearing mice [18]. It is previously known that patients with advanced cancer have malfunctional Th1 cells and a Th2-skewed immune system [19]. However, it is not known whether AbM contributed to rectify a possibly induced Th1/Th2 imbalance in the above-mentioned sepsis models in mice [1415]. The reason for the forceful and swift engagement of innate immunity when encountering an edible and harmless mushroom, such as AbM, is its sharing of pathogen-associated molecular patterns (PAMP) with other highly poisonous species. Such mushrooms and fungi are usually a health threat due to the action of their toxins, for example, muscimol from Amanita muscaria and the vasoconstrictor ergotamine from Claviceps purpurea, or invasion in immunodeficient patients (e.g., Aspergillus fumigatus) or normal individuals (e.g., Stachybotrys chartarum).

PAMP, such as β-glucans, which form the main cell wall skeleton in mushrooms and fungi and are their signature molecule, are recognized immediately by so-called pattern-recognition receptors (PRR) [26], such as TLR2, dectin-1, and CR3. One can exploit this immune reaction by using an innocent mushroom such as Agaricus blazei M to enhance the body's protection against serious diseases. Although AbM induced NF-κB activation via stimulation of TLR2 on cells in vitro [27], the AbM-based mushroom extract AndoSan had anti-inflammatory effect in inflammatory bowel disease patients in vivo [24]. In addition to monocytes, granulocytes, and DC, also NK cells bear such PRR [9] and are stimulated by AbM in vitro, for example, to induce increase in cytokine production, expression of the adhesion molecule CD11b on monocytes and granulocytes, and ROS and NO production in the latter cells [28]. An AbM extract also gave a dose-dependent increase in release of proinflammatory cytokines from HUVEC in culture [29]. Since human skin endothelial cells can express all 10 TLR genes [30], TLR-binding of AbM was probably one mechanism behind the above finding in HUVEC. This demonstrates that AbM also affects EC, which are important parttakers in the innate immune response. In a human study, AbM has been shown to increase NK cell activity in cancer patients [31]. Another important group of receptors on macrophages and other innate immune cells are cytosolic NOD (nucleotide-binding and oligomerization domain-) like receptors (NLR). These receptors detect conserved bacterial molecular signatures within the host cells, similar to recognition of β-glucans via surface receptors or so-called “danger signals,” alerting the immune system of hazardous environments [32], and “cross-talk” with TLR on DC [33].

It is also known that AbM can activate the alternative pathway of complement [34], giving binding of the CR3 ligand, iC3b, to particulate AbM and thus contribute to the AbM engagement of the phagocytic CR3. Moreover, the formation of complement activation split products and chemotaxins—C3a and C5a—when also the terminal complement pathway is activated will lead to their binding to C3a and C5a (CD88) receptors, respectively, and chemotaxis of the immune cells towards a C3a and C5a gradient and hence towards the AbM source.

Animal studies have shown that although β-glucans can enter the proximal small intestine, the specific β-1,3-glucan backbone is indigestible [35]. In vivo, AbM most probably engage Peyer's patches in the intestines both via their abundant β-glucans and other immunomodulating substances such as proteoglycans, which may be transported to macrophages and DC after being taken up from the gastrointestinal lumen by M cells. Another possibility is direct uptake of or stimulation by such substances of DC via their processes into the gastrointestinal lumen or by intestinal macrophages that may fragment the glucans for transport to the bone marrow and reticuloendothelial system for further release and stimulation of other immune cells [35]. Previously, AbM has in fact been shown to stimulate endothelial cells in vitro [29]. Sorimachi et al. [36] demonstrated that ethanol (50%) precipitation of water extracts of the AbM fruiting body or its supernatant, rather than of the mycelium, induced TNFα, IL-8, and NO secretion in bone-marrow-derived rat macrophages. Since the complex β-1,3-/1,6-glucansfrom AbM induced different cellular activities than the mainly glucose-containing β-1,4-glucan from its nonmedicinal cousin A. bisporus (the champignon) [37], both differences in content, structure, and branching of their β-glucans must be highly important for the mushrooms' biological properties. Especially, β-1,3-glucans seem to be essential for immunological activity [37]. Interestingly, also a low-molecular-weight polysaccharide isolated from AbM has been reported to suppress tumor growth and angiogenesis in vivo [38]. Pyroglutamate is found to be another such small substance [39]. Since also the anti-allergic effect of AbM extract seems to be owing to low-molecular-weight substances (Hetland G, unpublished results) in a mouse model, other smaller and simpler substances than β-glucans in AbM could very well be as important for the mushroom's biological effects.

Most probably the mushroom extract also affects the intestinal flora, which comprises 10 times more bacteria than cells in our entire body. Bacteria in the gut are known to produce essential vitamins and so forth, for example, bacteria that can ferment soy beans and produce K2 vitamin that is important for calcium metabolism [49]. It is probable that also intestinal bacteria either produce metabolites during digestion of AbM extract or produce analytes after themselves being stimulated by AbM. Such molecules may be biologically active after uptake from the gut and may affect the host.

The PAMP or NOD signature-bearing pathogens are annihilated immediately by the patrolling innate immune cells. Some of these like monocytes and DC—directors of the immune system orchestrating the linkage of innate and adaptive immunity—process antigens from the mushrooms/fungi and present them together with self HLA II molecules for CD4 T helper cells, thus engaging adaptive immunity against the intruders.

Recently, we examined AbM-stimulated MDCC and found a significantly increased production of various cytokines [50]. This agrees with microarray studies in vitro of promonocytic THP-1 cells incubated with the AbM extract, which showed upregulation for most of genes associated with immune function, including the gene for IL-23α subunit—a Th1 cytokine in the IL-12 family [10]. In fact, the AbM extract induced even higher production of cytokines TNFα, IL-1β and MCP-1, and G-CSF than did cells stimulated with an appropriate concentration of 0.5 μg/mL of LPS [50]. Hence, MDDC may in some respect be even more primed to defense against mushrooms/fungi than against Gram-negative bacteria. At our Department of Cellular Therapy, we construct autologous DC vaccines for clinical trials against various human cancers, by electroporating isolated cancer mRNA into dendritic cells harvested from the same patients. Benko et al. [33] have proposed the emerging of ligands of the innate recognition systems as new adjuvant candidates for vaccine design. In line with this school of thought, we are currently examining whether the above AbM extract may be used as immune adjuvant in such anti-cancer DC vaccines, similar to studies on DNA vaccines in the mouse against hepatitis B virus and mouth-and-foot diseases [5152]. In these vaccines, AbM was found to increase levels of antigen-specific antibodies as well as to promote proliferation of T cells, illustrating engagement of adaptive immunity. Figure 5 shows a cartoon of the proposed role of AbM in immune system modulation and the resulting disease control.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3168293/

https://pubmed.ncbi.nlm.nih.gov/19779112/

We also found that the highest dose of AbM increased interleukin-2 (IL-2) levels in splenocytes and that a medium dose increased interferon-γ. The levels of interleukin-4 (IL-4) were reduced or unchanged. T-helper type 1 cytokine levels were increased. AbM increased the humoral immune response and also affected the cellular immune response. These results provide evidence that AbM can modulate innate and adaptive immunity.

https://pubmed.ncbi.nlm.nih.gov/24292582/

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7285126/

 

Organic Astragalus

Strengthen Immune System

AM extract modulates the immune response against various pathological agents, including viruses, bacteria, and other germs. It boosts the immunity level and prevents the risk of different medical conditions. Medical studies reported that AM stimulates and increases immune cells' production, including T-lymphocytes, Natural Killer cells, and improved the level of certain antibodies such as IgG, IgA, and IgM. Thus, it strengthens both innate and adaptive immunity and supports immune responses against illnesses. Studies reported that AM root extracts could be used as immunological adjuncts in certain conditions for better immune response.

Antioxidant Activity

AM roots extract shows potent antioxidant activity and supports vital organ functions. It neutralizes free radicals and removes toxins from the body. Thus, it preserves cellular integrity and decreases the risk of medical conditions. Moreover, AM extract regulates the expression of natural antioxidant enzymes in the body, including superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Also, it removes reactive oxygen species from mitochondria and protects against oxidative damage. Thus, it improves cellular integrity and promotes cellular energy production.

Antiinflammatory Activity:

AM inhibits the production and secretion of various pro-inflammatory cytokines and decreases the risk of inflammation. Studies showed that it suppresses the production of leukotrienes, tumor necrosis factor-alpha, and nitric oxide. Plus, it also inhibits the cyclooxygenase and other enzymatic pathways responsible for inflammatory response. Thus, it relieves inflammation and decreases the pathogenesis of various underlying inflammatory conditions such as diabetes.

 

 

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6268577/

https://pubmed.ncbi.nlm.nih.gov/22695229/

Regulation Effect of APS on Immunity

APS regulates the immune function by enhancing the immune organ index, promoting the proliferation of immune cells, stimulating the release of cytokines, and affecting the secretion of immunoglobulin (Ig) and conduction of immune signals.

Influences on Immune Organs

Many studies showed that APS can act on various immune organs to increase organ weight, improve organ index, and promote the development of partial visceral organs. In a study investigating the effect of APS on H22 tumor-bearing mice, it was found that fluorouracil significantly inhibited tumor growth; the thymus was obviously degenerated, and the spleen was obviously swollen. After treatment with APS, the thymus and spleen showed obvious improvement compared with those observed in the model group (Yu et al., 2018). Recent studies showed that after treatment with APS in Lewis transplantable lung cancer mice, the spleen and thymic immune organ indices in APS group were higher than those determined in normal saline group. This effect may improve the function of immune organs by reducing the expression of vascular endothelial growth factor and epidermal growth factor receptor, thereby inhibiting the tumor. (Zhao et al., 2019). Similarly, the APS+polysaccharopeptide herbal formulation significantly improved the immune function of mice with Lewis lung cancer and increased the spleen and thymus indices (Zhou et al., 2018). Besides, APS antagonized thymus atrophy in rats with incremental load training (Hu and Gao, 2011). When combined with probiotics, APS markedly improved the spleen, bursa of fabricius, and thymus index in Gushi chicken, especially in chicks. APS was able to promote the maturation of immune organs (Zhao et al., 2012), and enhanced the immune function by increasing the weight of immune organs.

Influences on Immune Cells

The influences of APS on immune cells mainly include an increase the proliferation and differentiation of B lymphocytes and T lymphocytes, regulation of the balance in the T lymphocyte subgroup, and regulation of natural killer cells and macrophages. Dendritic cells (DCs) are key in activating the immune response. Studies have shown that APS facilitates the growth and maturation of DCs and their antigen-presenting capacity, as well as decreases the endocytosis activity of DCs. Moreover, APS can markedly promote the development and maturation of bone marrow-derived DCs (Shao et al., 2006). APS activates T cells by inducing the differentiation of DCs (Liu et al., 2011). A recent study found that APS is the first effective regulator of tumor M1/M2 macrophage polarization and an effective activator of DC maturation (Bamodu et al., 2019). APS activates the proliferation of B cells and macrophages and increases the production of cytokines. Furthermore, it can activate B lymphocytes through antigen receptors on B cell membranes rather than through the toll-like receptor 4 (TLR4) (Shao et al., 2004). An in vivo study of mice revealed that APS is able to increase complement (C3) deposition and the number of macrophages (Wang et al., 1989). In addition, some studies have shown that APS promotes humoral immune response by regulating the functional activity of natural killer and natural killer T cells (Xie et al., 2013).

Influences on Immune Molecules

Influences on Cytokines

APS exerts different effects on cytokines under different conditions. Under normal physiological conditions, it can promote cytokine production and enhance immunity. However, following an increase in cytokines as a result of an inflammatory response, APS can reduce inflammatory response factors and protect cells or the body (Table 1). Studies have shown that APS affects the secretion and production of cytokines; it is able to promote splenocytes to produce interleukin 2 (IL2), induce interferon (IFN), and promote the secretion of IL3, IL4, and IL6 (Deng et al., 2012). Recent studies have found that adenosine monophosphate (AMP), as a potential immunomodulator, improves the serum levels of IL11, tumor necrosis factor-α (TNF-α), and IFNγ, and enhances spermatogenesis and sperm quality in mice (Qiu and Cheng, 2019). APS promotes immune regulation by inducing the production of IL in human body. Following the application of APS, the production of IL10, IL12, and IL2 was found to be dose-dependent compared with the negative control (Yin et al., 2012). Besides, APS can upregulate the expression of TNF-α, lysozyme C, and IL1β in the spleen, gill, and kidney of the carp, which was also found to be dose-dependent (Yuan et al., 2008). APS can also improve the expression of IL2 and IL10 in the jejunum of cyclophosphamide broilers (Li S. et al., 2019). It has been shown that the levels of TNF-α, IL6, and IL1β were significantly increased in mice with colitis induced by saline in in vivo studies. However, the production of these inflammatory cytokines was markedly decreased in the groups treated with APS and dexamethasone treatment group (Lv et al., 2017). Another study of experimental colitis in rats treated with APS found that A high dose of APS (200 mg/kg) or dexamethasone could markedly downregulate the expression of IL1β and TNF-α and upregulate the protein expression of nuclear factors of activated T cells 4 mRNA. Although a low dose of APS (100 mg/kg) markedly downregulated IL1β, it had no significant impact on the expression of TNF-α protein (Yang et al., 2014). In vitro studies have investigated the effect of APS on the inflammatory reaction in lipopolysaccharide-infected (LPS-infected) Caco-2 cells. The results showed that APS could significantly downregulate the expression of TNF-α, IL1β, and IL8 in Caco-2 cells infected with LPS (Wang et al., 2013). APS also restrained the expression of TNF-α and IL1β by inhibiting the activation of nuclear factor kappa-B (NF-κB) in THP-1 macrophages, which is induced by LPS (Liu et al., 2017). After stimulating RAW264.7 cells with LPS or APS for 24 h, the levels of TNF-α and IL6 were significantly increased. It was found through reverse transcription-polymerase chain reaction that the mRNA expression of IL6, TNF-α, and inducible nitric oxide synthase was strongly increased after treatment with APS (Li et al., 2018). A study of tumor-bearing mice found that APS could enhance the immune function by increasing the levels of cytokines, such as IL2, IL6, IL12, and TNF-α (Xiao et al., 2009). In a similar study, APS upregulated the expression of TNF-α, IL12, and IL2, whereas it decreased the levels of IL10 and downregulated the expression of multidrug resistance 1 mRNA and P-glycoprotein in H22 tumor-bearing mice (Tian et al., 2012Yang et al., 2013). Another study revealed that APS can inhibit tumorous growth in tumor-bearing mice. The mechanism of this process may involve increasing the levels of TNF-α and IFNγ, and reducing those of IL10 and transforming growth factor-β (TGF-β) (Sun et al., 2014). A new study suggested that APS significantly improves cancer symptom clusters in patients with metastatic disease and reduces the expression of major proinflammatory cytokines, including IL1β, IL6, IL12, and IFNγ (Huang et al., 2019).

nfluences on Ig

The major role of APS in Ig is to mediate immunity through IgA, IgG, and IgM. APS increased the expression of IL2, IL3, IL4, IFNγ, IgM, and IgG, whereas it decreased that of IgE (Lu Y. et al., 2016). Animal experiments affirmed that the production of IgM antibody in aged mice (36 and 60 weeks) was increased following the administration of APS (Kajimura et al., 1997). The level of antibody IgG in the serum of mice infected with Listeria can be significantly increased by APS injection (Xiang et al., 2007). Similarly, the levels of serum IgA, IgM, and IgG in juvenile broilers fed with APS were higher than those reported in broilers without exposure to APS. However, the excessive dose of APS (> 1 g/kg) did not further improve the serum levels of IgA, IgM, and IgG in juvenile broilers (Wu, 2018). The study showed that oral administration of APS promoted the immune function of Newcastle disease-vaccinated chickens and the formation of IgA cells, and increased the secretion of secretory IgA, thus improving mucosal immunity in the jejunum (Shan et al., 2019).

Influences on Immune Signal Transduction

Intracellular signal transducers and immune signaling pathways play a key role in the process of immune regulation (Figure 2). Studies have shown that APS can increase the TLR4/NF-κB and Ca2+-cAMP signaling pathways in RAW264.7 cells (murine mononuclear macrophage leukemia cells) (Wang Z. et al., 2017). The mouse macrophages is activated by triggering the TLR4-mediated signaling pathways, upregulating the expression of phosphorylated-p38 (p-p38), p-extracellular signal-regulated kinase (p-ERK), and p-JNK, inducing inhibitor of IκB-α degradation and NF-κB translocation, and ultimately enhances nitric oxide and TNF-α (Wei et al., 2016). Recent research has found that APS prevents coxsackievirus B3-induced myocardial injury and inflammation by modulating the TLR4/NF-κBp65 signaling pathway (Liu T. et al., 2019). APS nanoparticles can protect against sepsis-induced cardiac dysfunction by inhibiting the TLR4/NF-κB pathway (Xu et al., 2019). APS supplementation in diet may regulate the immune function of piglets by activating the TLR4-mediated MyD88-dependent signaling pathway (Wang K.L. et al., 2019). APS can also inhibit the expression of thrombin-induced intercellular cell adhesion molecule-1 through blocking the NF-κB signal transduction in rat bone marrow endothelial progenitor cells, and upregulating the expression of vascular endothelial growth factor and its receptor (Zhang et al., 2016). The phosphatidylinositol 3-kinase/protein kinase (PI3K/AKT) signaling pathway regulates cell metabolism, growth, migration, and proliferation. Notably, endothelial nitric oxide synthase is a key enzyme in the regulation of endothelial nitric oxide production, and is regulated by the PI3K/AKT signaling pathway. Following the treatment of H9c2 cells with different concentrations of APS, the result showed that APS protected these cells from LPS-induced inflammatory injury. This effect may also be attributed to the downregulation of miR-127, as well as the adjustment of the JNK, NF-κB, and PI3K/AKT signaling pathways (Ren et al., 2018). APS can promote the proliferation and differentiation of bone marrow mesenchymal stem cells (BMSCs) by upregulating BMP9, during which overexpression of BMP9 activates the PI3K/AKT and Wnt/β-catenin signaling pathways (Li Q. et al., 2019). APS can also partially suppress pulmonary artery remodeling through endothelial nitric oxide synthase/nitric oxide and the NF-κB signaling pathway to improve monocrotaline-induced pulmonary hypertension (Yuan L.B. et al., 2017). In addition, APS can inhibit the expression of adhesion molecules, which is induced by TNF-α through blocking NF-κB signal transduction in human umbilical vein endothelial cells and suppressing the production of reactive oxygen species (Zhu Y.P. et al., 2013). APS activated the downstream PI3K/AKT pathway by inducing neuregulin 1 (NRG1), which enhanced the phosphorylation of PI3K and AKT (Chang et al., 2018). APS can improve muscle atrophy through AKT/mammalian target of rapamycin (AKT/mTOR), autophagy signal transduction, and ubiquitin proteasome; sodium-dependent neutral amino acid transporter (SNAT2) may be one of the latent targets (Lu L. et al., 2016). AMP-activated protein kinase (AMPK) activated by AMP is the most important substrate of liver kinase B1 (LKB1); it is able to sensitively perceive the levels of cellular energy and maintain homeostasis. LKB1/AMPK participates in the regulation of cell growth and cell cycle by regulating mTOR. The mTOR is an important kinase regulating cell growth in eukaryotes. In similar studies, the mTOR inhibitor rapamycin markedly eliminated the protective effect of APS on adriamycin-induced cardiac injury; APS may play a protective role by regulating LKB1/AMPK to regulate mTOR (Cao et al., 2017). In pathological conditions, APS can downregulate the activity of mTOR and protect cells. In a study investigating the effect of APS on iron-overloaded mice, APS activated the p38/mitogen-activated protein kinase (p38/MAPK) signal transduction pathway in vitro (Ren et al., 2016). The inhibitory effect of APS on autophagy may be regulated by a mTOR-independent signaling pathway. Moreover, APS reduces hydrogen peroxide-induced apoptosis in myoblast C2C12 cells by inhibiting the Caspase-3 signaling pathway (Yin et al., 2015). By modulating the MEK/ERK pathway to up-regulate Kruppel like factor 2 expression, it can also reduce hydrogen peroxide-induced cell damage in human umbilical vein endothelial cells effectively (Li D.T. et al., 2019). Recent studies have also found that APS can effectively inhibit experimental autoimmune encephalomyelitis-mediated immune response by downregulating proinflammatory cytokines, upregulating the co-stimulatory molecule PD-1/PD-Ls signaling pathway, and inhibiting T cells (Sun et al., 2019).


https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7105737/

https://pubmed.ncbi.nlm.nih.gov/26916911/

Astragalus membranaceus can inhibit the oxidant stress by up-regulating the antioxidant factors. Aqueous extract of Astragali radix decreased the myocardial infarction size and improved the cardiac function in a myocardial ischemic rat model, which is related to the antioxidant effects via maintaining the activity of superoxide dismutase (SOD), decreasing the production of malondialdehyde (MDA) and free radical levels, and reducing cell apoptosis [27]. Besides, Astragalus injection can decrease the reactive oxygen species (ROS) and MDA levels in a rat model of cerebral ischemia through up-regulating the expression of nuclear factor erythroid 2-related factor 2 (Nrf-2), SOD, catalase and glutathione peroxidase (GSH-Px) [28].

2.2 Immunomodulatory effects by extracts of Astragalus membranaceus

Astragalus membranaceus was used to promote immune function and as a tonic to build the stamina [29]. The aqueous extract of Astragali radix also has significant immunological adjuvant activity when compounded with human vaccines [30].

Astragalus membranaceus could affect the innate immune response. The aqueous extract of Astragali radix induced the activation and migration, and monocyte maturation of peripheral blood mononuclear cells [31]. In the macrophage cell line RAW 264.7, aqueous extract of Astragali radix reversed the increasing iNOS expression and NO production by lipopolysaccharide (LPS), and reduced the suppression of macrophage cell proliferation by methotrexate [32]. Another research found that in the macrophage cell line ANA-1, aqueous extract of Astragali radix inhibited cytokine production via depressing p38 MAPK and NF-κB signaling pathways induced by advanced glycation endproduct [33].

Astragalus membranaceus could also affect the acquired immune response. The aqueous extract of Astragali radix exhibits mitogenic activities on T-cell depleted populations, augments the antibody response, and restores the lymphocyte blastogenic response in aging mice [34]. And aqueous extract of Astragali radix activated CD4+ and CD8+ T cells of humans without influencing proliferation [35,36]. In addition, the ethanol extracts of Astragali Radix selectively alter Th1/Th2 cytokine secretion patterns of CD4+ T cells through enhancing the IL-4 and IL-10 levels in Th2 cells and reducing the levels of IL-2 and IFN-γ in Th1 cells [37,38]. Similarly, hydroalcoholic extract of Astragalus gypsicolus also modulated the balance of Th1/Th2 cytokines in allergic mice model [39].

Beyond that, aqueous extracts of Astragali radix promote myelopoiesis in myelosuppressed mice by improving the hematopoietic microenviroment, including enhancing the survival of bone-marrow-derived mesenchymal stem cells (BMSCs), proliferation of colony-forming unit-fibroblast through upregulation of granulocyte-macrophage colony stimulating factor and of bcl-2 expression in BMSCs [40].

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5758356/

https://pubmed.ncbi.nlm.nih.gov/2149722/

 

 

 

DNA Protective Activity of Gac Fruit

e DNA protective eect of Gac fruit against TK6 human lymphoblast cells, induced by H2O2 and ultraviolet UVC was investigated. In the previous study, it was found that TK6 exposure to 50 µM H2O2 for 5 min, produced massive oxida-tive DNA damage, while TK6 exposure to UVC signicantly enhanced DNA migration, when the TK6 cells were treated with Gac peel extract using ethanol 95% and 50%, thus leading to a signicant decrease in DNA damage.27

Antimicrobial Activity of Gac Fruit

Gac pulp (esh) and seed pulp (aril) were examined con-cerning their potential activity against dierent strains of both Gram-positive and Gram-negative bacteria. e results of such work indicated that water and ethanolic extraction of Gac pulp displayed higher antibacterial eect on Gram- positive strains than Gram-negative strain bacteria. Accordingly, the highest antibacterial activity was observed in the ethanolic extract of Gac esh against both strains, Micrococcus luteus 745 (20 mm) and M. luteus 884 (18.5 mm) inhibition zone.28 Another study assessed the antimicrobial activities from various parts of the Gac fruit (i.e. peel, pulp, and aril) against six pathogenic  bacteria  (Escherichia coli,  Staphylococcus aureus,  Bacillus cereus, Pseudomonas aeruginosa, Salmonella typhimurium, and Klebsiella pneumonia). Such results demonstrated that the data of this study conrmed that ethanolic extraction of various parts from Gac fruit had antimicrobial activity against all path-ogenic strains, as mentioned previously.29

Antiulcer Activity of Gac Fruit

It has also been documented that 7 and 14 days of treatment with an ethanolic extract of Gac seed, signicantly accelerates the healing of acetic acid via enhancing and upregulation of the angiogenesis, and increases the expression of mRNA as well as the vascular endothelial growth factor (VEGF), tested by real-time PCR and western blot.29

Anti-inammatory Activity of Gac Fruit

Surprisingly, Gac seed is found to be a rich source of saponins, which are known for their health benets.30

Two triterpenoid saponin compounds (Gypsogenin glycoside and Quillaic acid glycoside) were isolated and examined regarding their anti-in-ammatory activity. For example, compound 2 (Quillaic acid glycoside) which is a saponin compound that isolated from Gac seed demonstrated anti- inammatory eects in RAW 264.7 cells via inhibiting the lipopolysaccharide-induced expression of nitric oxide and IL-6 via the NF-κB pathway.

The antioxidant activity of Gac fruit has been examined using diphenyl-picrylhydrazyl (DPPH) radical scavenging, ferric reducing antioxidant power (FRAP), and 2,2' -azino-bis(3-ethylbenzothiazoline-6-sulphonic acid (ABTS). ese assays are widely used and accepted in evaluating the antioxidant proper-ties of dierent compounds in foods, plants, fruits, and herbs. Moreover,  dierent fractions of the Gac fruit at dierent matu-rity stages have been assessed regarding their potential antiox-idant FRAP, by applying DPPH assays. e results showed that Gac aril at the fully ripened stage had the highest FRAP value at 531.17 µmol/g. In the same study, the DPPH assay test.

https://www.researchgate.net/publication/330544853_Gac_Momordica_cochinchinensis_Spreng_fruit_and_its_potentiality_and_superiority_in-health_benefits