2,499
Views
1
CrossRef citations to date
0
Altmetric
Research Paper

Dynamized ultra-low dilution of Ruta graveolens disrupts plasma membrane organization and decreases migration of melanoma cancer cell

, , , , , , & show all
Pages 1-13 | Received 21 Jul 2022, Accepted 30 Nov 2022, Published online: 11 Dec 2022

References

  • Scarpa E, Mayor R. Collective cell migration in development. J Cell Biol. 2016;212(2):143–155.
  • Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2(6):442–454.
  • Eggermont AM, Spatz A, Robert C. Cutaneous melanoma. Lancet. 2014;383(9919):816–827.
  • Maverakis E, Cornelius L, Bowen G, et al. Metastatic melanoma – a review of current and future treatment options. Acta Derm Venereol. 2015;95(5):516–524.
  • Dos Santos Garrett NF M, da Costa Ac C, Barros Ferreira E, et al. Prevalence of dermatological toxicities in patients with melanoma undergoing immunotherapy: systematic review and meta-analysis. PloS One. 2021;16(8):e0255716.
  • Alrabadi NN, Abushukair HM, Ababneh OE, et al. Systematic review and meta-analysis efficacy and safety of immune checkpoint inhibitors in advanced melanoma patients with anti-PD-1 progression: a systematic review and meta-analysis. Clin Transl Oncol Off Publ Fed Span Oncol Soc Natl Cancer Inst Mex. 2021;23:1885–1904.
  • Bagot J-L, Legrand A, Theunissen I. Use of homeopathy in integrative oncology in Strasbourg, France: multi-center cross-sectional descriptive study of patients undergoing cancer treatment. Homeopathy. 2021;110(3):168–173. s-0040-1721065.
  • Rossi E, Vita A, Baccetti S, et al. Complementary and alternative medicine for cancer patients: results of the EPAAC survey on integrative oncology centres in Europe. Support Care Cancer. 2015;23(6):1795–1806.
  • Kathirvel P, Poonkodi K, Gomathi K, et al. GC-MS analysis and in vitro antioxidant activities of ruta graveolens l. From western ghats region –south INDIA. Asian J Pharm Clin Res. 2017;10(5):297.
  • Azalework HG, Jafri A S, Jafri A, et al. Phytochemical investigation, GC-MS profile and antimicrobial activity of a medicinal plant ruta graveolens l. From Ethiopia. Int J Pharm Pharm Sci. 2017;9(6):29.
  • Kamiński M, Kartanowicz R, Kamiński MM, et al. HPLC-DAD in identification and quantification of selected coumarins in crude extracts from plant cultures ofAmmi majus andRuta graveolens. J Sep Sci. 2003;26(14):1287–1291.
  • Ganeshpurkar A, Saluja AK. The pharmacological potential of rutin. Saudi Pharm J. 2017;25(2):149–164.
  • Martínez Conesa C, Vicente Ortega V, Yáñez Gascón MJ, et al. Treatment of metastatic melanoma B16F10 by the flavonoids tangeretin, rutin, and diosmin. J Agric Food Chem. 2005;53(17):6791–6797.
  • Seak C-J, Lin -C-C. Ruta Graveolens intoxication. Clin Toxicol. 2007;45(2):173–175.
  • Parray S, Bhat J, Ahmad N, et al. Ruta graveolens: from traditional system of medicine to modern pharmacology: an overview. Am. J. PharmTech Res. 2012;2:239–252.
  • Träger-Maury S, Tournigand C, Maindrault-Goebel F, et al. Use of complementary medicine by cancer patients in a French oncology department. Bull Cancer (Paris). 2007;94:1017–1025.
  • Frass M, Friehs H, Thallinger C, et al. Influence of adjunctive classical homeopathy on global health status and subjective wellbeing in cancer patients – a pragmatic randomized controlled trial. Complement Ther Med. 2015;23(3):309–317.
  • Frass M, Lechleitner P, Gründling C, et al. Homeopathic treatment as an add-on therapy may improve quality of life and prolong survival in patients with non-small cell lung cancer: a prospective, randomized, placebo-controlled, double-blind, three-arm, multicenter Study. Oncologist. 2020;25(12):e1930–e1955.
  • Rostock M, Naumann J, Guethlin C, et al. Classical homeopathy in the treatment of cancer patients - a prospective observational study of two independent cohorts. BMC Cancer. 2011;11(1):19.
  • Karp J-C, Sanchez C, Guilbert P, et al. Treatment with Ruta graveolens 5CH and Rhus toxicodendron 9CH may reduce joint pain and stiffness linked to aromatase inhibitors in women with early breast cancer: results of a pilot observational study. Homeopathy. 2016;105(4):299–308.
  • Frenkel M, Mishra BM, Sen S, et al. Cytotoxic effects of ultra-diluted remedies on breast cancer cells. Int J Oncol. 2010;36(2):395–403.
  • Sunila ES, Kuttan G. A preliminary study on antimetastatic activity of thuja occidentalis L. in mice model. Immunopharmacol Immunotoxicol. 2006;28(2):269–280.
  • MacLaughlin BW, Gutsmuths B, Pretner E, et al. Effects of homeopathic preparations on human prostate cancer growth in cellular and animal models. Integr Cancer Ther. 2006;5(4):362–372.
  • Saha S, Hossain DMS, Mukherjee S, et al. Calcarea carbonica induces apoptosis in cancer cells in p53-dependent manner via an immuno-modulatory circuit. BMC Complement Altern Med. 2013;13(1):230.
  • Arora S, Aggarwal A, Singla P, et al. Anti-proliferative effects of homeopathic medicines on human kidney, colon and breast cancer cells. Homeopathy J Fac Homeopathy. 2013;102(4):274–282.
  • Arora S, Tandon S. DNA fragmentation and cell cycle arrest: a hallmark of apoptosis induced by Ruta graveolens in human colon cancer cells. Homeopathy. 2015;104(1):36–47.
  • Fuselier C, Terryn C, Berquand A, et al. Low-diluted Phenacetinum disrupted the melanoma cancer cell migration. Sci Rep. 2019;9(1):9109.
  • Fuselier C, Quemener S, Dufay E, et al. Anti-Tumoral and anti-angiogenic effects of low-diluted phenacetinum on melanoma. Front Oncol. 2021;11:597503.
  • Maxfield FR, Wüstner D. Analysis of cholesterol trafficking with fluorescent probes. Methods cell biol [Internet]. Elsevier; 2012 [cited 2022 Mar 23]. p. 367–393. Available from: https://linkinghub.elsevier.com/retrieve/pii/B9780123864871000171.
  • Ogawa Y, Tanaka M. A fluorescent cholesterol analogue for observation of free cholesterol in the plasma membrane of live cells. Anal Biochem. 2016;492:49–55.
  • Adler NR, Kelly JW, Haydon A, et al. Clinicopathological characteristics and prognosis of patients with multiple primary melanomas. Br J Dermatol. Internet]. 2018 cited 2022 Mar 23;178. Available from.;(1). https://onlinelibrary.wiley.com/doi/10.1111/bjd.15855.
  • Cummins DL, Cummins JM, Pantle H, et al. Cutaneous malignant melanoma. Mayo Clin Proc. 2006;81(4):500–507.
  • Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–723.
  • Miller AJ, Mihm MC. Melanoma. N Engl J Med. 2006;355(1):51–65.
  • Bagot J-L, Karp J-C, Messerschmitt C, et al. Recommandations thérapeutiques de la Société Homéopathique Internationale de Soins de Support en Oncologie (SHISSO). Rev Homéopathie. 2017;8(4):183–191.
  • Gensbittel V, Kräter M, Harlepp S, et al. Mechanical adaptability of tumor cells in metastasis. Dev Cell. 2021;56(2):164–179.
  • Luo Q, Kuang D, Zhang B, et al. Cell stiffness determined by atomic force microscopy and its correlation with cell motility. Biochim Biophys Acta BBA - Gen Subj. 2016;1860(9):1953–1960.
  • Watanabe T, Kuramochi H, Takahashi A, et al. Higher cell stiffness indicating lower metastatic potential in B16 melanoma cell variants and in (−)-epigallocatechin gallate-treated cells. J Cancer Res Clin Oncol. 2012;138(5):859–866.
  • Weder G, Hendriks-Balk MC, Smajda R, et al. Increased plasticity of the stiffness of melanoma cells correlates with their acquisition of metastatic properties. Nanomed Nanotechnol Biol Med. 2014;10(1):141–148.
  • Kashani AS, Packirisamy M. Cancer cells optimize elasticity for efficient migration. R Soc Open Sci. 2020;7(10):200747.
  • Keren K. Cell motility: the integrating role of the plasma membrane. Eur Biophys J. 2011;40(9):1013–1027.
  • Sok M, Šentjurc M, Schara M, et al. Cell membrane fluidity and prognosis of lung cancer. Ann Thorac Surg. 2002;73(5):1567–1571.
  • Veronika M, Welsch R, Ng A, et al. Correlation of cell membrane dynamics and cell motility. BMC Bioinformatics. 2011;12(S13):S19.
  • Zalba S, ten Hagen Tlm, ten Hagen TLM. Cell membrane modulation as adjuvant in cancer therapy. Cancer Treat Rev. 2017;52:48–57.
  • Cooper GM. The cell: a molecular approach. 2. ed. Washington DC: ASM Press [u.a.]; 2000.
  • Sezgin E, Levental I, Mayor S, et al. The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat Rev Mol Cell Biol. 2017;18(6):361–374.
  • Nicolson G. Update of the 1972 singer-Nicolson fluid-mosaic model of membrane structure. Discoveries [Internet]. 2013 cited 2022 Mar 23]; Available from 2022 Mar 23: http://www.discoveriesjournals.org/D.2013.01.RA-Prof%20Nicolson.DOI.html.
  • Bleich HL, Boro ES, Cooper RA. Abnormalities of cell-membrane fluidity in the pathogenesis of disease. N Engl J Med. 1977;297(7):371–377.
  • Costa GA, de Souza SB, da Silva Teixeira LR, et al. Tumor cell cholesterol depletion and V-ATPase inhibition as an inhibitory mechanism to prevent cell migration and invasiveness in melanoma. Biochim Biophys Acta BBA - Gen Subj. 2018;1862(3):684–691.
  • Sherbet GV. Membrane fluidity and cancer metastasis. Pathobiology. 1989;57(4):198–205.
  • Hendrich A, Michalak K. Lipids as a target for drugs modulating multidrug resistance of cancer cells. Curr Drug Targets. 2003;4(1):23–30.
  • Epand RM, Thomas A, Brasseur R, et al. Cholesterol interaction with proteins that partition into membrane domains: an overview.In: Harris JR, editor. Cholest bind cholest transp proteins Internet]. Dordrecht: Springer Netherlands; 2010 [cited 2022 Mar 23]. p. 253–278. Available from. http://link.springer.com/10.1007/978-90-481-8622-8_9
  • Lindner R, Naim HY. Domains in biological membranes. Exp Cell Res. 2009;315(17):2871–2878.
  • Mollinedo F, Gajate C. Lipid rafts as major platforms for signaling regulation in cancer. Adv Biol Regul. 2015;57:130–146.
  • Pike LJ. Rafts defined: a report on the Keystone symposium on lipid rafts and cell function. J Lipid Res. 2006;47(7):1597–1598.
  • Ermilova I, Lyubartsev AP. Cholesterol in phospholipid bilayers: positions and orientations inside membranes with different unsaturation degrees. Soft Matter. 2019;15(1):78–93.
  • Chichili GR, Rodgers W. Cytoskeleton–membrane interactions in membrane raft structure. Cell Mol Life Sci. 2009;66(14):2319–2328.
  • Lingwood D, Simons K. Lipid rafts as a membrane-organizing principle. Science. 2010;327(5961):46–50.
  • Eytan GD, Regev R, Assaraf YG. Functional reconstitution of P-glycoprotein reveals an apparent near stoichiometric drug transport to ATP hydrolysis. J Biol Chem. 1996;271(6):3172–3178.
  • Hill WG, Zeidel ML. Reconstituting the barrier properties of a water-tight epithelial membrane by design of leaflet-specific liposomes. J Biol Chem. 2000;275(39):30176–30185.
  • Escriba PV, Ferrer-Montiel AV, Ferragut JA, et al. Role of membrane lipids in the interaction of daunomycin with plasma membranes from tumor cells: implications in drug-resistance phenomena. Biochemistry. 1990;29(31):7275–7282.
  • Levitan I, Fang Y, Rosenhouse-Dantsker A, et al. Cholesterol and ion channels In: Harris JR, editor. Cholest bind cholest transp proteins Internet]. Dordrecht: Springer Netherlands; 2010 [cited 2022 Mar 23]. p. 509–549. Available from: http://link.springer.com/10.1007/978-90-481-8622-8_19
  • Wei C, Wang X, Zheng M, et al. Calcium gradients underlying cell migration. Curr Opin Cell Biol. 2012;24(2):254–261.
  • Nebl T, Pestonjamasp KN, Leszyk JD, et al. Proteomic analysis of a detergent-resistant membrane skeleton from neutrophil plasma membranes. J Biol Chem. 2002;277(45):43399–43409.
  • Yanagida M, Nakayama H, Yoshizaki F, et al. Proteomic analysis of plasma membrane lipid rafts of HL-60 cells. Proteomics. 2007;7(14):2398–2409.
  • Sun M, Northup N, Marga F, et al. The effect of cellular cholesterol on membrane-cytoskeleton adhesion. J Cell Sci. 2007;120(13):2223–2231.
  • Wu L, Huang J, Yu X, et al. AFM of the ultrastructural and mechanical properties of lipid-raft-disrupted and/or cold-treated endothelial cells. J Membr Biol. 2014;247(2):189–200.
  • Levitan I. Paradoxical impact of cholesterol on lipid packing and cell stiffness. Front Biosci. 2016;21(6):1245–1259.
  • Brás MM, Radmacher M, Sousa SR, et al. Melanoma in the eyes of mechanobiology. Front Cell Dev Biol. 2020;8:54.
  • Bastatas L, Martinez-Marin D, Matthews J, et al. AFM nano-mechanics and calcium dynamics of prostate cancer cells with distinct metastatic potential. Biochim Biophys Acta BBA - Gen Subj. 2012;1820(7):1111–1120.