Preprint / Version 1

Doxycycline and Minocycline Drugs as a Treatment Proposal for Inhibition of ARDS and Inflammatory Cytokine Mediators Caused by COVID19

Authors

DOI:

https://doi.org/10.21467/preprints.43

Abstract

The novel coronavirus 2 (COVID 19) is a highly transmittable viral disease aroused in Wuhan, China at the end of 2019 and spreads around the world. The International Committee on Taxonomy of Viruses (ICTV) named it on February 11, 2020, as severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2). SARS-CoV-2 can infect humans as well likewise as animals and transmit from person to person. The World Health Organization declared that the novel coronavirus 2 pneumonia epidemic and was classified as a public health emergency of international attention on January 30, 2020. When COVID-19 infects the upper and lower respiratory tract it can give rise to mild or acute Respiratory Distress Syndrome (ARDS) with resultant release of cytokines like IL-1B, IL6, IL37, TNF alpha, and CCL2. we evaluated the effectiveness of doxycycline and minocycline as a tetracycline derivative to modulate serum levels of cytokines and we stand out their anti- inflammatory effect that can inhibit inflammation and pro-inflammation response that was caused by COVID-19, providing pertinent strategy.

Keywords:

COVID-19, SARS-CoV-2, ARDS, Tetracyclines, Cytokines

Downloads

Download data is not yet available.

References

Chopra I, Roberts M. Tetracycline antibiotics: mode of action, applications, molecular biology and epidemiology of bacterial resistance. Microbiol Mol Biol Rev 2001; 65:232–60.

Tauber SC, Nau R. Immunomodulatory properties of antibiotics. Current Molecular Pharmacology. 2008;1(1):68–79.

Lai AY, Todd KG. Hypoxia-activated microglial mediators of neuronal survival are differentially regulated by tetracyclines. GLIA. 2006;53(8):809–816.

Castro, J. E. Z., Vado-Solis, I., Perez-Osorio, C., & Fredeking, T. M. (2011). Modulation of Cytokine and Cytokine Receptor/Antagonist by Treatment with Doxycycline and Tetracycline in Patients with Dengue Fever. Clinical and Developmental Immunology, 2011, 1–5.

Lemaitre M, Guetard D, Henin Y, Montagnier L, Zerial A. Protective activity of tetracycline analogs against the cytopathic effect of the human immunodeficiency viruses in CEM cells. Res Virol 1990;141:5–16.

Szeto GL, Brice AK, Yang HC, Barber SA, Siliciano RF, Clements JE. Minocycline attenuates HIV infection and reactivation by suppressing cellular activation in human CD4+ T cells. J Infect Dis 2010;201:1132–40.

Atrasheuskaya AV, Fredeking TM, Ignatyev GM. Changes in immune parameters and their correction in human cases of tick-borne encephalitis. Clinical and Experimental Immunology. 2003;131(1):148–154.

Bethell DB, Flobbe K, Phuong CXT, et al. Pathophysiologic and prognostic role of cytokines in dengue hemorrhagic fever. Journal of Infectious Diseases. 1998;177(3):778–782.

Green S, Vaughn DW, Kalayanarooj S, et al. Early immune activation in acute dengue illness is related to development of plasma leakage and disease severity. Journal of Infectious Diseases. 1999;179(4):755–762.

Girardin E, Roux-Lombard P, Grau GE, Suter P, Gallati H, Dayer J-M. Imbalance between tumour necrosis factor-alpha and soluble TNF receptor concentrations in severe meningococcaemia. Immunology. 1992;76(1):20–23.

van Dissel JT, van Langevelde P, Westendorp RGJ, Kwappenberg K, Frölich M. Anti-inflammatory cytokine profile and mortality in febrile patients. Lancet. 1998;351(9107):950–953.

Dinarello CA. Anti-cytokine therapeutics and infections. Vaccine. 2003;21(2):S24–S34.

Clark IA, Budd AC, Alleva LM, Cowden WB. Human malarial disease: a consequence of inflammatory cytokine release. Malaria Journal. 2006;5:85–117.

Bozza FA, Cruz OG, Zagne SMO, et al. Multiplex cytokine profile from dengue patients: MIP-1beta and IFN-gamma as predictive factors for severity. BMC Infectious Diseases. 2008;8:86–93.

Green S, Vaughn DW, Kalayanarooj S, et al. Early immune activation in acute dengue illness is related to development of plasma leakage and disease severity. Journal of Infectious Diseases. 1999;179(4):755–762.

Castro, J. E. Z., Vado-Solis, I., Perez-Osorio, C., & Fredeking, T. M. (2011). Modulation of Cytokine and Cytokine Receptor/Antagonist by Treatment with Doxycycline and Tetracycline in Patients with Dengue Fever. Clinical and Developmental Immunology, 2011, 1–5.

Yang JM, Chen YF, Tu YY, Yen KR, Yang YL. Combinatorial computational approaches to identify tetracycline derivatives as flavivirus inhibitors. PLoS ONE. 2007;2(5):e428–e441.

X.N. Tang, Q. Wang, M.A. Koike, et al., Monitoring the protective effects of mino-cycline treatment with radiolabeled annexin V in an experimental model of focalcerebral ischemia, J. Nucl. Med. 48 (11) (2007) 1822–1828.

Giuliani F, Hader W, Yong VW. Minocycline attenuates T cell and microglia activity to impair cytokine production in T cell-microglia interaction. J Leukoc Biol 2005;78:135–43.

Popovic N, Schubart A, Goetz BD, Zhang SC, Linington C, Duncan ID. “Inhibition of autoimmune encephalomyelitis by a tetracycline“. Ann Neurol 2002;51:215–23.

Song Y, Wei EQ, Zhang WP, Zhang L, Liu JR, Chen Z. Minocycline protects PC12 cells from ischemic-like injury and inhibits 5- lipoxygenase activation. NeuroReport 2004;15:2181–4.

Chen M, Ona VO, Li M, Ferrano RJ, Fink KB, Zhu S. minocycline inhibits caspase – and caspase -3 expression and delays mortality in a transgenic mouse model of Huntington disease. Nat Med 2000;6:797–801.

Domerq M, Matute C. Neuroprotection of by tetracyclines. Trends Pharmacol Sci 2004;25:609–12.

Szeto GL, Brice AK, Yang HC, Barber SA, Siliciano RF, Clements JE. Minocycline attenuates HIV infection and reactivation by suppressing cellular activation in human CD4+ T cells. J Infect Dis 2010;201:1132–40.

J. Yrjänheikki, T. Tikka, R. Keinänen, G. Goldsteins, P.H. Chan, J. Koistinaho, Atetracycline derivative, minocycline, reduces inflammation and protects againstfocal cerebral ischemia with a wide therapeutic window, Proc. Natl. Acad. Sci. U. S.A. 96 (23) (1999) 13496–13500.

K. Hayakawa, K. Mishima, M. Nozako, et al., Delayed treatment with minocyclineameliorates neurologic impairment through activated microglia expressing a high-mobility group box-inhibiting mechanism, Stroke 39(3) (2008) 951–958.

T. Tikka, B.L. Fiebich, G. Goldsteins, R. Keinanen, J. Koistinaho, Minocycline, atetracycline derivative, is neuroprotective against excitotoxicity by inhibiting ac-tivation and proliferation of microglia, J. Neurosci. 21 (8) (2001) 2580–2588.

X.N. Tang, Q. Wang, M.A. Koike, et al., Monitoring the protective effects of mino-cycline treatment with radiolabeled annexin V in an experimental model of focalcerebral ischemia, J. Nucl. Med. 48 (11) (2007) 1822–1828.

R.O. Sanchez Mejia, V.O. Ona, M. Li, R.M. Friedlander, Minocycline reduces trau-matic brain injury-mediated caspase-1 activation, tissue damage, and neurologicaldysfunction, Neurosurgery 48 (6) (2001) 1393–1399.

Mohit Sodhi and Mahyar Etminan, Letter to the Editor, Therapeutic Potential for Tetracyclines in the Treatment of COVID-19.

Zakeri B, Wright GD. Chemical biology of tetracycline antibiotics. Biochem Cell Biol. 2008;86(2):124–136.

Humar, A., McGilvray, I., Phillips, M.J. and Levy, G.A. (2004), Severe acute respiratory syndrome and the liver. Hepatology, 39: 291-294

Phillips JM, Gallagher T, Weiss SR. Neurovirulent Murine Coronavirus JHM.SD Uses Cellular Zinc Metalloproteases for Virus Entry and Cell-Cell Fusion. J Virol. 2017;91(8):e01564-16

Rothan HA, Mohamed Z, Paydar M, Rahman NA, Yusof R. Inhibitory effect of doxycycline against dengue virus replication in vitro. Arch Virol. 2014; 159(4):711–718.

Sturtz FG. Antimurine retroviral effect of doxycycline. Methods Find Exp Clin Pharmacol. 1998; 20(8):643–647.

Henehan M, Montuno M, De Benedetto A. Doxycycline as an anti-inflammatory agent: updates in dermatology. J Eur Acad Dermatol Venereol. 2017; 31(11):1800–1808.

Yoshikawa T, Hill T, Li K, Peters CJ, Tseng CT. Severe acute respiratory syndrome (SARS) coronavirus-induced lung epithelial cytokines exacerbate SARS pathogenesis by modulating intrinsic functions of monocyte-derived macrophages and dendritic cells. J Virol. 2009; 83(7):3039–3048.

Kritas SK, Ronconi G, Caraffa A, Gallenga CE, Ross R, Conti P. Mast cells contribute to coronavirus-induced inflammation: new anti-inflammatory strategy [published online ahead of print, 2020 Feb 4]. J Biol Regul Homeost Agents. 2020; 34(1):10.23812/20- Editorial-Kritas.

Sandler C, Nurmi K, Lindstedt KA, et al. Chemically modified tetracyclines induce apoptosis in cultured mast cells. Int Immunopharmacol. 2005;5(11):1611–1621.

Sandler C, Ekokoski E, Lindstedt KA, et al. Chemically modified tetracycline (CMT)-3 inhibits histamine release and cytokine production in mast cells: possible involvement of protein kinase C. Inflamm Res. 2005;54(7):304–312

Griffin MO, Fricovsky E, Ceballos G, Villarreal F. Tetracyclines: a pleitropic family of compounds with promising therapeutic properties. Review of the literature. Am J Physiol Cell Physiol. 2010;299(3):C539–C548

Downloads

Posted

2020-04-25

Section

Coronavirus

Categories