COVID-19-Associated Sepsis: Potential Role of Phytochemicals as Functional Foods and Nutraceuticals
Abstract
:1. Introduction
2. Pathogenesis of COVID-19-Associated Sepsis
3. Current Treatment Approaches for COVID-19-Associated Sepsis and the Challenges
4. Importance of Phytochemicals as Functional Foods and Nutraceuticals
5. Importance of Phytochemicals as Potential Agents for COVID-19-Associated Sepsis
5.1. Quercetin
5.2. Curcumin
5.3. Luteolin
5.4. Apigenin
5.5. Resveratrol
5.6. Naringenin
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Cascella, M.; Rajnik, M.; Aleem, A.; Dulebohn, S.C.; Di Napoli, R. Features, Evaluation, and Treatment of Coronavirus (COVID-19). In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
- Maranatha, D.; Rahardjo, P.; Lusman, R. Evolution of chest CT scan manifestations in a patient recovered from COVID-19 severe pneumonia with acute respiratory distress syndrome. Respir. Med. Case Rep. 2021, 32, 101342. [Google Scholar] [CrossRef] [PubMed]
- Baghela, A.; An, A.; Zhang, P.; Acton, E.; Gauthier, J.; Brunet-Ratnasingham, E.; Blimkie, T.; Freue, G.C.; Kaufmann, D.; Lee, A.H.Y.; et al. Predicting severity in COVID-19 disease using sepsis blood gene expression signatures. Sci. Rep. 2023, 13, 1247. [Google Scholar] [CrossRef]
- Sulzbacher, M.M.; Sulzbacher, L.M.; Passos, F.R.; Bilibio, B.L.E.; Althaus, W.F.; Weizenmann, L.; de Oliveira, K.; Frizzo, M.N.; Ludwig, M.S.; Heck, T.G. A single dose of eHSP72 attenuates sepsis severity in mice. Sci. Rep. 2020, 10, 9198. [Google Scholar] [CrossRef]
- Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef] [PubMed]
- Zhu, J.; Bouzid, R.; Travert, B.; Geri, G.; Cohen, Y.; Picod, A.; Heming, N.; Rottman, M.; Joly-Laffargue, B.; Veyradier, A.; et al. Combined coagulation and inflammation markers as predictors of venous thrombo-embolism and death in COVID-19. Front. Med. 2024, 11, 1399335. [Google Scholar] [CrossRef]
- Ahmad, I.; Hamidullah. Efficacy of sarilumab and dexamethasone co-administration for lowering multiple blood biomarkers in the treatment of cytokine release syndrome in hospitalized COVID-19 patients. J. Pak. Med. Assoc. 2024, 74, 1345–1350. [Google Scholar] [CrossRef]
- Olwal, C.O.; Nganyewo, N.N.; Tapela, K.; Djomkam Zune, A.L.; Owoicho, O.; Bediako, Y.; Duodu, S. Parallels in Sepsis and COVID-19 Conditions: Implications for Managing Severe COVID-19. Front. Immunol. 2021, 12, 602848. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.; Huang, Y.; Han, N.; He, F.; Li, M.; Bian, Z.; Liu, J.; Sun, T.; Zhu, L. Quercetin suppresses NLRP3 inflammasome activation and attenuates histopathology in a rat model of spinal cord injury. Spinal Cord. 2016, 54, 592–596. [Google Scholar] [CrossRef]
- Wang, W.; Wang, C.; Ding, X.Q.; Pan, Y.; Gu, T.T.; Wang, M.X.; Liu, Y.L.; Wang, F.M.; Wang, S.J.; Kong, L.D. Quercetin and allopurinol reduce liver thioredoxin-interacting protein to alleviate inflammation and lipid accumulation in diabetic rats. Br. J. Pharmacol. 2013, 169, 1352–1371. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Zhang, J.; Ungvari, Z.; Zhang, C. Resveratrol improves endothelial function: Role of TNFalpha and vascular oxidative stress. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 1164–1171. [Google Scholar] [CrossRef]
- Zhang, H.; Morgan, B.; Potter, B.J.; Ma, L.; Dellsperger, K.C.; Ungvari, Z.; Zhang, C. Resveratrol improves left ventricular diastolic relaxation in type 2 diabetes by inhibiting oxidative/nitrative stress: In vivo demonstration with magnetic resonance imaging. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H985–H994. [Google Scholar] [CrossRef] [PubMed]
- Shahid, H.; Shahzad, M.; Shabbir, A.; Saghir, G. Immunomodulatory and Anti-Inflammatory Potential of Curcumin for the Treatment of Allergic Asthma: Effects on Expression Levels of Pro-inflammatory Cytokines and Aquaporins. Inflammation 2019, 42, 2037–2047. [Google Scholar] [CrossRef] [PubMed]
- Attallah, N.G.M.; El-Kadem, A.H.; Negm, W.A.; Elekhnawy, E.; El-Masry, T.A.; Elmongy, E.I.; Altwaijry, N.; Alanazi, A.S.; Al-Hamoud, G.A.; Ragab, A.E. Promising Antiviral Activity of Agrimonia pilosa Phytochemicals against Severe Acute Respiratory Syndrome Coronavirus 2 Supported with In Vivo Mice Study. Pharmaceuticals 2021, 14, 1313. [Google Scholar] [CrossRef] [PubMed]
- Kocak Tufan, Z.; Kayaaslan, B.; Mer, M. COVID-19 and Sepsis. Turk. J. Med. Sci. 2021, 51 (Suppl. S1), 3301–3311. [Google Scholar] [CrossRef] [PubMed]
- Chang, J.C. COVID-19 Sepsis: Pathogenesis and Endothelial Molecular Mechanisms Based on “Two-Path Unifying Theory” of Hemostasis and Endotheliopathy-Associated Vascular Microthrombotic Disease, and Proposed Therapeutic Approach with Antimicrothrombotic Therapy. Vasc. Health Risk Manag. 2021, 17, 273–298. [Google Scholar] [CrossRef] [PubMed]
- Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Jackson, C.B.; Mou, H.; Ojha, A.; Peng, H.; Quinlan, B.D.; Rangarajan, E.S.; Pan, A.; Vanderheiden, A.; Suthar, M.S.; et al. SARS-CoV-2 spike-protein D614G mutation increases virion spike density and infectivity. Nat. Commun. 2020, 11, 6013. [Google Scholar] [CrossRef] [PubMed]
- Hoffmann, M.; Kleine-Weber, H.; Pohlmann, S. A Multibasic Cleavage Site in the Spike Protein of SARS-CoV-2 Is Essential for Infection of Human Lung Cells. Mol. Cell 2020, 78, 779–784.e5. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Kaplan, N.; Wysocki, J.; Yang, W.; Lu, K.; Peng, H.; Batlle, D.; Lavker, R.M. The ACE2-deficient mouse: A model for a cytokine storm-driven inflammation. FASEB J. 2020, 34, 10505–10515. [Google Scholar] [CrossRef]
- Zhou, Y.; Fu, B.; Zheng, X.; Wang, D.; Zhao, C.; Qi, Y.; Sun, R.; Tian, Z.; Xu, X.; Wei, H. Pathogenic T-cells and inflammatory monocytes incite inflammatory storms in severe COVID-19 patients. Natl. Sci. Rev. 2020, 7, 998–1002. [Google Scholar] [CrossRef]
- Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef]
- Liu, J.; Li, S.; Liu, J.; Liang, B.; Wang, X.; Wang, H.; Li, W.; Tong, Q.; Yi, J.; Zhao, L.; et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine 2020, 55, 102763. [Google Scholar] [CrossRef]
- Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H.; et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Investig. 2020, 130, 2620–2629. [Google Scholar] [CrossRef]
- Zhang, H.; Zhou, P.; Wei, Y.; Yue, H.; Wang, Y.; Hu, M.; Zhang, S.; Cao, T.; Yang, C.; Li, M.; et al. Histopathologic Changes and SARS-CoV-2 Immunostaining in the Lung of a Patient With COVID-19. Ann. Intern. Med. 2020, 172, 629–632. [Google Scholar] [CrossRef]
- 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]
- Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 844–847. [Google Scholar] [CrossRef]
- Fan, Z.; Chen, L.; Li, J.; Cheng, X.; Yang, J.; Tian, C.; Zhang, Y.; Huang, S.; Liu, Z.; Cheng, J. Clinical Features of COVID-19-Related Liver Functional Abnormality. Clin. Gastroenterol. Hepatol. 2020, 18, 1561–1566. [Google Scholar] [CrossRef]
- Mao, L.; Jin, H.; Wang, M.; Hu, Y.; Chen, S.; He, Q.; Chang, J.; Hong, C.; Zhou, Y.; Wang, D.; et al. Neurologic Manifestations of Hospitalized Patients With Coronavirus Disease 2019 in Wuhan, China. JAMA Neurol. 2020, 77, 683–690. [Google Scholar] [CrossRef]
- Alexander, M.P.; Mangalaparthi, K.K.; Madugundu, A.K.; Moyer, A.M.; Adam, B.A.; Mengel, M.; Singh, S.; Herrmann, S.M.; Rule, A.D.; Cheek, E.H.; et al. Acute Kidney Injury in Severe COVID-19 Has Similarities to Sepsis-Associated Kidney Injury: A Multi-Omics Study. Mayo Clin. Proc. 2021, 96, 2561–2575. [Google Scholar] [CrossRef]
- Qian, Y.; Lei, T.; Patel, P.S.; Lee, C.H.; Monaghan-Nichols, P.; Xin, H.B.; Qiu, J.; Fu, M. Direct Activation of Endothelial Cells by SARS-CoV-2 Nucleocapsid Protein Is Blocked by Simvastatin. J. Virol. 2021, 95, e0139621. [Google Scholar] [CrossRef]
- Zafer, M.M.; El-Mahallawy, H.A.; Ashour, H.M. Severe COVID-19 and Sepsis: Immune Pathogenesis and Laboratory Markers. Microorganisms 2021, 9, 159. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Collazo, E.; Avendano-Ortiz, J.; Martin-Quiros, A.; Aguirre, L.A. Immune Response and COVID-19: A mirror image of Sepsis. Int. J. Biol. Sci. 2020, 16, 2479–2489. [Google Scholar] [CrossRef] [PubMed]
- Chen, R.; Sang, L.; Jiang, M.; Yang, Z.; Jia, N.; Fu, W.; Xie, J.; Guan, W.; Liang, W.; Ni, Z.; et al. Longitudinal hematologic and immunologic variations associated with the progression of COVID-19 patients in China. J. Allergy Clin. Immunol. 2020, 146, 89–100. [Google Scholar] [CrossRef] [PubMed]
- Diao, B.; Wang, C.; Tan, Y.; Chen, X.; Liu, Y.; Ning, L.; Chen, L.; Li, M.; Liu, Y.; Wang, G.; et al. Reduction and Functional Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19). Front. Immunol. 2020, 11, 827. [Google Scholar] [CrossRef]
- Kalpakci, Y.; Hacibekiroglu, T.; Trak, G.; Karacaer, C.; Demirci, T.; Kocayigit, H.; Sunu, C.; Varim, C.; Falay, M. Comparative evaluation of memory T cells in COVID-19 patients and the predictive role of CD4+CD8+ double positive T lymphocytes as a new marker. Rev. Assoc. Med. Bras. 2020, 66, 1666–1672. [Google Scholar] [CrossRef] [PubMed]
- Urra, J.M.; Cabrera, C.M.; Porras, L.; Rodenas, I. Selective CD8 cell reduction by SARS-CoV-2 is associated with a worse prognosis and systemic inflammation in COVID-19 patients. Clin. Immunol. 2020, 217, 108486. [Google Scholar] [CrossRef]
- Liu, R.; Wang, Y.; Li, J.; Han, H.; Xia, Z.; Liu, F.; Wu, K.; Yang, L.; Liu, X.; Zhu, C. Decreased T cell populations contribute to the increased severity of COVID-19. Clin. Chim. Acta 2020, 508, 110–114. [Google Scholar] [CrossRef]
- Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Moller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
- Fan, S.; Popli, S.; Chakravarty, S.; Chakravarti, R.; Chattopadhyay, S. Non-transcriptional IRF7 interacts with NF-kappaB to inhibit viral inflammation. J. Biol. Chem. 2024, 300, 107200. [Google Scholar] [CrossRef]
- Cusato, J.; Manca, A.; Palermiti, A.; Mula, J.; Costanzo, M.; Antonucci, M.; Trunfio, M.; Corcione, S.; Chiara, F.; De Vivo, E.D.; et al. COVID-19: A Possible Contribution of the MAPK Pathway. Biomedicines 2023, 11, 1459. [Google Scholar] [CrossRef]
- Salem, F.; Li, X.Z.; Hindi, J.; Casablanca, N.M.; Zhong, F.; El Jamal, S.M.; Haroon Al Rasheed, M.R.; Li, L.; Lee, K.; Chan, L.; et al. Activation of STAT3 signaling pathway in the kidney of COVID-19 patients. J. Nephrol. 2022, 35, 735–743. [Google Scholar] [CrossRef] [PubMed]
- Guan, G.W.; Gao, L.; Wang, J.W.; Wen, X.J.; Mao, T.H.; Peng, S.W.; Zhang, T.; Chen, X.M.; Lu, F.M. Exploring the mechanism of liver enzyme abnormalities in patients with novel coronavirus-infected pneumonia. Zhonghua Gan Zang Bing Za Zhi 2020, 28, 100–106. [Google Scholar] [CrossRef] [PubMed]
- Mantovani, A.; Beatrice, G.; Dalbeni, A. Coronavirus disease 2019 and prevalence of chronic liver disease: A meta-analysis. Liver Int. 2020, 40, 1316–1320. [Google Scholar] [CrossRef] [PubMed]
- Farahani, M.; Niknam, Z.; Mohammadi Amirabad, L.; Amiri-Dashatan, N.; Koushki, M.; Nemati, M.; Danesh Pouya, F.; Rezaei-Tavirani, M.; Rasmi, Y.; Tayebi, L. Molecular pathways involved in COVID-19 and potential pathway-based therapeutic targets. Biomed. Pharmacother. 2022, 145, 112420. [Google Scholar] [CrossRef] [PubMed]
- Appelberg, S.; Gupta, S.; Svensson Akusjarvi, S.; Ambikan, A.T.; Mikaeloff, F.; Saccon, E.; Vegvari, A.; Benfeitas, R.; Sperk, M.; Stahlberg, M.; et al. Dysregulation in Akt/mTOR/HIF-1 signaling identified by proteo-transcriptomics of SARS-CoV-2 infected cells. Emerg. Microbes Infect. 2020, 9, 1748–1760. [Google Scholar] [CrossRef]
- Tonai, K.; Ohta-Ogo, K.; Kainuma, S.; Tadokoro, N.; Tsukamoto, Y.; Hatakeyama, K.; Suzuki, T.; Fukushima, S. Case series: Successful salvage from refractory cardiogenic shock caused by COVID-19-associated myocardial injury with temporary ventricular assist devices. Eur. Heart J. Case Rep. 2024, 8, ytae308. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Li, M.; Wang, Z.; Sun, P.; Wei, S.; Zhang, C.; Wu, H.; Bai, H. Cardiovascular Risk After SARS-CoV-2 Infection Is Mediated by IL18/IL18R1/HIF-1 Signaling Pathway Axis. Front. Immunol. 2021, 12, 780804. [Google Scholar] [CrossRef] [PubMed]
- Espinosa, P.S.; Rizvi, Z.; Sharma, P.; Hindi, F.; Filatov, A. Neurological Complications of Coronavirus Disease (COVID-19): Encephalopathy, MRI Brain and Cerebrospinal Fluid Findings: Case 2. Cureus 2020, 12, e7930. [Google Scholar] [CrossRef]
- Scullen, T.; Keen, J.; Mathkour, M.; Dumont, A.S.; Kahn, L. Coronavirus 2019 (COVID-19)-Associated Encephalopathies and Cerebrovascular Disease: The New Orleans Experience. World Neurosurg. 2020, 141, e437–e446. [Google Scholar] [CrossRef]
- Group, R.C.; Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; et al. Dexamethasone in Hospitalized Patients with COVID-19. N. Engl. J. Med. 2021, 384, 693–704. [Google Scholar] [CrossRef]
- Shapiro, L.; Scherger, S.; Franco-Paredes, C.; Gharamti, A.; Henao-Martinez, A.F. Anakinra authorized to treat severe coronavirus disease 2019; Sepsis breakthrough or time to reflect? Front Microbiol 2023, 14, 1250483. [Google Scholar] [CrossRef] [PubMed]
- Mohseni Afshar, Z.; Tavakoli Pirzaman, A.; Hosseinzadeh, R.; Babazadeh, A.; Taghizadeh Moghadam, M.A.; Miri, S.R.; Sio, T.T.; Sullman, M.J.M.; Barary, M.; Ebrahimpour, S. Anticoagulant therapy in COVID-19: A narrative review. Clin. Transl. Sci. 2023, 16, 1510–1525. [Google Scholar] [CrossRef]
- Villar, J.; Ferrando, C.; Martinez, D.; Ambros, A.; Munoz, T.; Soler, J.A.; Aguilar, G.; Alba, F.; Gonzalez-Higueras, E.; Conesa, L.A.; et al. Dexamethasone treatment for the acute respiratory distress syndrome: A multicentre, randomised controlled trial. Lancet Respir. Med. 2020, 8, 267–276. [Google Scholar] [CrossRef] [PubMed]
- Cavalli, G.; De Luca, G.; Campochiaro, C.; Della-Torre, E.; Ripa, M.; Canetti, D.; Oltolini, C.; Castiglioni, B.; Tassan Din, C.; Boffini, N.; et al. Interleukin-1 blockade with high-dose anakinra in patients with COVID-19, acute respiratory distress syndrome, and hyperinflammation: A retrospective cohort study. Lancet Rheumatol. 2020, 2, e325–e331. [Google Scholar] [CrossRef]
- Salton, F.; Confalonieri, P.; Meduri, G.U.; Santus, P.; Harari, S.; Scala, R.; Lanini, S.; Vertui, V.; Oggionni, T.; Caminati, A.; et al. Prolonged Low-Dose Methylprednisolone in Patients With Severe COVID-19 Pneumonia. Open Forum Infect. Dis. 2020, 7, ofaa421. [Google Scholar] [CrossRef] [PubMed]
- Langarizadeh, M.A.; Ranjbar Tavakoli, M.; Abiri, A.; Ghasempour, A.; Rezaei, M.; Ameri, A. A review on function and side effects of systemic corticosteroids used in high-grade COVID-19 to prevent cytokine storms. EXCLI J. 2021, 20, 339–365. [Google Scholar] [CrossRef] [PubMed]
- Nelson, B.C.; Laracy, J.; Shoucri, S.; Dietz, D.; Zucker, J.; Patel, N.; Sobieszczyk, M.E.; Kubin, C.J.; Gomez-Simmonds, A. Clinical Outcomes Associated with Methylprednisolone in Mechanically Ventilated Patients with COVID-19. Clin. Infect. Dis. 2021, 72, e367–e372. [Google Scholar] [CrossRef] [PubMed]
- De Bosscher, K.; Vanden Berghe, W.; Vermeulen, L.; Plaisance, S.; Boone, E.; Haegeman, G. Glucocorticoids repress NF-kappaB-driven genes by disturbing the interaction of p65 with the basal transcription machinery, irrespective of coactivator levels in the cell. Proc. Natl. Acad. Sci. USA 2000, 97, 3919–3924. [Google Scholar] [CrossRef]
- Ho, K.S.; Narasimhan, B.; Difabrizio, L.; Rogers, L.; Bose, S.; Li, L.; Chen, R.; Sheehan, J.; El-Halabi, M.A.; Sarosky, K.; et al. Impact of corticosteroids in hospitalised COVID-19 patients. BMJ Open Respir. Res. 2021, 8, e000766. [Google Scholar] [CrossRef] [PubMed]
- Selvaraj, V.; Dapaah-Afriyie, K.; Finn, A.; Flanigan, T.P. Short-Term Dexamethasone in SARS-CoV-2 Patients. R I Med J. 2020, 103, 39–43. [Google Scholar]
- Kim, W.Y.; Kweon, O.J.; Cha, M.J.; Baek, M.S.; Choi, S.H. Dexamethasone may improve severe COVID-19 via ameliorating endothelial injury and inflammation: A preliminary pilot study. PLoS ONE 2021, 16, e0254167. [Google Scholar] [CrossRef] [PubMed]
- Edalatifard, M.; Akhtari, M.; Salehi, M.; Naderi, Z.; Jamshidi, A.; Mostafaei, S.; Najafizadeh, S.R.; Farhadi, E.; Jalili, N.; Esfahani, M.; et al. Intravenous methylprednisolone pulse as a treatment for hospitalised severe COVID-19 patients: Results from a randomised controlled clinical trial. Eur. Respir. J. 2020, 56, 2002808. [Google Scholar] [CrossRef] [PubMed]
- Mwangi, V.I.; Netto, R.L.A.; de Morais, C.E.P.; Silva, A.S.; Silva, B.M.; Lima, A.B.; Neves, J.C.F.; Borba, M.G.S.; Val, F.; de Almeida, A.C.G.; et al. Temporal patterns of cytokine and injury biomarkers in hospitalized COVID-19 patients treated with methylprednisolone. Front. Immunol. 2023, 14, 1229611. [Google Scholar] [CrossRef] [PubMed]
- Hatic, H.; Hearld, K.R.; Das, D.; Deshane, J. Clinical Outcomes in COVID-19 Patients Treated with Immunotherapy. Cancers 2022, 14, 5954. [Google Scholar] [CrossRef] [PubMed]
- Kooistra, E.J.; Waalders, N.J.B.; Grondman, I.; Janssen, N.A.F.; de Nooijer, A.H.; Netea, M.G.; van de Veerdonk, F.L.; Ewalds, E.; van der Hoeven, J.G.; Kox, M.; et al. Anakinra treatment in critically ill COVID-19 patients: A prospective cohort study. Crit. Care 2020, 24, 688. [Google Scholar] [CrossRef] [PubMed]
- Huet, T.; Beaussier, H.; Voisin, O.; Jouveshomme, S.; Dauriat, G.; Lazareth, I.; Sacco, E.; Naccache, J.M.; Bezie, Y.; Laplanche, S.; et al. Anakinra for severe forms of COVID-19: A cohort study. Lancet Rheumatol. 2020, 2, e393–e400. [Google Scholar] [CrossRef] [PubMed]
- Sheppard, M.; Laskou, F.; Stapleton, P.P.; Hadavi, S.; Dasgupta, B. Tocilizumab (Actemra). Hum. Vaccin. Immunother. 2017, 13, 1972–1988. [Google Scholar] [CrossRef]
- Keske, S.; Tekin, S.; Sait, B.; Irkoren, P.; Kapmaz, M.; Cimen, C.; Ugur, S.; Celebi, I.; Bakir, V.O.; Palaoglu, E.; et al. Appropriate use of tocilizumab in COVID-19 infection. Int. J. Infect. Dis. 2020, 99, 338–343. [Google Scholar] [CrossRef] [PubMed]
- Group, R.C. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): A randomised, controlled, open-label, platform trial. Lancet 2021, 397, 1637–1645. [Google Scholar] [CrossRef]
- Kewan, T.; Covut, F.; Al-Jaghbeer, M.J.; Rose, L.; Gopalakrishna, K.V.; Akbik, B. Tocilizumab for treatment of patients with severe COVID-19: A retrospective cohort study. EClinicalMedicine 2020, 24, 100418. [Google Scholar] [CrossRef]
- Tian, Y.; Pan, T.; Wen, X.; Ao, G.; Ma, Y.; Liu, X.; Liu, R.; Ran, H. Efficacy and Safety of Direct Oral Anticoagulants Compared with Heparin for Preventing Thromboembolism in Hospitalized Patients with COVID-19: A Systematic Review and Meta-Analysis. Clin. Appl. Thromb. Hemost. 2023, 29, 10760296231164355. [Google Scholar] [CrossRef] [PubMed]
- Hadid, T.; Kafri, Z.; Al-Katib, A. Coagulation and anticoagulation in COVID-19. Blood Rev. 2021, 47, 100761. [Google Scholar] [CrossRef] [PubMed]
- Shappell, C.N.; Klompas, M.; Rhee, C. Quantifying the Burden of Viral Sepsis During the Coronavirus Disease 2019 Pandemic and Beyond. Crit. Care Med. 2021, 49, 2140–2143. [Google Scholar] [CrossRef] [PubMed]
- Karakike, E.; Giamarellos-Bourboulis, E.J.; Kyprianou, M.; Fleischmann-Struzek, C.; Pletz, M.W.; Netea, M.G.; Reinhart, K.; Kyriazopoulou, E. Coronavirus Disease 2019 as Cause of Viral Sepsis: A Systematic Review and Meta-Analysis. Crit. Care Med. 2021, 49, 2042–2057. [Google Scholar] [CrossRef]
- Herrera-Van Oostdam, A.S.; Castaneda-Delgado, J.E.; Oropeza-Valdez, J.J.; Borrego, J.C.; Monarrez-Espino, J.; Zheng, J.; Mandal, R.; Zhang, L.; Soto-Guzman, E.; Fernandez-Ruiz, J.C.; et al. Immunometabolic signatures predict risk of progression to sepsis in COVID-19. PLoS ONE 2021, 16, e0256784. [Google Scholar] [CrossRef] [PubMed]
- Barasa, E.W.; Ouma, P.O.; Okiro, E.A. Assessing the hospital surge capacity of the Kenyan health system in the face of the COVID-19 pandemic. PLoS ONE 2020, 15, e0236308. [Google Scholar] [CrossRef] [PubMed]
- Marozoff, S.; Tan, J.; Lu, N.; Kirmani, A.; Loree, J.M.; Xie, H.; Lacaille, D.; Kopec, J.A.; Esdaile, J.M.; Corradetti, B.; et al. Effect of Immunosuppressive or Immunomodulatory Agents on Severe COVID-19 Outcomes: A Population-Based Cohort Study. ACR Open Rheumatol. 2023, 5, 685–693. [Google Scholar] [CrossRef] [PubMed]
- Petrovska, B.B. Historical review of medicinal plants’ usage. Pharmacogn. Rev. 2012, 6, 1–5. [Google Scholar] [CrossRef]
- Zhang, Y.J.; Gan, R.Y.; Li, S.; Zhou, Y.; Li, A.N.; Xu, D.P.; Li, H.B. Antioxidant Phytochemicals for the Prevention and Treatment of Chronic Diseases. Molecules 2015, 20, 21138–21156. [Google Scholar] [CrossRef]
- Shin, S.A.; Joo, B.J.; Lee, J.S.; Ryu, G.; Han, M.; Kim, W.Y.; Park, H.H.; Lee, J.H.; Lee, C.S. Phytochemicals as Anti-Inflammatory Agents in Animal Models of Prevalent Inflammatory Diseases. Molecules 2020, 25, 5932. [Google Scholar] [CrossRef]
- Sharma, B.R.; Kumar, V.; Gat, Y.; Kumar, N.; Parashar, A.; Pinakin, D.J. Microbial maceration: A sustainable approach for phytochemical extraction. 3 Biotech. 2018, 8, 401. [Google Scholar] [CrossRef] [PubMed]
- Palombo, E.A. Phytochemicals from traditional medicinal plants used in the treatment of diarrhoea: Modes of action and effects on intestinal function. Phytother. Res. 2006, 20, 717–724. [Google Scholar] [CrossRef] [PubMed]
- Kumar, A.; Nirmal, P.; Kumar, M.; Jose, A.; Tomer, V.; Oz, E.; Proestos, C.; Zeng, M.; Elobeid, T.; Sneha, K.; et al. Major Phytochemicals: Recent Advances in Health Benefits and Extraction Method. Molecules 2023, 28, 887. [Google Scholar] [CrossRef] [PubMed]
- Parihar, A.; Malviya, S.; Khan, R.; Kaushik, A.; Mostafavi, E. COVID-19 associated thyroid dysfunction and other comorbidities and its management using phytochemical-based therapeutics: A natural way. Biosci. Rep. 2023, 43, BSR20230293. [Google Scholar] [CrossRef] [PubMed]
- Chavda, V.P.; Vihol, D.; Mehta, B.; Shah, D.; Patel, M.; Vora, L.K.; Pereira-Silva, M.; Paiva-Santos, A.C. Phytochemical-loaded liposomes for anticancer therapy: An updated review. Nanomedicine 2022, 17, 547–568. [Google Scholar] [CrossRef] [PubMed]
- Pillai, S.S.; Mini, S. Attenuation of high glucose induced apoptotic and inflammatory signaling pathways in RIN-m5F pancreatic beta cell lines by Hibiscus rosa sinensis L. petals and its phytoconstituents. J. Ethnopharmacol. 2018, 227, 8–17. [Google Scholar] [CrossRef] [PubMed]
- Zhu, L.Y.; Wen, X.Y.; Xiang, Q.Y.; Guo, L.L.; Xu, J.; Zhao, S.P.; Liu, L. Comparison of the Reductions in LDL-C and Non-HDL-C Induced by the Red Yeast Rice Extract Xuezhikang Between Fasting and Non-fasting States in Patients with Coronary Heart Disease. Front. Cardiovasc. Med. 2021, 8, 674446. [Google Scholar] [CrossRef]
- Micek, A.; Godos, J.; Del Rio, D.; Galvano, F.; Grosso, G. Dietary Flavonoids and Cardiovascular Disease: A Comprehensive Dose-Response Meta-Analysis. Mol. Nutr. Food Res. 2021, 65, e2001019. [Google Scholar] [CrossRef]
- Zheng, Q.N.; Wang, J.; Zhou, H.B.; Niu, S.F.; Liu, Q.L.; Yang, Z.J.; Wang, H.; Zhao, Y.S.; Shi, S.L. Effectiveness of Amygdalus mongolica oil in hyperlipidemic rats and underlying antioxidant processes. J. Toxicol. Environ. Health A 2017, 80, 1193–1198. [Google Scholar] [CrossRef] [PubMed]
- Dominguez Diaz, L.; Fernandez-Ruiz, V.; Camara, M. The frontier between nutrition and pharma: The international regulatory framework of functional foods, food supplements and nutraceuticals. Crit. Rev. Food Sci. Nutr. 2020, 60, 1738–1746. [Google Scholar] [CrossRef]
- Damian, M.R.; Cortes-Perez, N.G.; Quintana, E.T.; Ortiz-Moreno, A.; Garfias Noguez, C.; Cruceno-Casarrubias, C.E.; Sanchez Pardo, M.E.; Bermudez-Humaran, L.G. Functional Foods, Nutraceuticals and Probiotics: A Focus on Human Health. Microorganisms 2022, 10, 1065. [Google Scholar] [CrossRef] [PubMed]
- Granato, D.; Barba, F.J.; Bursac Kovacevic, D.; Lorenzo, J.M.; Cruz, A.G.; Putnik, P. Functional Foods: Product Development, Technological Trends, Efficacy Testing, and Safety. Annu. Rev. Food Sci. Technol. 2020, 11, 93–118. [Google Scholar] [CrossRef] [PubMed]
- Timmers, S.; Konings, E.; Bilet, L.; Houtkooper, R.H.; van de Weijer, T.; Goossens, G.H.; Hoeks, J.; van der Krieken, S.; Ryu, D.; Kersten, S.; et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 2011, 14, 612–622. [Google Scholar] [CrossRef] [PubMed]
- Chen, K.H.; Cheng, M.L.; Jing, Y.H.; Chiu, D.T.; Shiao, M.S.; Chen, J.K. Resveratrol ameliorates metabolic disorders and muscle wasting in streptozotocin-induced diabetic rats. Am. J. Physiol. Endocrinol. Metab. 2011, 301, E853–E863. [Google Scholar] [CrossRef]
- Crandall, J.P.; Oram, V.; Trandafirescu, G.; Reid, M.; Kishore, P.; Hawkins, M.; Cohen, H.W.; Barzilai, N. Pilot study of resveratrol in older adults with impaired glucose tolerance. J. Gerontol. A Biol. Sci. Med. Sci. 2012, 67, 1307–1312. [Google Scholar] [CrossRef] [PubMed]
- Goldberg, D.M.; Yan, J.; Soleas, G.J. Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clin. Biochem. 2003, 36, 79–87. [Google Scholar] [CrossRef]
- Singh, V.K.; Arora, D.; Satija, N.K.; Khare, P.; Roy, S.K.; Sharma, P.K. Intricatinol synergistically enhances the anticancerous activity of cisplatin in human A549 cells via p38 MAPK/p53 signalling. Apoptosis 2017, 22, 1273–1286. [Google Scholar] [CrossRef] [PubMed]
- Fung, T.T.; Rexrode, K.M.; Mantzoros, C.S.; Manson, J.E.; Willett, W.C.; Hu, F.B. Mediterranean diet and incidence of and mortality from coronary heart disease and stroke in women. Circulation 2009, 119, 1093–1100. [Google Scholar] [CrossRef]
- Baum, L.; Ng, A. Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer’s disease animal models. J. Alzheimers Dis. 2004, 6, 367–377; discussion 369–443. [Google Scholar] [CrossRef]
- Pillai, S.S.; Mini, S. Hibiscus rosa sinensis Linn. Petals Modulates Glycogen Metabolism and Glucose Homeostasis Signalling Pathway in Streptozotocin-Induced Experimental Diabetes. Plant Foods Hum. Nutr. 2016, 71, 42–48. [Google Scholar] [CrossRef]
- Ahmed, M.B.; Islam, S.U.; Alghamdi, A.A.A.; Kamran, M.; Ahsan, H.; Lee, Y.S. Phytochemicals as Chemo-Preventive Agents and Signaling Molecule Modulators: Current Role in Cancer Therapeutics and Inflammation. Int. J. Mol. Sci. 2022, 23, 5765. [Google Scholar] [CrossRef]
- Jang, J.H.; Lee, T.J. Mechanisms of Phytochemicals in Anti-Inflammatory and Anti-Cancer. Int. J. Mol. Sci. 2023, 24, 7863. [Google Scholar] [CrossRef]
- Hartman, R.E.; Ross, D.M. Effects and mechanisms of actions of phytochemicals on Alzheimer’s disease neuropathology. Front. Biosci Elite Ed 2018, 10, 300–333. [Google Scholar] [CrossRef]
- Gamboa-Gomez, C.I.; Rocha-Guzman, N.E.; Gallegos-Infante, J.A.; Moreno-Jimenez, M.R.; Vazquez-Cabral, B.D.; Gonzalez-Laredo, R.F. Plants with potential use on obesity and its complications. EXCLI J. 2015, 14, 809–831. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Song, Y.; Chen, Z.; Leng, S.X. Connection between Systemic Inflammation and Neuroinflammation Underlies Neuroprotective Mechanism of Several Phytochemicals in Neurodegenerative Diseases. Oxid. Med. Cell Longev. 2018, 2018, 1972714. [Google Scholar] [CrossRef]
- Lv, O.; Wang, L.; Li, J.; Ma, Q.; Zhao, W. Effects of pomegranate peel polyphenols on lipid accumulation and cholesterol metabolic transformation in L-02 human hepatic cells via the PPARgamma-ABCA1/CYP7A1 pathway. Food Funct. 2016, 7, 4976–4983. [Google Scholar] [CrossRef] [PubMed]
- Jia, Y.; Kim, J.Y.; Jun, H.J.; Kim, S.J.; Lee, J.H.; Hoang, M.H.; Kim, H.S.; Chang, H.I.; Hwang, K.Y.; Um, S.J.; et al. Cyanidin is an agonistic ligand for peroxisome proliferator-activated receptor-alpha reducing hepatic lipid. Biochim. Biophys. Acta 2013, 1831, 698–708. [Google Scholar] [CrossRef]
- Moghadasian, M.H.; McManus, B.M.; Godin, D.V.; Rodrigues, B.; Frohlich, J.J. Proatherogenic and antiatherogenic effects of probucol and phytosterols in apolipoprotein E-deficient mice: Possible mechanisms of action. Circulation 1999, 99, 1733–1739. [Google Scholar] [CrossRef] [PubMed]
- Alikiaii, B.; Bagherniya, M.; Askari, G.; Johnston, T.P.; Sahebkar, A. The role of phytochemicals in sepsis: A mechanistic and therapeutic perspective. Biofactors 2021, 47, 19–40. [Google Scholar] [CrossRef]
- Usmani, J.; Khan, T.; Ahmad, R.; Sharma, M. Potential role of herbal medicines as a novel approach in sepsis treatment. Biomed. Pharmacother. 2021, 144, 112337. [Google Scholar] [CrossRef]
- Mahomoodally, M.F.; Aumeeruddy, M.Z.; Legoabe, L.J.; Dall’Acqua, S.; Zengin, G. Plants’ bioactive secondary metabolites in the management of sepsis: Recent findings on their mechanism of action. Front. Pharmacol. 2022, 13, 1046523. [Google Scholar] [CrossRef]
- Shapiro, H.; Lev, S.; Cohen, J.; Singer, P. Polyphenols in the prevention and treatment of sepsis syndromes: Rationale and pre-clinical evidence. Nutrition 2009, 25, 981–997. [Google Scholar] [CrossRef] [PubMed]
- Di Pierro, F.; Derosa, G.; Maffioli, P.; Bertuccioli, A.; Togni, S.; Riva, A.; Allegrini, P.; Khan, A.; Khan, S.; Khan, B.A.; et al. Possible Therapeutic Effects of Adjuvant Quercetin Supplementation Against Early-Stage COVID-19 Infection: A Prospective, Randomized, Controlled, and Open-Label Study. Int. J. Gen. Med. 2021, 14, 2359–2366. [Google Scholar] [CrossRef] [PubMed]
- Shanmugarajan, D.; Prabitha, P.; Kumar, B.R.P.; Suresh, B. Curcumin to inhibit binding of spike glycoprotein to ACE2 receptors: Computational modelling, simulations, and ADMET studies to explore curcuminoids against novel SARS-CoV-2 targets. RSC Adv. 2020, 10, 31385–31399. [Google Scholar] [CrossRef]
- Saber-Moghaddam, N.; Salari, S.; Hejazi, S.; Amini, M.; Taherzadeh, Z.; Eslami, S.; Rezayat, S.M.; Jaafari, M.R.; Elyasi, S. Oral nano-curcumin formulation efficacy in management of mild to moderate hospitalized coronavirus disease-19 patients: An open label nonrandomized clinical trial. Phytother. Res. 2021, 35, 2616–2623. [Google Scholar] [CrossRef]
- Lin, C.; Tsai, F.J.; Hsu, Y.M.; Ho, T.J.; Wang, G.K.; Chiu, Y.J.; Ha, H.A.; Yang, J.S. Study of Baicalin toward COVID-19 Treatment: In silico Target Analysis and in vitro Inhibitory Effects on SARS-CoV-2 Proteases. Biomed. Hub. 2021, 6, 122–137. [Google Scholar] [CrossRef] [PubMed]
- Yang, M.; Wei, J.; Huang, T.; Lei, L.; Shen, C.; Lai, J.; Yang, M.; Liu, L.; Yang, Y.; Liu, G.; et al. Resveratrol inhibits the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in cultured Vero cells. Phytother. Res. 2021, 35, 1127–1129. [Google Scholar] [CrossRef]
- Chang, Y.C.; Tsai, M.H.; Sheu, W.H.; Hsieh, S.C.; Chiang, A.N. The therapeutic potential and mechanisms of action of quercetin in relation to lipopolysaccharide-induced sepsis in vitro and in vivo. PLoS ONE 2013, 8, e80744. [Google Scholar] [CrossRef]
- Cui, W.; Hu, G.; Peng, J.; Mu, L.; Liu, J.; Qiao, L. Quercetin Exerted Protective Effects in a Rat Model of Sepsis via Inhibition of Reactive Oxygen Species (ROS) and Downregulation of High Mobility Group Box 1 (HMGB1) Protein Expression. Med. Sci. Monit. 2019, 25, 5795–5800. [Google Scholar] [CrossRef]
- Huang, W.; Tang, Y.; Li, L. HMGB1, a potent proinflammatory cytokine in sepsis. Cytokine 2010, 51, 119–126. [Google Scholar] [CrossRef]
- Sul, O.J.; Ra, S.W. Quercetin Prevents LPS-Induced Oxidative Stress and Inflammation by Modulating NOX2/ROS/NF-kB in Lung Epithelial Cells. Molecules 2021, 26, 6949. [Google Scholar] [CrossRef] [PubMed]
- Sang, A.; Wang, Y.; Wang, S.; Wang, Q.; Wang, X.; Li, X.; Song, X. Quercetin attenuates sepsis-induced acute lung injury via suppressing oxidative stress-mediated ER stress through activation of SIRT1/AMPK pathways. Cell Signal 2022, 96, 110363. [Google Scholar] [CrossRef]
- Wang, L.; Chen, J.; Wang, B.; Wu, D.; Li, H.; Lu, H.; Wu, H.; Chai, Y. Protective effect of quercetin on lipopolysaccharide-induced acute lung injury in mice by inhibiting inflammatory cell influx. Exp. Biol. Med. 2014, 239, 1653–1662. [Google Scholar] [CrossRef]
- Ding, W.; Zhang, W.; Chen, J.; Wang, M.; Ren, Y.; Feng, J.; Han, X.; Ji, X.; Nie, S.; Sun, Z. Protective mechanism of quercetin in alleviating sepsis-related acute respiratory distress syndrome based on network pharmacology and in vitro experiments. World J. Emerg. Med. 2024, 15, 111–120. [Google Scholar] [CrossRef]
- Meng, X.L.; Yu, M.M.; Liu, Y.C.; Gao, Y.L.; Chen, X.S.; Shou, S.T.; Chai, Y.F. Rutin Inhibits Cardiac Apoptosis and Prevents Sepsis-Induced Cardiomyopathy. Front. Physiol. 2022, 13, 834077. [Google Scholar] [CrossRef]
- Zhao, H.; Lin, X.; Chen, Q.; Wang, X.; Wu, Y.; Zhao, X. Quercetin inhibits the NOX2/ROS-mediated NF-kappaB/TXNIP signaling pathway to ameliorate pyroptosis of cardiomyocytes to relieve sepsis-induced cardiomyopathy. Toxicol. Appl. Pharmacol. 2023, 477, 116672. [Google Scholar] [CrossRef]
- Park, H.J.; Lee, S.J.; Cho, J.; Gharbi, A.; Han, H.D.; Kang, T.H.; Kim, Y.; Lee, Y.; Park, W.S.; Jung, I.D.; et al. Tamarixetin Exhibits Anti-inflammatory Activity and Prevents Bacterial Sepsis by Increasing IL-10 Production. J. Nat. Prod. 2018, 81, 1435–1443. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Luo, C.; Luo, M.; Zhou, S.; Kuang, G. Targets and mechanisms of Xuebijing in the treatment of acute kidney injury associated with sepsis: A network pharmacology-based study. Curr. Comput. Aided Drug Des. 2023, 20, 752–763. [Google Scholar] [CrossRef] [PubMed]
- Li, R.; Li, X.; Zhao, J.; Meng, F.; Yao, C.; Bao, E.; Sun, N.; Chen, X.; Cheng, W.; Hua, H.; et al. Mitochondrial STAT3 exacerbates LPS-induced sepsis by driving CPT1a-mediated fatty acid oxidation. Theranostics 2022, 12, 976–998. [Google Scholar] [CrossRef]
- Liu, Y.; Feng, L.; Hou, G.; Yao, L. Curcumin Elevates microRNA-183-5p via Cathepsin B-Mediated Phosphatidylinositol 3-Kinase/AKT Pathway to Strengthen Lipopolysaccharide-Stimulated Immune Function of Sepsis Mice. Contrast Media Mol. Imaging 2022, 2022, 6217234. [Google Scholar] [CrossRef]
- Chen, D.; Wang, H.; Cai, X. Curcumin interferes with sepsis-induced cardiomyocyte apoptosis via TLR1 inhibition. Rev. Port. Cardiol. 2023, 42, 209–221. [Google Scholar] [CrossRef] [PubMed]
- Hou, D.; Liao, H.; Hao, S.; Liu, R.; Huang, H.; Duan, C. Curcumin simultaneously improves mitochondrial dynamics and myocardial cell bioenergy after sepsis via the SIRT1-DRP1/PGC-1alpha pathway. Heliyon 2024, 10, e28501. [Google Scholar] [CrossRef] [PubMed]
- Rattis, B.A.C.; Piva, H.L.; Duarte, A.; Gomes, F.; Lellis, J.R.; Soave, D.F.; Ramos, S.G.; Tedesco, A.C.; Celes, M.R.N. Modulation of the mTOR Pathway by Curcumin in the Heart of Septic Mice. Pharmaceutics 2022, 14, 2277. [Google Scholar] [CrossRef] [PubMed]
- Poylin, V.; Fareed, M.U.; O’Neal, P.; Alamdari, N.; Reilly, N.; Menconi, M.; Hasselgren, P.O. The NF-kappaB inhibitor curcumin blocks sepsis-induced muscle proteolysis. Mediators Inflamm. 2008, 2008, 317851. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Lu, Y.; Zhao, L.; Hu, L.; Qiu, Q.; Zhang, Z.; Li, M.; Hong, G.; Wu, B.; Zhao, G.; et al. Curcumin attenuates sepsis-induced acute organ dysfunction by preventing inflammation and enhancing the suppressive function of Tregs. Int. Immunopharmacol. 2018, 61, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Xiao, X.; Yang, M.; Sun, D.; Sun, S. Curcumin protects against sepsis-induced acute lung injury in rats. J. Surg. Res. 2012, 176, e31–e39. [Google Scholar] [CrossRef]
- Xu, F.; Lin, S.H.; Yang, Y.Z.; Guo, R.; Cao, J.; Liu, Q. The effect of curcumin on sepsis-induced acute lung injury in a rat model through the inhibition of the TGF-beta1/SMAD3 pathway. Int. Immunopharmacol. 2013, 16, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.F.; Yang, C.W.; Liu, H.; Sui, S.G.; Li, X.D. Efficacy and Therapeutic Potential of Curcumin Against Sepsis-Induced Chronic Lung Injury in Male Albino Rats. J. Nutr. Health Aging 2017, 21, 307–313. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Wu, Q.; Lu, Y.; Meng, L.P.; Xu, X.L.; Wang, X.J.; Chen, W. Arginine-Glycine-Aspartic Acid-anchored Curcumin-based Nanotherapeutics Inhibit Pyroptosis-induced Cytokine Release Syndrome for In Vivo and In Vitro Sepsis Applications. Curr. Pharm. Des. 2023, 29, 283–294. [Google Scholar] [CrossRef] [PubMed]
- Teng, L.; Zhang, Y.; Chen, L.; Shi, G. Fabrication of a curcumin encapsulated bioengineered nano-cocktail formulation for stimuli-responsive targeted therapeutic delivery to enhance anti-inflammatory, anti-oxidant, and anti-bacterial properties in sepsis management. J. Biomater. Sci. Polym. Ed. 2023, 34, 1716–1740. [Google Scholar] [CrossRef]
- Naeini, F.; Tutunchi, H.; Razmi, H.; Mahmoodpoor, A.; Vajdi, M.; Sefidmooye Azar, P.; Najifipour, F.; Tarighat-Esfanjani, A.; Karimi, A. Does nano-curcumin supplementation improve hematological indices in critically ill patients with sepsis? A randomized controlled clinical trial. J. Food Biochem. 2022, 46, e14093. [Google Scholar] [CrossRef]
- Karimi, A.; Pourreza, S.; Vajdi, M.; Mahmoodpoor, A.; Sanaie, S.; Karimi, M.; Tarighat-Esfanjani, A. Evaluating the effects of curcumin nanomicelles on clinical outcome and cellular immune responses in critically ill sepsis patients: A randomized, double-blind, and placebo-controlled trial. Front. Nutr. 2022, 9, 1037861. [Google Scholar] [CrossRef] [PubMed]
- Rungsung, S.; Singh, T.U.; Perumalraja, K.; Mahobiya, A.; Sharma, M.; Lingaraju, M.C.; Parida, S.; Sahoo, M.; Kumar, D. Luteolin alleviates vascular dysfunctions in CLP-induced polymicrobial sepsis in mice. Pharmacol. Rep. 2022, 74, 1054–1068. [Google Scholar] [CrossRef] [PubMed]
- Liu, B.; Su, H. Luteolin improves vasoconstriction function and survival of septic mice via AMPK/NF-kappaB pathway. Heliyon 2023, 9, e13330. [Google Scholar] [CrossRef]
- Kotanidou, A.; Xagorari, A.; Bagli, E.; Kitsanta, P.; Fotsis, T.; Papapetropoulos, A.; Roussos, C. Luteolin reduces lipopolysaccharide-induced lethal toxicity and expression of proinflammatory molecules in mice. Am. J. Respir. Crit. Care Med. 2002, 165, 818–823. [Google Scholar] [CrossRef]
- Chen, D.; Bi, A.; Dong, X.; Jiang, Y.; Rui, B.; Liu, J.; Yin, Z.; Luo, L. Luteolin exhibits anti-inflammatory effects by blocking the activity of heat shock protein 90 in macrophages. Biochem. Biophys. Res. Commun. 2014, 443, 326–332. [Google Scholar] [CrossRef] [PubMed]
- Yan, L.; Liang, J.; Zhou, Y.; Huang, J.; Zhang, T.; Wang, X.; Yin, H. Switch Off “Parallel Circuit”: Insight of New Strategy of Simultaneously Suppressing Canonical and Noncanonical Inflammation Activation in Endotoxemic Mice. Adv. Biosyst. 2020, 4, e2000037. [Google Scholar] [CrossRef]
- Wu, B.; Song, H.; Fan, M.; You, F.; Zhang, L.; Luo, J.; Li, J.; Wang, L.; Li, C.; Yuan, M. Luteolin attenuates sepsis-induced myocardial injury by enhancing autophagy in mice. Int. J. Mol. Med. 2020, 45, 1477–1487. [Google Scholar] [CrossRef]
- Zhang, Y.; Zhang, J.; Ren, Y.; Li, T.; Bi, J.; Du, Z.; Wu, R. Luteolin Suppresses Sepsis-Induced Cold-Inducible RNA-Binding Protein Production and Lung Injury in Neonatal Mice. Shock 2021, 55, 268–273. [Google Scholar] [CrossRef]
- Rungsung, S.; Singh, T.U.; Rabha, D.J.; Kumar, T.; Cholenahalli Lingaraju, M.; Parida, S.; Paul, A.; Sahoo, M.; Kumar, D. Luteolin attenuates acute lung injury in experimental mouse model of sepsis. Cytokine 2018, 110, 333–343. [Google Scholar] [CrossRef]
- Zhang, Z.H.; Yang, H.X.; Jin, Q.; Wu, Y.L.; Cui, Z.Y.; Shang, Y.; Liu, J.; Zhan, Z.Y.; Lian, L.H.; Nan, J.X. Luteolin attenuates hepatic injury in septic mice by regulating P2X7R-based HMGB1 release. Food Funct. 2021, 12, 10714–10727. [Google Scholar] [CrossRef] [PubMed]
- Xin, S.B.; Yan, H.; Ma, J.; Sun, Q.; Shen, L. Protective Effects of Luteolin on Lipopolysaccharide-Induced Acute Renal Injury in Mice. Med. Sci. Monit. 2016, 22, 5173–5180. [Google Scholar] [CrossRef] [PubMed]
- Yang, R.; Yang, H.; Wei, J.; Li, W.; Yue, F.; Song, Y.; He, X.; Hu, K. Mechanisms Underlying the Effects of Lianhua Qingwen on Sepsis-Induced Acute Lung Injury: A Network Pharmacology Approach. Front. Pharmacol. 2021, 12, 717652. [Google Scholar] [CrossRef] [PubMed]
- Cicek, M.; Unsal, V.; Doganer, A.; Demir, M. Investigation of oxidant/antioxidant and anti-inflammatory effects of apigenin on apoptosis in sepsis-induced rat lung. J. Biochem. Mol. Toxicol. 2021, 35, e22743. [Google Scholar] [CrossRef] [PubMed]
- Karamese, M.; Erol, H.S.; Albayrak, M.; Findik Guvendi, G.; Aydin, E.; Aksak Karamese, S. Anti-oxidant and anti-inflammatory effects of apigenin in a rat model of sepsis: An immunological, biochemical, and histopathological study. Immunopharmacol. Immunotoxicol. 2016, 38, 228–237. [Google Scholar] [CrossRef] [PubMed]
- Zhang, T.; Yan, T.; Du, J.; Wang, S.; Yang, H. Apigenin attenuates heart injury in lipopolysaccharide-induced endotoxemic model by suppressing sphingosine kinase 1/sphingosine 1-phosphate signaling pathway. Chem. Biol. Interact. 2015, 233, 46–55. [Google Scholar] [CrossRef]
- Li, F.; Lang, F.; Zhang, H.; Xu, L.; Wang, Y.; Zhai, C.; Hao, E. Apigenin Alleviates Endotoxin-Induced Myocardial Toxicity by Modulating Inflammation, Oxidative Stress, and Autophagy. Oxid. Med. Cell Longev. 2017, 2017, 2302896. [Google Scholar] [CrossRef]
- Berkoz, M.; Unal, S.; Karayakar, F.; Yunusoglu, O.; Ozkan-Yilmaz, F.; Ozluer-Hunt, A.; Aslan, A. Prophylactic effect of myricetin and apigenin against lipopolysaccharide-induced acute liver injury. Mol. Biol. Rep. 2021, 48, 6363–6373. [Google Scholar] [CrossRef]
- Zhou, R.J.; Zhao, Y.; Fan, K.; Xie, M.L. Protective effect of apigenin on d-galactosamine/LPS-induced hepatocellular injury by increment of Nrf-2 nucleus translocation. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 929–936. [Google Scholar] [CrossRef]
- Zhou, R.J.; Ye, H.; Wang, F.; Wang, J.L.; Xie, M.L. Apigenin inhibits d-galactosamine/LPS-induced liver injury through upregulation of hepatic Nrf-2 and PPARgamma expressions in mice. Biochem. Biophys. Res. Commun. 2017, 493, 625–630. [Google Scholar] [CrossRef]
- He, Y.; Xia, Z.; Yu, D.; Wang, J.; Jin, L.; Huang, D.; Ye, X.; Li, X.; Zhang, B. Hepatoprotective effects and structure-activity relationship of five flavonoids against lipopolysaccharide/d-galactosamine induced acute liver failure in mice. Int. Immunopharmacol. 2019, 68, 171–178. [Google Scholar] [CrossRef] [PubMed]
- Li, K.; He, Z.; Wang, X.; Pineda, M.; Chen, R.; Liu, H.; Ma, K.; Shen, H.; Wu, C.; Huang, N.; et al. Apigenin C-glycosides of Microcos paniculata protects lipopolysaccharide induced apoptosis and inflammation in acute lung injury through TLR4 signaling pathway. Free Radic. Biol. Med. 2018, 124, 163–175. [Google Scholar] [CrossRef] [PubMed]
- Li, K.C.; Ho, Y.L.; Hsieh, W.T.; Huang, S.S.; Chang, Y.S.; Huang, G.J. Apigenin-7-glycoside prevents LPS-induced acute lung injury via downregulation of oxidative enzyme expression and protein activation through inhibition of MAPK phosphorylation. Int. J. Mol. Sci. 2015, 16, 1736–1754. [Google Scholar] [CrossRef] [PubMed]
- Patil, R.H.; Babu, R.L.; Naveen Kumar, M.; Kiran Kumar, K.M.; Hegde, S.M.; Nagesh, R.; Ramesh, G.T.; Sharma, S.C. Anti-Inflammatory Effect of Apigenin on LPS-Induced Pro-Inflammatory Mediators and AP-1 Factors in Human Lung Epithelial Cells. Inflammation 2016, 39, 138–147. [Google Scholar] [CrossRef] [PubMed]
- Cardenas, H.; Arango, D.; Nicholas, C.; Duarte, S.; Nuovo, G.J.; He, W.; Voss, O.H.; Gonzalez-Mejia, M.E.; Guttridge, D.C.; Grotewold, E.; et al. Dietary Apigenin Exerts Immune-Regulatory Activity in Vivo by Reducing NF-kappaB Activity, Halting Leukocyte Infiltration and Restoring Normal Metabolic Function. Int. J. Mol. Sci. 2016, 17, 323. [Google Scholar] [CrossRef] [PubMed]
- Duarte, S.; Arango, D.; Parihar, A.; Hamel, P.; Yasmeen, R.; Doseff, A.I. Apigenin protects endothelial cells from lipopolysaccharide (LPS)-induced inflammation by decreasing caspase-3 activation and modulating mitochondrial function. Int. J. Mol. Sci. 2013, 14, 17664–17679. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.X.; Li, C.X.; Kakar, M.U.; Khan, M.S.; Wu, P.F.; Amir, R.M.; Dai, D.F.; Naveed, M.; Li, Q.Y.; Saeed, M.; et al. Resveratrol (RV): A pharmacological review and call for further research. Biomed. Pharmacother. 2021, 143, 112164. [Google Scholar] [CrossRef]
- Weiskirchen, S.; Weiskirchen, R. Resveratrol: How Much Wine Do You Have to Drink to Stay Healthy? Adv. Nutr. 2016, 7, 706–718. [Google Scholar] [CrossRef]
- Bi, X.L.; Yang, J.Y.; Dong, Y.X.; Wang, J.M.; Cui, Y.H.; Ikeshima, T.; Zhao, Y.Q.; Wu, C.F. Resveratrol inhibits nitric oxide and TNF-alpha production by lipopolysaccharide-activated microglia. Int. Immunopharmacol. 2005, 5, 185–193. [Google Scholar] [CrossRef]
- Pan, W.; Yu, H.; Huang, S.; Zhu, P. Resveratrol Protects against TNF-alpha-Induced Injury in Human Umbilical Endothelial Cells through Promoting Sirtuin-1-Induced Repression of NF-KB and p38 MAPK. PLoS ONE 2016, 11, e0147034. [Google Scholar] [CrossRef]
- Sebai, H.; Ben-Attia, M.; Sani, M.; Aouani, E.; Ghanem-Boughanmi, N. Protective effect of resveratrol in endotoxemia-induced acute phase response in rats. Arch. Toxicol. 2009, 83, 335–340. [Google Scholar] [CrossRef] [PubMed]
- Quan, H.; Yin, M.; Kim, J.; Jang, E.A.; Yang, S.H.; Bae, H.B.; Jeong, S. Resveratrol suppresses the reprogramming of macrophages into an endotoxin-tolerant state through the activation of AMP-activated protein kinase. Eur. J. Pharmacol. 2021, 899, 173993. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Zhang, Z.; Zhuo, Y.; Cui, L.; Li, C.; Li, D.; Zhang, S.; Cui, N.; Wang, X.; Gao, H. Resveratrol alleviates sepsis-induced acute lung injury by suppressing inflammation and apoptosis of alveolar macrophage cells. Am. J. Transl. Res. 2018, 10, 1961–1975. [Google Scholar] [PubMed]
- Wang, C.; Yuan, J.; Du, J. Resveratrol alleviates acute lung injury through regulating PLSCR-3-mediated mitochondrial dysfunction and mitophagy in a cecal ligation and puncture model. Eur. J. Pharmacol. 2021, 913, 174643. [Google Scholar] [CrossRef]
- Zeng, Y.; Cao, G.; Lin, L.; Zhang, Y.; Luo, X.; Ma, X.; Aiyisake, A.; Cheng, Q. Resveratrol Attenuates Sepsis-Induced Cardiomyopathy in Rats through Anti-Ferroptosis via the Sirt1/Nrf2 Pathway. J. Invest. Surg. 2023, 36, 2157521. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.S.; Zhao, H.L.; Yang, G.M.; Zang, J.T.; Zheng, D.Y.; Duan, C.Y.; Kuang, L.; Zhu, Y.; Wu, Y.; Li, T.; et al. Role of resveratrol in protecting vasodilatation function in septic shock rats and its mechanism. J. Trauma. Acute Care Surg. 2019, 87, 1336–1345. [Google Scholar] [CrossRef]
- Holthoff, J.H.; Wang, Z.; Seely, K.A.; Gokden, N.; Mayeux, P.R. Resveratrol improves renal microcirculation, protects the tubular epithelium, and prolongs survival in a mouse model of sepsis-induced acute kidney injury. Kidney Int. 2012, 81, 370–378. [Google Scholar] [CrossRef]
- Gan, Y.; Tao, S.; Cao, D.; Xie, H.; Zeng, Q. Protection of resveratrol on acute kidney injury in septic rats. Hum. Exp. Toxicol. 2017, 36, 1015–1022. [Google Scholar] [CrossRef]
- Wang, B.; Wang, Y.; Xu, K.; Zeng, Z.; Xu, Z.; Yue, D.; Li, T.; Luo, J.; Liu, J.; Yuan, J. Resveratrol alleviates sepsis-induced acute kidney injury by deactivating the lncRNA MALAT1/MiR-205 axis. Cent. Eur. J. Immunol. 2021, 46, 295–304. [Google Scholar] [CrossRef]
- Manchope, M.F.; Casagrande, R.; Verri, W.A., Jr. Naringenin: An analgesic and anti-inflammatory citrus flavanone. Oncotarget 2017, 8, 3766–3767. [Google Scholar] [CrossRef]
- Ye, G.; Wang, M.; Liu, D.; Cheng, L.; Yin, X.; Zhang, Q.; Liu, W. Mechanism of Naringenin Blocking the Protection of LTB4/BLT1 Receptor Against Septic Cardiac Dysfunction. Ann. Clin. Lab. Sci. 2020, 50, 769–774. [Google Scholar] [PubMed]
- Yu, D.H.; Ma, C.H.; Yue, Z.Q.; Yao, X.; Mao, C.M. Protective effect of naringenin against lipopolysaccharide-induced injury in normal human bronchial epithelium via suppression of MAPK signaling. Inflammation 2015, 38, 195–204. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Wang, N.; Fan, S.; Zheng, X.; Yang, Y.; Zhu, Y.; Lu, Y.; Chen, Q.; Zhou, H.; Zheng, J. The citrus flavonoid naringenin confers protection in a murine endotoxaemia model through AMPK-ATF3-dependent negative regulation of the TLR4 signalling pathway. Sci. Rep. 2016, 6, 39735. [Google Scholar] [CrossRef] [PubMed]
- Zhao, M.; Li, C.; Shen, F.; Wang, M.; Jia, N.; Wang, C. Naringenin ameliorates LPS-induced acute lung injury through its anti-oxidative and anti-inflammatory activity and by inhibition of the PI3K/AKT pathway. Exp. Ther. Med. 2017, 14, 2228–2234. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.J.; Qiao, W.; Xiao, Y.J.; Ren, W.D. Layer-specific strain for assessing the effect of naringin on systolic myocardial dysfunction induced by sepsis and its underlying mechanisms. J. Int. Med. Res. 2021, 49, 300060520986369. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Gao, M.; Yang, B.; Zhang, H.; Wang, K.; Liu, Z.; Xiao, X.; Yang, M. Naringin attenuates MLC phosphorylation and NF-kappaB activation to protect sepsis-induced intestinal injury via RhoA/ROCK pathway. Biomed. Pharmacother. 2018, 103, 50–58. [Google Scholar] [CrossRef]
- Sun, L.J.; Qiao, W.; Xiao, Y.J.; Cui, L.; Wang, X.; Ren, W.D. Naringin mitigates myocardial strain and the inflammatory response in sepsis-induced myocardial dysfunction through regulation of PI3K/AKT/NF-kappaB pathway. Int. Immunopharmacol. 2019, 75, 105782. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.L.; Yang, B.C.; Gao, M.; Xiao, X.F.; Zhao, S.P.; Liu, Z.L. Naringin improves sepsis-induced intestinal injury by modulating macrophage polarization via PPARgamma/miR-21 axis. Mol. Ther. Nucleic Acids 2021, 25, 502–514. [Google Scholar] [CrossRef] [PubMed]
- Abian, O.; Ortega-Alarcon, D.; Jimenez-Alesanco, A.; Ceballos-Laita, L.; Vega, S.; Reyburn, H.T.; Rizzuti, B.; Velazquez-Campoy, A. Structural stability of SARS-CoV-2 3CLpro and identification of quercetin as an inhibitor by experimental screening. Int. J. Biol. Macromol. 2020, 164, 1693–1703. [Google Scholar] [CrossRef]
- Gu, Y.Y.; Zhang, M.; Cen, H.; Wu, Y.F.; Lu, Z.; Lu, F.; Liu, X.S.; Lan, H.Y. Quercetin as a potential treatment for COVID-19-induced acute kidney injury: Based on network pharmacology and molecular docking study. PLoS ONE 2021, 16, e0245209. [Google Scholar] [CrossRef]
- Roy, A.V.; Chan, M.; Banadyga, L.; He, S.; Zhu, W.; Chretien, M.; Mbikay, M. Quercetin inhibits SARS-CoV-2 infection and prevents syncytium formation by cells co-expressing the viral spike protein and human ACE2. Virol. J. 2024, 21, 29. [Google Scholar] [CrossRef] [PubMed]
- Lee, C.C.E.; Ali, K.; Connell, D.; Mordi, I.R.; George, J.; Lang, E.M.; Lang, C.C. COVID-19-Associated Cardiovascular Complications. Diseases 2021, 9, 47. [Google Scholar] [CrossRef] [PubMed]
- Fang, J.Y.; Huang, K.Y.; Wang, T.H.; Lin, Z.C.; Chen, C.C.; Chang, S.Y.; Chen, E.L.; Chao, T.L.; Yang, S.C.; Yang, P.C.; et al. Development of nanoparticles incorporated with quercetin and ACE2-membrane as a novel therapy for COVID-19. J. Nanobiotechnol. 2024, 22, 169. [Google Scholar] [CrossRef]
- Xu, L.; Liu, J.; Lu, M.; Yang, D.; Zheng, X. Liver injury during highly pathogenic human coronavirus infections. Liver Int. 2020, 40, 998–1004. [Google Scholar] [CrossRef]
- Di Pierro, F.; Iqtadar, S.; Khan, A.; Ullah Mumtaz, S.; Masud Chaudhry, M.; Bertuccioli, A.; Derosa, G.; Maffioli, P.; Togni, S.; Riva, A.; et al. Potential Clinical Benefits of Quercetin in the Early Stage of COVID-19: Results of a Second, Pilot, Randomized, Controlled and Open-Label Clinical Trial. Int. J. Gen. Med. 2021, 14, 2807–2816. [Google Scholar] [CrossRef] [PubMed]
- Di Pierro, F.; Khan, A.; Iqtadar, S.; Mumtaz, S.U.; Chaudhry, M.N.A.; Bertuccioli, A.; Derosa, G.; Maffioli, P.; Togni, S.; Riva, A.; et al. Quercetin as a possible complementary agent for early-stage COVID-19: Concluding results of a randomized clinical trial. Front. Pharmacol. 2022, 13, 1096853. [Google Scholar] [CrossRef]
- Shohan, M.; Nashibi, R.; Mahmoudian-Sani, M.R.; Abolnezhadian, F.; Ghafourian, M.; Alavi, S.M.; Sharhani, A.; Khodadadi, A. The therapeutic efficacy of quercetin in combination with antiviral drugs in hospitalized COVID-19 patients: A randomized controlled trial. Eur. J. Pharmacol. 2022, 914, 174615. [Google Scholar] [CrossRef] [PubMed]
- Ujjan, I.D.; Khan, S.; Nigar, R.; Ahmed, H.; Ahmad, S.; Khan, A. The possible therapeutic role of curcumin and quercetin in the early-stage of COVID-19-Results from a pragmatic randomized clinical trial. Front. Nutr. 2022, 9, 1023997. [Google Scholar] [CrossRef]
- Rondanelli, M.; Perna, S.; Gasparri, C.; Petrangolini, G.; Allegrini, P.; Cavioni, A.; Faliva, M.A.; Mansueto, F.; Patelli, Z.; Peroni, G.; et al. Promising Effects of 3-Month Period of Quercetin Phytosome(R) Supplementation in the Prevention of Symptomatic COVID-19 Disease in Healthcare Workers: A Pilot Study. Life 2022, 12, 66. [Google Scholar] [CrossRef]
- Maurya, V.K.; Kumar, S.; Prasad, A.K.; Bhatt, M.L.B.; Saxena, S.K. Structure-based drug designing for potential antiviral activity of selected natural products from Ayurveda against SARS-CoV-2 spike glycoprotein and its cellular receptor. Virusdisease 2020, 31, 179–193. [Google Scholar] [CrossRef]
- Huynh, T.; Wang, H.; Luan, B. In Silico Exploration of the Molecular Mechanism of Clinically Oriented Drugs for Possibly Inhibiting SARS-CoV-2’s Main Protease. J. Phys. Chem. Lett. 2020, 11, 4413–4420. [Google Scholar] [CrossRef] [PubMed]
- Suravajhala, R.; Parashar, A.; Choudhir, G.; Kumar, A.; Malik, B.; Nagaraj, V.A.; Padmanaban, G.; Polavarapu, R.; Suravajhala, P.; Kishor, P.B.K. Molecular docking and dynamics studies of curcumin with COVID-19 proteins. Netw. Model. Anal. Health Inform. Bioinform. 2021, 10, 44. [Google Scholar] [CrossRef] [PubMed]
- Shafiee, A.; Athar, M.M.T.; Shahid, A.; Ghafoor, M.S.; Ayyan, M.; Zahid, A.; Cheema, H.A. Curcumin for the treatment of COVID-19 patients: A meta-analysis of randomized controlled trials. Phytother. Res. 2023, 37, 1167–1175. [Google Scholar] [CrossRef] [PubMed]
- Valizadeh, H.; Abdolmohammadi-Vahid, S.; Danshina, S.; Ziya Gencer, M.; Ammari, A.; Sadeghi, A.; Roshangar, L.; Aslani, S.; Esmaeilzadeh, A.; Ghaebi, M.; et al. Nano-curcumin therapy, a promising method in modulating inflammatory cytokines in COVID-19 patients. Int. Immunopharmacol. 2020, 89 Part B, 107088. [Google Scholar] [CrossRef]
- Tahmasebi, S.; Saeed, B.Q.; Temirgalieva, E.; Yumashev, A.V.; El-Esawi, M.A.; Navashenaq, J.G.; Valizadeh, H.; Sadeghi, A.; Aslani, S.; Yousefi, M.; et al. Nanocurcumin improves Treg cell responses in patients with mild and severe SARS-CoV-2. Life Sci. 2021, 276, 119437. [Google Scholar] [CrossRef] [PubMed]
- Ahmadi, R.; Salari, S.; Sharifi, M.D.; Reihani, H.; Rostamiani, M.B.; Behmadi, M.; Taherzadeh, Z.; Eslami, S.; Rezayat, S.M.; Jaafari, M.R.; et al. Oral nano-curcumin formulation efficacy in the management of mild to moderate outpatient COVID-19: A randomized triple-blind placebo-controlled clinical trial. Food Sci. Nutr. 2021, 9, 4068–4075. [Google Scholar] [CrossRef] [PubMed]
- Ahmad Merza Mohammad, T. Combining nano-curcumin with catechin improves COVID-19-infected patient’s inflammatory conditions. Hum. Immunol. 2023, 84, 471–483. [Google Scholar] [CrossRef] [PubMed]
- Hassaniazad, M.; Eftekhar, E.; Inchehsablagh, B.R.; Kamali, H.; Tousi, A.; Jaafari, M.R.; Rafat, M.; Fathalipour, M.; Nikoofal-Sahlabadi, S.; Gouklani, H.; et al. A triple-blind, placebo-controlled, randomized clinical trial to evaluate the effect of curcumin-containing nanomicelles on cellular immune responses subtypes and clinical outcome in COVID-19 patients. Phytother. Res. 2021, 35, 6417–6427. [Google Scholar] [CrossRef] [PubMed]
- Askari, G.; Bagherniya, M.; Kiani, Z.; Alikiaii, B.; Mirjalili, M.; Shojaei, M.; Hassanizadeh, S.; Vajdi, M.; Feizi, A.; Majeed, M.; et al. Evaluation of Curcumin-Piperine Supplementation in COVID-19 Patients Admitted to the Intensive Care: A Double-Blind, Randomized Controlled Trial. Adv. Exp. Med. Biol. 2023, 1412, 413–426. [Google Scholar] [CrossRef]
- Widjaja, S.S.; Rusdiana, R.; Amelia, R. Curcumin: Boosting the immunity of COVID-19-vaccinated populations. J. Adv. Pharm. Technol. Res. 2022, 13, 187–190. [Google Scholar] [CrossRef]
- Fessler, S.N.; Chang, Y.; Liu, L.; Johnston, C.S. Curcumin Confers Anti-Inflammatory Effects in Adults Who Recovered from COVID-19 and Were Subsequently Vaccinated: A Randomized Controlled Trial. Nutrients 2023, 15, 1548. [Google Scholar] [CrossRef]
- Yu, R.; Chen, L.; Lan, R.; Shen, R.; Li, P. Computational screening of antagonists against the SARS-CoV-2 (COVID-19) coronavirus by molecular docking. Int. J. Antimicrob. Agents 2020, 56, 106012. [Google Scholar] [CrossRef]
- Munafo, F.; Donati, E.; Brindani, N.; Ottonello, G.; Armirotti, A.; De Vivo, M. Quercetin and luteolin are single-digit micromolar inhibitors of the SARS-CoV-2 RNA-dependent RNA polymerase. Sci. Rep. 2022, 12, 10571. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Yang, C.; Xia, J.; Li, N.; Xiong, W. Luteolin is a potential inhibitor of COVID-19: An in silico analysis. Medicine 2023, 102, e35029. [Google Scholar] [CrossRef] [PubMed]
- Alzaabi, M.M.; Hamdy, R.; Ashmawy, N.S.; Hamoda, A.M.; Alkhayat, F.; Khademi, N.N.; Al Joud, S.M.A.; El-Keblawy, A.A.; Soliman, S.S.M. Flavonoids are promising safe therapy against COVID-19. Phytochem. Rev. 2022, 21, 291–312. [Google Scholar] [CrossRef]
- Shawan, M.; Halder, S.K.; Hasan, M.A. Luteolin and abyssinone II as potential inhibitors of SARS-CoV-2: An in silico molecular modeling approach in battling the COVID-19 outbreak. Bull. Natl. Res. Cent. 2021, 45, 27. [Google Scholar] [CrossRef]
- Xie, Y.Z.; Peng, C.W.; Su, Z.Q.; Huang, H.T.; Liu, X.H.; Zhan, S.F.; Huang, X.F. A Practical Strategy for Exploring the Pharmacological Mechanism of Luteolin Against COVID-19/Asthma Comorbidity: Findings of System Pharmacology and Bioinformatics Analysis. Front. Immunol. 2021, 12, 769011. [Google Scholar] [CrossRef]
- Di Stadio, A.; Gallina, S.; Cocuzza, S.; De Luca, P.; Ingrassia, A.; Oliva, S.; Sireci, F.; Camaioni, A.; Ferreli, F.; Mercante, G.; et al. Treatment of COVID-19 olfactory dysfunction with olfactory training, palmitoylethanolamide with luteolin, or combined therapy: A blinded controlled multicenter randomized trial. Eur. Arch. Otorhinolaryngol. 2023, 280, 4949–4961. [Google Scholar] [CrossRef] [PubMed]
- D’Ascanio, L.; Vitelli, F.; Cingolani, C.; Maranzano, M.; Brenner, M.J.; Di Stadio, A. Randomized clinical trial “olfactory dysfunction after COVID-19: Olfactory rehabilitation therapy vs. intervention treatment with Palmitoylethanolamide and Luteolin”: Preliminary results. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 4156–4162. [Google Scholar] [CrossRef]
- Versace, V.; Ortelli, P.; Dezi, S.; Ferrazzoli, D.; Alibardi, A.; Bonini, I.; Engl, M.; Maestri, R.; Assogna, M.; Ajello, V.; et al. Co-ultramicronized palmitoylethanolamide/luteolin normalizes GABA(B)-ergic activity and cortical plasticity in long COVID-19 syndrome. Clin. Neurophysiol. 2023, 145, 81–88. [Google Scholar] [CrossRef]
- De Luca, P.; Camaioni, A.; Marra, P.; Salzano, G.; Carriere, G.; Ricciardi, L.; Pucci, R.; Montemurro, N.; Brenner, M.J.; Di Stadio, A. Effect of Ultra-Micronized Palmitoylethanolamide and Luteolin on Olfaction and Memory in Patients with Long COVID: Results of a Longitudinal Study. Cells 2022, 11, 2552. [Google Scholar] [CrossRef] [PubMed]
- Farhat, A.; Ben Hlima, H.; Khemakhem, B.; Ben Halima, Y.; Michaud, P.; Abdelkafi, S.; Fendri, I. Apigenin analogues as SARS-CoV-2 main protease inhibitors: In-silico screening approach. Bioengineered 2022, 13, 3350–3361. [Google Scholar] [CrossRef] [PubMed]
- Hossain, M.A.; Rahman, M.H.; Sultana, H.; Ahsan, A.; Rayhan, S.I.; Hasan, M.I.; Sohel, M.; Somadder, P.D.; Moni, M.A. An integrated in-silico Pharmaco-BioInformatics approaches to identify synergistic effects of COVID-19 to HIV patients. Comput. Biol. Med. 2023, 155, 106656. [Google Scholar] [CrossRef] [PubMed]
- Jo, S.; Kim, S.; Shin, D.H.; Kim, M.S. Inhibition of SARS-CoV 3CL protease by flavonoids. J. Enzyme Inhib. Med. Chem. 2020, 35, 145–151. [Google Scholar] [CrossRef] [PubMed]
- Muhammad, S.; Hassan, S.H.; Al-Sehemi, A.G.; Shakir, H.A.; Khan, M.; Irfan, M.; Iqbal, J. Exploring the new potential antiviral constituents of Moringa oliefera for SARS-CoV-2 pathogenesis: An in silico molecular docking and dynamic studies. Chem. Phys. Lett. 2021, 767, 138379. [Google Scholar] [CrossRef]
- Xiong, Y.; Zhu, G.H.; Wang, H.N.; Hu, Q.; Chen, L.L.; Guan, X.Q.; Li, H.L.; Chen, H.Z.; Tang, H.; Ge, G.B. Discovery of naturally occurring inhibitors against SARS-CoV-2 3CL(pro) from Ginkgo biloba leaves via large-scale screening. Fitoterapia 2021, 152, 104909. [Google Scholar] [CrossRef] [PubMed]
- Patel, H.; Chandra, S.; Alexander, S.; Soble, J.; Williams, K.A., Sr. Plant-Based Nutrition: An Essential Component of Cardiovascular Disease Prevention and Management. Curr. Cardiol. Rep. 2017, 19, 104. [Google Scholar] [CrossRef] [PubMed]
- Albohy, A.; Zahran, E.M.; Abdelmohsen, U.R.; Salem, M.A.; Al-Warhi, T.; Al-Sanea, M.M.; Abelyan, N.; Khalil, H.E.; Desoukey, S.Y.; Fouad, M.A.; et al. Multitarget in silico studies of Ocimum menthiifolium, family Lamiaceae against SARS-CoV-2 supported by molecular dynamics simulation. J. Biomol. Struct. Dyn. 2022, 40, 4062–4072. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.; Chen, D.Y.; Scartelli, C.; Xie, H.; Merrill-Skoloff, G.; Yang, M.; Sun, L.; Saeed, M.; Flaumenhaft, R. Plant flavonoid inhibition of SARS-CoV-2 main protease and viral replication. iScience 2023, 26, 107602. [Google Scholar] [CrossRef]
- Chaves, O.A.; Fintelman-Rodrigues, N.; Wang, X.; Sacramento, C.Q.; Temerozo, J.R.; Ferreira, A.C.; Mattos, M.; Pereira-Dutra, F.; Bozza, P.T.; Castro-Faria-Neto, H.C.; et al. Commercially Available Flavonols Are Better SARS-CoV-2 Inhibitors than Isoflavone and Flavones. Viruses 2022, 14, 1458. [Google Scholar] [CrossRef]
- Ter Ellen, B.M.; Dinesh Kumar, N.; Bouma, E.M.; Troost, B.; van de Pol, D.P.I.; van der Ende-Metselaar, H.H.; Apperloo, L.; van Gosliga, D.; van den Berge, M.; Nawijn, M.C.; et al. Resveratrol and Pterostilbene Inhibit SARS-CoV-2 Replication in Air-Liquid Interface Cultured Human Primary Bronchial Epithelial Cells. Viruses 2021, 13, 1335. [Google Scholar] [CrossRef] [PubMed]
- Pasquereau, S.; Nehme, Z.; Haidar Ahmad, S.; Daouad, F.; Van Assche, J.; Wallet, C.; Schwartz, C.; Rohr, O.; Morot-Bizot, S.; Herbein, G. Resveratrol Inhibits HCoV-229E and SARS-CoV-2 Coronavirus Replication In Vitro. Viruses 2021, 13, 354. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Li, J.; Song, S.; Xiao, Z.; Chen, X.; Huang, B.; Sun, M.; Su, G.; Zhou, D.; Wang, G.; et al. Effective inhibition of coronavirus replication by Polygonum cuspidatum. Front. Biosci. Landmark Ed. 2021, 26, 789–798. [Google Scholar] [CrossRef] [PubMed]
- Xiao, Z.; Ye, Q.; Duan, X.; Xiang, T. Network Pharmacology Reveals That Resveratrol Can Alleviate COVID-19-Related Hyperinflammation. Dis. Markers 2021, 2021, 4129993. [Google Scholar] [CrossRef]
- McCreary, M.R.; Schnell, P.M.; Rhoda, D.A. Randomized double-blind placebo-controlled proof-of-concept trial of resveratrol for outpatient treatment of mild coronavirus disease (COVID-19). Sci. Rep. 2022, 12, 10978. [Google Scholar] [CrossRef]
- Prasetyo, W.E.; Purnomo, H.; Sadrini, M.; Wibowo, F.R.; Firdaus, M.; Kusumaningsih, T. Identification of potential bioactive natural compounds from Indonesian medicinal plants against 3-chymotrypsin-like protease (3CL(pro)) of SARS-CoV-2: Molecular docking, ADME/T, molecular dynamic simulations, and DFT analysis. J. Biomol. Struct. Dyn. 2023, 41, 4467–4484. [Google Scholar] [CrossRef]
- Xia, Q.D.; Xun, Y.; Lu, J.L.; Lu, Y.C.; Yang, Y.Y.; Zhou, P.; Hu, J.; Li, C.; Wang, S.G. Network pharmacology and molecular docking analyses on Lianhua Qingwen capsule indicate Akt1 is a potential target to treat and prevent COVID-19. Cell Prolif. 2020, 53, e12949. [Google Scholar] [CrossRef]
- Aleebrahim-Dehkordi, E.; Ghoshouni, H.; Koochaki, P.; Esmaili-Dehkordi, M.; Aleebrahim, E.; Chichagi, F.; Jafari, A.; Hanaei, S.; Heidari-Soureshjani, E.; Rezaei, N. Targeting the vital non-structural proteins (NSP12, NSP7, NSP8 and NSP3) from SARS-CoV-2 and inhibition of RNA polymerase by natural bioactive compound naringenin as a promising drug candidate against COVID-19. J. Mol. Struct. 2023, 1287, 135642. [Google Scholar] [CrossRef]
- Clementi, N.; Scagnolari, C.; D’Amore, A.; Palombi, F.; Criscuolo, E.; Frasca, F.; Pierangeli, A.; Mancini, N.; Antonelli, G.; Clementi, M.; et al. Naringenin is a powerful inhibitor of SARS-CoV-2 infection in vitro. Pharmacol. Res. 2021, 163, 105255. [Google Scholar] [CrossRef]
- Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of Biological Importance of Quercetin: A Bioactive Flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar] [CrossRef]
- Mullen, W.; Edwards, C.A.; Crozier, A. Absorption, excretion and metabolite profiling of methyl-, glucuronyl-, glucosyl- and sulpho-conjugates of quercetin in human plasma and urine after ingestion of onions. Br. J. Nutr. 2006, 96, 107–116. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Yin, O.Q.; Zuo, Z.; Chow, M.S. Pharmacokinetics and modeling of quercetin and metabolites. Pharm. Res. 2005, 22, 892–901. [Google Scholar] [CrossRef] [PubMed]
- de Vries, J.H.; Hollman, P.C.; Meyboom, S.; Buysman, M.N.; Zock, P.L.; van Staveren, W.A.; Katan, M.B. Plasma concentrations and urinary excretion of the antioxidant flavonols quercetin and kaempferol as biomarkers for dietary intake. Am. J. Clin. Nutr. 1998, 68, 60–65. [Google Scholar] [CrossRef] [PubMed]
- de Vries, J.H.; Hollman, P.C.; van Amersfoort, I.; Olthof, M.R.; Katan, M.B. Red wine is a poor source of bioavailable flavonols in men. J. Nutr. 2001, 131, 745–748. [Google Scholar] [CrossRef] [PubMed]
- Riva, A.; Ronchi, M.; Petrangolini, G.; Bosisio, S.; Allegrini, P. Improved Oral Absorption of Quercetin from Quercetin Phytosome(R), a New Delivery System Based on Food Grade Lecithin. Eur. J. Drug Metab. Pharmacokinet. 2019, 44, 169–177. [Google Scholar] [CrossRef] [PubMed]
- Aghababaei, F.; Hadidi, M. Recent Advances in Potential Health Benefits of Quercetin. Pharmaceuticals 2023, 16, 20. [Google Scholar] [CrossRef]
- Qi, W.; Qi, W.; Xiong, D.; Long, M. Quercetin: Its Antioxidant Mechanism, Antibacterial Properties and Potential Application in Prevention and Control of Toxipathy. Molecules 2022, 27, 6545. [Google Scholar] [CrossRef]
- Di Petrillo, A.; Orru, G.; Fais, A.; Fantini, M.C. Quercetin and its derivates as antiviral potentials: A comprehensive review. Phytother. Res. 2022, 36, 266–278. [Google Scholar] [CrossRef]
- Warren, C.A.; Paulhill, K.J.; Davidson, L.A.; Lupton, J.R.; Taddeo, S.S.; Hong, M.Y.; Carroll, R.J.; Chapkin, R.S.; Turner, N.D. Quercetin may suppress rat aberrant crypt foci formation by suppressing inflammatory mediators that influence proliferation and apoptosis. J. Nutr. 2009, 139, 101–105. [Google Scholar] [CrossRef]
- Ghosh, N.; Chakraborty, T.; Mallick, S.; Mana, S.; Singha, D.; Ghosh, B.; Roy, S. Synthesis, characterization and study of antioxidant activity of quercetin-magnesium complex. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2015, 151, 807–813. [Google Scholar] [CrossRef]
- Wang, S.; Yao, J.; Zhou, B.; Yang, J.; Chaudry, M.T.; Wang, M.; Xiao, F.; Li, Y.; Yin, W. Bacteriostatic Effect of Quercetin as an Antibiotic Alternative In Vivo and Its Antibacterial Mechanism In Vitro. J. Food Prot. 2018, 81, 68–78. [Google Scholar] [CrossRef] [PubMed]
- Edwards, R.L.; Lyon, T.; Litwin, S.E.; Rabovsky, A.; Symons, J.D.; Jalili, T. Quercetin reduces blood pressure in hypertensive subjects. J. Nutr. 2007, 137, 2405–2411. [Google Scholar] [CrossRef] [PubMed]
- Egert, S.; Bosy-Westphal, A.; Seiberl, J.; Kurbitz, C.; Settler, U.; Plachta-Danielzik, S.; Wagner, A.E.; Frank, J.; Schrezenmeir, J.; Rimbach, G.; et al. Quercetin reduces systolic blood pressure and plasma oxidised low-density lipoprotein concentrations in overweight subjects with a high-cardiovascular disease risk phenotype: A double-blinded, placebo-controlled cross-over study. Br. J. Nutr. 2009, 102, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
- Lee, T.J.; Kim, O.H.; Kim, Y.H.; Lim, J.H.; Kim, S.; Park, J.W.; Kwon, T.K. Quercetin arrests G2/M phase and induces caspase-dependent cell death in U937 cells. Cancer Lett. 2006, 240, 234–242. [Google Scholar] [CrossRef] [PubMed]
- Chou, C.C.; Yang, J.S.; Lu, H.F.; Ip, S.W.; Lo, C.; Wu, C.C.; Lin, J.P.; Tang, N.Y.; Chung, J.G.; Chou, M.J.; et al. Quercetin-mediated cell cycle arrest and apoptosis involving activation of a caspase cascade through the mitochondrial pathway in human breast cancer MCF-7 cells. Arch. Pharm. Res. 2010, 33, 1181–1191. [Google Scholar] [CrossRef]
- Gugliandolo, E.; Peritore, A.F.; D’Amico, R.; Licata, P.; Crupi, R. Evaluation of Neuroprotective Effects of Quercetin against Aflatoxin B1-Intoxicated Mice. Animals 2020, 10, 898. [Google Scholar] [CrossRef]
- Ben Salem, I.; Prola, A.; Boussabbeh, M.; Guilbert, A.; Bacha, H.; Lemaire, C.; Abid-Essefi, S. Activation of ER stress and apoptosis by alpha- and beta-zearalenol in HCT116 cells, protective role of Quercetin. Neurotoxicology 2016, 53, 334–342. [Google Scholar] [CrossRef] [PubMed]
- Nieman, D.C.; Henson, D.A.; Maxwell, K.R.; Williams, A.S.; McAnulty, S.R.; Jin, F.; Shanely, R.A.; Lines, T.C. Effects of quercetin and EGCG on mitochondrial biogenesis and immunity. Med. Sci. Sports Exerc. 2009, 41, 1467–1475. [Google Scholar] [CrossRef]
- Denny Joseph, K.M.; Muralidhara. Enhanced neuroprotective effect of fish oil in combination with quercetin against 3-nitropropionic acid induced oxidative stress in rat brain. Prog. Neuropsychopharmacol. Biol. Psychiatry 2013, 40, 83–92. [Google Scholar] [CrossRef]
- Karimi, A.; Naeini, F.; Asghari Azar, V.; Hasanzadeh, M.; Ostadrahimi, A.; Niazkar, H.R.; Mobasseri, M.; Tutunchi, H. A comprehensive systematic review of the therapeutic effects and mechanisms of action of quercetin in sepsis. Phytomedicine 2021, 86, 153567. [Google Scholar] [CrossRef]
- Gasmi, A.; Mujawdiya, P.K.; Lysiuk, R.; Shanaida, M.; Peana, M.; Gasmi Benahmed, A.; Beley, N.; Kovalska, N.; Bjorklund, G. Quercetin in the Prevention and Treatment of Coronavirus Infections: A Focus on SARS-CoV-2. Pharmaceuticals 2022, 15, 1049. [Google Scholar] [CrossRef]
- Lee, S.; Yu, Y.; Trimpert, J.; Benthani, F.; Mairhofer, M.; Richter-Pechanska, P.; Wyler, E.; Belenki, D.; Kaltenbrunner, S.; Pammer, M.; et al. Virus-induced senescence is a driver and therapeutic target in COVID-19. Nature 2021, 599, 283–289. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Li, S.; Liu, J.; Cheng, J.; Kang, L.; Li, W.; Zhong, Y.; Wei, C.; Fu, L.; Qi, J.; et al. Bioactive compounds from Huashi Baidu decoction possess both antiviral and anti-inflammatory effects against COVID-19. Proc. Natl. Acad. Sci. USA 2023, 120, e2301775120. [Google Scholar] [CrossRef]
- Imran, M.; Thabet, H.K.; Alaqel, S.I.; Alzahrani, A.R.; Abida, A.; Alshammari, M.K.; Kamal, M.; Diwan, A.; Asdaq, S.M.B.; Alshehri, S. The Therapeutic and Prophylactic Potential of Quercetin against COVID-19: An Outlook on the Clinical Studies, Inventive Compositions, and Patent Literature. Antioxidants 2022, 11, 876. [Google Scholar] [CrossRef]
- Nelson, K.M.; Dahlin, J.L.; Bisson, J.; Graham, J.; Pauli, G.F.; Walters, M.A. The Essential Medicinal Chemistry of Curcumin. J. Med. Chem. 2017, 60, 1620–1637. [Google Scholar] [CrossRef] [PubMed]
- Sharifi-Rad, J.; Rayess, Y.E.; Rizk, A.A.; Sadaka, C.; Zgheib, R.; Zam, W.; Sestito, S.; Rapposelli, S.; Neffe-Skocinska, K.; Zielinska, D.; et al. Turmeric and Its Major Compound Curcumin on Health: Bioactive Effects and Safety Profiles for Food, Pharmaceutical, Biotechnological and Medicinal Applications. Front. Pharmacol. 2020, 11, 01021. [Google Scholar] [CrossRef] [PubMed]
- Dempe, J.S.; Scheerle, R.K.; Pfeiffer, E.; Metzler, M. Metabolism and permeability of curcumin in cultured Caco-2 cells. Mol. Nutr. Food Res. 2013, 57, 1543–1549. [Google Scholar] [CrossRef]
- Holder, G.M.; Plummer, J.L.; Ryan, A.J. The metabolism and excretion of curcumin (1,7-bis-(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione) in the rat. Xenobiotica 1978, 8, 761–768. [Google Scholar] [CrossRef]
- Pfeiffer, E.; Hoehle, S.I.; Walch, S.G.; Riess, A.; Solyom, A.M.; Metzler, M. Curcuminoids form reactive glucuronides in vitro. J. Agric. Food Chem. 2007, 55, 538–544. [Google Scholar] [CrossRef]
- Metzler, M.; Pfeiffer, E.; Schulz, S.I.; Dempe, J.S. Curcumin uptake and metabolism. Biofactors 2013, 39, 14–20. [Google Scholar] [CrossRef]
- Garcea, G.; Berry, D.P.; Jones, D.J.; Singh, R.; Dennison, A.R.; Farmer, P.B.; Sharma, R.A.; Steward, W.P.; Gescher, A.J. Consumption of the putative chemopreventive agent curcumin by cancer patients: Assessment of curcumin levels in the colorectum and their pharmacodynamic consequences. Cancer Epidemiol. Biomark. Prev. 2005, 14, 120–125. [Google Scholar] [CrossRef]
- Shoba, G.; Joy, D.; Joseph, T.; Majeed, M.; Rajendran, R.; Srinivas, P.S. Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med. 1998, 64, 353–356. [Google Scholar] [CrossRef]
- Verma, S.P.; Salamone, E.; Goldin, B. Curcumin and genistein, plant natural products, show synergistic inhibitory effects on the growth of human breast cancer MCF-7 cells induced by estrogenic pesticides. Biochem. Biophys. Res. Commun. 1997, 233, 692–696. [Google Scholar] [CrossRef]
- Anand, P.; Kunnumakkara, A.B.; Newman, R.A.; Aggarwal, B.B. Bioavailability of curcumin: Problems and promises. Mol. Pharm. 2007, 4, 807–818. [Google Scholar] [CrossRef] [PubMed]
- Bisht, S.; Feldmann, G.; Soni, S.; Ravi, R.; Karikar, C.; Maitra, A.; Maitra, A. Polymeric nanoparticle-encapsulated curcumin (“nanocurcumin”): A novel strategy for human cancer therapy. J. Nanobiotechnol. 2007, 5, 3. [Google Scholar] [CrossRef]
- Suresh, D.; Srinivasan, K. Studies on the in vitro absorption of spice principles--curcumin, capsaicin and piperine in rat intestines. Food Chem. Toxicol. 2007, 45, 1437–1442. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Braiteh, F.S.; Kurzrock, R. Liposome-encapsulated curcumin: In vitro and in vivo effects on proliferation, apoptosis, signaling, and angiogenesis. Cancer 2005, 104, 1322–1331. [Google Scholar] [CrossRef]
- Ma, Z.; Shayeganpour, A.; Brocks, D.R.; Lavasanifar, A.; Samuel, J. High-performance liquid chromatography analysis of curcumin in rat plasma: Application to pharmacokinetics of polymeric micellar formulation of curcumin. Biomed. Chromatogr. 2007, 21, 546–552. [Google Scholar] [CrossRef]
- Swamy, A.V.; Gulliaya, S.; Thippeswamy, A.; Koti, B.C.; Manjula, D.V. Cardioprotective effect of curcumin against doxorubicin-induced myocardial toxicity in albino rats. Indian J. Pharmacol. 2012, 44, 73–77. [Google Scholar] [CrossRef]
- Murugan, P.; Pari, L. Influence of tetrahydrocurcumin on hepatic and renal functional markers and protein levels in experimental type 2 diabetic rats. Basic Clin. Pharmacol. Toxicol. 2007, 101, 241–245. [Google Scholar] [CrossRef]
- Belcaro, G.; Cesarone, M.R.; Dugall, M.; Pellegrini, L.; Ledda, A.; Grossi, M.G.; Togni, S.; Appendino, G. Efficacy and safety of Meriva(R), a curcumin-phosphatidylcholine complex, during extended administration in osteoarthritis patients. Altern. Med. Rev. 2010, 15, 337–344. [Google Scholar]
- Lopez-Lazaro, M. Anticancer and carcinogenic properties of curcumin: Considerations for its clinical development as a cancer chemopreventive and chemotherapeutic agent. Mol. Nutr. Food Res. 2008, 52 (Suppl. S1), S103–S127. [Google Scholar] [CrossRef]
- Sreejayan; Rao, M.N. Nitric oxide scavenging by curcuminoids. J. Pharm. Pharmacol. 1997, 49, 105–107. [Google Scholar] [CrossRef]
- Brouet, I.; Ohshima, H. Curcumin, an anti-tumour promoter and anti-inflammatory agent, inhibits induction of nitric oxide synthase in activated macrophages. Biochem. Biophys. Res. Commun. 1995, 206, 533–540. [Google Scholar] [CrossRef]
- De, R.; Kundu, P.; Swarnakar, S.; Ramamurthy, T.; Chowdhury, A.; Nair, G.B.; Mukhopadhyay, A.K. Antimicrobial activity of curcumin against Helicobacter pylori isolates from India and during infections in mice. Antimicrob. Agents Chemother. 2009, 53, 1592–1597. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.K.; Choi, G.J.; Lee, H.S. Fungicidal property of Curcuma longa L. rhizome-derived curcumin against phytopathogenic fungi in a greenhouse. J. Agric. Food Chem. 2003, 51, 1578–1581. [Google Scholar] [CrossRef] [PubMed]
- Si, X.; Wang, Y.; Wong, J.; Zhang, J.; McManus, B.M.; Luo, H. Dysregulation of the ubiquitin-proteasome system by curcumin suppresses coxsackievirus B3 replication. J. Virol. 2007, 81, 3142–3150. [Google Scholar] [CrossRef] [PubMed]
- Gupta, S.C.; Patchva, S.; Aggarwal, B.B. Therapeutic roles of curcumin: Lessons learned from clinical trials. AAPS J. 2013, 15, 195–218. [Google Scholar] [CrossRef] [PubMed]
- Nirmala, C.; Puvanakrishnan, R. Protective role of curcumin against isoproterenol induced myocardial infarction in rats. Mol. Cell Biochem. 1996, 159, 85–93. [Google Scholar] [CrossRef]
- Fiala, M.; Liu, P.T.; Espinosa-Jeffrey, A.; Rosenthal, M.J.; Bernard, G.; Ringman, J.M.; Sayre, J.; Zhang, L.; Zaghi, J.; Dejbakhsh, S.; et al. Innate immunity and transcription of MGAT-III and Toll-like receptors in Alzheimer’s disease patients are improved by bisdemethoxycurcumin. Proc. Natl. Acad. Sci. USA 2007, 104, 12849–12854. [Google Scholar] [CrossRef]
- Holt, P.R.; Katz, S.; Kirshoff, R. Curcumin therapy in inflammatory bowel disease: A pilot study. Dig. Dis. Sci. 2005, 50, 2191–2193. [Google Scholar] [CrossRef]
- Chandran, B.; Goel, A. A randomized, pilot study to assess the efficacy and safety of curcumin in patients with active rheumatoid arthritis. Phytother. Res. 2012, 26, 1719–1725. [Google Scholar] [CrossRef]
- Chuengsamarn, S.; Rattanamongkolgul, S.; Luechapudiporn, R.; Phisalaphong, C.; Jirawatnotai, S. Curcumin extract for prevention of type 2 diabetes. Diabetes Care 2012, 35, 2121–2127. [Google Scholar] [CrossRef]
- Soni, K.B.; Kuttan, R. Effect of oral curcumin administration on serum peroxides and cholesterol levels in human volunteers. Indian. J. Physiol. Pharmacol. 1992, 36, 273–275. [Google Scholar]
- Vieira, B.M.; Caetano, M.A.F.; de Carvalho, M.T.; Dos Santos Arruda, F.; Tome, F.D.; de Oliveira, J.F.; Soave, D.F.; Pereira, J.X.; Celes, M.R.N. Impacts of Curcumin Treatment on Experimental Sepsis: A Systematic Review. Oxid. Med. Cell Longev. 2023, 2023, 2252213. [Google Scholar] [CrossRef]
- Shojaei, M.; Foshati, S.; Abdi, M.; Askari, G.; Sukhorukov, V.N.; Bagherniya, M.; Sahebkar, A. The effectiveness of nano-curcumin on patients with COVID-19: A systematic review of clinical trials. Phytother. Res. 2023, 37, 1663–1677. [Google Scholar] [CrossRef]
- Hatamipour, M.; Sahebkar, A.; Alavizadeh, S.H.; Dorri, M.; Jaafari, M.R. Novel nanomicelle formulation to enhance bioavailability and stability of curcuminoids. Iran. J. Basic. Med. Sci. 2019, 22, 282–289. [Google Scholar] [CrossRef]
- Taheri, Y.; Sharifi-Rad, J.; Antika, G.; Yilmaz, Y.B.; Tumer, T.B.; Abuhamdah, S.; Chandra, S.; Saklani, S.; Kilic, C.S.; Sestito, S.; et al. Paving Luteolin Therapeutic Potentialities and Agro-Food-Pharma Applications: Emphasis on In Vivo Pharmacological Effects and Bioavailability Traits. Oxid. Med. Cell Longev. 2021, 2021, 1987588. [Google Scholar] [CrossRef] [PubMed]
- Hostetler, G.L.; Ralston, R.A.; Schwartz, S.J. Flavones: Food Sources, Bioavailability, Metabolism, and Bioactivity. Adv. Nutr. 2017, 8, 423–435. [Google Scholar] [CrossRef] [PubMed]
- Hu, M.; Chen, J.; Lin, H. Metabolism of flavonoids via enteric recycling: Mechanistic studies of disposition of apigenin in the Caco-2 cell culture model. J. Pharmacol. Exp. Ther. 2003, 307, 314–321. [Google Scholar] [CrossRef] [PubMed]
- Thilakarathna, S.H.; Rupasinghe, H.P. Flavonoid bioavailability and attempts for bioavailability enhancement. Nutrients 2013, 5, 3367–3387. [Google Scholar] [CrossRef]
- Miyashita, A.; Ito, J.; Parida, I.S.; Syoji, N.; Fujii, T.; Takahashi, H.; Nakagawa, K. Improving water dispersibility and bioavailability of luteolin using microemulsion system. Sci. Rep. 2022, 12, 11949. [Google Scholar] [CrossRef]
- Lo, S.; Leung, E.; Fedrizzi, B.; Barker, D. Syntheses of mono-acylated luteolin derivatives, evaluation of their antiproliferative and radical scavenging activities and implications on their oral bioavailability. Sci. Rep. 2021, 11, 12595. [Google Scholar] [CrossRef]
- Liu, J.; Sun, Y.; Cheng, M.; Liu, Q.; Liu, W.; Gao, C.; Feng, J.; Jin, Y.; Tu, L. Improving Oral Bioavailability of Luteolin Nanocrystals by Surface Modification of Sodium Dodecyl Sulfate. AAPS PharmSciTech 2021, 22, 133. [Google Scholar] [CrossRef]
- Zhang, M.; Wang, R.; Tian, J.; Song, M.; Zhao, R.; Liu, K.; Zhu, F.; Shim, J.H.; Dong, Z.; Lee, M.H. Targeting LIMK1 with luteolin inhibits the growth of lung cancer in vitro and in vivo. J. Cell Mol. Med. 2021, 25, 5560–5571. [Google Scholar] [CrossRef]
- Ziyan, L.; Yongmei, Z.; Nan, Z.; Ning, T.; Baolin, L. Evaluation of the anti-inflammatory activity of luteolin in experimental animal models. Planta Med. 2007, 73, 221–226. [Google Scholar] [CrossRef]
- Guo, Y.; Liu, Y.; Zhang, Z.; Chen, M.; Zhang, D.; Tian, C.; Liu, M.; Jiang, G. The Antibacterial Activity and Mechanism of Action of Luteolin Against Trueperella pyogenes. Infect. Drug Resist. 2020, 13, 1697–1711. [Google Scholar] [CrossRef]
- Men, X.; Li, S.; Cai, X.; Fu, L.; Shao, Y.; Zhu, Y. Antiviral Activity of Luteolin against Pseudorabies Virus In Vitro and In Vivo. Animals 2023, 13, 761. [Google Scholar] [CrossRef]
- Harris, G.K.; Qian, Y.; Leonard, S.S.; Sbarra, D.C.; Shi, X. Luteolin and chrysin differentially inhibit cyclooxygenase-2 expression and scavenge reactive oxygen species but similarly inhibit prostaglandin-E2 formation in RAW 264.7 cells. J. Nutr. 2006, 136, 1517–1521. [Google Scholar] [CrossRef] [PubMed]
- Jang, C.H.; Moon, N.; Oh, J.; Kim, J.S. Luteolin Shifts Oxaliplatin-Induced Cell Cycle Arrest at G(0)/G(1) to Apoptosis in HCT116 Human Colorectal Carcinoma Cells. Nutrients 2019, 11, 770. [Google Scholar] [CrossRef]
- Dong, R.; Huang, R.; Shi, X.; Xu, Z.; Mang, J. Exploration of the mechanism of luteolin against ischemic stroke based on network pharmacology, molecular docking and experimental verification. Bioengineered 2021, 12, 12274–12293. [Google Scholar] [CrossRef] [PubMed]
- Goyal, A.; Solanki, K.; Verma, A. Luteolin: Nature’s promising warrior against Alzheimer’s and Parkinson’s disease. J. Biochem. Mol. Toxicol. 2024, 38, e23619. [Google Scholar] [CrossRef]
- Kou, J.J.; Shi, J.Z.; He, Y.Y.; Hao, J.J.; Zhang, H.Y.; Luo, D.M.; Song, J.K.; Yan, Y.; Xie, X.M.; Du, G.H.; et al. Luteolin alleviates cognitive impairment in Alzheimer’s disease mouse model via inhibiting endoplasmic reticulum stress-dependent neuroinflammation. Acta Pharmacol. Sin. 2022, 43, 840–849. [Google Scholar] [CrossRef] [PubMed]
- Radziejewska, I.; Borzym-Kluczyk, M.; Leszczynska, K. Luteolin alters MUC1 extracellular domain, sT antigen, ADAM-17, IL-8, IL-10 and NF-kappaB expression in Helicobacter pylori-infected gastric cancer CRL-1739 cells: A preliminary study. Biomed. Rep. 2021, 14, 19. [Google Scholar] [CrossRef]
- Xue, J.; Ye, J.; Xia, Z.; Cheng, B. Effect of luteolin on apoptosis, MAPK and JNK signaling pathways in guinea pig chondrocyte with osteoarthritis. Cell. Mol. Biol. 2019, 65, 91–95. [Google Scholar] [CrossRef]
- Kim, S.H.; Saba, E.; Kim, B.K.; Yang, W.K.; Park, Y.C.; Shin, H.J.; Han, C.K.; Lee, Y.C.; Rhee, M.H. Luteolin attenuates airway inflammation by inducing the transition of CD4(+)CD25(-) to CD4(+)CD25(+) regulatory T cells. Eur. J. Pharmacol. 2018, 820, 53–64. [Google Scholar] [CrossRef]
- Zhang, Z.T.; Zhang, D.Y.; Xie, K.; Wang, C.J.; Xu, F. Luteolin activates Tregs to promote IL-10 expression and alleviating caspase-11-dependent pyroptosis in sepsis-induced lung injury. Int. Immunopharmacol. 2021, 99, 107914. [Google Scholar] [CrossRef]
- Zhao, L.; Zhou, Z.; Zhu, C.; Fu, Z.; Yu, D. Luteolin alleviates myocardial ischemia reperfusion injury in rats via Siti1/NLRP3/NF-kappaB pathway. Int. Immunopharmacol. 2020, 85, 106680. [Google Scholar] [CrossRef]
- Li, B.; Du, P.; Du, Y.; Zhao, D.; Cai, Y.; Yang, Q.; Guo, Z. Luteolin alleviates inflammation and modulates gut microbiota in ulcerative colitis rats. Life Sci. 2021, 269, 119008. [Google Scholar] [CrossRef]
- Wang, I.C.; Lin, J.H.; Lee, W.S.; Liu, C.H.; Lin, T.Y.; Yang, K.T. Baicalein and luteolin inhibit ischemia/reperfusion-induced ferroptosis in rat cardiomyocytes. Int. J. Cardiol. 2023, 375, 74–86. [Google Scholar] [CrossRef]
- Qiao, X.R.; Feng, T.; Zhang, D.; Zhi, L.L.; Zhang, J.T.; Liu, X.F.; Pan, Y.; Xu, J.W.; Cui, W.J.; Dong, L. Luteolin alleviated neutrophilic asthma by inhibiting IL-36gamma secretion-mediated MAPK pathways. Pharm. Biol. 2023, 61, 165–176. [Google Scholar] [CrossRef]
- Vajdi, M.; Karimi, A.; Karimi, M.; Abbasalizad Farhangi, M.; Askari, G. Effects of luteolin on sepsis: A comprehensive systematic review. Phytomedicine 2023, 113, 154734. [Google Scholar] [CrossRef]
- Shankar, E.; Goel, A.; Gupta, K.; Gupta, S. Plant flavone apigenin: An emerging anticancer agent. Curr. Pharmacol. Rep. 2017, 3, 423–446. [Google Scholar] [CrossRef]
- Ng, S.P.; Wong, K.Y.; Zhang, L.; Zuo, Z.; Lin, G. Evaluation of the first-pass glucuronidation of selected flavones in gut by Caco-2 monolayer model. J. Pharm. Pharm. Sci. 2004, 8, 1–9. [Google Scholar] [PubMed]
- Liu, Y.; Hu, M. Absorption and metabolism of flavonoids in the caco-2 cell culture model and a perused rat intestinal model. Drug Metab. Dispos. 2002, 30, 370–377. [Google Scholar] [CrossRef]
- Chen, P.; Chen, F.; Guo, Z.; Lei, J.; Zhou, B. Recent advancement in bioeffect, metabolism, stability, and delivery systems of apigenin, a natural flavonoid compound: Challenges and perspectives. Front. Nutr. 2023, 10, 1221227. [Google Scholar] [CrossRef]
- Wang, S.W.; Kulkarni, K.H.; Tang, L.; Wang, J.R.; Yin, T.; Daidoji, T.; Yokota, H.; Hu, M. Disposition of flavonoids via enteric recycling: UDP-glucuronosyltransferase (UGT) 1As deficiency in Gunn rats is compensated by increases in UGT2Bs activities. J. Pharmacol. Exp. Ther. 2009, 329, 1023–1031. [Google Scholar] [CrossRef]
- Gradolatto, A.; Basly, J.P.; Berges, R.; Teyssier, C.; Chagnon, M.C.; Siess, M.H.; Canivenc-Lavier, M.C. Pharmacokinetics and metabolism of apigenin in female and male rats after a single oral administration. Drug Metab. Dispos. 2005, 33, 49–54. [Google Scholar] [CrossRef] [PubMed]
- Zhao, X.; Wang, Z. A pH-sensitive microemulsion-filled gellan gum hydrogel encapsulated apigenin: Characterization and in vitro release kinetics. Colloids Surf. B Biointerfaces 2019, 178, 245–252. [Google Scholar] [CrossRef] [PubMed]
- Banerjee, K.; Banerjee, S.; Mandal, M. Enhanced chemotherapeutic efficacy of apigenin liposomes in colorectal cancer based on flavone-membrane interactions. J. Colloid. Interface Sci. 2017, 491, 98–110. [Google Scholar] [CrossRef] [PubMed]
- Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kregiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino, E.; et al. The Therapeutic Potential of Apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef]
- Kashyap, P.; Shikha, D.; Thakur, M.; Aneja, A. Functionality of apigenin as a potent antioxidant with emphasis on bioavailability, metabolism, action mechanism and in vitro and in vivo studies: A review. J. Food Biochem. 2022, 46, e13950. [Google Scholar] [CrossRef]
- Yoon, J.H.; Kim, M.Y.; Cho, J.Y. Apigenin: A Therapeutic Agent for Treatment of Skin Inflammatory Diseases and Cancer. Int. J. Mol. Sci. 2023, 24, 1498. [Google Scholar] [CrossRef]
- Yan, X.; Qi, M.; Li, P.; Zhan, Y.; Shao, H. Apigenin in cancer therapy: Anti-cancer effects and mechanisms of action. Cell Biosci. 2017, 7, 50. [Google Scholar] [CrossRef]
- Wang, M.; Firrman, J.; Liu, L.; Yam, K. A Review on Flavonoid Apigenin: Dietary Intake, ADME, Antimicrobial Effects, and Interactions with Human Gut Microbiota. Biomed. Res. Int. 2019, 2019, 7010467. [Google Scholar] [CrossRef]
- Muhammed, T.M.; Jalil, A.T.; Taher, W.M.; Aminov, Z.; Alsaikhan, F.; Ramirez-Coronel, A.A.; Ramaiah, P.; Farhood, B. The Effects of Apigenin in the Treatment of Diabetic Nephropathy: A Systematic Review of Non-clinical Studies. Mini Rev. Med. Chem. 2024, 24, 341–354. [Google Scholar] [CrossRef] [PubMed]
- Ihim, S.A.; Kaneko, Y.K.; Yamamoto, M.; Yamaguchi, M.; Kimura, T.; Ishikawa, T. Apigenin Alleviates Endoplasmic Reticulum Stress-Mediated Apoptosis in INS-1 beta-Cells. Biol. Pharm. Bull. 2023, 46, 630–635. [Google Scholar] [CrossRef] [PubMed]
- Khaled, M.; Salama, R.A.M.; Aboughalia, A.; Tarek, M.; Mohamed Fawzy, N. Apigenin ameliorates genitourinary dysfunction in a type 1 diabetic rat model via Drp1 modulation. Sci. Rep. 2024, 14, 5754. [Google Scholar] [CrossRef]
- Liu, H.J.; Fan, Y.L.; Liao, H.H.; Liu, Y.; Chen, S.; Ma, Z.G.; Zhang, N.; Yang, Z.; Deng, W.; Tang, Q.Z. Apigenin alleviates STZ-induced diabetic cardiomyopathy. Mol. Cell Biochem. 2017, 428, 9–21. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Huang, P.; Wang, X.; Ma, Y.; Tong, J.; Li, J.; Ding, H. Apigenin analogs as alpha-glucosidase inhibitors with antidiabetic activity. Bioorg Chem. 2024, 143, 107059. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Li, X.; Wang, H. Protective Roles of Apigenin Against Cardiometabolic Diseases: A Systematic Review. Front. Nutr. 2022, 9, 875826. [Google Scholar] [CrossRef] [PubMed]
- Myoung, H.J.; Kim, G.; Nam, K.W. Apigenin isolated from the seeds of Perilla frutescens britton var crispa (Benth.) inhibits food intake in C57BL/6J mice. Arch. Pharm. Res. 2010, 33, 1741–1746. [Google Scholar] [CrossRef] [PubMed]
- Su, T.; Huang, C.; Yang, C.; Jiang, T.; Su, J.; Chen, M.; Fatima, S.; Gong, R.; Hu, X.; Bian, Z.; et al. Apigenin inhibits STAT3/CD36 signaling axis and reduces visceral obesity. Pharmacol. Res. 2020, 152, 104586. [Google Scholar] [CrossRef]
- Ohno, M.; Shibata, C.; Kishikawa, T.; Yoshikawa, T.; Takata, A.; Kojima, K.; Akanuma, M.; Kang, Y.J.; Yoshida, H.; Otsuka, M.; et al. The flavonoid apigenin improves glucose tolerance through inhibition of microRNA maturation in miRNA103 transgenic mice. Sci. Rep. 2013, 3, 2553. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Fang, X.; Shi, J.; Li, X.; Xie, M.; Liu, X. Apigenin attenuates pulmonary hypertension by inducing mitochondria-dependent apoptosis of PASMCs via inhibiting the hypoxia inducible factor 1alpha-KV1.5 channel pathway. Chem. Biol. Interact. 2020, 317, 108942. [Google Scholar] [CrossRef] [PubMed]
- Yamagata, K.; Tagawa, C.; Matsufuji, H.; Chino, M. Dietary apigenin regulates high glucose and hypoxic reoxygenation-induced reductions in apelin expression in human endothelial cells. J. Nutr. Biochem. 2012, 23, 929–936. [Google Scholar] [CrossRef] [PubMed]
- Cai, M.; Ma, Y.; Zhang, W.; Wang, S.; Wang, Y.; Tian, L.; Peng, Z.; Wang, H.; Qingrong, T. Apigenin-7-O-beta-D-(-6''-p-coumaroyl)-Glucopyranoside Treatment Elicits Neuroprotective Effect against Experimental Ischemic Stroke. Int. J. Biol. Sci. 2016, 12, 42–52. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Xu, S.; Duan, H.; Zhu, Z.; Yang, Z.; Cao, J.; Zhao, Y.; Huang, Z.; Wu, Q.; Duan, J. A novel, highly-water-soluble apigenin derivative provides neuroprotection following ischemia in male rats by regulating the ERK/Nrf2/HO-1 pathway. Eur. J. Pharmacol. 2019, 855, 208–215. [Google Scholar] [CrossRef] [PubMed]
- Pang, Q.; Zhao, Y.; Chen, X.; Zhao, K.; Zhai, Q.; Tu, F. Apigenin Protects the Brain against Ischemia/Reperfusion Injury via Caveolin-1/VEGF In Vitro and In Vivo. Oxid. Med. Cell Longev. 2018, 2018, 7017204. [Google Scholar] [CrossRef] [PubMed]
- Tu, F.; Pang, Q.; Huang, T.; Zhao, Y.; Liu, M.; Chen, X. Apigenin Ameliorates Post-Stroke Cognitive Deficits in Rats Through Histone Acetylation-Mediated Neurochemical Alterations. Med. Sci. Monit. 2017, 23, 4004–4013. [Google Scholar] [CrossRef]
- Zick, S.M.; Wright, B.D.; Sen, A.; Arnedt, J.T. Preliminary examination of the efficacy and safety of a standardized chamomile extract for chronic primary insomnia: A randomized placebo-controlled pilot study. BMC Complement. Altern. Med. 2011, 11, 78. [Google Scholar] [CrossRef]
- Shoara, R.; Hashempur, M.H.; Ashraf, A.; Salehi, A.; Dehshahri, S.; Habibagahi, Z. Efficacy and safety of topical Matricaria chamomilla L. (chamomile) oil for knee osteoarthritis: A randomized controlled clinical trial. Complement. Ther. Clin. Pract. 2015, 21, 181–187. [Google Scholar] [CrossRef]
- Amsterdam, J.D.; Shults, J.; Soeller, I.; Mao, J.J.; Rockwell, K.; Newberg, A.B. Chamomile (Matricaria recutita) may provide antidepressant activity in anxious, depressed humans: An exploratory study. Altern. Ther. Health Med. 2012, 18, 44–49. [Google Scholar]
- Mao, J.J.; Xie, S.X.; Keefe, J.R.; Soeller, I.; Li, Q.S.; Amsterdam, J.D. Long-term chamomile (Matricaria chamomilla L.) treatment for generalized anxiety disorder: A randomized clinical trial. Phytomedicine 2016, 23, 1735–1742. [Google Scholar] [CrossRef]
- Zhu, L.; Zhang, H.; Zhang, X.; Xia, L.; Zhang, J. Research progress on antisepsis effect of apigenin and its mechanism of action. Heliyon 2023, 9, e22290. [Google Scholar] [CrossRef]
- Patel, U.; Desai, K.; Dabhi, R.C.; Maru, J.J.; Shrivastav, P.S. Bioprospecting phytochemicals of Rosmarinus officinalis L. for targeting SARS-CoV-2 main protease (M(pro)): A computational study. J. Mol. Model. 2023, 29, 161. [Google Scholar] [CrossRef]
- Samec, D.; Karalija, E.; Sola, I.; Vujcic Bok, V.; Salopek-Sondi, B. The Role of Polyphenols in Abiotic Stress Response: The Influence of Molecular Structure. Plants 2021, 10, 118. [Google Scholar] [CrossRef]
- Gambini, J.; Ingles, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of Resveratrol: In Vitro and In Vivo Studies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxid. Med. Cell Longev. 2015, 2015, 837042. [Google Scholar] [CrossRef]
- Marel, A.K.; Lizard, G.; Izard, J.C.; Latruffe, N.; Delmas, D. Inhibitory effects of trans-resveratrol analogs molecules on the proliferation and the cell cycle progression of human colon tumoral cells. Mol. Nutr. Food Res. 2008, 52, 538–548. [Google Scholar] [CrossRef]
- Wang, L.X.; Heredia, A.; Song, H.; Zhang, Z.; Yu, B.; Davis, C.; Redfield, R. Resveratrol glucuronides as the metabolites of resveratrol in humans: Characterization, synthesis, and anti-HIV activity. J. Pharm. Sci. 2004, 93, 2448–2457. [Google Scholar] [CrossRef]
- Miksits, M.; Wlcek, K.; Svoboda, M.; Kunert, O.; Haslinger, E.; Thalhammer, T.; Szekeres, T.; Jager, W. Antitumor activity of resveratrol and its sulfated metabolites against human breast cancer cells. Planta Med. 2009, 75, 1227–1230. [Google Scholar] [CrossRef] [PubMed]
- Arunachalam, G.; Yao, H.; Sundar, I.K.; Caito, S.; Rahman, I. SIRT1 regulates oxidant- and cigarette smoke-induced eNOS acetylation in endothelial cells: Role of resveratrol. Biochem. Biophys. Res. Commun. 2010, 393, 66–72. [Google Scholar] [CrossRef] [PubMed]
- Zimmermann-Franco, D.C.; Esteves, B.; Lacerda, L.M.; Souza, I.O.; Santos, J.A.D.; Pinto, N.C.C.; Scio, E.; da Silva, A.D.; Macedo, G.C. In vitro and in vivo anti-inflammatory properties of imine resveratrol analogues. Bioorg. Med. Chem. 2018, 26, 4898–4906. [Google Scholar] [CrossRef] [PubMed]
- Campagna, M.; Rivas, C. Antiviral activity of resveratrol. Biochem. Soc. Trans. 2010, 38 Pt 1, 50–53. [Google Scholar] [CrossRef] [PubMed]
- Lu, C.C.; Lai, H.C.; Hsieh, S.C.; Chen, J.K. Resveratrol ameliorates Serratia marcescens-induced acute pneumonia in rats. J. Leukoc. Biol. 2008, 83, 1028–1037. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Xie, Q.; Chen, Z.; Ni, H.; Xia, L.; Zhao, Q.; Chen, Z.; Chen, P. Resveratrol suppresses the invasion and migration of human gastric cancer cells via inhibition of MALAT1-mediated epithelial-to-mesenchymal transition. Exp. Ther. Med. 2019, 17, 1569–1578. [Google Scholar] [CrossRef] [PubMed]
- Ashrafizadeh, M.; Rafiei, H.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Anti-tumor activity of resveratrol against gastric cancer: A review of recent advances with an emphasis on molecular pathways. Cancer Cell Int. 2021, 21, 66. [Google Scholar] [CrossRef] [PubMed]
- Shang, X.; Lin, K.; Yu, R.; Zhu, P.; Zhang, Y.; Wang, L.; Xu, J.; Chen, K. Resveratrol Protects the Myocardium in Sepsis by Activating the Phosphatidylinositol 3-Kinases (PI3K)/AKT/Mammalian Target of Rapamycin (mTOR) Pathway and Inhibiting the Nuclear Factor-kappaB (NF-kappaB) Signaling Pathway. Med. Sci. Monit. 2019, 25, 9290–9298. [Google Scholar] [CrossRef] [PubMed]
- de Souza Andrade, M.M.; Leal, V.N.C.; Fernandes, I.G.; Gozzi-Silva, S.C.; Beserra, D.R.; Oliveira, E.A.; Teixeira, F.M.E.; Yendo, T.M.; Sousa, M.; Teodoro, W.R.; et al. Resveratrol Downmodulates Neutrophil Extracellular Trap (NET) Generation by Neutrophils in Patients with Severe COVID-19. Antioxidants 2022, 11, 1690. [Google Scholar] [CrossRef]
- Li, J.; Zeng, X.; Yang, F.; Wang, L.; Luo, X.; Liu, R.; Zeng, F.; Lu, S.; Huang, X.; Lei, Y.; et al. Resveratrol: Potential Application in Sepsis. Front. Pharmacol. 2022, 13, 821358. [Google Scholar] [CrossRef]
- L’Heureux, M.; Sternberg, M.; Brath, L.; Turlington, J.; Kashiouris, M.G. Sepsis-Induced Cardiomyopathy: A Comprehensive Review. Curr. Cardiol. Rep. 2020, 22, 35. [Google Scholar] [CrossRef] [PubMed]
- Den Hartogh, D.J.; Tsiani, E. Antidiabetic Properties of Naringenin: A Citrus Fruit Polyphenol. Biomolecules 2019, 9, 99. [Google Scholar] [CrossRef] [PubMed]
- Ravetti, S.; Garro, A.G.; Gaitan, A.; Murature, M.; Galiano, M.; Brignone, S.G.; Palma, S.D. Naringin: Nanotechnological Strategies for Potential Pharmaceutical Applications. Pharmaceutics 2023, 15, 863. [Google Scholar] [CrossRef] [PubMed]
- Felgines, C.; Texier, O.; Morand, C.; Manach, C.; Scalbert, A.; Regerat, F.; Remesy, C. Bioavailability of the flavanone naringenin and its glycosides in rats. Am. J. Physiol. Gastrointest. Liver Physiol. 2000, 279, G1148–G1154. [Google Scholar] [CrossRef]
- Fang, T.; Wang, Y.; Ma, Y.; Su, W.; Bai, Y.; Zhao, P. A rapid LC/MS/MS quantitation assay for naringin and its two metabolites in rats plasma. J. Pharm. Biomed. Anal. 2006, 40, 454–459. [Google Scholar] [CrossRef] [PubMed]
- Du, Y.; Ma, J.; Fan, Y.; Wang, X.; Zheng, S.; Feng, J.; Li, J.; Fan, Z.; Li, G.; Ye, Q. Naringenin: A Promising Therapeutic Agent against Organ Fibrosis. Oxid. Med. Cell Longev. 2021, 2021, 1210675. [Google Scholar] [CrossRef] [PubMed]
- Orrego-Lagaron, N.; Martinez-Huelamo, M.; Vallverdu-Queralt, A.; Lamuela-Raventos, R.M.; Escribano-Ferrer, E. High gastrointestinal permeability and local metabolism of naringenin: Influence of antibiotic treatment on absorption and metabolism. Br. J. Nutr. 2015, 114, 169–180. [Google Scholar] [CrossRef] [PubMed]
- Salehi, B.; Fokou, P.V.T.; Sharifi-Rad, M.; Zucca, P.; Pezzani, R.; Martins, N.; Sharifi-Rad, J. The Therapeutic Potential of Naringenin: A Review of Clinical Trials. Pharmaceuticals 2019, 12, 11. [Google Scholar] [CrossRef]
- Bajgai, B.; Suri, M.; Singh, H.; Hanifa, M.; Bhatti, J.S.; Randhawa, P.K.; Bali, A. Naringin: A flavanone with a multifaceted target against sepsis-associated organ injuries. Phytomedicine 2024, 130, 155707. [Google Scholar] [CrossRef]
- Alberca, R.W.; Teixeira, F.M.E.; Beserra, D.R.; de Oliveira, E.A.; Andrade, M.M.S.; Pietrobon, A.J.; Sato, M.N. Perspective: The Potential Effects of Naringenin in COVID-19. Front. Immunol. 2020, 11, 570919. [Google Scholar] [CrossRef]
- Bi, C.; Jiang, Y.; Fu, T.; Hao, Y.; Zhu, X.; Lu, Y. Naringin inhibits lipopolysaccharide-induced damage in human umbilical vein endothelial cells via attenuation of inflammation, apoptosis and MAPK pathways. Cytotechnology 2016, 68, 1473–1487. [Google Scholar] [CrossRef]
- Nicolescu, A.; Babota, M.; Barros, L.; Rocchetti, G.; Lucini, L.; Tanase, C.; Mocan, A.; Bunea, C.I.; Crisan, G. Bioaccessibility and bioactive potential of different phytochemical classes from nutraceuticals and functional foods. Front. Nutr. 2023, 10, 1184535. [Google Scholar] [CrossRef] [PubMed]
- Selby-Pham, S.N.B.; Miller, R.B.; Howell, K.; Dunshea, F.; Bennett, L.E. Physicochemical properties of dietary phytochemicals can predict their passive absorption in the human small intestine. Sci. Rep. 2017, 7, 1931. [Google Scholar] [CrossRef] [PubMed]
- Sulfikkarali, N.; Krishnakumar, N.; Manoharan, S.; Nirmal, R.M. Chemopreventive efficacy of naringenin-loaded nanoparticles in 7,12-dimethylbenz(a)anthracene induced experimental oral carcinogenesis. Pathol. Oncol. Res. 2013, 19, 287–296. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.N.; Cao, Y.N.; Sun, J.; Liang, Z.; Wu, Q.; Cui, S.H.; Zhi, D.F.; Guo, S.T.; Zhen, Y.H.; Zhang, S.B. Anti-breast cancer activity of resveratrol encapsulated in liposomes. J. Mater. Chem. B 2020, 8, 27–37. [Google Scholar] [CrossRef]
- Madaan, K.; Lather, V.; Pandita, D. Evaluation of polyamidoamine dendrimers as potential carriers for quercetin, a versatile flavonoid. Drug Deliv. 2016, 23, 254–262. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Cui, C.; Wei, F.; Lv, H. Improved solubility and oral bioavailability of apigenin via Soluplus/Pluronic F127 binary mixed micelles system. Drug Dev. Ind. Pharm. 2017, 43, 1276–1282. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Luo, C.; Yang, G.; Wei, X.; Liu, D.; Zhou, S. A Luteolin-Loaded Electrospun Fibrous Implantable Device for Potential Therapy of Gout Attacks. Macromol. Biosci. 2016, 16, 1598–1609. [Google Scholar] [CrossRef]
Phytochemical | Sources | Experimental Models | Physiological Effects | References |
---|---|---|---|---|
Quercetin | Citrus fruits, apples, onions, broccoli, parsley, tea, red wine, olive oil, grapes, dark cherries, and dark berries. | In vitro: LPS-treated cells (RAW264.7, human alveolar epithelial A549 cells, murine lung epithelial cells, macrophages, and H9C2 cells). In vivo: CLP in rats and mice; LPS-treated mice; E. coli K1 treatment in mice. In silico: Network pharmacology. |
| [119,120,121,122,123,124,125,126,127,128,129] |
Curcumin | Turmeric | In vitro: LPS-treated cells (HL-1 cells, RAW264.7) In vivo: LPS-treated mice and rats; CLP in mice. In silico: Gene expression matrix. Clinical trial: Human patients. |
| [130,131,132,133,134,135,136,137,138,139,140,141,142,143] |
Luteolin | Celery, parsley, broccoli, onion leaves, carrots, peppers, cabbages, apple skins, and chrysanthemum flowers. | In vitro: LPS-treated cells (RAW264.7 macrophages, peritoneal macrophages, HepG2 cells). In vivo: CLP in mice; LPS-treated mice; CSI in mouse pups. In silico: Network pharmacology. |
| [144,145,146,147,148,149,150,151,152,153,154] |
Apigenin | Chamomile, parsley, celery, onions, oranges, thyme, oregano, and basil. | In vitro: LPS-treated cells (H9c2 cells, RAW264.7 macrophages, human lung A549 cells, bovine aortic endothelial cells). In vivo: LPS-treated mice and rats. |
| [155,156,157,158,159,160,161,162,163,164,165,166,167] |
Resveratrol | Grapes, raspberries, blueberries, plums, peanuts, roots, stems, leaves, and teas. | In vitro: LPS-treated cells (rat cortical microglia, mouse microglial cell line N9, human umbilical endothelial cells, macrophages, MH-S cells). In vivo: LPS-treated rats; CLP in mice. |
| [168,169,170,171,172,173,174,175,176,177,178,179,180] |
Naringenin | Grapes, lemons, oranges, bergamot, and tomatoes. | In vitro: Human bronchial epithelium; LPS-stimulated murine macrophages; MODE-K cells. In vivo: LPS-treated mice and rats. |
| [181,182,183,184,185,186,187,188,189] |
Phytochemical | Experimental Models | Pharmacological Effects | References |
---|---|---|---|
Quercetin | In vitro: Antiviral activity; activity-based experimental screening; SARS-CoV-2-infected green monkey kidney Vero E6 cells and human colon carcinoma Caco-2 cells; human embryonic kidney HEK293 co-expressing SARS-CoV-2 spike (S) protein and ACE2; H1975-ACE2; 293T-ACE2; BEAS-2B-ACE2 cells. In vivo: SARS-CoV-2-infected hamsters and mice. In silico: Molecular docking; network pharmacology; protein–protein interaction network. Clinical trials: Human patients. |
| [190,191,192,193,194,195,196,197,198,199,200] |
Curcumin | In silico: Network pharmacology; molecular docking. Clinical trials: Human patients. |
| [116,201,202,203,204,205,206,207,208,209,210,211,212] |
Luteolin | In silico: Network pharmacology; molecular docking. Clinical trials: Human patients. |
| [213,214,215,216,217,218,219,220,221,222] |
Apigenin | In vitro: SARS-CoV-2-infected African green monkey kidney Vero E6 cells; human lung epithelial Calu-3 cells. In silico: Network pharmacology; molecular docking. |
| [223,224,225,226,227,228,229,230,231] |
Resveratrol | In vitro: Human primary bronchial epithelial cells; Vero cells; MRC-5. In silico: Network pharmacology Clinical trials: Human patients. |
| [118,232,233,234,235,236] |
Naringenin | In vitro: Vero E6 cells In silico: Molecular docking; network pharmacology. |
| [201,237,238,239,240] |
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. |
© 2024 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
de Souza Goncalves, B.; Sangani, D.; Nayyar, A.; Puri, R.; Irtiza, M.; Nayyar, A.; Khalyfa, A.; Sodhi, K.; Pillai, S.S. COVID-19-Associated Sepsis: Potential Role of Phytochemicals as Functional Foods and Nutraceuticals. Int. J. Mol. Sci. 2024, 25, 8481. https://doi.org/10.3390/ijms25158481
de Souza Goncalves B, Sangani D, Nayyar A, Puri R, Irtiza M, Nayyar A, Khalyfa A, Sodhi K, Pillai SS. COVID-19-Associated Sepsis: Potential Role of Phytochemicals as Functional Foods and Nutraceuticals. International Journal of Molecular Sciences. 2024; 25(15):8481. https://doi.org/10.3390/ijms25158481
Chicago/Turabian Stylede Souza Goncalves, Bruno, Darshan Sangani, Aleen Nayyar, Raghav Puri, Mahir Irtiza, Asma Nayyar, Abdelnaby Khalyfa, Komal Sodhi, and Sneha S. Pillai. 2024. "COVID-19-Associated Sepsis: Potential Role of Phytochemicals as Functional Foods and Nutraceuticals" International Journal of Molecular Sciences 25, no. 15: 8481. https://doi.org/10.3390/ijms25158481
APA Stylede Souza Goncalves, B., Sangani, D., Nayyar, A., Puri, R., Irtiza, M., Nayyar, A., Khalyfa, A., Sodhi, K., & Pillai, S. S. (2024). COVID-19-Associated Sepsis: Potential Role of Phytochemicals as Functional Foods and Nutraceuticals. International Journal of Molecular Sciences, 25(15), 8481. https://doi.org/10.3390/ijms25158481