The Impact of Curcumin on Immune Response: An Immunomodulatory Strategy to Treat Sepsis
Abstract
:1. Introduction
Immunodeficiencies and Infections
2. Curcumin and Infections
2.1. Curcumin and Its Effects on the Immune System
2.2. Other Immune-Mediated Effects of Curcumin on Infections
3. Curcumin and Virus Infections
3.1. Curcumin and Human Immunodeficiency Virus
3.2. Curcumin and SARS-CoV 2 Infection
3.3. Curcumin and Enterovirus
3.4. Effect of Curcumin on Other Viral Infections
4. Curcumin and Bacterial Infection
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Zanza, C.; Caputo, G.; Tornatore, G.; Romenskaya, T.; Piccioni, A.; Franceschi, F.; Artico, M.; Taurone, S.; Savioli, G.; Longhitano, Y. Cellular Immuno-Profile in Septic Human Host: A Scoping Review. Biology 2022, 11, 1626. [Google Scholar] [CrossRef] [PubMed]
- Almalki, W.H. The sepsis induced defective aggravation of immune cells: A translational science underling chemico-biological interactions from altered bioenergetics and/or cellular metabolism to organ dysfunction. Mol. Cell Biochem. 2021, 476, 2337–2344. [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]
- Hotchkiss, R.S.; Monneret, G.; Payen, D. Immunosuppression in sepsis: A novel understanding of the disorder and a new therapeutic approach. Lancet Infect. Dis. 2013, 13, 260–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hotchkiss, R.S.; Monneret, G.; Payen, D. Sepsis-induced immunosuppression: From cellular dysfunctions to immunotherapy. Nat. Rev. Immunol. 2013, 13, 862–874. [Google Scholar] [CrossRef] [Green Version]
- Hotchkiss, R.S.; Moldawer, L.L.; Opal, S.M.; Reinhart, K.; Turnbull, I.R.; Vincent, J.L. Sepsis and septic shock. Nat. Rev. Dis. Primers 2016, 2, 16045. [Google Scholar] [CrossRef] [Green Version]
- Gül, F.; Arslantaş, M.K.; Cinel, İ.; Kumar, A. Changing definitions of sepsis. Turk. J. Anaesthesiol. Reanim. 2017, 45, 129–138. [Google Scholar] [CrossRef] [PubMed]
- Venet, F.; Monneret, G. Advances in the Understanding and Treatment of Sepsis-Induced Immunosuppression. Nat. Rev. Nephrol 2018, 14, 121–137. [Google Scholar] [CrossRef] [PubMed]
- Puck, J.M. Newborn screening for severe combined immunodeficiency and T-cell lymphopenia. Immunol. Rev. 2019, 287, 241–252. [Google Scholar] [CrossRef]
- Mogensen, T.H. Genetic susceptibility to viral disease in humans. Clin. MicroBiol. Infect. 2022, 28, 1411–1416. [Google Scholar] [CrossRef]
- Casanova, J.L.; Abel, L. Primary immunodeficiencies: A field in its infancy. Science 2007, 317, 617–619. [Google Scholar] [CrossRef] [Green Version]
- Chellapandian, D.; Chitty-Lopez, M.; Leiding, J.W. Precision therapy for the treatment of primary immunodysregulatory diseases. Immunol. Allergy Clin. 2020, 40, 511–526. [Google Scholar] [CrossRef] [PubMed]
- Bousfha, A.; Jeddane, L.; Picard, C.; Al-Herz, W.; Ailal, F.; Chatila, T.; Cunningham-Rundles, C.; Etzioni, A.; Franco, J.L.; Holland, S.M.; et al. Human inborn errors of immunity: 2019 update of the IUIS phenotypical classification. J. Clin. Immunol. 2020, 40, 66–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonilla, F.A.; Khan, D.A.; Ballas, Z.K.; Chinen, J.; Frank, M.M.; Hsu, J.T.; Keller, M.; Kobrynski, L.J.; Komarow, H.D.; Mazer, B.; et al. Practice parameter for the diagnosis and management of primary immunodeficiency. Immunol. Allergy Clin. N. Am. 2015, 136, 1186–1205. [Google Scholar] [CrossRef] [Green Version]
- Dorsey, M.J.; Dvorak, C.C.; Cowan, M.J.; Puck, J.M. Treatment of infants identified as having severe combined immunodeficiency by means of newborn screening. J. Allergy Clin. Immunol. 2017, 139, 733–742. [Google Scholar] [CrossRef] [Green Version]
- Ballow, M.; Paris, K.; de la Morena, M. Should antibiotic prophylaxis be routinely used in patients with antibody-mediated primary immunodeficiency? J. Allergy Clin. Immunol. Pract. 2018, 6, 421–426. [Google Scholar] [CrossRef]
- Tangye, S.G.; Al-Herz, W.; Bousfiha, A.; Cunningham-Rundles, C.; Franco, J.L.; Holland, S.M.; Klein, C.; Morio, T.; Oksenhendler, E.; Picard, C.; et al. The Ever-Increasing Array of Novel Inborn Errors of Immunity: An Interim Update by the IUIS Committee. J. Clin. Immunol. 2021, 41, 666–679. [Google Scholar] [CrossRef] [PubMed]
- Tuano, K.S.; Seth, N.; Chinen, J. Secondary immunodeficiencies: An overview. Ann. Allergy Asthma Immunol. 2021, 127, 617–626. [Google Scholar] [CrossRef]
- Lee, W.J.; Lee, T.A.; Calip, G.S.; Suda, K.J.; Briars, L.; Schumock, G.T. Risk of Serious Bacterial Infection Associated With Tumor Necrosis Factor-Alpha Inhibitors in Children and Young Adults With Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2018, 24, 883–891. [Google Scholar] [CrossRef]
- McAtee, C.L.; Lubega, J.; Underbrink, K.; Curry, K.; Msaouel, P.; Barrow, M. Association of Rituximab Use With Adverse Events in Children, Adolescents, and Young Adults. JAMA Netw. Open 2021, 4, e2036321. [Google Scholar] [CrossRef]
- Allegra, A.; Tonacci, A.; Musolino, C.; Pioggia, G.; Gangemi, S. Secondary Immunodeficiency in Hematological Malignancies: Focus on Multiple Myeloma and Chronic Lymphocytic Leukemia. Front. Immunol. 2021, 12, 738915. [Google Scholar] [CrossRef]
- Relja, B.; Land, W.G. Damage-associated molecular patterns in trauma. Eur. J. Trauma Emerg. Surg. 2020, 46, 751–775. [Google Scholar] [CrossRef] [Green Version]
- Deya-Martinez, A.; Flinn, A.M.; Gennery, A.R. Neonatal thymectomy in children accelerating the immunologic clock? J. Allergy Clin. Immunol. 2020, 146, 236–243. [Google Scholar] [CrossRef]
- Gudmundsdottir, J.; Óskarsdóttir, S.; Skogberg, G.; Lindgren, S.; Lundberg, V.; Berglund, M.; Lundell, A.C.; Berggren, H.; Fasth, A.; Telemo, E.; et al. Early thymectomy leads to premature immunologic ageing: An 18-year follow-up. J. Allergy Clin. Immunol. 2016, 138, 1439–1443.e10. [Google Scholar] [CrossRef] [Green Version]
- Silva, S.L.; Albuquerque, A.; Amaral, A.J.; Li, Q.Z.; Mota, C.; Cheynier, R.; Victorino, R.M.M.; Pereira-Santos, M.C.; Sousa, A.E. Autoimmunity and allergy control in adults submitted to complete thymectomy early in infancy. PLoS ONE 2017, 12, e0180385. [Google Scholar] [CrossRef] [PubMed]
- Nedeva, C.; Menassa, J.; Puthalakath, H. Sepsis: Inflammation Is a Necessary Evil. Front. Cell Dev. Biol. 2019, 7, 108. [Google Scholar] [CrossRef] [Green Version]
- Delano, M.J.; Ward, P.A. The immune system’s role in sepsis progression, resolution, and long-term outcome. Immunol. Rev. 2016, 274, 330–353. [Google Scholar] [CrossRef] [Green Version]
- Vu, C.T.B.; Thammahong, A.; Leelahavanichkul, A.; Ritprajak, P. Alteration of macrophage immune phenotype in a murine sepsis model is associated with susceptibility to secondary fungal infection. Asian Pac. J. Allergy Immunol. 2022, 40, 162–171. [Google Scholar] [CrossRef]
- Otto, G.P.; Sossdorf, M.; Claus, R.A.; Rödel, J.; Menge, K.; Reinhart, K.; Bauer, M.; Riedemann, N.C. The late phase of sepsis is characterized by an increased microbiological burden and death rate. Crit. Care 2011, 15, R183. [Google Scholar] [CrossRef] [Green Version]
- Misra, A.K.; Levy, M.M.; Ward, N.S. Biomarkers of Immunosuppression. Crit. Care Clin. 2020, 36, 167–176. [Google Scholar] [CrossRef]
- Rubio, I.; Osuchowski, M.F.; Shankar-Hari, M.; Skirecki, T.; Winkler, M.S.; Lachmann, G.; La Rosée, P.; Monneret, G.; Venet, F.; Bauer, M.; et al. Current Gaps in Sepsis Immunology: New Opportunities for Translational Research. Lancet Infect. Dis. 2019, 19, e422–e436. [Google Scholar] [CrossRef] [PubMed]
- Yao, R.Q.; Ren, C.; Wang, J.N.; Wu, G.S.; Zhu, X.M.; Xia, Z.F.; Yao, Y.M. Publication Trends of Research on Sepsis and Host Immune Response During 1999-2019: A 20-Year Bibliometric Analysis. Int. J. Biol. Sci. 2020, 16, 27–37. [Google Scholar] [CrossRef]
- Venet, F.; Lukaszewicz, A.C.; Payen, D.; Hotchkiss, R.; Monneret, G. Monitoring the Immune Response in Sepsis: A Rational Approach to Administration of Immunoadjuvant Therapies. Curr. Opin. Immunol. 2013, 25, 477–483. [Google Scholar] [CrossRef] [Green Version]
- Ong, D.S.Y.; Bonten, M.J.M.; Spitoni, C.; Verduyn Lunel, F.M.; Frencken, J.F.; Horn, J.; Schultz, M.J.; van der Poll, T.; Klein Klouwenberg, P.M.C.; Cremer, O.L. Epidemiology of Multiple Herpes Viremia in Previously Immunocompetent Patients With Septic Shock. Clin. Infect. Dis. 2017, 64, 1204–1210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van der Poll, T.; Shankar-Hari, M.; Wiersinga, W.J. The Immunology of Sepsis. Immunity 2021, 54, 2450–2464. [Google Scholar] [CrossRef]
- Yao, R.Q.; Ren, C.; Xia, Z.F.; Yao, Y.M. Organelle-Specific Autophagy in Inflammatory Diseases: A Potential Therapeutic Target Underlying the Quality Control of Multiple Organelles. Autophagy 2021, 17, 385–401. [Google Scholar] [CrossRef] [Green Version]
- Ren, C.; Yao, R.Q.; Zhang, H.; Feng, Y.W.; Yao, Y.M. Sepsis-Associated Encephalopathy: A Vicious Cycle of Immunosuppression. J. Neuroinflamm. 2020, 17, 14. [Google Scholar] [CrossRef] [PubMed]
- Seeley, J.J.; Ghosh, S. Molecular Mechanisms of Innate Memory and Tolerance to LPS. J. Leukoc. Biol. 2017, 101, 107–119. [Google Scholar] [CrossRef]
- Cao, C.; Ma, T.; Chai, Y.F.; Shou, S.T. The role of regulatory T cells in immune dysfunction during sepsis. World J. Emerg. Med. 2015, 6, 5–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Delano, M.J.; Ward, P.A. Sepsis-induced immune dysfunction: Can immune therapies reduce mortality? J. Clin. Investig. 2016, 126, 23–31. [Google Scholar] [CrossRef] [Green Version]
- Kumar, V. T cells and their immunometabolism: A novel way to understanding sepsis immunopathogenesis and future therapeutics. Eur. J. Cell Biol. 2018, 97, 379–392. [Google Scholar] [CrossRef] [Green Version]
- Gharamti, A.A.; Samara, O.; Monzon, A.; Montalbano, G.; Scherger, S.; DeSanto, K.; Chastain, D.B.; Sillau, S.; Montoya, J.G.; Franco-Paredes, C.; et al. Proinflammatory cytokines levels in sepsis and healthy volunteers, and tumor necrosis factor-alpha associated sepsis mortality: A systematic review and meta-analysis. Cytokine 2022, 158, 156006. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Zhang, H.; Chen, C.; Qiao, K.; Li, Z.; Han, J.; Han, X.; Li, K.; Lai, K.; Liu, N.; et al. Biomimetic Immunosuppressive Exosomes that Inhibit Cytokine Storms Contribute to the Alleviation of Sepsis. Adv. Mater. 2022, 34, e2108476. [Google Scholar] [CrossRef] [PubMed]
- Rittirsch, D.; Flierl, M.A.; Ward, P.A. Harmful molecular mechanisms in sepsis. Nat. Rev. Immunol. 2008, 8, 776–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Elinav, E.; Thaiss, C.A.; Flavell, R.A. Analysis of microbiota alterations in inflammasome-deficient mice. Methods Mol. Biol. 2013, 1040, 185–194. [Google Scholar]
- Zhang, X.; Fan, C.; Zhang, H.; Zhao, Q.; Liu, Y.; Xu, C.; Xie, Q.; Wu, X.; Yu, X.; Zhang, J.; et al. MLKL and FADD are critical for suppressing progressive lymphoproliferative disease and activating the NLRP3 inflammasome. Cell Rep. 2016, 16, 3247–3259. [Google Scholar] [CrossRef] [Green Version]
- Conos, S.A.; Chen, K.W.; de Nardo, D.; Hara, H.; Whitehead, L.; Núñez, G.; Masters, S.L.; Murphy, J.M.; Schroder, K.; Vaux, D.L.; et al. Active MLKL triggers the NLRP3 inflammasome in a cell-intrinsic manner. Proc. Natl. Acad. Sci. USA 2017, 114, E961–E969. [Google Scholar] [CrossRef] [Green Version]
- Gutierrez, K.D.; Davis, M.A.; Daniels, B.P.; Olsen, T.M.; Ralli-Jain, P.; Tait, S.W.; Gale, M., Jr.; Oberst, A. MLKL activation triggers NLRP3-mediated processing and release of IL-1beta independently of gasdermin-D. J. Immunol. 2017, 198, 2156–2164. [Google Scholar] [CrossRef] [Green Version]
- Sharma, A.A.; Jen, R.; Kan, B.; Sharma, A.; Marchant, E.; Tang, A.; Gadawski, I.; Senger, C.; Skoll, A.; Turvey, S.E.; et al. Impaired NLRP3 inflammasome activity during fetal development regulates IL-1β production in human monocytes. Eur. J. Immunol. 2015, 45, 238–249. [Google Scholar] [CrossRef] [Green Version]
- Stouch, A.N.; Mccoy, A.M.; Greer, R.M.; Lakhdari, O.; Yull, F.E.; Blackwell, T.S.; Hoffman, H.M.; Prince, L.S. IL-1β and inflammasome activity link inflammation to abnormal fetal airway development. J. Immunol. 2016, 196, 3411–3420. [Google Scholar] [CrossRef] [Green Version]
- Kumar, V. Inflammasomes: Pandora’s box for sepsis. J. Inflamm. Res. 2018, 11, 477–502. [Google Scholar] [CrossRef] [PubMed]
- Shankar-Hari, M.; Phillips, G.S.; Levy, M.L.; Seymour, C.W.; Liu, V.X.; Deutschman, C.S.; Angus, D.C.; Rubenfeld, G.D.; Singer, M. Developing a new definition and assessing new clinical criteria for septic shock: For the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 775–787. [Google Scholar] [CrossRef] [Green Version]
- Dhawan, B.N. Anti-Viral Activity of Indian Plants. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 2012, 82, 209–224. [Google Scholar] [CrossRef]
- Prasad, S.; Tyagi, A.K.; Aggarwal, B.B. Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: The golden pigment from golden spice. Cancer Res. Treat. 2014, 46, 2–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hewlings, S.J.; Kalman, D.S. Curcumin: A review of its’ effects on human health. Foods 2017, 6, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allegra, A.; Innao, V.; Russo, S.; Gerace, D.; Alonci, A.; Musolino, C. Anticancer Activity of Curcumin and Its Analogues: Preclinical and Clinical Studies. Cancer Investig. 2017, 35, 1–22. [Google Scholar] [CrossRef]
- Allegra, A.; Speciale, A.; Molonia, M.S.; Guglielmo, L.; Musolino, C.; Ferlazzo, G.; Costa, G.; Saija, A.; Cimino, F. Curcumin ameliorates the in vitro efficacy of carfilzomib in human multiple myeloma U266 cells targeting p53 and NF-κB pathways. Toxicol. In Vitro 2018, 47, 186–194. [Google Scholar] [CrossRef]
- Ettari, R.; Previti, S.; Di Chio, C.; Maiorana, S.; Allegra, A.; Schirmeister, T.; Zappalà, M. Drug Synergism: Studies of Combination of RK-52 and Curcumin against Rhodesain of Trypanosoma brucei rhodesiense. ACS Med. Chem. Lett. 2020, 11, 806–810. [Google Scholar] [CrossRef] [PubMed]
- Ettari, R.; Previti, S.; Maiorana, S.; Allegra, A.; Schirmeister, T.; Grasso, S.; Zappalà, M. Drug combination studies of curcumin and genistein against rhodesain of Trypanosoma brucei rhodesiense. Nat. Prod. Res. 2019, 33, 3577–3581. [Google Scholar] [CrossRef]
- Ferreira, L.L.C.; Abreu, M.P.; Costa, C.B.; Leda, P.O.; Behrens, M.D.; Dos Santos, E.P. Curcumin and Its Analogs as a Therapeutic Strategy in Infections Caused by RNA Genome Viruses. Food Environ. Virol. 2022, 14, 120–137. [Google Scholar] [CrossRef]
- Hussain, Y.; Alam, W.; Ullah, H.; Dacrema, M.; Daglia, M.; Khan, H.; Arciola, C.R. Antimicrobial Potential of Curcumin: Therapeutic Potential and Challenges to Clinical Applications. Antibiotics 2022, 11, 322. [Google Scholar] [CrossRef]
- Vogel, A.; Pelletier, J. Examen chimique de la racine de Curcuma. J. Pharm. 1815, 1, 289–300. [Google Scholar]
- Gupta, S.C.; Patchva, S.; Koh, W.; Aggarwal, B.B. Discovery of curcumin, a component of golden spice, and its miraculous biological activities. Clin. Exp. Pharmacol. Physiol. 2012, 39, 283–299. [Google Scholar] [CrossRef]
- Ouyang, W.; Rutz, S.; Crellin, N.K.; Valdez, P.A.; Hymowitz, S.G. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu. Rev. Immunol. 2011, 29, 71–109. [Google Scholar] [CrossRef]
- Alroqi, F.J.; Chatila, T.A. T Regulatory cell biology in health and disease. Curr. Allergy Asthma Rep. 2016, 16, 27. [Google Scholar] [CrossRef]
- Momtazi-Borojeni, A.A.; Haftcheshmeh, S.M.; Esmaeili, S.A.; Johnston, T.P.; Abdollahi, E.; Sahebkar, A. Curcumin: A natural modulator of immune cells in systemic lupus erythematosus. Autoimmun. Rev. 2018, 17, 125–135. [Google Scholar] [CrossRef]
- Bose, S.; Panda, A.K.; Mukherjee, S.; Sa, G. Curcumin and tumor immune-editing: Resurrecting the immune system. Cell Div. 2015, 10, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Musolino, C.; Allegra, A.; Pioggia, G.; Gangemi, S. Immature myeloid-derived suppressor cells: A bridge between inflammation and cancer (Review). Oncol. Rep. 2017, 37, 671–683. [Google Scholar] [CrossRef] [Green Version]
- Liao, F.; Liu, L.; Luo, E.; Hu, J. Curcumin enhances anti-tumor immune response in tongue squamous cell carcinoma. Arch. Oral Biol. 2018, 92, 32–37. [Google Scholar] [CrossRef]
- Xu, B.; Yu, L.; Zhao, L.Z. Curcumin up regulates T helper 1 cells in patients with colon cancer. Am. J. Transl. Res. 2017, 9, 1866–1875. [Google Scholar] [PubMed]
- Zou, J.Y.; Su, C.H.; Luo, H.H.; Lei, Y.Y.; Zeng, B.; Zhu, H.S.; Chen, Z.G. Curcumin converts Foxp3+regulatory T cells to T helper 1 cells in patients with lung cancer. J. Cell Biochem. 2018, 119, 1420–1428. [Google Scholar] [CrossRef] [PubMed]
- Mathew, D.; Hsu, W.-L. Antiviral potential of curcumin. J. Funct. Foods 2018, 40, 692–699. [Google Scholar] [CrossRef]
- Youns, M.; Fathy, G.M. Upregulation of extrinsic apoptotic pathway in curcumin-mediated antiproliferative effect on human pancreatic carcinogenesis. J. Cell Biochem. 2013, 114, 2654–2665. [Google Scholar] [CrossRef] [PubMed]
- Shinde, P.; Kuhikar, R.; Kulkarni, R.; Khan, N.; Limaye, L.; Kale, V. Curcumin restores the engraftment capacity of aged hematopoietic stem cells and also reduces PD-1 expression on cytotoxic T cells. J. Tissue Eng. Regen. Med. 2021, 15, 388–400. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.G.; Veena, M.S.; Basak, S.K.; Han, E.; Tajima, T.; Gjertson, D.W.; Starr, J.; Eidelman, O.; Pollard, H.B.; Srivastava, M.; et al. Curcumin treatment suppresses IKKbeta kinase activity of salivary cells of patients with head and neck cancer: A pilot study. Clin. Cancer Res. 2011, 17, 5953–5961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Epstein, J.; Docena, G.; MacDonald, T.T.; Sanderson, I.R. Curcumin suppresses p38 mitogen-activated protein kinase activation, reduces IL-1beta and matrix metalloproteinase-3 and enhances IL-10 in the mucosa of children and adults with inflammatory bowel disease. Br J. Nutr. 2010, 103, 824–832. [Google Scholar] [CrossRef] [Green Version]
- Khajehdehi, P.; Pakfetrat, M.; Javidnia, K.; Azad, F.; Malekmakan, L.; Nasab, M.H.; Dehghanzadeh, G. Oral supplementation of turmeric attenuates proteinuria, transforming growth factor-beta and interleukin 8 levels in patients with overt type 2 diabetic nephropathy: A randomized, double-blind and placebo-controlled study. Scand. J. Urol. Nephrol. 2011, 45, 365–370. [Google Scholar] [CrossRef]
- James, J.S. Curcumin: Clinical trial finds no antiviral effect. AIDS Treat. News. 1996, 242, 1–2. [Google Scholar]
- Kalpravidh, R.W.; Siritanaratkul, N.; Insain, P.; Charoensakdi, R.; Panichkul, N.; Hatairaktham, S.; Srichairatanakool, S.; Phisalaphong, C.; Rachmilewitz, E.; Fucharoen, S. Improvement in oxidative stress and antioxidant parameters in beta-thalassemia/Hb E patients treated with curcuminoids. Clin. Biochem. 2010, 43, 424–429. [Google Scholar] [CrossRef]
- Bhattacharyya, S.; Md Sakib Hossain, D.; Mohanty, S.; Sankar Sen, G.; Chattopadhyay, S.; Banerjee, S.; Chakraborty, J.; Das, K.; Sarkar, D.; Das, T.; et al. Curcumin reverses T cell-mediated adaptive immune dysfunctions in tumor-bearing hosts. Cell Mol. Immunol. 2010, 7, 306–315. [Google Scholar] [CrossRef] [Green Version]
- Milano, F.; Mari, L.; van de Luijtgaarden, W.; Parikh, K.; Calpe, S.; Krishnadath, K.K. Nano-curcumin inhibits proliferation of esophageal adenocarcinoma cells and enhances the T cell mediated immune response. Front. Oncol. 2013, 3, 137. [Google Scholar] [CrossRef] [Green Version]
- Luo, F.; Song, X.; Zhang, Y.; Chu, Y. Low-dose curcumin leads to the inhibition of tumor growth via enhancing CTL-mediated antitumor immunity. Int. Immunopharmacol. 2011, 11, 1234–1240. [Google Scholar] [CrossRef]
- Lee, H.H.; Cho, H. Improved anti-cancer effect of curcumin on breast cancer cells by increasing the activity of natural killer cells. J. MicroBiol. Biotechnol. 2018, 28, 874–882. [Google Scholar] [CrossRef]
- Halder, R.C.; Almasi, A.; Sagong, B.; Leung, J.; Jewett, A.; Fiala, M. Curcuminoids and ω-3 fatty acids with anti-oxidants potentiate cytotoxicity of natural killer cells against pancreatic ductal adenocarcinoma cells and inhibit interferon γ production. Front. Physiol. 2015, 22, 6. [Google Scholar] [CrossRef]
- Zhang, H.G.; Kim, H.; Liu, C.; Yu, S.; Wang, J.; Grizzle, W.E.; Kimberly, R.P.; Barnes, S. Curcumin reverses breast tumor exosomes mediated immune suppression of NK cell tumor cytotoxicity. Biochim. Biophys. Acta 2007, 1773, 1116–1123. [Google Scholar] [CrossRef] [Green Version]
- Shi, J.; Zhao, Y.; Wang, Y.; Gao, W.; Ding, J.; Li, P.; Hu, L.; Shao, F. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 2014, 514, 187–192. [Google Scholar] [CrossRef]
- Salomao, R.; Brunialti, M.K.; Rapozo, M.M.; Baggio-Zappia, G.L.; Galanos, C.; Freudenberg, M. Bacterial sensing, cell signaling, and modulation of the immune response during sepsis. Shock 2012, 38, 227–242. [Google Scholar] [CrossRef] [Green Version]
- 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. Mediat. Inflamm. 2008, 2008, 317851. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Lukita-Atmadja, W.; Ito, Y.; Baker, G.L.; McCuskey, R.S. Effect of curcuminoids as anti-inflammatory agents on the hepatic microvascular response to endotoxin. Shock 2002, 17, 399–403. [Google Scholar] [CrossRef]
- Yang, C.; Wu, K.; Li, S.H.; You, Q. Protective effect of curcumin against cardiac dysfunction in sepsis rats. Pharm. Biol. 2013, 51, 482–487. [Google Scholar] [CrossRef] [Green Version]
- Gupta, S.C.; Prasad, S.; Kim, J.H.; Patchva, S.; Webb, L.J.; Priyadarsini, I.K.; Aggarwal, B.B. Multitargeting by curcumin as revealed by molecular interaction studies. Nat. Prod. Rep. 2011, 28, 1937–1955. [Google Scholar] [CrossRef]
- Wang, J.; Wang, H.; Zhu, R.; Liu, Q.; Fei, J.; Wang, S. Antiinflammatory activity of curcumin-loaded solid lipid nanoparticles in IL-1β transgenic mice subjected to the lipopolysaccharide-induced sepsis. Biomaterials 2015, 53, 475–483. [Google Scholar] [CrossRef]
- Zhong, W.; Qian, K.; Xiong, J.; Ma, K.; Wang, A.; Zou, Y. Curcumin alleviates lipopolysaccharide induced sepsis and liver failure by suppression of oxidative stress-related inflammation via PI3K/AKT and NF-κB related signaling. Biomed. Pharmacother. 2016, 83, 302–313. [Google Scholar] [CrossRef]
- Hafez, M.H.; El-Kazaz, S.E.; Alharthi, B.; Ghamry, H.I.; Alshehri, M.A.; Sayed, S.; Shukry, M.; El-Sayed, Y.S. The Impact of Curcumin on Growth Performance, Growth-Related Gene Expression, Oxidative Stress, and Immunological Biomarkers in Broiler Chickens at Different Stocking Densities. Animals 2022, 2, 958. [Google Scholar] [CrossRef]
- Bhoopathy, S.; Inbakandan, D.; Rajendran, T.; Chandrasekaran, K.; Prabha, S.B.; Reddy, B.A.; Kasilingam, R.; RameshKumar, V.; Dharani, G. Dietary supplementation of curcumin-loaded chitosan nanoparticles stimulates immune response in the white leg shrimp Litopenaeus vannamei challenged with Vibrio harveyi. Fish Shellfish Immunol. 2021, 117, 188–191. [Google Scholar] [CrossRef]
- Abdel-Tawwab, M.; Eissa, E.H.; Tawfik, W.A.; Abd Elnabi, H.E.; Saadony, S.; Bazina, W.K.; Ahmed, R.A. Dietary curcumin nanoparticles promoted the performance, antioxidant activity, and humoral immunity, and modulated the hepatic and intestinal histology of Nile tilapia fingerlings. Fish Physiol. Biochem. 2022, 48, 585–601. [Google Scholar] [CrossRef]
- Allegra, A.; Alonci, A.; Campo, S.; Penna, G.; Petrungaro, A.; Gerace, D.; Musolino, C. Circulating microRNAs: New biomarkers in diagnosis, prognosis and treatment of cancer (review). Int. J. Oncol. 2012, 41, 1897–1912. [Google Scholar] [CrossRef] [Green Version]
- Xu, C.; Xu, J.; Lu, L.; Tian, W.; Ma, J.; Wu, M. Identification of key genes and novel immune infiltration-associated biomarkers of sepsis. Innate Immun. 2020, 26, 666–682. [Google Scholar] [CrossRef]
- Dang, C.P.; Leelahavanichkul, A. Over-expression of miR-223 induces M2 macrophage through glycolysis alteration and attenuates LPS-induced sepsis mouse model, the cell-based therapy in sepsis. PLoS ONE 2020, 15, e0236038. [Google Scholar] [CrossRef]
- Ma, F.; Liu, F.; Ding, L.; You, M.; Yue, H.; Zhou, Y.; Hou, Y. Anti-inflammatory effects of curcumin are associated with down regulating microRNA-155 in LPS-treated macrophages and mice. Pharm. Biol. 2017, 55, 1263–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed]
- WHO. The Top 10 Causes of Death; World Health Organization (WHO) Report; WHO: Geneva, Switzerland, 2018. [Google Scholar]
- Campbell-Yesufu, O.T.; Gandhi, R.T. Update on human immunodeficiency virus (HIV)-2 infection. Clin. Infect. Dis. 2011, 52, 780–787. [Google Scholar] [CrossRef] [Green Version]
- van Tienen, C.; van der Loeff, M.S. Epidemiology of HIV-2 Infection in West Africa. In Encyclopedia of AIDS; Hope, T.J., Richman, D.D., Stevenson, M., Eds.; Springer: New York, NY, USA, 2018; pp. 513–523. [Google Scholar]
- Lodi, S.; Phillips, A.; Touloumi, G.; Geskus, R.; Meyer, L.; Thiébaut, R.; Pantazis, N.; Amo, J.D.; Johnson, A.M.; Babiker, A.; et al. Time from human immunodeficiency virus seroconversion to reaching CD4+ cell count thresholds <200, <350, and <500 Cells/mm³: Assessment of need following changes in treatment guidelines. Clin. Infect. Dis. 2011, 53, 817–825. [Google Scholar] [CrossRef] [Green Version]
- Ali, A.; Banerjea, A.C. Curcumin inhibits HIV-1 by promoting Tat protein degradation. Sci. Rep. 2016, 6, 27539–27639. [Google Scholar] [CrossRef] [Green Version]
- Jordan, W.C.; Drew, C.R. Curcumin–a natural herb with anti-HIV activity. J. Natl. Med. Assoc. 1996, 88, 333–433. [Google Scholar]
- Gandapu, U.; Chaitanya, R.K.; Kishore, G.; Reddy, R.C.; Kondapi, A.K. Curcumin-loaded apotransferrin nanoparticles provide efficient cellular uptake and effectively inhibit HIV-1 replication in vitro. PLoS ONE 2011, 6, e23388. [Google Scholar] [CrossRef] [Green Version]
- Vajragupta, O.; Boonchoong, P.; Morris, G.M.; Olson, A.J. Active site binding modes of curcumin in HIV-1 protease and integrase. Bioorg. Med. Chem. Lett. 2005, 15, 3364–3368. [Google Scholar] [CrossRef]
- Mazumder, A.; Neamati, N.; Sunder, S.; Schulz, J.; Pertz, H.; Eich, E.; Pommier, Y. Curcumin analogs with altered potencies against HIV-1 integrase as probes for biochemical mechanisms of drug action. J. Med. Chem. 1997, 40, 3057–3063. [Google Scholar] [CrossRef]
- Mazumder, A.; Raghavan, K.; Weinstein, J.; Kohn, K.W.; Pommier, Y. Inhibition of human immunodeficiency virus type-1 integrase by curcumin. Biochem. Pharmacol. 1995, 49, 1165–1170. [Google Scholar] [CrossRef] [PubMed]
- Karn, J.; Stoltzfus, C.M. Transcriptional and posttranscriptional regulation of HIV-1 gene expression. Cold Spring Harb. Perspect. Med. 2012, 2, a006916. [Google Scholar] [CrossRef] [PubMed]
- Prasad, S.; Tyagi, A.K. Curcumin and its analogues: A potential natural compound against HIV infection and AIDS. Food Funct. 2015, 6, 3412–3419. [Google Scholar] [CrossRef] [PubMed]
- Balasubramanyam, K.; Varier, R.A.; Altaf, M.; Swaminathan, V.; Siddappa, N.B.; Ranga, U.; Kundu, T.K. Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription. J. Biol. Chem. 2004, 279, 51163–51171. [Google Scholar] [CrossRef]
- Bhullar, K.S.; Jha, A.; Youssef, D.; Rupasinghe, H.P. Curcumin and its carbocyclic analogs: Structure-activity in relation to antioxidant and selected biological properties. Molecules 2013, 18, 5389–5404. [Google Scholar] [CrossRef] [Green Version]
- Available online: www.ClinicalTrial.gov (accessed on 1 September 2022).
- Lu, R.; Zhao, X.; Li, J.; Niu, P.; Yang, B.; Wu, H.; Wang, W.; Song, H.; Huang, B.; Zhu, N.; et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor binding. Lancet 2020, 395, 565-574. [Google Scholar] [CrossRef] [Green Version]
- Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef]
- Allegra, A.; Di Gioacchino, M.; Tonacci, A.; Musolino, C.; Gangemi, S. Immunopathology of SARS-CoV-2 Infection: Immune Cells and Mediators, Prognostic Factors, and Immune-Therapeutic Implications. Int. J. Mol. Sci. 2020, 21, 4782. [Google Scholar] [CrossRef]
- Murdaca, G.; Paladin, F.; Tonacci, A.; Isola, S.; Allegra, A.; Gangemi, S. The Potential Role of Cytokine Storm Pathway in the Clinical Course of Viral Respiratory Pandemic. Biomedicines 2021, 9, 1688. [Google Scholar] [CrossRef]
- Allegra, A.; Innao, V.; Allegra, A.G.; Musolino, C. Coagulopathy and thromboembolic events in patients with SARS-CoV-2 infection: Pathogenesis and management strategies. Ann. Hematol. 2020, 99, 1953–1965. [Google Scholar] [CrossRef]
- Mrityunjaya, M.; Pavithra, V.; Neelam, R.; Janhavi, P.; Halami, P.M.; Ravindra, P.V. Immune-boosting, antioxidant and anti-inflammatory food supplements targeting pathogenesis of COVID-19. Front. Immunol. 2020, 11, 570122. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Liu, L. Curcumin alleviates macrophage activation and lung inflammation induced by influenza virus infection through inhibiting the NF-κB signaling pathway. Influenza Other Respir. Viruses 2017, 11, 457–463. [Google Scholar] [CrossRef]
- Dhar, S.; Bhattacharjee, P. Promising role of curcumin against viral diseases emphasizing COVID-19 management: A review on the mechanistic insights with reference to host-pathogen interaction and immunomodulation. J. Funct. Foods 2021, 82, 104503. [Google Scholar] [CrossRef] [PubMed]
- Khanna, K.; Kohli, S.K.; Kaur, R.; Bhardwaj, A.; Bhardwaj, V.; Ohri, P.; Sharma, A.; Ahmad, A.; Bhardwaj, R.; Ahmad, P. Herbal immune-boosters: Substantial warriors of pandemic COVID-19 battle. Phytomedicine 2021, 85, 153361. [Google Scholar] [CrossRef] [PubMed]
- Jamiu, A.T.; Aruwa, C.E.; Abdulakeem, I.A.; Ayokunnun Ajao, A.; Sabiu, S. Phytotherapeutic evidence against coronaviruses and prospects for COVID-19. Pharmacogn. J. 2020, 12, 1252–1267. [Google Scholar] [CrossRef]
- Rattis, B.A.; Ramos, S.G.; Celes, M.R. Curcumin as a potential treatment for COVID-19. Front. Pharmacol. 2021, 12, 675287. [Google Scholar] [CrossRef] [PubMed]
- Kritis, P.; Karampela, I.; Kokoris, S.; Dalamaga, M. The combination of bromelain and curcumin as an immune-boosting nutraceutical in the prevention of severe COVID-19. Metabol. Open 2020, 8, 100066. [Google Scholar] [CrossRef] [PubMed]
- Pollard, A.J.; Bijker, E.M. A guide to vaccinology: From basic principles to new developments. Nat. Rev. Immunol. 2021, 21, 83–100. [Google Scholar] [CrossRef] [PubMed]
- Diks, A.M.; Overduin, L.A.; van Leenen, L.D.; Slobbe, L.; Jolink, H.; Visser, L.G.; van Dongen, J.J.M.; Berkowska, M.A. B-Cell immunophenotyping to predict vaccination outcome in the immunocompromised—A systematic review. Front. Immunol. 2021, 12, 690328. [Google Scholar] [CrossRef]
- Luo, W.; Yin, Q. B cell response to vaccination. Immunol. Investig. 2021, 50, 780–801. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Thimmulappa, R.K.; Mudnakudu-Nagaraju, K.K.; Shivamallu, C.; Subramaniam, K.J.T.; Radhakrishnan, A.; Bhojraj, S.; Kuppusamy, G. Antiviral and Immunomodulatory Activity of Curcumin: A Case for Prophylactic Therapy for COVID-19. Heliyon 2021, 7, e06350. [Google Scholar] [CrossRef]
- Vahedian-Azimi, A.; Abbasifard, M.; Rahimi-Bashar, F.; Guest, P.C.; Majeed, M.; Mohammadi, A.; Banach, M.; Jamialahmadi, T.; Shikar, A. Effectiveness of curcumin on outcomes of hospitalized COVID-19 patients: A systematic review of clinical trials. Nutrients 2022, 14, 256. [Google Scholar] [CrossRef]
- Liu, L.; Zhao, H.; Zhang, Y.; Wang, J.; Che, Y.; Dong, C.; Zhang, X.; Na, R.; Shi, H.; Jiang, L.; et al. Neonatal rhesus monkey is a potential animal model for studying pathogenesis of EV71 infection. Virology 2011, 412, 91–100. [Google Scholar] [CrossRef] [PubMed]
- Yip, C.C.; Lau, S.K.; Lo, J.Y.; Chan, K.H.; Woo, P.C.; Yuen, K.Y. Genetic characterization of EV71 isolates from 2004 to 2010 reveals predominance and persistent circulation of the newly proposed genotype D and recent emergence of a distinct lineage of subgenotype C2 in Hong Kong. Virol. J. 2013, 10, 222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, Z.L.; Mao, Q.Y.; Wang, Y.P.; Zhu, F.C.; Li, J.X.; Yao, X.; Gao, F.; Wu, X.; Xu, M.; Wang, J.Z. Progress on the research and development of inactivated EV71 whole-virus vaccines. Hum. Vaccin. Immunother. 2013, 9, 1701–1705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mao, Q.; Dong, C.; Li, X.; Gao, Q.; Guo, Z.; Yao, X.; Wang, Y.; Gao, F.; Li, F.; Xu, M.; et al. Comparative analysis of the immunogenicity and protective effects of inactivated EV71 vaccines in mice. PLoS ONE 2012, 7, e46043. [Google Scholar] [CrossRef]
- Permanent, B.; Kiener, T.K.; Meng, T.; Tan, Y.R.; Jia, Q.; Chow, V.T.; Kwang, J. Induction of protective immune responses against EV71 in mice by baculovirus encoding a novel expression cassette for capsid protein VP1. Antivir. Res. 2012, 95, 311–315. [Google Scholar]
- Qin, Y.; Lin, L.; Chen, Y.; Wu, S.; Si, X.; Wu, H.; Zhai, X.; Wang, Y.; Tong, L.; Pan, B.; et al. Curcumin inhibits the replication of enterovirus 71 in vitro. Acta Pharm. Sin. B 2014, 4, 284–294. [Google Scholar] [CrossRef] [Green Version]
- Teng, C.F.; Yu, C.H.; Chang, H.Y.; Hsieh, W.C.; Wu, T.H.; Lin, J.H.; Wu, H.C.; Jeng, L.B.; Su, I.J. Chemopreventive Effect of Phytosomal Curcumin on Hepatitis B Virus-Related Hepatocellular Carcinoma in A Transgenic Mouse Model. Sci. Rep. 2019, 9, 10338. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.; Kim, K.H.; Kim, H.Y.; Cho, H.K.; Sakamoto, N.; Cheong, J. Curcumin inhibits hepatitis C virus replication via suppressing the Akt-SREBP-1 pathway. FEBS Lett. 2010, 584, 707–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martins, C.V.; da Silva, D.L.; Neres, A.T.; Magalhães, T.F.; Watanabe, G.A.; Modolo, L.V.; Sabino, A.A.; de Fátima, A.; de Resende, M.A. Curcumin as a promising antifungal of clinical interest. J. Antimicrob. Chemother. 2009, 63, 337–339. [Google Scholar] [CrossRef] [PubMed]
- Colpitts, C.C.; Schang, L.M.; Rachmawati, H.; Frentzen, A.; Pfaender, S.; Behrendt, P.; Brown, R.J.; Bankwitz, D.; Steinmann, J.; Ott, M.; et al. Turmeric curcumin inhibits entry of all hepatitis C virus genotypes into human liver cells. Gut 2014, 63, 1137–1149. [Google Scholar] [CrossRef]
- Dogra, A.; Bhatt, S.; Magotra, A.; Sharma, A.; Kotwal, P.; Gour, A.; Wazir, P.; Singh, G.; Nandi, U. Intervention of curcumin on oral pharmacokinetics of daclatasvir in rat: A possible risk for long-term use. Phytother. Res. 2018, 32, 1967–1974. [Google Scholar] [CrossRef] [Green Version]
- Krishnaswamy, K. Turmeric: The Salt of Orient Is the Spice of Life; Allied Publishers Private Ltd.: Mumbai, India, 2008; pp. 175–178. [Google Scholar]
- Kutluay, S.B.; Doroghazi, J.; Roemer, M.E.; Triezenberg, S.J. Curcumin inhibits herpes simplex virus immediate-early gene expression by a mechanism independent of p300/CBP histone acetyltransferase activity. Virology 2008, 373, 239–247. [Google Scholar] [CrossRef]
- Harikumar, K.B.; Kuttan, G.; Kuttan, R. Inhibition of progression of erythroleukemia induced by Friend’s virus in BALBC mice by natural products—Berberine, curcumin and picroliv. J. Exp. Ther. Oncol. 2008, 7, 275–284. [Google Scholar]
- Harikumar, K.B.; Kuttan, R. Antiviral activity of Phyllanthus amarus and curcumin. Amla Res. Bull. 2006, 26, 198–205. [Google Scholar]
- Berk, A.J. Functions of adenovirus E1A. Cancer Surv. 1986, 5, 367–387. [Google Scholar] [PubMed]
- Jennings, M.R.; Parks, R.J. Antiviral Effects of Curcumin on Adenovirus Replication. Microorganisms 2020, 8, 1524. [Google Scholar] [CrossRef]
- Zuccotti, G.V.; Trabattoni, D.; Morelli, M.; Borgo NoVo, S.; Schneider, L.; Clerical, M. Immune modulation by lactoferrin and curcumin in children with recurrent respiratory infections. J. Biol. Regul. Homeost. Agents. 2009, 23, 119–123. [Google Scholar] [PubMed]
- Sharma, R.A.; Euden, S.A.; Platton, S.L.; Cooke, D.N.; Shafayat, A.; Hewitt, H.R.; Marczylo, T.H.; Morgan, B.; Hemingway, D.; Plummer, S.M.; et al. Phase I clinical trial of oral curcumin: Biomarkers of systemic activity and compliance. Clin. Cancer Res. 2004, 10, 6847–6854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, T.; Mazzoli, S.; Bechi, A.; Addonisio, P.; Mondaini, N.; Pagliai, R.C.; Bartoletti, R. Serenoa repens associated with urtica dioica (ProstaMEV) and curcumin and quercitin (FlogMEV) extracts are able to improve the efficacy of prulifloxacin in bacterial prostatitis patients: Results from a prospective randomised study. Int. J. Antimicrob. Agents 2009, 33, 549–553. [Google Scholar] [CrossRef] [PubMed]
- Jayhawk, S.M.; Saklani, A. Challenges and Opportunities in Drug Discovery from Plants. Curr. Sci. 2007, 92, 1251–1257. [Google Scholar] [CrossRef]
- Allegra, A.; Tonacci, A.; Pioggia, G.; Musolino, C.; Gangemi, S. Anticancer Activity of Rosmarinus officinalis L.: Mechanisms of Action and Therapeutic Potentials. Nutrients 2020, 12, 1739. [Google Scholar] [CrossRef] [PubMed]
- Siviero, A.; Gallo, E.; Maggini, V.; Gori, L.; Mugelli, A.; Firenzuoli, F.; Vannacci, A. Curcumin, a golden spice with a low bioavailability. J. Herbal. Med. 2015, 5, 57–70. [Google Scholar] [CrossRef]
- Moniruzzaman, M.; Min, T. Curcumin, curcumin nanoparticles and curcumin nanospheres: A review on their pharmacodynamics based on monogastric farm animal, poultry and fish nutrition. Pharmaceutics 2020, 12, 447. [Google Scholar] [CrossRef] [PubMed]
- Nair, M.; Jayant, R.D.; Kaushik, A.; Sagar, V. Getting into the brain: Potential of nanotechnology in the management of Neuro AIDS. Adv. Drug Del. Rev. 2016, 103, 202–217. [Google Scholar] [CrossRef]
- Hanafy, N.A.N.; El-Kemary, M.A. Silymarin/curcumin loaded albumin nanoparticles coated by chitosan as muco-inhalable delivery system observing anti-inflammatory and anti COVID-19 characterizations in oleic acid triggered lung injury and in vitro COVID-19 experiment. Int. J. Biol. Macromol. 2022, 198, 101–110. [Google Scholar] [CrossRef]
- Xu, J.; Yang, X.; Ji, J.; Gao, Y.; Qiu, N.; Xi, Y.; Liu, A.; Zhai, G. RVG-functionalized reduction sensitive micelles for the effective accumulation of doxorubicin in brain. J. Nanobiotechnol. 2021, 19, 251. [Google Scholar] [CrossRef] [PubMed]
- Badria, F.A.; Abdelaziz, A.E.; Hassan, A.H.; Elgazar, A.A.; Mazyed, E.A. Development of Provesicular Nanodelivery System of Curcumin as a Safe and Effective Antiviral Agent: Statistical Optimization, In Vitro Characterization, and Antiviral Effectiveness. Molecules 2020, 25, 5668. [Google Scholar] [CrossRef] [PubMed]
- Kumar, V.; Kumar, R.; Jain, V.K.; Nagpal, S. Preparation and characterization of nanocurcumin based hybrid virosomes as a drug delivery vehicle with enhanced anticancerous activity and reduced toxicity. Sci. Rep. 2021, 11, 368. [Google Scholar] [CrossRef]
- Fotticchia, T.; Vecchione, R.; Scognamiglio, P.L.; Guarnieri, D.; Calcagno, V.; Di Natale, C.; Attanasio, C.; De Gregorio, M.; Di Cicco, C.; Quagliariello, V.; et al. Enhanced Drug Delivery into Cell Cytosol via Glycoprotein H-Derived Peptide Conjugated Nanoemulsions. ACS Nano 2017, 11, 9802–9813. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.-Y.; Lin, J.-Y.; Chen, W.-Y.; Luo, L.; Wei-Guan Diau, E.; Chen, Y.-C. Functional Gold Nanoclusters as Antimicrobial Agents for Antibiotic-Resistant Bacteria. Nanomedicine 2010, 5, 755–764. [Google Scholar] [CrossRef] [PubMed]
- Lao, C.D.; Ruffin, M.T.; Normal, D.; Heath, D.D.; Murray, S.I.; Bailey, J.M.; Boggs, M.E.; Crowell, J.; Rock, C.L.; Brenner, D.E. Dose escalation of a curcuminoid formulation. BMC Complement Altern. Med. 2006, 6, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laorodphun, P.; Cherngwelling, R.; Panya, A.; Arjinajarn, P. Curcumin protects rats against gentamicin-induced nephrotoxicity by amelioration of oxidative stress, endoplasmic reticulum stress and apoptosis. Pharm. Biol. 2022, 60, 491–500. [Google Scholar] [CrossRef] [PubMed]
- Allegra, A.; Casciaro, M.; Lo Presti, E.; Musolino, C.; Gangemi, S. Harnessing Unconventional T Cells and Innate Lymphoid Cells to Prevent and Treat Hematological Malignancies: Prospects for New Immunotherapy. Biomolecules 2022, 12, 754. [Google Scholar] [CrossRef]
Virus | Effect | Type of Study | Ref. |
---|---|---|---|
HIV | Increase in CD4 + cells. Greater perception of well-being. | In vivo | [108] |
Effects on gp120 binding, integrase, topoisomerase II. Reduced HIV proliferation. | In vitro | [109,110,111,112,113] | |
Effect on Tat and Rev proteins. Reduced HIV proliferation. | In vitro | [114,115] | |
Effect on p300CREB-CBP histone acetyltransferases. | In vivo and in vitro | [116] | |
Effect on oxidative stress and HIV-1 proteases. | In vitro | [117] | |
SARS-COV 2 | Free radical deactivation. | In vitro | [124] |
Effect on inflammatory transcription elements, reduced expression of proinflammatory cytokines. | In vitro | [125,126] | |
Enhanced antibodies production after vaccination. | In vivo | [134] | |
Effect on IL-6 trans signal, NF-kB. Prevention of severe pneumonia. | In vivo | [135] | |
Enterovirus | Reduction of proteasome activity. Effect on p53 and p21 protein. | In vitro | [144] |
HCV | Reduction of inflammatory cytokines (IL-4, IL-6, IL-8, TNF. Increased expression of IL-10 and soluble intercellular adhesion molecule 1. | In vitro | [145] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Allegra, A.; Mirabile, G.; Ettari, R.; Pioggia, G.; Gangemi, S. The Impact of Curcumin on Immune Response: An Immunomodulatory Strategy to Treat Sepsis. Int. J. Mol. Sci. 2022, 23, 14710. https://doi.org/10.3390/ijms232314710
Allegra A, Mirabile G, Ettari R, Pioggia G, Gangemi S. The Impact of Curcumin on Immune Response: An Immunomodulatory Strategy to Treat Sepsis. International Journal of Molecular Sciences. 2022; 23(23):14710. https://doi.org/10.3390/ijms232314710
Chicago/Turabian StyleAllegra, Alessandro, Giuseppe Mirabile, Roberta Ettari, Giovanni Pioggia, and Sebastiano Gangemi. 2022. "The Impact of Curcumin on Immune Response: An Immunomodulatory Strategy to Treat Sepsis" International Journal of Molecular Sciences 23, no. 23: 14710. https://doi.org/10.3390/ijms232314710
APA StyleAllegra, A., Mirabile, G., Ettari, R., Pioggia, G., & Gangemi, S. (2022). The Impact of Curcumin on Immune Response: An Immunomodulatory Strategy to Treat Sepsis. International Journal of Molecular Sciences, 23(23), 14710. https://doi.org/10.3390/ijms232314710