COVID-19 and Thymoquinone: Clinical Benefits, Cure, and Challenges
Abstract
:1. Introduction
2. Methods
3. Possible Curative Efficacy of TQ for the Inflammation and Multiple Organ Failure Associated with COVID-19
4. Oxidative Stress Associated with COVID-19 and the Antioxidant Effect of TQ
5. Cardiopulmonary Protective Effect of TQ
6. Neuroprotective Effect of TQ can Overcome the Neurologic/Cognitive Manifestations Associated with COVID-19
7. Hepatorenal Protective Effect of TQ against COVID-19
8. Gastrointestinal Dysfunction Associated with COVID-19 and the Gastroprotective Effect of TQ
COVID-19 Complications | Thymoquinone | References |
---|---|---|
Inflammation and cytokine release syndrome (CRS) |
| [22,30,31,32] |
Oxidative damage |
| [39,40] |
Cardiac injury |
| [41,47,48,49] |
Pulmonary damage |
| [53,54,55] |
Neurological disease |
| [60,61,62] |
Liver injury |
| [67,68,69] |
Kidney injury |
| [72,74,75] |
GIT injury |
| [78] |
9. Thymoquinone Block SARS-CoV-2 Entry into Cells
10. Limitations of Clinical Application of TQ
11. New Insights into the Future of TQ Clinical Application for Treatment of COVID-19
12. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Ranjbar, M.; Rahimi, A.; Baghernejadan, Z.; Ghorbani, A.; Khorramdelazad, H. Role of CCL2/CCR2 axis in the pathogenesis of COVID-19 and possible Treatments: All options on the Table. Int. Immunopharmacol. 2022, 113, 109325. [Google Scholar] [CrossRef]
- Sharma, A.; Ahmad Farouk, I.; Lal, S.K. COVID-19: A Review on the Novel Coronavirus Disease Evolution, Transmission, Detection, Control and Prevention. Viruses 2021, 13, 202. [Google Scholar] [CrossRef]
- Baj, J.; Karakula-Juchnowicz, H.; Teresinski, G.; Buszewicz, G.; Ciesielka, M.; Sitarz, E.; Forma, A.; Karakula, K.; Flieger, W.; Portincasa, P.; et al. COVID-19: Specific and Non-Specific Clinical Manifestations and Symptoms: The Current State of Knowledge. J. Clin. Med. 2020, 9, 1753. [Google Scholar] [CrossRef]
- Gabutti, G.; d’Anchera, E.; Sandri, F.; Savio, M.; Stefanati, A. Coronavirus: Update related to the current outbreak of COVID-19. Infect. Dis. Ther. 2020, 9, 241–253. [Google Scholar] [CrossRef]
- Grant, M.C.; Geoghegan, L.; Arbyn, M.; Mohammed, Z.; McGuinness, L.; Clarke, E.L.; Wade, R.G. The prevalence of symptoms in 24,410 adults infected by the novel coronavirus (SARS-CoV-2; COVID-19): A systematic review and meta-analysis of 148 studies from 9 countries. PLoS ONE 2020, 15, e0234765. [Google Scholar] [CrossRef]
- Ismail, M.; Al-Naqeep, G.; Chan, K.W. Nigella sativa thymoquinone-rich fraction greatly improves plasma antioxidant capacity and expression of antioxidant genes in hypercholesterolemic rats. Free Radic. Biol. Med. 2010, 48, 664–672. [Google Scholar] [CrossRef]
- Chen, X.; Laurent, S.; Onur, O.A.; Kleineberg, N.N.; Fink, G.R.; Schweitzer, F.; Warnke, C. A systematic review of neurological symptoms and complications of COVID-19. J. Neurol. 2021, 268, 392–402. [Google Scholar] [CrossRef]
- Pennisi, M.; Lanza, G.; Falzone, L.; Fisicaro, F.; Ferri, R.; Bella, R. SARS-CoV-2 and the Nervous System: From Clinical Features to Molecular Mechanisms. Int. J. Mol. Sci. 2020, 21, 5475. [Google Scholar] [CrossRef]
- Wiersinga, W.J.; Rhodes, A.; Cheng, A.C.; Peacock, S.J.; Prescott, H.C. Pathophysiology, Transmission, Diagnosis, and Treatment of Coronavirus Disease 2019 (COVID-19): A Review. JAMA 2020, 324, 782–793. [Google Scholar] [CrossRef]
- El–Dakhakhny, M. Studies on the chemical constitution of Egyptian Nigella sativa L. seeds. Ii1) the essential oil. Planta Med. 1963, 11, 465–470. [Google Scholar] [CrossRef]
- Cobourne-Duval, M.K.; Taka, E.; Mendonca, P.; Bauer, D.; Soliman, K. The antioxidant effects of thymoquinone in activated BV-murine microglial cells. Neurochem. Res. 2017, 41, 3227–3238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oyero, O.G.; Toyama, M.; Mitsuhiro, N.; Onifade, A.A.; Hidaka, A.; Okamoto, M.; Baba, M. Selective Inhibition of Hepatitis C Virus Replication by Alpha-Zam, a Nigella Sativa Seed Formulation. Afr. J. Tradit. Complement. Altern. Med. 2016, 13, 144–148. [Google Scholar] [CrossRef]
- Umar, S.; Shah, M.A.A.; Munir, M.T.; Yaqoob, M.; Fiaz, M.; Anjum, S.; Kaboudi, K.; Bouzouaia, M.; Younus, M.; Nisa, Q.; et al. Synergistic effects of thymoquinone and curcumin on immune response and antiviral activity against avian influenza virus (H9N2) in turkeys. Poult. Sci. 2016, 95, 1513–1520. [Google Scholar] [CrossRef] [PubMed]
- Zihlif, M.A.; Mahmoud, I.S.; Ghanim, M.T.; Zreikat, M.S.; Alrabadi, N.; Imraish, A.; Odeh, F.; Abbas, M.A.; Ismail, S.I. Thymoquinone efficiently inhibits the survival of EBV-infected B cells and alters EBV gene expression. Integr. Cancer Ther. 2013, 12, 257–263. [Google Scholar] [CrossRef] [PubMed]
- Khader, M.; Eckl, P.M. Thymoquinone: An emerging natural drug with a wide range of medical applications. Iran. J. Basic Med. Sci. 2014, 17, 950–957. [Google Scholar] [PubMed]
- Back, D.; Marzolini, C.; Hodge, C.; Marra, F.; Boyle, A.; Gibbons, S.; Burger, D.; Khoo, S. COVID-19 treatment in patients with comorbidities: Awareness of drug-drug interactions. Br. J. Clin. Pharmacol. 2021, 87, 212–213. [Google Scholar] [CrossRef]
- Bersanelli, M. Controversies about COVID-19 and anticancer treatment with immune checkpoint inhibitors. Immunotherapy 2020, 12, 269–273. [Google Scholar] [CrossRef] [Green Version]
- Vivarelli, S.; Falzone, L.; Torino, F.; Scandurra, G.; Russo, G.; Bordonaro, R.; Pappalardo, F.; Spandidos, D.A.; Raciti, G.; Libra, M. Immune-checkpoint inhibitors from cancer to COVID19: A promising avenue for the treatment of patients with COVID19 (Review). Int. J. Oncol. 2021, 58, 145–157. [Google Scholar] [CrossRef]
- Romanzi, A.; Moroni, R.; Rongoni, E.; Scolaro, R.; La Regina, D.; Mongelli, F.; Putorti, A.; Rossi, F.; Zanardo, M.; Vannelli, A. The management of “fragile” and suspected COVID-19 surgical patients during pandemic: An Italian single-center experience. Minerva Chir. 2020, 75, 320–327. [Google Scholar] [CrossRef]
- Hannan, M.A.; Rahman, M.A.; Sohag, A.A.M.; Uddin, M.J.; Dash, R.; Sikder, M.H.; Rahman, M.S.; Timalsina, B.; Munni, Y.A.; Sarker, P.P.; et al. Black Cumin (Nigella sativa L.): A Comprehensive Review on Phytochemistry, Health Benefits, Molecular Pharmacology, and Safety. Nutrients 2021, 13, 1784. [Google Scholar] [CrossRef]
- Witika, B.A.; Makoni, P.A.; Mweetwa, L.L.; Ntemi, P.V.; Chikukwa, M.T.R.; Matafwali, S.K.; Mwila, C.; Mudenda, S.; Katandula, J.; Walker, R.B. Nano-Biomimetic Drug Delivery Vehicles: Potential Approaches for COVID-19 Treatment. Molecules 2020, 25, 5952. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Liao, R.; Qiang, Z.; Zhang, C. Pro-inflammatory cytokine-driven PI3K/Akt/Sp1 signalling and H2S production facilitates the pathogenesis of severe acute pancreatitis. Biosci. Rep. 2017, 37, BSR20160483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panigrahy, D.; Gilligan, M.M.; Huang, S.; Gartung, A.; Cortes-Puch, I.; Sime, P.J.; Phipps, R.P.; Serhan, C.N.; Hammock, B.D. Inflammation resolution: A dual-pronged approach to averting cytokine storms in COVID-19? Cancer Metastasis Rev. 2020, 39, 337–340. [Google Scholar] [CrossRef] [PubMed]
- Kell, D.B.; Heyden, E.L.; Pretorius, E. The Biology of Lactoferrin, an Iron-Binding Protein That Can Help Defend Against Viruses and Bacteria. Front. Immunol. 2020, 11, 1221. [Google Scholar] [CrossRef]
- Bunt, S.K.; Yang, L.; Sinha, P.; Clements, V.K.; Leips, J.; Ostrand-Rosenberg, S. Reduced inflammation in the tumor microenvironment delays the accumulation of myeloid-derived suppressor cells and limits tumor progression. Cancer Res. 2007, 67, 10019–10026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shah, A. Novel Coronavirus-Induced NLRP3 Inflammasome Activation: A Potential Drug Target in the Treatment of COVID-19. Front. Immunol. 2020, 11, 1021. [Google Scholar] [CrossRef]
- Barkat, M.A.; Ahmad, J.; Khan, M.A.; Beg, S.; Ahmad, F.J. Insights into the targeting potential of thymoquinone for therapeutic intervention against triple-negative breast cancer. Curr. Drug Targets 2018, 19, 70–80. [Google Scholar] [CrossRef]
- Gibellini, L.; De Biasi, S.; Paolini, A.; Borella, R.; Boraldi, F.; Mattioli, M.; Lo Tartaro, D.; Fidanza, L.; Caro-Maldonado, A.; Meschiari, M.; et al. Altered bioenergetics and mitochondrial dysfunction of monocytes in patients with COVID-19 pneumonia. EMBO Mol. Med. 2020, 12, e13001. [Google Scholar] [CrossRef]
- Yang, L.; Nilsson-Payant, B.E.; Han, Y.; Jaffré, F.; Zhu, J.; Wang, P.; Zhang, T.; Redmond, D.; Houghton, S.; Møller, R.; et al. Cardiomyocytes recruit monocytes upon SARS-CoV-2 infection by secreting CCL2. Stem Cell Rep. 2021, 16, 2274–2288. [Google Scholar] [CrossRef]
- Arjumand, S.; Shahzad, M.; Shabbir, A.; Yousaf, M.Z. Thymoquinone attenuates rheumatoid arthritis by downregulating TLR2, TLR4, TNF-alpha, IL-1, and NFkappaB expression levels. Biomed. Pharmacother. 2019, 111, 958–963. [Google Scholar] [CrossRef]
- Houghton, P.J.; Zarka, R.; de las Heras, B.; Hoult, J.R. Fixed oil of Nigella sativa and derived thymoquinone inhibit eicosanoid generation in leukocytes and membrane lipid peroxidation. Planta Med. 1995, 61, 33–36. [Google Scholar] [CrossRef] [PubMed]
- Koshak, A.E.; Koshak, E.A.; Mobeireek, A.F.; Badawi, M.A.; Wali, S.O.; Malibary, H.M.; Atwah, A.F.; Alhamdan, M.M.; Almalki, R.A.; Madani, T.A. Nigella sativa for the treatment of COVID-19: An open-label randomized controlled clinical trial. Complement. Ther. Med. 2021, 61, 102769. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Chen, Z. The pathophysiological role of mitochondrial oxidative stress in lung diseases. J. Transl. Med. 2017, 15, 207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jean, S.S.; Lee, P.I.; Hsueh, P.R. Treatment options for COVID-19: The reality and challenges. J. Microbiol. Immunol. Infect. 2020, 53, 436–443. [Google Scholar] [CrossRef]
- Olsson, M.; Zhivotovsky, B. Caspases and cancer. Cell Death Differ. 2011, 18, 1441–1449. [Google Scholar] [CrossRef] [Green Version]
- Khan, M.A.; Tania, M.; Wei, C.; Mei, Z.; Fu, S.; Cheng, J.; Xu, J.; Fu, J. Thymoquinone inhibits cancer metastasis by downregulating TWIST1 expression to reduce epithelial to mesenchymal transition. Oncotarget 2015, 6, 19580–19591. [Google Scholar] [CrossRef] [Green Version]
- Elbarbry, F.; Ragheb, A.; Marfleet, T.; Shoker, A. Modulation of hepatic drug metabolizing enzymes by dietary doses of thymoquinone in female New Zealand White rabbits. Phytother. Res. 2012, 26, 1726–1730. [Google Scholar] [CrossRef]
- Periyanayagam, S.; Arumugam, G.; Ravikumar, A.; Ganesan, V.S. Thymoquinone ameliorates NLRP3-mediated inflammation in the pancreas of albino Wistar rats fed ethanol and high-fat diet. J. Basic Clin. Physiol. Pharmacol. 2015, 26, 623–632. [Google Scholar] [CrossRef]
- Holgersen, E.M.; Gandhi, S.; Zhou, Y.; Kim, J.; Vaz, B.; Bogojeski, J.; Bugno, M.; Shalev, Z.; Cheung-Ong, K.; Goncalves, J.; et al. Transcriptome-Wide Off-Target Effects of Steric-Blocking Oligonucleotides. Nucleic Acid Ther. 2021, 31, 392–403. [Google Scholar] [CrossRef]
- Goncalves, T.L.; Erthal, F.; Corte, C.L.; Muller, L.G.; Piovezan, C.M.; Nogueira, C.W.; Rocha, J.B. Involvement of oxidative stress in the pre-malignant and malignant states of cervical cancer in women. Clin. Biochem. 2005, 38, 1071–1075. [Google Scholar] [CrossRef]
- Guo, T.; Fan, Y.; Chen, M.; Wu, X.; Zhang, L.; He, T.; Wang, H.; Wan, J.; Wang, X.; Lu, Z. Cardiovascular Implications of Fatal Outcomes of Patients With Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020, 5, 811–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parvu, S.; Müller, K.; Dahdal, D.; Cosmin, I.; Christodorescu, R.; Duda-Seiman, D.; Man, D.; Sharma, A.; Dragoi, R.; Baneu, P.; et al. COVID-19 and cardiovascular manifestations. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 4509–4519. [Google Scholar] [CrossRef] [PubMed]
- Lindner, D.; Fitzek, A.; Bräuninger, H.; Aleshcheva, G.; Edler, C.; Meissner, K.; Scherschel, K.; Kirchhof, P.; Escher, F.; Schultheiss, H.P.; et al. Association of Cardiac Infection With SARS-CoV-2 in Confirmed COVID-19 Autopsy Cases. JAMA Cardiol. 2020, 5, 1281–1285. [Google Scholar] [CrossRef] [PubMed]
- Shi, S.; Qin, M.; Shen, B.; Cai, Y.; Liu, T.; Yang, F.; Gong, W.; Liu, X.; Liang, J.; Zhao, Q.; et al. Association of Cardiac Injury With Mortality in Hospitalized Patients With COVID-19 in Wuhan, China. JAMA Cardiol. 2020, 5, 802–810. [Google Scholar] [CrossRef] [Green Version]
- Shah, K.S.; Yang, E.H.; Maisel, A.S.; Fonarow, G.C. The Role of Biomarkers in Detection of Cardio-toxicity. Curr. Oncol. Rep. 2017, 19, 42. [Google Scholar] [CrossRef] [Green Version]
- Zilinyi, R.; Czompa, A.; Czegledi, A.; Gajtko, A.; Pituk, D.; Lekli, I.; Tosaki, A. The Cardioprotective Effect of Metformin in Doxorubicin-Induced Cardiotoxicity: The Role of Autophagy. Molecules 2018, 23, 1184. [Google Scholar] [CrossRef] [Green Version]
- Su, Y.; Qu, Y.; Zhao, F.; Li, H.; Mu, D.; Li, X. Regulation of autophagy by the nuclear factor kappaB signaling pathway in the hippocampus of rats with sepsis. J. Neuroinflammation 2015, 12, 116. [Google Scholar] [CrossRef] [Green Version]
- Ojha, S.; Azimullah, S.; Mohanraj, R.; Sharma, C.; Yasin, J.; Arya, D.S.; Adem, A. Thymoquinone Protects against Myocardial Ischemic Injury by Mitigating Oxidative Stress and Inflammation. Evid. Based Complement. Altern. Med. 2015, 2015, 143629. [Google Scholar] [CrossRef] [Green Version]
- Khan, M.A.; Younus, H. Potential Implications of Black Seed and its Principal Constituent Thymoquinone in the Treatment of COVID-19 Patients. Curr. Pharm. Biotechnol. 2021, 22, 1315–1324. [Google Scholar] [CrossRef]
- Singhal, T. A Review of Coronavirus Disease-2019 (COVID-19). Indian J. Pediatr. 2020, 87, 281–286. [Google Scholar] [CrossRef]
- Kanter, M. Thymoquinone attenuates lung injury induced by chronic toluene exposure in rats. Toxicol. Ind. Health 2011, 27, 387–395. [Google Scholar] [CrossRef] [PubMed]
- Pourgholamhossein, F.; Sharififar, F.; Rasooli, R.; Pourgholi, L.; Nakhaeipour, F.; Samareh-Fekri, H.; Iranpour, M.; Mandegary, A. Thymoquinone effectively alleviates lung fibrosis induced by paraquat herbicide through down-regulation of pro-fibrotic genes and inhibition of oxidative stress. Environ. Toxicol. Pharmacol. 2016, 45, 340–345. [Google Scholar] [CrossRef] [PubMed]
- Colak, M.; Kalemci, S.; Alpaydin, A.O.; Karacam, V.; Meteoglu, I.; Yilmaz, O.; Itil, B.O. Efficacy of thymoquinone in the treatment of experimental lipopolysaccharide-induced acute lung injury. Kardiochirurgia Torakochirurgia Pol. 2020, 17, 65–69. [Google Scholar] [CrossRef]
- Boskabady, M.; Khazdair, M.R.; Bargi, R.; Saadat, S.; Memarzia, A.; Mohammadian Roshan, N.; Hosseini, M.; Askari, V.R.; Boskabady, M.H. Thymoquinone Ameliorates Lung Inflammation and Pathological Changes Observed in Lipopolysaccharide-Induced Lung Injury. Evid. Based Complement. Altern. Med. 2021, 2021, 6681729. [Google Scholar] [CrossRef] [PubMed]
- Khazdair, M.R.; Ghafari, S.; Sadeghi, M. Possible therapeutic effects of Nigella sativa and its thymoquinone on COVID-19. Pharm. Biol. 2021, 59, 696–703. [Google Scholar] [CrossRef]
- Lu, Y.; Li, X.; Geng, D.; Mei, N.; Wu, P.Y.; Huang, C.C.; Jia, T.; Zhao, Y.; Wang, D.; Xiao, A.; et al. Cerebral Micro-Structural Changes in COVID-19 Patients—An MRI-based 3-month Follow-up Study. EClinicalMedicine 2020, 25, 100484. [Google Scholar] [CrossRef] [PubMed]
- Filatov, A.; Sharma, P.; Hindi, F.; Espinosa, P.S. Neurological Complications of Coronavirus Disease (COVID-19): Encephalopathy. Cureus 2020, 12, e7352. [Google Scholar] [CrossRef] [Green Version]
- Farkhondeh, T.; Samarghandian, S.; Azimin-Nezhad, M.; Samini, F. Effect of chrysin on nociception in formalin test and serum levels of noradrenalin and corticosterone in rats. Int. J. Clin. Exp. Med. 2015, 8, 2465–2470. [Google Scholar]
- Cárdenas-Rodríguez, N.; Bandala, C.; Vanoye-Carlo, A.; Ignacio-Mejía, I.; Gómez-Manzo, S.; Hernández-Cruz, E.Y.; Pedraza-Chaverri, J.; Carmona-Aparicio, L.; Hernández-Ochoa, B. Use of Antioxidants for the Neuro-Therapeutic Management of COVID-19. Antioxidants 2021, 10, 971. [Google Scholar] [CrossRef]
- Wang, Y.; Gao, H.; Zhang, W.; Zhang, W.; Fang, L. Thymoquinone inhibits lipopolysaccharide-induced inflammatory mediators in BV2 microglial cells. Int. Immunopharmacol. 2015, 26, 169–173. [Google Scholar] [CrossRef]
- Mosley, R.L.; Benner, E.J.; Kadiu, I.; Thomas, M.; Boska, M.D.; Hasan, K.; Laurie, C.; Gendelman, H.E. Neuroinflammation, Oxidative Stress and the Pathogenesis of Parkinson’s Disease. Clin. Neurosci. Res. 2006, 6, 261–281. [Google Scholar] [CrossRef]
- Alhebshi, A.H.; Gotoh, M.; Suzuki, I. Thymoquinone protects cultured rat primary neurons against amyloid beta-induced neurotoxicity. Biochem. Biophys. Res. Commun. 2013, 433, 362–367. [Google Scholar] [CrossRef] [PubMed]
- Parohan, M.; Yaghoubi, S.; Seraji, A. Liver injury is associated with severe coronavirus disease 2019 (COVID-19) infection:A systematic review and meta-analysis of retrospective studies. Hepatol. Res. 2020, 50, 924–935. [Google Scholar] [PubMed]
- Abd El Rhman, M.M.; Shafik, N.S.; Maher, A.; Hemdan, S.B.; Abd Elhamed, R.M. Liver Injuries in COVID-19 infected patients. Sohag Med. J. 2019, 12, 223–230. [Google Scholar]
- Maizels, R.M.; McSorley, H.J. Regulation of the host immune system by helminth parasites. J. Allergy Clin. Immunol. 2016, 138, 666–675. [Google Scholar] [CrossRef] [Green Version]
- Nili-Ahmadabadi, A.; Tavakoli, F.; Hasanzadeh, G.; Rahimi, H.; Sabzevari, O. Protective effect of pretreatment with thymoquinone against Aflatoxin B(1) induced liver toxicity in mice. Daru 2011, 19, 282–287. [Google Scholar]
- Bai, T.; Yang, Y.; Wu, Y.L.; Jiang, S.; Lee, J.J.; Lian, L.H.; Nan, J.X. Thymoquinone alleviates thioacetamide-induced hepatic fibrosis and inflammation by activating LKB1-AMPK signaling pathway in mice. Int. Immunopharmacol. 2014, 19, 351–357. [Google Scholar] [CrossRef]
- Raghunandhakumar, S.; Paramasivam, A.; Senthilraja, S.; Naveenkumar, C.; Asokkumar, S.; Binuclara, J.; Jagan, S.; Anandakumar, P.; Devaki, T. Thymoquinone inhibits cell proliferation through regulation of G1/S phase cell cycle transition in N-nitrosodiethylamine-induced experimental rat hepatocellular carcinoma. Toxicol. Lett. 2013, 223, 60–72. [Google Scholar] [CrossRef]
- Nagi, M.N.; Almakki, H.A. Thymoquinone supplementation induces quinone reductase and glutathione transferase in mice liver: Possible role in protection against chemical carcinogenesis and toxicity. Phytother. Res. 2009, 23, 1295–1298. [Google Scholar] [CrossRef]
- Gabarre, P.; Dumas, G.; Dupont, T.; Darmon, M.; Azoulay, E.; Zafrani, L. Acute kidney injury in critically ill patients with COVID-19. Intensive Care Med. 2020, 46, 1339–1348. [Google Scholar] [CrossRef]
- Li, A.; Zhang, W.; Zhang, L.; Liu, Y.; Li, K.; Du, G.; Qin, X. Elucidating the time-dependent changes in the urinary metabolome under doxorubicin-induced nephrotoxicity. Toxicol. Lett. 2020, 319, 204–212. [Google Scholar] [CrossRef]
- Fouda, A.M.; Daba, M.H.; Dahab, G.M.; Sharaf El-Din, O.A. Thymoquinone ameliorates renal oxidative damage and proliferative response induced by mercuric chloride in rats. Basic Clin. Pharmacol. Toxicol. 2008, 103, 109–118. [Google Scholar] [CrossRef] [PubMed]
- Sayed-Ahmed, M.M.; Nagi, M.N. Thymoquinone supplementation prevents the development of gentamicin-induced acute renal toxicity in rats. Clin. Exp. Pharmacol. Physiol. 2007, 34, 399–405. [Google Scholar] [CrossRef] [PubMed]
- Guo, L.P.; Liu, S.X.; Yang, Q.; Liu, H.Y.; Xu, L.L.; Hao, Y.H.; Zhang, X.Q. Effect of Thymoquinone on Acute Kidney Injury Induced by Sepsis in BALB/c Mice. Biomed Res. Int. 2020, 2020, 1594726. [Google Scholar] [CrossRef] [PubMed]
- Ince, S.; Kucukkurt, I.; Demirel, H.H.; Turkmen, R.; Sever, E. Thymoquinone attenuates cypermethrin induced oxidative stress in Swiss albino mice. Pestic. Biochem. Physiol. 2012, 104, 229–235. [Google Scholar] [CrossRef]
- Su, S.; Shen, J.; Zhu, L.; Qiu, Y.; He, J.S.; Tan, J.Y.; Iacucci, M.; Ng, S.C.; Ghosh, S.; Mao, R.; et al. Involvement of digestive system in COVID-19: Manifestations, pathology, management and challenges. Ther. Adv. Gastroenterol. 2020, 13, 1756284820934626. [Google Scholar] [CrossRef] [PubMed]
- Randhawa, M.A.; Alenazy, A.K.; Alrowaili, M.G.; Basha, J. An active principle of Nigella sativa L., thymoquinone, showing significant antimicrobial activity against anaerobic bacteria. J. Intercult. Ethnopharmacol. 2017, 6, 97–101. [Google Scholar] [CrossRef]
- Goel, S.; Mishra, P. Thymoquinone inhibits biofilm formation and has selective antibacterial activity due to ROS generation. Appl. Microbiol. Biotechnol. 2018, 102, 1955–1967. [Google Scholar] [CrossRef]
- Ali, S.; Alam, M.; Khatoon, F.; Fatima, U.; Elasbali, A.M.; Adnan, M.; Islam, A.; Hassan, M.I.; Snoussi, M.; De Feo, V. Natural products can be used in therapeutic management of COVID-19: Probable mechanistic insights. Biomed. Pharmacother. 2022, 147, 112658. [Google Scholar] [CrossRef]
- Sommer, A.P.; Försterling, H.D.; Naber, K.G. Thymoquinone: Shield and sword against SARS-CoV-2. Precis. Nanomed. 2020, 3, 541–548. [Google Scholar] [CrossRef]
- Amin, B.; Hosseinzadeh, H. Black Cumin (Nigella sativa) and Its Active Constituent, Thymoquinone: An Overview on the Analgesic and Anti-inflammatory Effects. Planta Med. 2016, 82, 8–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salata, C.; Calistri, A.; Parolin, C.; Baritussio, A.; Palu, G. Antiviral activity of cationic amphiphilic drugs. Expert Rev. Anti Infect. Ther. 2017, 15, 483–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Omar, S.; Bouziane, I.; Bouslama, Z.; Djemel, A. In-silico identification of potent inhibitors of COVID-19 main protease (Mpro) and Angiotensin converting enzyme 2 (ACE2) from natural products: Quercetin, Hispidulin, and Cirsimaritin exhibited better potential inhibition than Hydroxy- Chloroquine against COVID-19 main protease active site and ACE2. Biol. Med. Chem. 2020, 5, 000189. [Google Scholar]
- Elfiky, A.A. Natural products may interfere with SARS-CoV-2 attachment to the host cell. J. Biomol. Struct. Dyn. 2020, 3, 110–117. [Google Scholar] [CrossRef] [PubMed]
- Badary, O.A.; Hamza, M.S.; Tikamdas, R. Thymoquinone: A Promising Natural Compound with Potential Benefits for COVID-19 Prevention and Cure. Drug Des. Dev. Ther. 2021, 15, 1819–1833. [Google Scholar] [CrossRef]
- El-Najjar, N.; Chatila, M.; Moukadem, H.; Vuorela, H.; Ocker, M.; Gandesiri, M.; Schneider-Stock, R.; Gali-Muhtasib, H. Reactive oxygen species mediate thymoquinone-induced apoptosis and activate ERK and JNK signaling. Apoptosis 2010, 15, 183–195. [Google Scholar] [CrossRef]
- Mashayekhi-Sardoo, H.; Rezaee, R.; Karimi, G. An overview of in vivo toxicological profile of Thymoquinone. Toxin Rev. 2018, 39, 115–122. [Google Scholar] [CrossRef]
- Khader, M.; Bresgen, N.; Eckl, P.M. In vitro toxicological properties of thymoquinone. Food Chem. Toxicol. 2009, 47, 129–133. [Google Scholar] [CrossRef]
- Badary, O.A.; Al-Shabanah, O.A.; Nagi, M.N.; Al-Bekairi, A.M.; Elmazar, M. Acute and subchronic toxicity of thymoquinone in mice. Drug Dev. Res. 1998, 44, 56–61. [Google Scholar] [CrossRef]
- Al-Ali, A.; Alkhawajah, A.A.; Randhawa, M.A.; Shaikh, N.A. Oral and intraperitoneal LD50 of thymoquinone, an active principle of Nigella sativa, in mice and rats. J. Ayub Med. Coll. Abbottabad 2008, 20, 25–27. [Google Scholar]
- Mansour, M.A.; Ginawi, O.T.; El-Hadiyah, T.; El-Khatib, A.S.; Al-Shabanah, O.A.; Al-Sawaf, H.A. Effects of volatile oil constituents of Nigella sativa on carbon tetrachloride-induced hepatotoxicity in mice: Evidence for antioxidant effects of thymoquinone. Res. Commun. Mol. Pathol. Pharmacol. 2001, 110, 239–251. [Google Scholar] [PubMed]
- Harzallah, H.J.; Grayaa, R.; Kharoubi, W.; Maaloul, A.; Hammami, M.; Mahjoub, T. Thymoquinone, the Nigella sativa bioactive compound, prevents circulatory oxidative stress caused by 1,2-dimethylhydrazine in erythrocyte during colon postinitiation carcinogenesis. Oxid. Med. Cell. Longev. 2012, 2012, 854065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- El-Sayed, S.A.E.; Rizk, M.A.; Yokoyama, N.; Igarashi, I. Evaluation of the in vitro and in vivo inhibitory effect of thymoquinone on piroplasm parasites. Parasit. Vectors 2019, 12, 37. [Google Scholar] [CrossRef] [PubMed]
- Goyal, S.N.; Prajapati, C.P.; Gore, P.R.; Patil, C.R.; Mahajan, U.B.; Sharma, C.; Talla, S.P.; Ojha, S.K. Therapeutic Potential and Pharmaceutical Development of Thymoquinone: A Multitargeted Molecule of Natural Origin. Front. Pharmacol. 2017, 8, 656. [Google Scholar] [CrossRef]
- Salmani, J.M.; Asghar, S.; Lv, H.; Zhou, J. Aqueous solubility and degradation kinetics of the phytochemical anticancer thymoquinone; probing the effects of solvents, pH and light. Molecules 2014, 19, 5925–5939. [Google Scholar] [CrossRef]
- Tubesha, Z.; Abu Bakar, Z.; Ismail, M. Characterization and stability evaluation of thymoquinone nanoemulsions prepared by high-pressure homogenization. J. Nanomater. 2013, 2013, 453290. [Google Scholar] [CrossRef] [Green Version]
- El-Far, A.H.; Al Jaouni, S.K.; Li, W.; Mousa, S.A. Protective Roles of Thymoquinone Nanoformulations: Potential Nanonutraceuticals in Human Diseases. Nutrients 2018, 10, 1369. [Google Scholar] [CrossRef] [Green Version]
- Ballout, F.; Habli, Z.; Rahal, O.N.; Fatfat, M.; Gali-Muhtasib, H. Thymoquinone-based nanotechnology for cancer therapy: Promises and challenges. Drug Discov. Today 2018, 23, 1089–1098. [Google Scholar] [CrossRef]
- Ong, Y.S.; Yazan, L.S.; Ng, W.K.; Noordin, M.M.; Sapuan, S.; Foo, J.B.; Tor, Y.S. Acute and subacute toxicity profiles of thymoquinone-loaded nanostructured lipid carrier in BALB/c mice. Int. J. Nanomed. 2016, 11, 5905–5915. [Google Scholar] [CrossRef] [Green Version]
- Ng, W.K.; Saiful Yazan, L.; Yap, L.H.; Wan Nor Hafiza, W.A.G.; How, C.W.; Abdullah, R. Thymoquinone-loaded nanostructured lipid carrier exhibited cytotoxicity towards breast cancer cell lines (MDA-MB-231 and MCF-7) and cervical cancer cell lines (HeLa and SiHa). BioMed Res. Int. 2015, 2015, 263131. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharya, S.; Ahir, M.; Patra, P.; Mukherjee, S.; Ghosh, S.; Mazumdar, M.; Chattopadhyay, S.; Das, T.; Chattopadhyay, D.; Adhikary, A. PEGylated-thymoquinone-nanoparticle mediated retardation of breast cancer cell migration by deregulation of cytoskeletal actin polymerization through miR-34a. Biomaterials 2015, 51, 91–107. [Google Scholar] [CrossRef] [PubMed]
- Ozer, E.K.; Goktas, M.T.; Toker, A.; Pehlivan, S.; Bariskaner, H.; Ugurluoglu, C.; Iskit, A.B. Thymoquinone protects against the sepsis induced mortality, mesenteric hypoperfusion, aortic dysfunction and multiple organ damage in rats. Pharmacol. Rep. 2017, 69, 683–690. [Google Scholar] [CrossRef] [PubMed]
- Shaarani, S.; Hamid, S.S.; Mohd Kaus, N.H. The Influence of Pluronic F68 and F127 Nanocarrier on Physicochemical Properties, In vitro Release, and Antiproliferative Activity of Thymoquinone Drug. Pharmacogn. Res. 2017, 9, 12–20. [Google Scholar] [CrossRef] [Green Version]
- Fakhria, A.; Gilani, S.J.; Imam, S.S. Formulation of thymoquinone loaded chitosan nano vesicles: In-vitro evaluation and in-vivo anti-hyperlipidemic assessment. J. Drug Deliv. Sci. Technol. 2019, 50, 339–346. [Google Scholar] [CrossRef]
- Mohammadabadi, M.R.; Mozafari, M.R. Enhanced efficacy and bioavailability of thymo- quinone using nanoliposomal dosage form. J. Drug Deliv. Sci. Technol. 2018, 47, 445–453. [Google Scholar] [CrossRef]
- Ramachandran, S.; Thangarajan, S. A novel therapeutic application of solid lipid nanoparticles encapsulated thymoquinone (TQ-SLNs) on 3-nitroproponic acid induced Huntington’s disease-like symptoms in wistar rats. Chem. Biol. Interact. 2016, 256, 25–36. [Google Scholar] [CrossRef]
- Tubesha, Z.; Imam, M.U.; Mahmud, R.; Ismail, M. Study on the potential toxicity of a thymoquinone-rich fraction nanoemulsion in Sprague Dawley rats. Molecules 2013, 18, 7460–7472. [Google Scholar] [CrossRef] [Green Version]
- Velho-Pereira, R.; Japtap, A.; Elaridi, J. Diodistribution and scintigraphic evaluation of microemulsion formulations of technetium-99m-radiolabeled-thymoquinone. J. Chem. Pharm. Res. 2017, 9, 188–198. [Google Scholar]
- Fahmy, H.M.; Fathy, M.M.; Abd-Elbadia, R.A.; Elshemey, W.M. Targeting of Thymoquinone-loaded mesoporous silica nanoparticles to different brain areas: In vivo study. Life Sci. 2019, 222, 94–102. [Google Scholar] [CrossRef]
- Hamed, S.F.; Shaaban, H.A.; Ramadan, A.A.; Edris, A.E. Potentials of enhancing the physico-chemical and functional characteristics of Nigella sativa oil by using the screw pressing technique for extraction. Grasas Y Aceites 2017, 68, e188–e196. [Google Scholar] [CrossRef] [Green Version]
- El-Tahir, K.A.-A.; Al-Bekairi, M. Some cardiovascular effects of the dethymoquinonated Nigella sativa volatile oil and its major components α-pinene and p-cymene in rats. Saudi Pharm. J. 2003, 11, 104–110. [Google Scholar]
- Edris, A. Isolation of Pure Thymoquinone Crystals from the Volatile Oil Fraction of Nigella sativa Using Nanometric Method; Pending Patent No. 2017/1753; Academy of Scientific Research and Technology, Assigned to the National Research Center: Cairo, Egypt, 2017. [Google Scholar]
- Dockal, E.R.; Cass, Q.B.; Brocksom, T.J.; Brocksom, U.; Corrěa, A.G. A simple and efficient synthesis of thymoquinone and methyl P-benzoquinone. Synth. Commun. 1985, 15, 1033–1036. [Google Scholar] [CrossRef]
- Johnson-Ajinwo, O.R.; Ullah, I.; Mbye, H.; Richardson, A.; Horrocks, P.; Li, W.W. The synthesis and evaluation of thymoquinone analogs as anti-ovarian cancer and antimalarial agents. Bioorganic Med. Chem. Lett. 2018, 28, 1219–1222. [Google Scholar] [CrossRef] [PubMed]
- Frohlich, T.; Reiter, C.; Saeed, M.E.M.; Hutterer, C.; Hahn, F.; Leidenberger, M.; Friedrich, O.; Kappes, B.; Marschall, M.; Efferth, T.; et al. Synthesis of Thymoquinone-Artemisinin Hybrids: New Potent Antileukemia, Antiviral, and Antimalarial Agents. ACS Med. Chem. Lett. 2018, 9, 534–539. [Google Scholar] [CrossRef]
- Frohlich, T.; Ndreshkjana, B.; Muenzner, J.K.; Reiter, C.; Hofmeister, E.; Mederer, S.; Fatfat, M.; El-Baba, C.; Gali-Muhtasib, H.; Schneider-Stock, R.; et al. Synthesis of Novel Hybrids of Thymoquinone and Artemisinin with High Activity and Selectivity Against Colon Cancer. ChemMedChem 2017, 12, 226–234. [Google Scholar] [CrossRef]
- Yusufi, M.; Banerjee, S.; Mohammad, M.; Khatal, S.; Venkateswara Swamy, K.; Khan, E.M.; Aboukameel, A.; Sarkar, F.H.; Padhye, S. Synthesis, characterization and anti-tumor activity of novel thymoquinone analogs against pancreatic cancer. Bioorg. Med. Chem. Lett. 2013, 23, 3101–3104. [Google Scholar] [CrossRef]
- Ahmad, A.; Husain, A.; Mujeeb, M.; Khan, S.A.; Najmi, A.K.; Siddique, N.A.; Damanhouri, Z.A.; Anwar, F. A review on therapeutic potential of Nigella sativa: A miracle herb. Asian Pac. J. Trop. Biomed. 2013, 3, 337–352. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
El-Sayed, S.A.E.-S.; Rizk, M.A. COVID-19 and Thymoquinone: Clinical Benefits, Cure, and Challenges. BioMed 2023, 3, 59-76. https://doi.org/10.3390/biomed3010005
El-Sayed SAE-S, Rizk MA. COVID-19 and Thymoquinone: Clinical Benefits, Cure, and Challenges. BioMed. 2023; 3(1):59-76. https://doi.org/10.3390/biomed3010005
Chicago/Turabian StyleEl-Sayed, Shimaa Abd El-Salam, and Mohamed Abdo Rizk. 2023. "COVID-19 and Thymoquinone: Clinical Benefits, Cure, and Challenges" BioMed 3, no. 1: 59-76. https://doi.org/10.3390/biomed3010005
APA StyleEl-Sayed, S. A. E. -S., & Rizk, M. A. (2023). COVID-19 and Thymoquinone: Clinical Benefits, Cure, and Challenges. BioMed, 3(1), 59-76. https://doi.org/10.3390/biomed3010005