370
Views
4
CrossRef citations to date
0
Altmetric
Perspective

Application of emetine in SARS-CoV-2 treatment: regulation of p38 MAPK signaling pathway for preventing emetine-induced cardiac complications

, & ORCID Icon
Pages 2379-2386 | Received 07 Mar 2022, Accepted 07 Jul 2022, Published online: 19 Jul 2022

References

  • Wang A, Sun Y, Liu Q, et al. Low dose of emetine as potential anti-SARS-CoV-2 virus therapy: preclinical in vitro inhibition and in vivo pharmacokinetic evidences. Mol Biomed. 2020;1:1–9.
  • Ellinger B, Bojkova D, Zaliani A, et al. A SARS-CoV-2 cytopathicity dataset generated by high-content screening of a large drug repurposing collection. Sci Data. 2021;8:1–10.
  • Kumar R, Afsar M, Khandelwal N, et al. Emetine suppresses SARS-CoV-2 replication by inhibiting interaction of viral mRNA with eIF4E. Antiviral Res. 2021;189:105056.
  • Ren PX, Shang WJ, and Yin WC, et al. A multi-targeting drug design strategy for identifying potent anti-SARS-CoV-2 inhibitors. Acta Pharmacol Sin. 2022 Feb;43(2):483–493.
  • Bojkova D, Klann K, Koch B, et al. Proteomics of SARS-CoV-2-infected host cells reveals therapy targets. Nature. 2020;583:469–472.
  • Valipour M. Different aspects of emetine’s capabilities as a highly potent SARS-CoV-2 inhibitor against COVID-19. ACS Pharmacol Transl Sci. 2022;5:387–399.
  • Choy K-T, Wong AY-L, Kaewpreedee P, et al. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antiviral Res. 2020;178:104786.
  • Luo Y, Yu F, Zhou M, et al. Engineering a reliable and convenient SARS-CoV-2 replicon system for analysis of viral RNA synthesis and screening of antiviral inhibitors. Mbio. 2021;12:e02754–20.
  • Dyall J, Coleman CM, Venkataraman T, et al. Repurposing of clinically developed drugs for treatment of Middle East respiratory syndrome coronavirus infection. Antimicrob Agents Chemother. 2014;58:4885–4893.
  • Shen L, Niu J, Wang C, et al. High-throughput screening and identification of potent broad-spectrum inhibitors of coronaviruses. J Virol. 2019;93:e00023–19.
  • Ko M, Chang SY, and Byun SY, et al. Screening of FDA-approved drugs using a MERS-CoV clinical isolate from South Korea identifies potential therapeutic options for COVID-19. BioRxiv. 2021;13(4):651.
  • Yang C-W, Peng -T-T, Hsu H-Y, et al. Repurposing old drugs as antiviral agents for coronaviruses. Biomed J. 2020;43:368–374.
  • Yin Low J, Chen K, Wu K, et al. Antiviral activity of emetine dihydrochloride against dengue virus infection. J Antivir Antiretrovir. 2009;1:062.
  • Mukhopadhyay R, Roy S, Venkatadri R, et al. Efficacy and mechanism of action of low dose emetine against human cytomegalovirus. PLoS Pathog. 2016;12:e1005717.
  • Valadão ALC, Abreu CM, Dias JZ, et al. Natural plant alkaloid (emetine) inhibits HIV-1 replication by interfering with reverse transcriptase activity. Molecules. 2015;20:11474–11489.
  • Abate C, Niso M, and Abatematteo FS, et al. PB28, the sigma-1 and sigma-2 receptors modulator with potent anti–SARS-CoV-2 activity: a review about its pharmacological properties and structure affinity relationships. Front Pharmacol. 2020;11:589810.
  • MacGibeny MA, Koyuncu OO, Wirblich C, et al. Retrograde axonal transport of rabies virus is unaffected by interferon treatment but blocked by emetine locally in axons. PLoS Pathog. 2018;14:e1007188.
  • Andersen PI, Krpina K, Ianevski A, et al. Novel antiviral activities of obatoclax, emetine, niclosamide, brequinar, and homoharringtonine. Viruses. 2019;11:964.
  • Khandelwal N, Chander Y, Rawat KD, et al. Emetine inhibits replication of RNA and DNA viruses without generating drug-resistant virus variants. Antiviral Res. 2017;144:196–204.
  • Tay MZ, Poh CM, Rénia L, et al. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol. 2020;20:363–374.
  • Lowery SA, Sariol A, Perlman S. Innate immune and inflammatory responses to SARS-CoV-2: implications for COVID-19. Cell Host & Microbe; 2021.
  • Valipour M, Zarghi A, Ebrahimzadeh MA, et al. Therapeutic potential of chelerythrine as a multi-purpose adjuvant for the treatment of COVID-19. Cell Cycle. 2021;20:2321–2336.
  • Miller SC, Huang R, Sakamuru S, et al. Identification of known drugs that act as inhibitors of NF-κB signaling and their mechanism of action. Biochem Pharmacol. 2010;79:1272–1280.
  • Shah V, Ferguson J, Hunsaker L, et al. Cardiac glycosides inhibit LPS-induced activation of pro-inflammatory cytokines in whole blood through an NF-κB-dependent mechanism. Int J Appl Res Nat Prod. 2011;4:11–19.
  • Bleasel MD, Peterson GM. Emetine is not ipecac: considerations for its use as treatment for SARS-CoV2. Pharmaceuticals. 2020;13:428.
  • Ryan JJ, Melendres-Groves L, Zamanian RT, et al. Care of patients with pulmonary arterial hypertension during the coronavirus (COVID-19) pandemic. Pulm Circ. 2020;10:2045894020920153.
  • Forrester SJ, Kikuchi DS, Hernandes MS, et al. Reactive oxygen species in metabolic and inflammatory signaling. Circ Res. 2018;122:877–902.
  • Honda T, Hirakawa Y, Nangaku M. The role of oxidative stress and hypoxia in renal disease. Kidney Res Clin Pract. 2019;38:414.
  • Giussani DA, Niu Y, and Herrera EA, et al. Heart disease link to fetal hypoxia and oxidative stress. Adv Fetal Neonat Physiol. 2014;814:77–87.
  • Siddique MAH, Satoh K, Kurosawa R, et al. Identification of emetine as a therapeutic agent for pulmonary arterial hypertension: novel effects of an old drug. Arterioscler Thromb Vasc Biol. 2019;39:2367–2385.
  • Dresser LP, Massey EW, Johnson EE, et al. Ipecac myopathy and cardiomyopathy. J Neurol Neurosurg. 1993;56:560–562.
  • Hopf NJ, Goebel HH. Experimental emetine myopathy: enzyme histochemical, electron microscopic, and immunomorphological studies. Acta Neuropathol. 1993;85:414–418.
  • Halbig L, Gutmann L, Goebel H, et al. Ultrastructural pathology in emetine-induced myopathy. Acta Neuropathol. 1988;75:577–582.
  • Thyagarajan D, Day BJ, Wodak J, et al. Emetine myopathy in a patient with an eating disorder. Med j Aust. 1993;159:757–760.
  • Fan S, Zhen Q, Chen C, et al. Clinical efficacy of low-dose emetine for patients with COVID-19: a real-world study. J Bio-X Res. 2021;4:53–59.
  • Dubick MA, Yang WC. Effects of chronic emetine treatment on mitochondrial function. J Pharm Sci. 1981;70:343–345.
  • Yang WC, Dubick M. Mechanism of emetine cardiotoxicity. Pharmacol Ther. 1980;10:15–26.
  • Dack S, Moloshok RE. Cardiac manifestations of toxic action of emetine hydrochloride in amebic dysentery. Arch Internal Med. 1947;79:228–238.
  • Banerjea J. The effect of emetine therapy on the cardiovascular system. J Assoc Phys Ind. 1966;14:349–364.
  • Kini P, Venugopal N, Santhamma K, et al. The effect of emetine on the electrocardiogram and the serum transaminases. J Assoc Phys Ind. 1969;17:457–461.
  • Ho PC, Dweik R, Cohen MC. Rapidly reversible cardiomyopathy associated with chronic ipecac ingestion. Clin Cardiol. 1998;21:780–784.
  • Khan MY, Haider B, Thind I. Emetine-induced cardiomyopathy in rabbits. J Submicro Cytol. 1983;15:495–507.
  • Beller BM, Mongillo S. Observations on the mechanism of emetine poisoning of myocardial tissue. Circ Res. 1968;22:501–505.
  • Brem TH, Konwaler BE. Fatal myocarditis due to emetine hydrochloride. Am Heart J. 1955;50:476–481.
  • Mastrangelo MJ, Grage TB, Bellet RE, et al. A phase I study of emetine hydrochloride (NSC 33669) in solid tumors. Cancer. 1973;31:1170–1175.
  • Kane R, Cohen M, Broder L, et al. Phase I-II evaluation of emetine (NSC-33669) in the treatment of epidermoid bronchogenic carcinoma. Cancer Chemother Rep. 1975;59:1171–1172.
  • Siddiqui S, Firat D, Olshin S. Phase II study of emetine (NSC-33669) in the treatment of solid tumors. Cancer Chemother Rep. 1973;57:423–428.
  • Wang DY, Salem J-E, Cohen JV, et al. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol. 2018;4:1721–1728.
  • Ni J, Zhang L, Zhang X. Marked elevation of creatine phosphokinase alone caused by sintilimab-beware of hypothyroid myopathy. Eur J Cancer. 2020;128:57–59.
  • Hale KK, Trollinger D, Rihanek M, et al. Differential expression and activation of p38 mitogen-activated protein kinase α, β, γ, and δ in inflammatory cell lineages. J Immunol. 1999;162:4246–4252.
  • Kyriakis JM, Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev. 2001;81:807–869.
  • Yokota T, Wang Y. p38 MAP kinases in the heart. Gene. 2016;575:369–376.
  • Liang Q, Molkentin JD. Redefining the roles of p38 and JNK signaling in cardiac hypertrophy: dichotomy between cultured myocytes and animal models. J Mol Cell Cardiol. 2003;35:1385–1394.
  • Nemoto S, Sheng Z, Lin A. Opposing effects of Jun kinase and p38 mitogen-activated protein kinases on cardiomyocyte hypertrophy. Mol Cell Biol. 1998;18:3518–3526.
  • Zechner D, Thuerauf DJ, Hanford DS, et al. A role for the p38 mitogen-activated protein kinase pathway in myocardial cell growth, sarcomeric organization, and cardiac-specific gene expression. J Cell Biol. 1997;139:115–127.
  • Engel FB, Hsieh PC, and Lee RT, et al. FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction. Proc Natl Acad Sci. USA. 2006;103:15546–15551.
  • Engel FB, Schebesta M, Duong MT, et al. p38 MAP kinase inhibition enables proliferation of adult mammalian cardiomyocytes. Genes Dev. 2005;19:1175–1187.
  • Wang Y, Huang S, Sah VP, et al. Cardiac muscle cell hypertrophy and apoptosis induced by distinct members of the p38 mitogen-activated protein kinase family. J Biol Chem. 1998;273:2161–2168.
  • Kim JK, Pedram A, Razandi M, et al. Estrogen prevents cardiomyocyte apoptosis through inhibition of reactive oxygen species and differential regulation of p38 kinase isoforms. J Biol Chem. 2006;281:6760–6767.
  • Liu H, Pedram A, Kim JK. Oestrogen prevents cardiomyocyte apoptosis by suppressing p38α-mediated activation of p53 and by down-regulating p53 inhibition on p38β. Cardiovasc Res. 2011;89:119–128.
  • Kroemer G. The proto-oncogene Bcl-2 and its role in regulating apoptosis. Nat Med. 1997;3:614–620.
  • Antonsson B, Martinou J-C. The Bcl-2 protein family. Exp Cell Res. 2000;256:50–57.
  • Boon-Unge K, Yu Q, Zou T, et al. Emetine regulates the alternative splicing of Bcl-x through a protein phosphatase 1-dependent mechanism. Chem Biol. 2007;14:1386–1392.
  • Sun Q, Yogosawa S, Iizumi Y, et al. The alkaloid emetine sensitizes ovarian carcinoma cells to cisplatin through downregulation of bcl-xL. Int J Oncol. 2015;46:389–394.
  • Kaiser RA, Bueno OF, Lips DJ, et al. Targeted inhibition of p38 mitogen-activated protein kinase antagonizes cardiac injury and cell death following ischemia-reperfusion in vivo. J Biol Chem. 2004;279:15524–15530.
  • Sharov VG, Todor A, Suzuki G, et al. Hypoxia, angiotensin‐II, and norepinephrine mediated apoptosis is stimulus specific in canine failed cardiomyocytes: a role for p38 MAPK, Fas‐L and cyclin D1. Eur J Heart Fail. 2003;5:121–129.
  • Kim JH, Cho EB, Lee J, et al. Emetine inhibits migration and invasion of human non-small-cell lung cancer cells via regulation of ERK and p38 signaling pathways. Chem Biol Interact. 2015;242:25–33.
  • Westermarck J, Li S-P, Kallunki T, et al. p38 mitogen-activated protein kinase-dependent activation of protein phosphatases 1 and 2A inhibits MEK1 and MEK2 activity and collagenase 1 (MMP-1) gene expression. Mol Cell Biol. 2001;21:2373–2383.
  • Munshi HG, Wu YI, Mukhopadhyay S, et al. Differential regulation of membrane type 1-matrix metalloproteinase activity by ERK 1/2-and p38 MAPK-modulated tissue inhibitor of metalloproteinases 2 expression controls transforming growth factor-β1-induced pericellular collagenolysis. J Biol Chem. 2004;279:39042–39050.
  • Son J, Lee SY. Emetine exerts anticancer effects in U2OS human osteosarcoma cells via activation of p38 and inhibition of ERK, JNK, and β‐catenin signaling pathways. J Biochem Mol Toxicol. 2021;35:e22868.
  • Ma XL, Kumar S, Gao F, et al. Inhibition of p38 mitogen-activated protein kinase decreases cardiomyocyte apoptosis and improves cardiac function after myocardial ischemia and reperfusion. Circulation. 1999;99:1685–1691.
  • Auger-Messier M, Accornero F, Goonasekera SA, et al. Unrestrained p38 MAPK activation in Dusp1/4 double-null mice induces cardiomyopathy. Circ Res. 2013;112:48–56.
  • Croons V, Martinet W, Herman AG, et al. The protein synthesis inhibitor anisomycin induces macrophage apoptosis in rabbit atherosclerotic plaques through p38 mitogen-activated protein kinase. J Pharmacol Exp Ther. 2009;329:856–864.
  • Marber MS, Rose B, Wang Y. The p38 mitogen-activated protein kinase pathway—a potential target for intervention in infarction, hypertrophy, and heart failure. J Mol Cell Cardiol. 2011;51:485–490.
  • Barros LF, Young M, Saklatvala J, et al. Evidence of two mechanisms for the activation of the glucose transporter GLUT1 by anisomycin: p38 (MAP kinase) activation and protein synthesis inhibition in mammalian cells. J Physiol. 1997;504:517–525.
  • Li H, Mittal A, Paul PK, et al. Tumor necrosis factor-related weak inducer of apoptosis augments matrix metalloproteinase 9 (MMP-9) production in skeletal muscle through the activation of nuclear factor-κB-inducing kinase and p38 mitogen-activated protein kinase: a potential role of MMP-9 in myopathy. J Biol Chem. 2009;284:4439–4450.
  • Page TH, Brown A, Timms EM, et al. Inhibitors of p38 suppress cytokine production in rheumatoid arthritis synovial membranes: does variable inhibition of interleukin‐6 production limit effectiveness in vivo? Arthritis Rheumatism. 2010;62:3221–3231.
  • Meng Q, Bhandary B, Osinska H, et al. MMI‐0100 inhibits cardiac fibrosis in a mouse model overexpressing cardiac myosin binding protein C. J Am Heart Assoc. 2017;6:e006590.
  • Xu L, Yates CC, Lockyer P, et al. MMI-0100 inhibits cardiac fibrosis in myocardial infarction by direct actions on cardiomyocytes and fibroblasts via MK2 inhibition. J Mol Cell Cardiol. 2014;77:86–101.
  • Brown DI, Cooley BC, Quintana MT, et al. Nebulized delivery of the MAPKAP kinase 2 peptide inhibitor MMI-0100 protects against ischemia-induced systolic dysfunction. Int J Pept Res Ther. 2016;22:317–324.
  • Barbour AM, Sarov-Blat L, Cai G, et al. Safety, tolerability, pharmacokinetics and pharmacodynamics of losmapimod following a single intravenous or oral dose in healthy volunteers. Br J Clin Pharmacol. 2013;76(1):99–106.
  • Cheriyan J, Webb AJ, Sarov-Blat L, et al. Inhibition of p38 mitogen-activated protein kinase improves nitric oxide–mediated vasodilatation and reduces inflammation in hypercholesterolemia. Circulation. 2011;123:515–523.
  • Christie JD, Vaslef S, Chang PK, et al. A randomized dose-escalation study of the safety and anti-inflammatory activity of the p38 mitogen-activated protein kinase inhibitor dilmapimod in severe trauma subjects at risk for acute respiratory distress syndrome. Crit Care Med. 2015;43:1859–1869.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.