research article

Homeopathic Viscum Album at Potencies D3 and 200CH Presents Cytokine Modulatory Effect Produced by in vitro Culture of Mesenchymal Stem Cells

Authors: Ana Catarina Viana Valle1*, Hilana dos Santos Sena Brunel2, Patricia Furtado Malard2, Carla Lujan Villarroel2, Rosângela Vieira Andrade3

*Corresponding Author: Ana Catarina Viana Valle, IDIS/Lamasson, Doctor Izao Soares Institute, Postal Code: 14015-050, Sao Paulo, Brazil

1IDIS/Lamasson, Doctor Izao Soares Institute, Sao Paulo, Brazil

2Bio Innova Laboratory, R. 25 Sul, lote 30, Brasilia, Distrito Federal, Brazil

3Genomics Science and Biotechnology Post Graduation Program, Catholic University of Brasilia, SGAN 916, Asa Norte, Brazil

Received Date: 12 May 2022

Accepted Date: 23 May 2022

Published Date: 27 May 2022

Citation: Valle ACV, Brunel HSS, Malard PF, Villarroel CL, Andrade RV (2022) Homeopathic Viscum Album at Potencies D3 and 200CH Presents Cytokine Modulatory Effect Produced by in vitro Culture of Mesenchymal Stem Cells. Curr Res Cmpl Alt Med 6: 155. DOI: https://doi.org/10.29011/2577-2201.100055

Abstract

Viscum Album (VA) has been widely used as adjuvant therapy in cancer. Its anticancer activity has been attributed to a combination of cytotoxic, immunomodulatory, and anti-inflammatory properties, induction of apoptosis, and inhibition of angiogenesis. This study aimed to evaluate the in vitro immunologic effects of homeopathic VA on mesenchymal stem cells. Two concentrations of VA in the potencies D3 and 200CH were used: 20 and 30 mL/mL. The levels of the pro-inflammatory cytokines TNF-α, IFN-γ, IL-6, and IL-1b produced by the cells were measured after a 48h-incubation period with VA. The concentrations of VA D3 and VA200CH used did not induce changes in TNF-α and IFN-γ responses but decreased the values of IL-6 and IL-1b. This result demonstrates that VAD3 and VA200CH may play an important role as anti-inflammatory and immunomodulatory medicines.

Introduction

Viscum Album (VA) is a hemiparasitic plant of the Loranthaceae family used to treat many diseases in traditional and folk medicines. Anticancer, immunomodulatory, antidiabetic, neuropharmacological, antibacterial, hepatoprotective, cardiac, and antifungal properties comprise the most studied properties of VA extracts [1].

Extracts produced from VA are used for treating cancer within concepts of complementary medicine, especially anthroposophical medicine. Several studies have reported the modulatory effect on the natural

immune system of VA extracts. These findings may explain the therapeutic effects observed in some patients with cancer, such as prolonged overall survival, improved quality of life, and reduction of adverse events associated with conventional anticancer treatments [2,3]. Studies have also found that homeopathic VA can reduce the viability of cancer cells in vitro [4,5].

According to Gardin [45], VA can enhance the immune response and restore cellular and humoral immunosuppressed systems. Its bi-directional activity in cancer treatment has been widely documented. Initially, it affects the quality of life of cancer patients by showing a supportive effect on cancer-related fatigue, insomnia, exhaustion, nausea, vomiting, appetite, depression, anxiety, pain, physical functioning, and side effects of traditional treatments [6,7]. However, the molecular and cellular mechanisms behind the beneficial effects of VA encompass the antitumor activity by cytotoxic properties, induction of apoptosis [8,9], and inhibition of angiogenesis [10]. Findings indicate that adjuvant treatment of cancer patients with VA can be associated with better survival [11,12].

Its extracts contain various substances, such as lectins, viscotoxins, alkaloids, lignans, sterols, fatty acids, flavonoids, terpenoids, phenolic acids, and phenylpropanoids. Lectins are the major biologically active components of the VA extracts. They are known to induce apoptosis in lymphocytes and tumor cells, and at toxic concentrations, they can also stimulate the release of cytokines [13].

Cytokines are small and soluble proteins or glycoproteins that mediate cell-to-cell communication. They are secreted by various immune and nonimmune cells and are responsible for growth, differentiation, and inflammatory or anti-inflammatory signals to diverse cell types [14,15]. Cytokine-mediated effects dominate the fields of inflammation, immunology, atherosclerosis, and cancer [16]. Several cytokines limit tumor cell growth by a direct antiproliferative or pro-apoptotic activity or indirectly by stimulating the cytotoxic activity of immune cells against tumor cells [14].

Cytokines like IFN-γ or TNF play a critical role in creating an immunogenic microenvironment and, consequently, fighting against metastatic cancer. IFN-γ is produced predominantly by NK cells and natural killer T (NKT) cells as an innate immune response. TNF-α is a pleiotropic cytokine involved in multiple homeostatic and pathological mechanisms. It is mainly produced by macrophages, T lymphocytes, and NK cells. It is a multifunctional cytokine involved in apoptosis, cell survival, inflammation, and immunity [17].

IL-6 is also a pleiotropic cytokine, and the increase in its expression has been linked to the pathogenesis of several disorders such as chronic inflammatory diseases, autoimmune diseases, and tumor development. This feature could be explained by its influence on the cellular metabolism and several immune and physiological processes, such as antigen-specific immune responses, inflammation, hematopoiesis, apoptosis, and differentiation [18,19,20]. Excessive IL-1β production contributes to autoimmune [21] and causes autoinflammatory diseases [22]. In addition, in the case of chronic

inflammation, sustained IL1β may promote both tumor induction and, later on, also tumor propagation by different mechanisms [23]. This cytokine has also contributed to the severity of inflammatory diseases [24,25].

Clinical trials have increasingly assessed the safety and efficacy of cytokine-based drugs in recent years due to a renewed interest in the antitumor properties of cytokines. Consequently, cytokines have been studied as single agents and combined with other immunomodulatory drugs [14].

Therefore, cytokines are being measured and monitored more routinely in clinical laboratories due to their enhanced use as therapeutics, targets for modulation of undesirable inflammation, biomarkers of inflammation, and diagnostic markers [15].

The objective of this study was to assess the in vitro immunologic effects of homeopathic VA on mesenchymal stem cells by dosing IL-6, IL-1b, TNF-a, and IFN-g.

Methods

Cell culture

Mesenchymal stem cells (MSC) derived from adipose tissue of two donors were obtained from a commercial cell bank. Cells were cultured with Dulbecco’s Modified Eagle Medium (DMEM) added with 10% fetal bovine serum and 0.02% amikacin (all from the Sigma-Aldrich® brand) following the Data Sheet methodology.

Preparation of VAD3 and VA200CH

The Mother Tincture was the starting point for preparing the tested substance (VAD3 and VA200CH). As described in the Brazilian Homeopathic Pharmacopoeia, the Hahnemannian Decimal Method was used. Using a sterile isotonic solution, 1 part of the active ingredient was mixed with 9 parts of the inert ingredient and succussed 100 times, yielding VAD1 (1×10−1). Then, 1 part of VAD1 was used with 9 parts of the inert ingredient and succussed 100 times, yielding VAD2 (1×10−2). The successive dilution continued till VAD3 and VA200CH were obtained. These products were then bottled in 1.1 mL ampoules.

Cytokines analyses

Cells were plated in 6-well plates (1x105 cells per well) with culture medium (DMEM with 10% FBS) and incubated at 37oC and 5% CO2 for 24 hours. After this period, the treatment of cells was initiated with the culture medium containing different concentrations of homeopathic VA (VAD3 and VA200CH) at 20 and 30 mL/mL of culture medium, followed by incubation for 48 hours in the conditions previously described. Control wells were maintained containing cells and culture medium with no addition of VA.

After 48 hours of culture, the medium was collected and frozen to further quantify cytokines IL-6, IL-1b, TNF-α, and IFN-γ. Analyzes were performed in triplicate wells, and the dosage was calculated automatically based on the kit standards. The cytokines dosages (pg/mL) were performed by the sandwich ELISA method with commercial kits (Prepotech mini-ABTS), following the datasheet provided by the manufacturer. The standards and samples were distributed in 96-well plates. Reading was performed in a spectrophotometer at a 405 nm wavelength and correction at 650 nm.

Statistical Analysis

Data are presented as means and standard deviations. The one-way analysis of variance was used to compare each group with the control. Results were considered significant at P < 0.05. All statistical analyses were performed GraphPad Prism version 7.0.4 for Windows, GraphPad Software, San Diego, California USA, “www.graphpad.com”.

Results

In our study, mesenchymal stem cells from two donors were cultured in the presence of different concentrations of homeopathic VA. The cytokine dosages in the supernatant were measured, and the mean and standard deviation was calculated as presented in Table 1.

The present study results demonstrate that the VA concentrations of 20 and 30 mL/mL were not sufficient to induce changes in TNF-α and IFN-γ responses in mesenchymal stem cells. However, it is possible to observe a decreasing trend of these cytokines with these treatments (Figure 1).

The pro-inflammatory cytokines IL-6 and IL-1b decreased (P<0.05) in the supernatant when VA was added to the culture medium (Figure 2), meaning that the cells produced less IL-6 and IL-1b.

Discussion

Our study demonstrates that different concentrations of VAD3 and VA200CH (20 and 30 mL/mL) were not sufficient to induce significant changes in TNF-α and IFN-γ but decreased the pro-inflammatory cytokines IL-6 and IL-1b produced by mesenchymal stem cells.

Cytokines, including interleukins, interferons, and tumor necrosis factors, have various pro- and anti-inflammatory effects in the body through several biochemical pathways and interactions [26]. TNF-α is a pro-inflammatory cytokine critical for cell communication during host defense, inflammation, and organogenesis. TNF-α plays a central role at the beginning of the inflammatory reactions of the innate immune system. This cytokine can cause chronic inflammation if not regulated and septic shock when high acute concentrations are generated. In a study with mice in which TNF-α mRNA stability was increased, they developed arthritis, cachexia, and inflammatory bowel disease [27]. These mouse experiments relate directly to recent studies in chronic human diseases since deregulated local production of TNF-α is characteristic of diseases such as rheumatoid arthritis [28] and Crohn’s disease [29]. Treatment with TNF-α antagonists is effective in most patients. In our study, the non-alteration of TNF-α observed in cells treated with VAD3 and VA200CH suggests that these medicines may bring benefits with the potential to regulate the immune system, avoiding the high release of TNF-α. Many chronic inflammatory diseases are associated with an increased risk of cancer, as these cell mutations arise at sites of chronic inflammation and chemical mediators of inflammation are found in many cancers [30, 31] and one of the major chemical mediators implicated in inflammation-associated cancers is TNF-α. There is substantial evidence that TNF-α is involved in the promotion and progression of cancers in humans since, when produced in the tumor microenvironment, TNF-α can act as an endogenous tumor promoter [32]. Orosz [33] showed increased lung metastases of an experimental fibrosarcoma after pre-treatment of animals with TNF-α. Alternatively, lung metastases may be reduced by adding an anti-TNF-α antibody, neutralizing endogenous TNF-α levels. Therefore, medicines that regulate the release of this factor in vitro, such as VAD3 and VA200CH, may be important as aids in treating cancer.

Another cytokine studied in this study, IFN-γ, promotes both protective immune responses and immune-pathological processes. In response to inflammatory mediators, including IFN-γ, tumor cells initiate an immune regulation program [34,35]. Under the influence of chronic inflammation associated with cancer, IFN-γ can thus induce the expression of immune regulatory molecules in tumor cells. Immunogenic cancers such as melanoma, prostate cancer, breast cancer, leukemia, and HPV-infected high-grade cervical intraepithelial neoplasia can stimulate IFN-γ production upon involvement with immune cells [36]. In addition, immunogenic cell death is widely observed in solid tumors and induces the STING molecule (IFN/TMEM173 gene stimulator) that stimulates interferon production, including IFN-γ [37,38]. IFN-γ-mediated increase in various molecules such as CD38 in chronic lymphocytic leukemia and CD74 in melanoma can directly contribute to tumor cell proliferation and survival [39,40]. A medicine that regulates the production of IFN-γ, not allowing the production of high levels of this cytokine, may be important in treating cancers. By not stimulating the in vitro production of this molecule, VAD3 and VA200CH may become essential tools to be better studied in the complementary treatment against tumors.

The decrease of pro-inflammatory cytokines in tumor environments may be one of the critical mechanisms for cancer control. IL-6 is a pleiotropic cytokine and can influence several immune and physiological processes, such as inflammation, antigen-specific immune responses, hematopoiesis, apoptosis, differentiation, and cellular metabolism [18,19,20]. Increased IL-6 expression has been linked to the pathogenesis of several disorders, such as chronic inflammatory diseases, autoimmune diseases, and tumor development [41]. Also, the IL-6/IL-6R axis contributes to the progression of several diseases, and inhibition of this axis is highly effective against diseases such as rheumatoid arthritis (RA), Castleman disease, and cytokine release syndrome [42]. The low in vitro production of this cytokine by mesenchymal stem cells stimulated by the homeopathic medicines VAD3 and VA200CH may be an important factor since this medicine is used in the integrative treatment of some types of cancers [43, 44] and due its role as an immune system stimulator and ability to reestablish immune-suppressed cellular and humoral systems [45].

In conjunction with the low production of IL-6, our study also demonstrated the low IL-1β production, another cytokine related to inflammation and cancer cases. Excessive IL-1β production contributes to autoimmune [21] and causes autoinflammatory diseases [22]. Sustained IL-1β caused by chronic inflammation may promote both tumor induction, tumor propagation [23] and contrtibute to the severity of inflammatory diseases [24,25]. For example, elevated IL-1 signaling was demonstrated to cause neuronal cell death [46]. Of note, high IL-1β production has been observed in patients who have epilepsy [47], stroke [48], Alzheimer’s disease [49], and other neurological disorders [50].

There is a relationship between the cytokines presented in this study: the innate pro-inflammatory cytokines, including IL1, TNF-α, and IL-6, are crucial to resolving acute inflammations. However, high levels of innate cytokines, as apparent in chronic inflammation, may promote tumor development by driving sustained NF-κB activation [51] and mitogen-activated protein kinase (MAPK) activity [52]. The driving mechanism is the highlevel production of IL-1β by plasma cells which induces IL-6 in stromal cells [53]. IL-6, in turn, promotes the development of malign plasma cells. Increased levels of IL-1β in body fluids are correlated in experimental tumor models and cancer patients with bad prognosis, carcinogenesis, and experimental tumor models and cancer patients with bad prognosis, carcinogenesis, and invasiveness of the tumor [54]. The key mechanisms by which IL1β promotes tumor development are driving chronic non-resolved inflammation [55], endothelial cell activation [56], and chronic tumor angiogenesis [54] and

induction of immunosuppressive cells. Altogether, these mechanisms account for suppression of the adaptive immunity, tumor promotion and metastasis suppression of the adaptive immunity, tumor promotion, and metastasis [55]. In this context, it is vital to downregulate the production of these cytokines to prevent or control cancer diseases, as VAD3 and VA200CH possibly did in our in vitro study.

Conclusion

Since TNF-α, IFN-γ, IL-6, and IL-1b have pro-inflammatory effects, the non-alteration of the tumoral factor and the interferon proteins favors the role of VAD3 and VA200CH as an antiinflammatory medicine. Also, the decrease in IL-6 and IL-1b produced by cells treated with VA demonstrates that VAD3 and VA200CH may play an important role as anti-inflammatory and immunomodulatory medicines. Despite the interesting in vitro results observed, more in vivo studies should be performed to record the direct relationship of the effect of these medicines against the various types of cancers.

Figures


Figure 1: TNF-α and IFN-γ responses to VAD3 and VA200CH. Data presented by mean and standard deviations.


Figure 2: IL-6 and IL-1b responses to VAD3 and VA200CH. Data presented by mean and standard deviations. *P<0.05.

References

  1. Szurpnicka A, Kowalczuk A, Szterk A (2020) Biological activity of mistletoe: in vitro and in vivo studies and mechanisms of action. Arch Pharm Res 43: 593–629.
  2. Kienle GS, Kiene H (2010) Influence of Viscum album L (European mistletoe) extracts on quality of life in cancer patients: a systematic review of controlled clinical studies. Integr Cancer Ther 9:142-157.
  3. Evans M, Bryant S, Huntley AL, Feder G (2016) Cancer patients’ experiences of using mistletoe (Viscum album): a qualitative systematic review and synthesis. J Altern and Complement Med 22: 134-144.
  4. Valle Szurpnicka A, Zjawiony JK, Szterk A (2019) Therapeutic potential of mistletoe in CNS-related neurological disorders and the chemical composition of Viscum species. J Ethnopharmacol 231: 241-252.
  5. Valle ACV, Aguiar LR, Brunel HSS, Malard PF, Andrade RV (2020) Homoeopathic Viscum album extract inhibits the growth of osteosarcoma cells. Journal of Integrated Standardized Homoeopathy 3: 59-63.
  6. Oei SL, Thronicke A, Kröz M, von Trott P, Schad F, et al. (2020) Impact of Oncological Therapy and Viscum album L Treatment on CancerRelated Fatigue and Internal Coherence in Nonmetastasized Breast Cancer Patients. Integr Cancer Ther 19:1534735420917211.
  7. Valle ACV, Carvalho AC (2021) Homeopathic viscum album in the treatment of cutaneous melanoma in a dog (canis familiaris) –case Paripex - Indian Journal of Research 10.
  8. Mavrikou S, Tsekouras V, Karageorgou MA, Moschopoulou G, Kintzios S (2020) Anticancer and biochemical effects of Viscum album protein extracts on HeLa cells. Plant Cell Tiss Organ Cult 140: 369-378.
  9. Kwon Y-S, Chun S-Y, Kim M-K, Nan H-Y, Lee C-H, et al. (2021) Mistletoe Extract Targets the STAT3-FOXM1 Pathway to Induce Apoptosis and Inhibits Metastasis in Breast Cancer Cells. Am J Chi Med 49: 487-504.
  10. Elluru SR, Duong Van Huyen JP, Delignat S, Prost F, Heudes D, et (2009) Antiangiogenic properties of Viscum album extracts are associated with endotelial cytotoxicity. Anticancer Res 29:2945–2950.
  11. Reynel M, Villegas Y, Werthmann PG, Kiene H, Kienle GS (2020) Longterm survival of a patient with an inoperable thymic neuroendocrine tumor stage IIIa under sole treatment with Viscum album extract: A CARE compliant clinical case report. Medicine 99: e18990.
  12. Ostermann T, Appelbaum S, Poier D, Boehm K, Raak C, et al. (2020) A Systematic Review and Meta-Analysis on the Survival of Cancer Patients Treated with a Fermented Viscum album Extract (Iscador): An Update of Findings. Complement Med Res 27: 260-271.
  13. Stein GM, Pfüller U, Schietzel M, Büssing A (2000) Toxic proteins from European mistletoe (Viscum album): increase of intracellular IL-4 but decrease of IFN-gamma in apoptotic cells. Anticancer Res 20:1673-1678.
  14. Berraondo P, Sanmamed MF, Ochoa MC, Etxeberria I, Aznar MA, et al. (2019) Cytokines in clinical cancer immunotherapy. Br J Cancer 120: 6-15.
  15. Knight V, Long T, Meng QH, Linden MA, Rhoads DD (2020) Variability in the Laboratory Measurement of Cytokines. Arch Pathol Lab Med 144:1230-1233.
  16. Dinarello CA. (2007) Historical insights into cytokines. Eur J Immunol 37(Suppl 1): S34-45.
  17. Atzeni F, Sarzi-Puttini P (2013) Tumor Necrosis Factor. Brenner’s Encyclopedia of Genetics (Second Edition), Academic Press 229-231.
  18. Chalaris A, Garbers C, Rabe B, Rose-John S, Scheller J (2011) The soluble interleukin 6 receptor: generation and role in inflammation and Eur J Cell Biol 90:484–494.
  19. Hunter CA, Jones SA (2015) IL-6 as a keystone cy- tokine in health and disease. Nat Immunol 16:448-457.
  20. Kishimoto T (2010) IL-6: from its discovery to clin- ical applications. Int Immunol 22: 347-352.
  21. Cordero MD, Alcocer-Gomez E, Ryffel B (2018) Gain of function mutation and inflammasome driven diseases in human and mouse J Autoimmun 91: 13-22.
  22. Dinarello CA (2014) An expanding role for interleukin-1 blockade from gout to cancer. Mol Med 20 (Suppl. 1): S43–S58.
  23. Garlanda C, Dinarello CA, Mantovani A (2013) The interleukin-1 family: Back to the future. Immunity 39: 1003-1018.
  24. Dinarello CA, van der Meer JW (2013) Treating inflammation by blocking interleukin-1 in humans. Semin Immunol 25: 469-484.
  25. Abbate A, Canada JM, Van Tassell BW, Wise CM, Dinarello CA (2014) Interleukin-1blockade in rheumatoid arthritis and heart failure: A missed opportunity?. Int J Cardiol 171: e125–e126.
  26. Monastero RN, Pentyala MS (2017) Cytokines as Biomarkers and Their Respective Clinical Cutoff Levels. Int J Inflam 2017:4309485.
  27. Kontoyiannis D, Pasparakis M, Pizarro TT, Cominelli F, Kollias, G (1999) Impaired on/off regulation of TNF biosynthesis in mice lacking TNF AU-rich elements: Implications for joint and gut-associated Immunity 10: 387-398.
  28. Feldmann M (2002) Development of anti-TNF therapy for rheumatoid Nat Rev Immunol 2: 364-371.
  29. Reich K, Nestle FO, Papp K, Ortonne J-P, Evans R, et al. (2005) Infliximab induction and maintenance therapy for moderate-to-severe psoriasis: A phase III, multicentre, double-blind trial. Lancet 366: 1367
  30. Balkwill F, Mantovani A (2001) Inflammation and cancer: Back to Lancet 357: 539-545.
  31. Balkwill F, Charles KA, Mantovani A (2005) Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 7: 211-217.
  32. Balkwill F (2002) Tumor necrosis factor or tumor promoting factor? Cytokine Growth Factor Rev 13: 135-141.
  33. Orosz P, Echtenacher B, Falk W, Ruschoff J, Weber D, et al. (1993) Enhancement of experimental metastasis by tumor necrosis factor. J Exp Med 177: 1391-1398.
  34. Matzinger P (2002) The danger model: a renewed sense of self. Science 296: 301-305.
  35. Gilroy D, De Maeyer R (2015) New insights into the resolution of Semin Immunol 27: 161-168.
  36. Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, et al. (2013) Up- regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med 5:
  37. White MJ, McArthur K, Metcalf D, Lane RM, Cambier JC, et al. (2014) Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 159: 1549-1562.
  38. Temizoz B, Kuroda E, Ohata K, Jounai N, Ozasa K, et al. (2015) TLR9 and STING agonists synergistically induce innate and adaptive type-II Eur J Immunol 45: 1159-1169.
  39. Burgler S, Gimeno A, Parente-Ribes A, Wang D, Os A, et al. (2015) Chronic lymphocytic leukemia cells express CD38 in response to Th1 cell- derived IFN-gamma by a T-bet-dependent mechanism. J Immunol 194: 827-835.
  40. Tanese K, Hashimoto Y, Berkova Z, Wang Y, Samaniego F, et al. (2015) Cell surface CD74-MIF interactions drive melanoma survival in response to interferon-gamma. J Invest Dermatol 135: 2775-2784.
  41. Heinrich PC, Behrmann I, Haan S, Hermanns HM, Muller-Newen, et (2003) Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J 374: 1-20.
  42. Kang S, Tanaka T, Narazaki M, Kishimoto T (2019) Targeting tnterleukin-6 signaling in clinic. Immunity 50:1007-1023.
  43. Valle ACV, Lima L, Andrade RV, Bonamin L, Brunel H, et al. (2020) Use of Viscum album in the Integrative Treatment of Cholangiocarcinoma in a Dog (Canis familiaris)- Case Report. Adv Complement Alt Med. 5: 000619.2020.
  44. Valle ACV, Carvalho AC (2021) Homeopathic Viscum Album on the Treatment of Scamous Cell Carcinoma Lesion in a Dog (Canis familiaris) – Case Report. Integr J Vet Biosc 5: 1-3.
  45. Gardin NE (2009) Immunological response to mistletoe (Viscum albumL.) in cancer patients: a four-case series. Phytother Res 23: 407-411.
  46. Murray KN, Parry-Jones AR, Allan SM (2015) Interleukin-1 and acute brain injury. Front Cell Neurosci 9:18.
  47. Tan CC, Zhang JG, Tan MS, Chen H, Meng DW, et al. (2015) NLRP1 inflammasome is activated in patients with medial temporal lobe epilepsy and contributes to neuronal pyroptosis in amygdala kindlinginduced rat model. J Neuroinflamm 12:18.
  48. Fann DY, Lim YA, Cheng YL, Lok KZ, Chunduri P, et al. (2018) Evidence that NF-kB and MAPK signaling promotes NLRP inflammasome activation in neurons following ischemic stroke. Mol Neurobiol 55: 1082-1096.
  49. Tan MS, Yu JT, Jiang T, Zhu XC, Tan L (2013) The NLRP3 inflammasome in Alzheimer’s disease. Mol Neurobiol 48: 875-882.
  50. Mamik MK, Power C (2017) Inflammasomes in neurological diseases: Emerging pathogenic and therapeutic concepts. Brain 140: 2273
  51. Huang Y, Chen R, Zhou J (2016) E2F1 and NF-κB: Key mediators of inflammation-associated cancers and potential therapeutic targets. Curr Cancer Drug Targets 16: 765-772.
  52. Kim EK, Choi EJ (2015) Compromised MAPK signaling in human diseases: An update. Arch Toxicol 89: 867-882.
  53. Costes V, Portier M, Lu ZY, Rossi JF, Bataille R, et al. (1998) Interleukin-1 in multiple myeloma: Producer cells and their role in the control of IL-6 production. Br J Haematol 103: 1152-1160.
  54. Apte RN, Krelin Y, Song X, Dotan S, Recih E, et al. (2006) Effects of micro-environment- and malignant cell-derived interleukin-1 in carcinogenesis, tumour invasiveness and tumour-host interactions. Eur J Cancer 42: 751-759.
  55. Mantovani A, Barajon I, Garlanda C (2018) IL-1 and IL 1 regulatory pathways in cancer progression and therapy. Immunol Rev 281: 57
  56. Steel JL, Terhorst L, Collins KP,  DA,  Vodovotz Y, et al. (2018) Prospective analyses of cytokine mediation of sleep and survival in the context of advanced cancer. Psychosom Med 80: 483-491.
  57. Valle ACV, Brunel HSS, Malard PF, Cunha AC, Andrade RV (2019) Citotoxicity of Ultradiluted Viscum Album (1x10-400) in a Lineage of Human Osteosarcoma. Adv Complement Alt Med. 5: ACAM.000618.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Current Research in Complementary & Alternative Medicine

cara menggunakan pola slot mahjongrtp tertinggi hari inislot mahjong ways 1pola gacor olympus hari inipola gacor starlight princessslot mahjong ways 2strategi olympustrik mahjong ways 2trik olympus hari inirtp koi gatertp pragmatic tertinggicheat jackpot mahjongpg soft link gamertp jackpotelemen sakti mahjongpola maxwin mahjongslot olympus mudah mainrtp live starlightrumus slot mahjongmahjong scatter hitamslot pragmaticjam gacor mahjongpola gacor mahjongstrategi maxwin olympusslot jamin menangrtp slot gacorscatter wild banditopola slot mahjongstrategi maxwin sweet bonanzartp slot terakuratkejutan scatter hitamslot88 resmimaxwin olympuspola mahjong pgsoftretas mahjong waystrik mahjongtrik slot olympusewallet modal recehpanduan pemula slotpg soft primadona slottercheat mahjong androidtips dewa slot mahjongslot demo mahjonghujan scatter olympusrtp caishen winsrtp sweet bonanzamahjong vs qilinmaxwin x5000 starlight princessmahjong wins x1000rtp baru wild scatterpg soft trik maxwinamantotorm1131