Analgesics
Antiandrogens
Azvudine
Bromhexine
Budesonide
Colchicine
Conv. Plasma
Curcumin
Famotidine
Favipiravir
Fluvoxamine
Hydroxychlor..
Ivermectin
Lifestyle
Melatonin
Metformin
Minerals
Molnupiravir
Monoclonals
Naso/orophar..
Nigella Sativa
Nitazoxanide
Paxlovid
Quercetin
Remdesivir
Thermotherapy
Vitamins
More

Other
Feedback
Home
Top
Abstract
All ivermectin studies
Meta analysis
 
Feedback
Home
next
study
previous
study
c19ivm.org COVID-19 treatment researchIvermectinIvermectin (more..)
Melatonin Meta
Metformin Meta
Azvudine Meta
Bromhexine Meta Molnupiravir Meta
Budesonide Meta
Colchicine Meta
Conv. Plasma Meta Nigella Sativa Meta
Curcumin Meta Nitazoxanide Meta
Famotidine Meta Paxlovid Meta
Favipiravir Meta Quercetin Meta
Fluvoxamine Meta Remdesivir Meta
Hydroxychlor.. Meta Thermotherapy Meta
Ivermectin Meta

All Studies   Meta Analysis    Recent:   

Effect of Ivermectin and Atorvastatin on Nuclear Localization of Importin Alpha and Drug Target Expression Profiling in Host Cells from Nasopharyngeal Swabs of SARS-CoV-2- Positive Patients

Segatori et al., Viruses, doi:10.3390/v13102084
Oct 2021  
  Post
  Facebook
Share
  Source   PDF   All Studies   Meta AnalysisMeta
Ivermectin for COVID-19
4th treatment shown to reduce risk in August 2020
 
*, now known with p < 0.00000000001 from 101 studies, recognized in 22 countries.
No treatment is 100% effective. Protocols combine complementary and synergistic treatments. * >10% efficacy in meta analysis with ≥3 clinical studies.
3,900+ studies for 60+ treatments. c19ivm.org
Gene expression analysis of nasopharyngeal swabs of COVID-19 positive and negative patients, and in vitro study supporting the use of ivermectin and atorvastatin for COVID-19, and the efficacy of ivermectin at clinically relevant dosages.
Experiments showed that ivermectin and atorvastatin halted NF-κB activation, impaired importin and Rho GTPases gene expression, and inhibited importin α nuclear accumulation. Authors note that ivermectin and atorvastatin's targetting of importin-mediated nuclear trafficking may also indicate applicability to other infections including dengue fever, zika, and influenza.
Authors show that an ivermectin concentration as low as 0.2μM for 24h produced a similar effect on the inhibition of importin α nuclear to cytoplasmic distribution as that of a 2.5μM for 1h. This suggests that a sustained exposure to lower concentrations could interfere with the host cell machinery that SARS-CoV-2 requires for replication. Experiments also indicate improved results with the combination of ivermectin and atorvastatin.
Ivermectin, better known for antiparasitic activity, is a broad spectrum antiviral with activity against many viruses including H7N7 Götz, Dengue Jitobaom, Tay, Wagstaff, HIV-1 Wagstaff, Simian virus 40 Wagstaff (B), Zika Barrows, Jitobaom, Yang, West Nile Yang, Yellow Fever Mastrangelo, Varghese, Japanese encephalitis Mastrangelo, Chikungunya Varghese, Semliki Forest virus Varghese, Human papillomavirus Li, Epstein-Barr Li, BK Polyomavirus Bennett, and Sindbis virus Varghese.
Ivermectin inhibits importin-α/β-dependent nuclear import of viral proteins Götz, Kosyna, Wagstaff, Wagstaff (B), inhibits SARS-CoV-2 3CLpro Mody, shows spike-ACE2 disruption at 1nM with microfluidic diffusional sizing Fauquet, binds to glycan sites on the SARS-CoV-2 spike protein preventing interaction with blood and epithelial cells and inhibiting hemagglutination Boschi, Scheim, exhibits dose-dependent inhibition of lung injury Abd-Elmawla, Ma, may inhibit SARS-CoV-2 via IMPase inhibition Jitobaom, may inhibit SARS-CoV-2 induced formation of fibrin clots resistant to degradation Vottero, may inhibit SARS-CoV-2 RdRp activity Parvez (B), may be beneficial for COVID-19 ARDS by blocking GSDMD and NET formation Liu (C), shows protection against inflammation, cytokine storm, and mortality in an LPS mouse model sharing key pathological features of severe COVID-19 DiNicolantonio, Zhang, may be beneficial in severe COVID-19 by binding IGF1 to inhibit the promotion of inflammation, fibrosis, and cell proliferation that leads to lung damage Zhao, may minimize SARS-CoV-2 induced cardiac damage Liu, Liu (B), increases Bifidobacteria which play a key role in the immune system Hazan, has immunomodulatory Munson and anti-inflammatory DiNicolantonio (B), Yan properties, and has an extensive and very positive safety profile Descotes.
Segatori et al., 15 Oct 2021, peer-reviewed, 11 authors.
In Vitro studies are an important part of preclinical research, however results may be very different in vivo.
This PaperIvermectinAll
Effect of Ivermectin and Atorvastatin on Nuclear Localization of Importin Alpha and Drug Target Expression Profiling in Host Cells from Nasopharyngeal Swabs of SARS-CoV-2- Positive Patients
Valeria Inés Segatori, Juan Garona, Lorena Grisel Caligiuri, Juan Bizzotto, Rosario Lavignolle, Ayelén Toro, Pablo Sanchis, Eduardo Spitzer, Alejandro Krolewiecki, Geraldine Gueron, Daniel Fernando Alonso
Viruses, doi:10.3390/v13102084
Nuclear transport and vesicle trafficking are key cellular functions involved in the pathogenesis of RNA viruses. Among other pleiotropic effects on virus-infected host cells, ivermectin (IVM) inhibits nuclear transport mechanisms mediated by importins and atorvastatin (ATV) affects actin cytoskeleton-dependent trafficking controlled by Rho GTPases signaling. In this work, we first analyzed the response to infection in nasopharyngeal swabs from SARS-CoV-2-positive and -negative patients by assessing the gene expression of the respective host cell drug targets importins and Rho GTPases. COVID-19 patients showed alterations in KPNA3, KPNA5, KPNA7, KPNB1, RHOA, and CDC42 expression compared with non-COVID-19 patients. An in vitro model of infection with Poly(I:C), a synthetic analog of viral double-stranded RNA, triggered NF-κB activation, an effect that was halted by IVM and ATV treatment. Importin and Rho GTPases gene expression was also impaired by these drugs. Furthermore, through confocal microscopy, we analyzed the effects of IVM and ATV on nuclear to cytoplasmic importin α distribution, alone or in combination. Results showed a significant inhibition of importin α nuclear accumulation under IVM and ATV treatments. These findings confirm transcriptional alterations in importins and Rho GTPases upon SARS-CoV-2 infection and point to IVM and ATV as valid drugs to impair nuclear localization of importin α when used at clinically-relevant concentrations.
Supplementary Materials: The following are available online at www.mdpi.com/article/10.3390/v13102084/s1; Table S1 : Patient demographics for GSE152075 dataset. Figure S1 : Gene expression in COVID-19 and non-COVID-19 patients categorized by sex. Figure S2 : Gene expression of importins and Rho GTPases in A549 cells and ferrets. Figure S3 : Importin and Rho GTPases gene expression analysis in A549 and HMVEC cells. Author Contributions: Conceptualization, D.F.A., G.G., and E.S.; Methodology, V.I.S., J.G., L.G.C., J.B., R.L., A.T., and P.S.; Software, J.B., R.L., A.T., and P.S.; Validation, J.B., R.L., A.T., and P.S.; Formal analysis, V.I.S., J.G.,J.B., R.L., A.T., and P.S.; Investigation, V.I.S., J.G., J.B., R.L., A.T., and P.S.; Data curation, J.B., R.L., A.T., and P.S.; Writing-original draft preparation, V.I.S., J.G., G.G., and D.F.A.; Writing-review & editing, V.I.S., J.G., J.B., R.L., A.T., P.S., E.S., A.K., G.G., and D.F.A.; Supervision, D.F.A. and G.G.; Project administration, V.I.S. and J.G.; Funding acquisition, G.G., D.F.A., and A.K. All authors have read and agreed to the published version of the manuscript.
References
Alonso, Farina, Repurposing of host-based therapeutic agents for the treatment of coronavirus disease 2019 (COVID-19): A link between antiviral and anticancer mechanisms?, Int. J. Antimicrob. Agents, doi:10.1016/j.ijantimicag.2020.106125
Atkinson, Audsley, Lieu, Marsh, Thomas et al., Recognition by host nuclear transport proteins drives disorder-to-order transition in Hendra virus V, Sci. Rep, doi:10.1038/s41598-017-18742-8
Audsley, Jans, Moseley, Nucleocytoplasmic trafficking of Nipah virus W protein involves multiple discrete interactions with the nuclear import and export machinery, Biochem. Biophys. Res. Commun, doi:10.1016/j.bbrc.2016.09.043
Bello, Elucidation of the inhibitory activity of ivermectin with host nuclear importin α and several SARS-CoV-2 targets, J. Biomol. Struct. Dyn, doi:10.1080/07391102.2021.1911857
Bizzotto, Sanchis, Abbate, Lage-Vickers, Lavignolle et al., SARS-CoV-2 Infection Boosts MX1 Antiviral Effector in COVID-19 Patients, iScience, doi:10.1016/j.isci.2020.101585
Blanco-Melo, Nilsson-Payant, Liu, Uhl, Hoagland et al., Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19, Cell, doi:10.1016/j.cell.2020.04.026
Boerma, Fu, Wang, Loose, Bartolozzi et al., Comparative gene expression profiling in three primary human cell lines after treatment with a novel inhibitor of Rho kinase or atorvastatin, Blood Coagul. Fibrinolysis Int. J. Haemost. Thromb, doi:10.1097/MBC.0b013e32830b2891
Caly, Druce, Catton, Jans, Wagstaff, The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro, Antivir. Res, doi:10.1016/j.antiviral.2020.104787
Chi, Wang, Huang, Stamnes, Chen, Roles of Rho GTPases in Intracellular Transport and Cellular Transformation, Int. J. Mol. Sci, doi:10.3390/ijms14047089
Core, R: A Language and Environment for Statistical Computing; R Foundation for Statistical Computing
De Melo, Lazarini, Larrous, Feige, Kornobis et al., Attenuation of clinical and immunological outcomes during SARS-CoV-2 infection by ivermectin, EMBO Mol. Med, doi:10.15252/emmm.202114122
Errecalde, Lifschitz, Vecchioli, Ceballos, Errecalde et al., Safety and Pharmacokinetic Assessments of a Novel Ivermectin Nasal Spray Formulation in a Pig Model, J. Pharm. Sci, doi:10.1016/j.xphs.2021.01.017
Farina, Bublik, Alonso, Gomez, Lovastatin alters cytoskeleton organization and inhibits experimental metastasis of mammary carcinoma cells, Clin. Exp. Metastasis, doi:10.1023/a:1020355621043
Fedson, Opal, Rordam, Hiding in Plain Sight: An Approach to Treating Patients with Severe COVID-19 Infection, mBio, doi:10.1128/mBio.00398-20
Fernández-Iglesias, Fuente, Gil-Peña, Alonso-Duran, García-Bengoa et al., A simple method based on confocal microscopy and thick sections recognizes seven subphases in growth plate chondrocytes, Sci. Rep, doi:10.1038/s41598-020-63978-6
Fortier, Kent, Ashdown, Poole, Boksa et al., The viral mimic, polyinosinic:polycytidylic acid, induces fever in rats via an interleukin-1-dependent mechanism, Am. J. Physiol. Regul. Integr. Comp. Physiol, doi:10.1152/ajpregu.00293.2004
Garona, Pifano, Orlando, Pastrian, Iannucci et al., The novel desmopressin analogue [V4Q5]dDAVP inhibits angiogenesis, tumour growth and metastases in vasopressin type 2 receptor-expressing breast cancer models, Int. J. Oncol, doi:10.3892/ijo.2015.2952
Garona, Pifano, Pastrian, Gomez, Ripoll et al., Addition of vasopressin synthetic analogue [V(4)Q(5)]dDAVP to standard chemotherapy enhances tumour growth inhibition and impairs metastatic spread in aggressive breast tumour models, Clin. Exp. Metastasis, doi:10.1007/s10585-016-9799-5
Gower, Peeples, Collins, Graham, RhoA Is Activated During Respiratory Syncytial Virus Infection, Virology, doi:10.1006/viro.2001.0891
Gu, Zhao, Jin, Song, Zhi et al., Cytokine Signature Induced by SARS-CoV-2 Spike Protein in a Mouse Model, Front. Immunol, doi:10.3389/fimmu.2020.621441
Guzzo, Furtek, Porras, Chen, Tipping et al., Safety, tolerability, and pharmacokinetics of escalating high doses of ivermectin in healthy adult subjects, J. Clin. Pharmacol, doi:10.1177/009127002401382731
Haji Aghajani, Moradi, Azhdari Tehrani, Amini, Pourheidar et al., Promising effects of atorvastatin on mortality and need for mechanical ventilation in patients with severe COVID-19; a retrospective cohort study, Int. J. Clin. Pract, doi:10.1111/ijcp.14434
Harris, Sridhar, Peng, Phillips, Cohn et al., Double-stranded RNA induces molecular and inflammatory signatures that are directly relevant to COPD, Mucosal Immunol, doi:10.1038/mi.2012.86
Hill, Garratt, Levi, Falconer, Ellis et al., Expression of Concern: "Meta-analysis of Randomized Trials of Ivermectin to Treat SARS-CoV-2 Infection, Open Forum Infect. Dis, doi:10.1093/ofid/ofab394
Huang, Stuart, Takeda, D'agnillo, Golding et al., I:C) Induces Human Lung Endothelial Barrier Dysfunction by Disrupting Tight Junction Expression of Claudin-5, PLoS ONE, doi:10.1371/journal.pone.0160875
Jans, Wagstaff, Ivermectin as a Broad-Spectrum Host-Directed Antiviral: The Real Deal?, Cells, doi:10.3390/cells9092100
Kassambara, Based Publication Ready Plots. R package version 0
Kern, Schöning, Chaccour, Hammann, Modeling of SARS-CoV-2 Treatment Effects for Informed Drug Repurposing, Front. Pharmacol, doi:10.3389/fphar.2021.625678
Krolewiecki, Lifschitz, Moragas, Travacio, Valentini et al., Antiviral effect of high-dose ivermectin in adults with COVID-19: A proof-of-concept randomized trial, EClinicalMedicine, doi:10.1016/j.eclinm.2021.100959
Lever, Park, Mulhern, Jackson, Comolli et al., Comprehensive evaluation of poly(I:C) induced inflammatory response in an airway epithelial model, Physiol. Rep, doi:10.14814/phy2.12334
Lieberman, Peddu, Xie, Shrestha, Huang et al., In vivo antiviral host transcriptional response to SARS-CoV-2 by viral load, sex, and age, PLoS Biol, doi:10.1371/journal.pbio.3000849
Livak, Schmittgen, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method, Methods, doi:10.1006/meth.2001.1262
Love, Huber, Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, doi:10.1186/s13059-014-0550-8
López-Medina, López, Hurtado, Dávalos, Ramirez et al., Effect of Ivermectin on Time to Resolution of Symptoms Among Adults With Mild COVID-19: A Randomized Clinical Trial, JAMA, doi:10.1001/jama.2021.3071
Menna, Parera, Cardama, Alonso, Gomez et al., Enhanced cytostatic activity of statins in mouse mammary carcinoma cells overexpressing β2-chimaerin, Mol. Med. Rep, doi:10.3892/mmr_00000068
Miyamoto, Saiwaki, Yamashita, Yasuda, Kotera et al., Cellular stresses induce the nuclear accumulation of importin alpha and cause a conventional nuclear import block, J. Cell Biol, doi:10.1083/jcb.200312008
Poon, Oro, Dias, Zhang, Jans, Apoptin nuclear accumulation is modulated by a CRM1-recognized nuclear export signal that is active in normal but not in tumor cells, Cancer Res, doi:10.1158/0008-5472.can-05-1370
Pushpakom, Iorio, Eyers, Escott, Hopper et al., Drug repurposing: Progress, challenges and recommendations, Nat. Rev. Drug Discov, doi:10.1038/nrd.2018.168
Sanchis, Lavignolle, Abbate, Lage-Vickers, Vazquez et al., Analysis workflow of publicly available RNA-sequencing datasets, STAR Protoc, doi:10.1016/j.xpro.2021.100478
Schloerke, Cook, Larmarange, Francois, Marbach et al., Extension to "ggplot2
Serafin, Bottega, Foletto, Da Rosa, Hörner et al., Drug repositioning is an alternative for the treatment of coronavirus COVID-19, Int. J. Antimicrob. Agents, doi:10.1016/j.ijantimicag.2020.105969
Shukla, Misra, Antiviral Effects of Ivermectin in COVID-19-Clinically Plausible?, Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis, doi:10.1016/j.ijid.2021.06.048
Siedner, Ivermectin for the treatment of COVID-19 disease: Too good to pass up or too good to be true? Open, Forum Infect. Dis, doi:10.1093/ofid/ofab318
Tay, Fraser, Chan, Moreland, Rathore et al., Nuclear localization of dengue virus (DENV) 1-4 non-structural protein 5; protection against all 4 DENV serotypes by the inhibitor Ivermectin, Antivir. Res, doi:10.1016/j.antiviral.2013.06.002
Wagstaff, Sivakumaran, Heaton, Harrich, Jans, Ivermectin is a specific inhibitor of importin α/βmediated nuclear import able to inhibit replication of HIV-1 and dengue virus, Biochem. J, doi:10.1042/bj20120150
Wickham, Elegant Graphics for Data Analysis
Yan, Ci, Chen, Chen, Li et al., Anti-inflammatory effects of ivermectin in mouse model of allergic asthma, Inflamm. Res, doi:10.1007/s00011-011-0307-8
Zeidler, Karpinski, SARS-CoV, MERS-CoV, SARS-CoV-2 Comparison of Three Emerging Coronaviruses, Jundishapur J. Microbiol, doi:10.5812/jjm.103744
Zhang, Song, Ci, An, Ju et al., Ivermectin inhibits LPS-induced production of inflammatory cytokines and improves LPS-induced survival in mice, Inflamm. Res, doi:10.1007/s00011-008-8007-8
Loading..
Please send us corrections, updates, or comments. c19early involves the extraction of 100,000+ datapoints from thousands of papers. Community updates help ensure high accuracy. Treatments and other interventions are complementary. All practical, effective, and safe means should be used based on risk/benefit analysis. No treatment or intervention is 100% available and effective for all current and future variants. We do not provide medical advice. Before taking any medication, consult a qualified physician who can provide personalized advice and details of risks and benefits based on your medical history and situation. FLCCC and WCH provide treatment protocols.
  or use drag and drop   
Submit