Clinical Research Progress of Small Molecule Compounds Targeting Nrf2 for Treating Inflammation-Related Diseases
Abstract
:1. Introduction to Nrf2 Function
2. Research Progress of Nrf2 in Inflammation-Related Diseases
2.1. Nrf2 and Coronavirus Disease 2019 (COVID-19)
2.2. Nrf2 and Rheumatoid Arthritis
2.3. Nrf2 and Alzheimer’s Disease
2.4. Nrf2 and Parkinson’s Disease
2.5. Nrf2 and Lupus Erythematosus
2.6. Nrf2 and Liver Injury
2.6.1. Role of Nrf2 in Nonalcoholic Fatty Liver Disease (NAFLD)
2.6.2. Role of Nrf2 in Viral Hepatitis
2.6.3. Role of Nrf2 in Primary Biliary Cholangitis
2.6.4. Role of Nrf2 in Liver Fibrosis
3. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
ACE2 | angiotensin-converting enzyme 2 |
AD | Alzheimer’s disease |
AIH | autoimmune hepatitis |
AILI | APAP-induced liver injury |
ALD | alcoholic liver disease |
ALT | alanine aminotransferase |
AMPK | adenosine 5‘-monophosphate (AMP)-activated protein kinase |
Ang II | angiotensin II |
APAP | acetaminophen |
APP/PS1 | amyloid precursor protein plus presenilin-1 |
ARE | antioxidant response element |
ASH | alcoholic steatohepatitis |
AST | aspartate aminotransferase |
ATP | adenosine triphosphate |
Aβ | β-amyloid |
BACE | β-site amyloid-precursor-cleaving enzyme |
BMDCs | bone-marrow-derived dendritic cells |
CAT | catalase |
cGMP | cyclic guanosine 3′, 5′-monophosphate |
CIA | collagen-induced arthritis |
CNS | central nervous system |
COVID-19 | Coronavirus Disease 2019 |
COX-2 | Cyclooxygenase-2 |
Cul3 | CULLIN3 |
CYP 450 | cytochrome P450 |
CYP2E1 | cytochrome P450 2E1 |
DAPI | 4′,6-diamidino-2-phenylindole |
DFT | density functional theory |
DILI | drug-induced liver injury |
DMF | Dimethyl fumarate |
DPPH | 2,2-diphenyl-1-picrylhydrazyl |
dTHP-1 | Human THP-1 cells differentiated to the macrophage phenotype |
EC | Epieatechin |
ECG | Epieatechin gallate |
EGC | Epigalloeatechin |
EGCG | (−)-epigallocatechin-3-gallate |
FDA | Food and Drug Administration |
GCL | glutamate cysteine ligase |
GSH-Px | glutathione peroxidase |
GST | glutathione-S-transferases |
IC50 | half maximal inhibitory concentration |
IDMF | di-(methyl fumarate) |
IFN-I | I interferons |
IL-17 | interleukin-17 |
IL-1β | interleukin 1 beta |
IL-6 | interleukin 6 |
iNOS | inducible nitric oxide synthase |
GCLC | catalytic subunit of glutamate–cysteine ligase |
GCLM | gutamate-cysteine ligase modifier subunitl |
GSH | glutathione |
GSK-3 | glycogen synthase kinase 3 |
HAS | hydroxy-α-sanshool |
HBV | hepatitis B |
HBx | hepatitis B x protein |
HCC | hepatocellular carcinoma |
HCV | hepatitis C |
HFD | high-fat diet |
HMOX-1 | heme-oxigenase-1 |
HO-1 | heme-oxigenase-1heme-oxigenase-1 |
HSC | hepatic stellate cell ICC intrahepatic cholangiocarcinoma |
Keap1 | Kelch-like ECH-associated protein 1 |
LN | lupus nephritis |
LPS | Lipopolysaccharide |
LSEC | liver sinusoidal endothelial cell |
MAF | musculoaponeurotic fibrosarcoma |
MAO | Monoamine oxidase |
MS | multiple sclerosis |
NAFLD | non-alcoholic fatty liver disease |
NAPQI | N-acetyl-1,4-benzoquinone imine |
NDP52 | nuclear dot protein 52 |
NF-κB | nuclear factor kappa B |
NLR | nucleotide-binding domain leucine-rich repeat-related |
NLRP3 | nucleotide-binding domain leucine-rich repeat-related (NLR) family pyrin do-main-containing 3 |
NOX | NADPH-oxidase |
NQO1 | NAD (P) H-quinone oxidoreductase 1 |
Nrf2 | nuclear factor E2-related factor 2 |
NS3 | nonstructural protein 3 |
NS5A | nonstructural protein 5A |
OSI | oxidative stress index |
PBC | primary biliary cholangitis PSC primary sclerosing cholangitis |
PD | Parkinson’s disease |
PTEN | phosphatase and tensin homolog |
PINK | phosphatase and tensin homolog (PTEN)-induced kinase |
PSC | primary sclerosing cholangitis |
qPCR | Quantitative Real-time PCR |
RNA | Ribonucleic Acid |
RA | Rheumatoid arthritis |
ROS | reactive oxygen species |
SARS-CoV-2 | severe acute respiratory syndrome coronavirus 2 |
SFN | Sulforaphane |
SLE | systemic lupus erythematosus |
Socs3 | suppresses cytokine signaling 3 |
SOD | superoxide dismutase |
STAT3 | Signal transducer and activator of transcription 3 |
TAS | total antioxidant status |
Th17 | T helper type 17 |
TLR7 | Toll-like receptor 7 |
TLR9 | Toll-like receptor 9 |
TMPRSS2 | transmembrane protease serine 2 |
TNF-α | tumor necrosis factor alpha |
TrX-1 | thioredoxin-1 |
TRX | thioredoxin |
TrxR1 | thioredoxin reductase 1 |
TOS | total oxidant status |
UDCA | ursodeoxycholic acid |
α-syn | α-synuclein |
γ-GCS | γ-glutamyl cysteine synthetase |
References
- Xie, J.; Gorle, N.; Vandendriessche, C.; Van Imschoot, G.; Van Wonterghem, E.; Van Cauwenberghe, C.; Parthoens, E.; Van Hamme, E.; Lippens, S.; Van Hoecke, L.; et al. Low-grade peripheral inflammation affects brain pathology in the App(NL-G-F)mouse model of Alzheimer’s disease. Acta Neuropathol. Commun. 2021, 9. [Google Scholar] [CrossRef]
- La Vitola, P.; Balducci, C.; Baroni, M.; Artioli, L.; Santamaria, G.; Castiglioni, M.; Cerovic, M.; Colombo, L.; Caldinelli, L.; Pollegioni, L.; et al. Peripheral inflammation exacerbates alpha-synuclein toxicity and neuropathology in Parkinson’s models. Neuropathol. Appl. Neurobiol. 2021, 47, 43–60. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.P.; Hahn, B.H.; Bischoff, D.S. Identification and Contribution of Inflammation-Induced Novel MicroRNA in the Pathogenesis of Systemic Lupus Erythematosus. Front. Immunol. 2022, 13, 848149. [Google Scholar] [CrossRef]
- Lanquetin, A.; Leclercq, S.; de Timary, P.; Segobin, S.; Naveau, M.; Coulbault, L.; Maccioni, P.; Lorrai, I.; Colombo, G.; Vivien, D.; et al. Role of inflammation in alcohol-related brain abnormalities: A translational study. Brain Commun. 2021, 3, fcab154. [Google Scholar] [CrossRef] [PubMed]
- Del Valle, D.M.; Kim-Schulze, S.; Huang, H.-H.; Beckmann, N.D.; Nirenberg, S.; Wang, B.; Lavin, Y.; Swartz, T.H.; Madduri, D.; Stock, A.; et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat. Med. 2020, 26, 1636–1643. [Google Scholar] [CrossRef]
- Jiang, X.; Zhou, X.; Yu, X.; Chen, X.; Hu, X.; Lu, J.; Zhao, H.; Cao, Q.; Gu, Y.; Yang, Y.; et al. High expression of nuclear NRF2 combined with NFE2L2 alterations predicts poor prognosis in esophageal squamous cell carcinoma patients. Mod. Pathol. 2022, 35, 929–937. [Google Scholar] [CrossRef] [PubMed]
- Gessl, I.; Aletaha, D.; Mandl, P. Response to: ‘Correspondence on ‘Role of joint damage, malalignment and inflammation in articular tenderness in rheumatoid arthritis, psoriatic arthritis and osteoarthritis’’ by Dumoulin et al. Ann. Rheum. Dis. 2021. [Google Scholar] [CrossRef] [PubMed]
- Kopacz, A.; Kloska, D.; Forman, H.J.; Jozkowicz, A.; Grochot-Przeczek, A. Beyond repression of Nrf2: An update on Keap1. Free Radic. Biol. Med. 2020, 157, 63–74. [Google Scholar] [CrossRef]
- Gao, L.; Kumar, V.; Vellichirammal, N.N.; Park, S.-Y.; Rudebush, T.L.; Yu, L.; Son, W.-M.; Pekas, E.J.; Wafi, A.M.; Hong, J.; et al. Functional, proteomic and bioinformatic analyses of Nrf2-and Keap1-null skeletal muscle. J. Physiol. Lond. 2020, 598, 5427–5451. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Yang, C.; Elsheikh, N.A.H.; Li, C.; Yang, F.; Wang, G.; Li, L. HO-1 reduces heat stress-induced apoptosis in bovine granulosa cells by suppressing oxidative stress. Aging 2019, 11, 5535–5547. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Zheng, J.; Wang, M.; Zhang, J.; Tian, T.; Wang, Z.; Yuan, S.; Liu, L.; Zhu, P.; Gu, F.; et al. NQO1 promotes an aggressive phenotype in hepatocellular carcinoma via amplifying ERK-NRF2 signaling. Cancer Sci. 2021, 112, 641–654. [Google Scholar] [CrossRef]
- Mohar, I.; Botta, D.; White, C.C.; McConnachie, L.A.; Kavanagh, T.J. Glutamate cysteine ligase (GCL) transgenic and gene-targeted mice for controlling glutathione synthesis. Curr. Protoc. Toxicol. 2009, 39, 6.16.1–6.16.20. [Google Scholar] [CrossRef]
- Ayna, A.; Khosnaw, L.; Temel, Y.; Ciftci, M. Antibiotics as Inhibitor of Glutathione S-transferase: Biological Evaluation and Molecular Structure Studies. Curr. Drug Metab. 2021, 22, 308–314. [Google Scholar] [CrossRef] [PubMed]
- Sakamoto, T.; Imai, H. Hydrogen peroxide produced by superoxide dismutase SOD-2 activates sperm in Caenorhabditis elegans. J. Biol. Chem. 2017, 292, 14804–14813. [Google Scholar] [CrossRef]
- Kobayashi, S.; Ikeda, Y.; Shigeno, Y.; Konno, H.; Fujii, J. γ-Glutamylcysteine synthetase and γ-glutamyl transferase as differential enzymatic sources of γ-glutamylpeptides in mice. Amino Acids 2020, 52, 555–566. [Google Scholar] [CrossRef]
- Yuan, J.; Wang, Z.; Wang, B.; Mei, H.; Zhai, X.; Zhuang, Z.; Chen, M.; Zhang, Y. Non-Specific Immunity Associated Gut Microbiome in Aristichthys nobilis under Different Rearing Strategies. Genes 2021, 12, 916. [Google Scholar] [CrossRef] [PubMed]
- Bartolini, D.; Arato, I.; Mancuso, F.; Giustarini, D.; Bellucci, C.; Vacca, C.; Aglietti, M.C.; Stabile, A.M.; Rossi, R.; Cruciani, G.; et al. Melatonin modulates Nrf2 activity to protect porcine pre-pubertal Sertoli cells from the abnormal H2O2 generation and reductive stress effects of cadmium. J. Pineal Res. 2022, 73, e12806. [Google Scholar] [CrossRef] [PubMed]
- Kumar, H.; Kumar, R.M.; Bhattacharjee, D.; Somanna, P.; Jain, V. Role of Nrf2 Signaling Cascade in Breast Cancer: Strategies and Treatment. Front. Pharmacol. 2022, 13, 720076. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Donquiles, C.; Alonso-Molero, J.; Fernandez-Villa, T.; Vilorio-Marqués, L.; Molina, A.J.; Martín, V. The NRF2 transcription factor plays a dual role in colorectal cancer: A systematic review. PLoS ONE 2017, 12, e0177549. [Google Scholar] [CrossRef] [PubMed]
- Zinatizadeh, M.R.; Schock, B.; Chalbatani, G.M.; Zarandi, P.K.; Jalali, S.A.; Miri, S.R. The Nuclear Factor Kappa B (NF-kB) signaling in cancer development and immune diseases. Genes Dis. 2021, 8, 287–297. [Google Scholar] [CrossRef] [PubMed]
- Al Riyami, N.; Sheik, S. COVID-19 and Pregnancy: A narrative review of maternal and perinatal outcomes. Sultan Qaboos Univ. Med. J. 2022, 22, 167–178. [Google Scholar] [CrossRef]
- Jacob, S.; Kapadia, R.; Soule, T.; Luo, H.; Schellenberg, K.L.; Douville, R.N.; Pfeffer, G. Neuromuscular Complications of SARS-CoV-2 and Other Viral Infections. Front. Neurol. 2022, 13, 914411. [Google Scholar] [CrossRef]
- Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krueger, 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]
- Liskova, A.; Samec, M.; Koklesova, L.; Samuel, S.M.; Zhai, K.; Al-Ishaq, R.K.; Abotaleb, M.; Nosal, V.; Kajo, K.; Ashrafizadeh, M.; et al. Flavonoids against the SARS-CoV-2 induced inflammatory storm. Biomed. Pharmacother. 2021, 138, 111430. [Google Scholar] [CrossRef]
- Gumus, H.; Erat, T.; Ozturk, I.; Demir, A.; Koyuncu, I. Oxidative stress and decreased Nrf2 level in pediatric patients with COVID-19. J. Med. Virol. 2022, 94, 2259–2264. [Google Scholar] [CrossRef]
- Lin, C.Y.; Yao, C.A. Potential Role of Nrf2 Activators with Dual Antiviral and Anti-Inflammatory Properties in the Management of Viral Pneumonia. Infect. Drug Resist. 2020, 13, 1735–1741. [Google Scholar] [CrossRef]
- Zhu, Y.; Xie, D.-Y. Docking Characterization and in vitro Inhibitory Activity of Flavan-3-ols and Dimeric Proanthocyanidins Against the Main Protease Activity of SARS-CoV-2. Front. Plant Sci. 2020, 11, 601316. [Google Scholar] [CrossRef] [PubMed]
- Kesic, M.J.; Simmons, S.O.; Bauer, R.; Jaspers, I. Nrf2 expression modifies influenza A entry and replication in nasal epithelial cells. Free Radic. Biol. Med. 2011, 51, 444–453. [Google Scholar] [CrossRef]
- Xiang, Q.; Cheng, L.; Zhang, R.; Liu, Y.; Wu, Z.; Zhang, X. Tea Polyphenols Prevent and Intervene in COVID-19 through Intestinal Microbiota. Foods 2022, 11, 506. [Google Scholar] [CrossRef]
- Zhang, Z.; Zhang, X.; Bi, K.; He, Y.; Yan, W.; Yang, C.S.; Zhang, J. Potential protective mechanisms of green tea polyphenol EGCG against COVID-19. Trends Food Sci. Technol. 2021, 114, 11–24. [Google Scholar] [CrossRef]
- Uchiyama, Y.; Suzuki, T.; Mochizuki, K.; Goda, T. Dietary supplementation with (-)-epigallocatechin-3-gallate reduces inflammatory response in adipose tissue of non-obese type 2 diabetic Goto-Kakizaki (GK) rats. J. Agric. Food Chem. 2013, 61, 11410–11417. [Google Scholar] [CrossRef]
- Dai, W.; Lou, N.; Xie, D.; Hu, Z.; Song, H.; Lu, M.; Shang, D.; Wu, W.; Peng, J.; Yin, P.; et al. N-Ethyl-2-Pyrrolidinone-Substituted Flavan-3-Ols with Anti-inflammatory Activity in Lipopolysaccharide-Stimulated Macrophages Are Storage-Related Marker Compounds for Green Tea. J. Agric Food Chem. 2020, 68, 12164–12172. [Google Scholar] [CrossRef]
- Jang, M.; Park, Y.-I.; Cha, Y.-E.; Park, R.; Namkoong, S.; Lee, J.I.; Park, J. Tea Polyphenols EGCG and Theaflavin Inhibit the Activity of SARS-CoV-2 3CL-Protease In Vitro. Evid. Based Complement. Altern. Med. 2020, 2020, 5630838. [Google Scholar] [CrossRef]
- Hurst, B.L.; Dickinson, D.; Hsu, S. Epigallocatechin-3-Gallate (EGCG) Inhibits SARS-CoV-2 Infection in Primate Epithelial Cells: (A Short Communication). Microbiol. Infect. Dis. 2021, 5, 1–6. [Google Scholar] [CrossRef]
- Qizhen, D.; Heyuan, J. Pharmacology and Application of Tea Pigment. China Tea 1997, 05, 36–37. [Google Scholar]
- Ali, M.; Bonay, M.; Vanhee, V.; Vinit, S.; Deramaudt, T.B. Comparative effectiveness of 4 natural and chemical activators of Nrf2 on inflammation, oxidative stress, macrophage polarization, and bactericidal activity in an in vitro macrophage infection model. PLoS ONE 2020, 15, e0234484. [Google Scholar] [CrossRef]
- Mohanty, S.; Sahoo, A.K.; Konkimalla, V.B.; Pal, A.; Si, S.C. Naringin in Combination with Isothiocyanates as Liposomal Formulations Potentiates the Anti-inflammatory Activity in Different Acute and Chronic Animal Models of Rheumatoid Arthritis. ACS Omega 2020, 5, 28319–28332. [Google Scholar] [CrossRef]
- Gasparello, J.; D’Aversa, E.; Papi, C.; Gambari, L.; Grigolo, B.; Borgatti, M.; Finotti, A.; Gambari, R. Sulforaphane inhibits the expression of interleukin-6 and interleukin-8 induced in bronchial epithelial IB3-1 cells by exposure to the SARS-CoV-2 Spike protein. Phytomedicine 2021, 87, 153583. [Google Scholar] [CrossRef]
- Maina, S.; Misinzo, G.; Bakari, G.; Kim, H.-Y. Human, Animal and Plant Health Benefits of Glucosinolates and Strategies for Enhanced Bioactivity: A Systematic Review. Molecules 2020, 25, 3682. [Google Scholar] [CrossRef]
- Wang, T.; Dai, F.; Li, G.-H.; Chen, X.-M.; Li, Y.-R.; Wang, S.-Q.; Ren, D.-M.; Wang, X.-N.; Lou, H.-X.; Zhou, B.; et al. Trans-4,4′-dihydroxystilbene ameliorates cigarette smoke-induced progression of chronic obstructive pulmonary disease via inhibiting oxidative stress and inflammatory response. Free Radic. Biol. Med. 2020, 152, 525–539. [Google Scholar] [CrossRef]
- Moustafa, P.E.; Abdelkader, N.F.; El Awdan, S.A.; El-Shabrawy, O.A.; Zaki, H.F. Extracellular Matrix Remodeling and Modulation of Inflammation and Oxidative Stress by Sulforaphane in Experimental Diabetic Peripheral Neuropathy. Inflammation 2018, 41, 1460–1476. [Google Scholar] [CrossRef]
- Sangeeta, M.; Abhisek, P.; Badireenath, K.V.; Sudam, C.S. Anti-Inflammatory and Anti-Granuloma Activity of Sulforaphane, a Naturally Occurring Isothiocyanate from Broccoli (Brassica Oleracea). Asian J. Pharm. Clin. Res. 2018, 11, 411–416. [Google Scholar]
- Ordonez, A.A.; Bullen, C.K.; Villabona-Rueda, A.F.; Thompson, E.A.; Turner, M.L.; Merino, V.F.; Yan, Y.; Kim, J.; Davis, S.L.; Komm, O.; et al. Sulforaphane exhibits antiviral activity against pandemic SARS-CoV-2 and seasonal HCoV-OC43 coronaviruses in vitro and in mice. Commun. Biol. 2022, 5, 242. [Google Scholar] [CrossRef]
- Boehm, J.; Davis, R.; Murar, C.E.; Li, T.; McCleland, B.; Dong, S.; Yan, H.; Kerns, J.; Moody, C.J.; Wilson, A.J.; et al. Discovery of a crystalline sulforaphane analog with good solid-state stability and engagement of the Nrf2 pathway in vitro and in vivo. Bioorganic Med. Chem. 2019, 27, 579–588. [Google Scholar] [CrossRef]
- Xin, M.; Wang, L.; Mou, X.; Zhao, F. Synthesis, Anti-inflammatory Activity and Molecula Docking of Resveratrol Amide Derivatives. J. Yantai Univ. Nat. Sci. Eng. Ed. 2022, 35, 1–10. [Google Scholar] [CrossRef]
- Wicinski, M.; Socha, M.; Walczak, M.; Wodkiewicz, E.; Malinowski, B.; Rewerski, S.; Gorski, K.; Pawlak-Osinska, K. Beneficial Effects of Resveratrol Administration-Focus on Potential Biochemical Mechanisms in Cardiovascular Conditions. Nutrients 2018, 10, 1813. [Google Scholar] [CrossRef]
- de Ligt, M.; Hesselink, M.K.C.; Jorgensen, J.; Hoebers, N.; Blaak, E.E.; Goossens, G.H. Resveratrol supplementation reduces ACE2 expression in human adipose tissue. Adipocyte 2021, 10, 408–411. [Google Scholar] [CrossRef]
- Jiang, Z.X.; Zhang, H.; Gao, J.; Yu, H.; Han, R.F.; Zhu, L.; Chen, X.; Fan, Q.; Hao, P.; Wang, L.M.; et al. ACE2 Expression Is Upregulated in Inflammatory Corneal Epithelial Cells and Attenuated by Resveratrol. Investig. Ophthalmol. Vis. Sci. 2021, 62, 25. [Google Scholar] [CrossRef]
- ter Ellen, B.M.; Kumar, N.D.; 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]
- Peng, W.; Ma, Y.-Y.; Zhang, K.; Zhou, A.-Y.; Zhang, Y.; Wang, H.; Du, Z.; Zhao, D.-G. Synthesis and Biological Evaluation of Novel Resveratrol-NSAID Derivatives as Anti-inflammatory Agents. Chem. Pharm. Bull. 2016, 64, 609–615. [Google Scholar] [CrossRef]
- Pei, L.; Xu, Q. Structure-Antioxidant Activity Relationship of Resveratrol and its Analogues. Nat. Prod. Res. Dev. 2017, 29, 1277–1283+1306. [Google Scholar] [CrossRef]
- Cooke, M.L.; Fyfe, M.C.T.; Teobald, B.J. Preparation of fumarate compounds with antiinflammatory properties. Eurasian Patent WO2022038365A2, 24 February 2022. [Google Scholar]
- Gold, R.; Linker, R.A.; Stangel, M. Fumaric acid and its esters: An emerging treatment for multiple sclerosis with antioxidative mechanism of action. Clin. Immunol. 2012, 142, 44–48. [Google Scholar] [CrossRef] [PubMed]
- Olagnier, D.; Farahani, E.; Thyrsted, J.; Blay-Cadanet, J.; Herengt, A.; Idorn, M.; Hait, A.; Hernaez, B.; Knudsen, A.; Iversen, M.B.; et al. SARS-CoV2-mediated suppression of NRF2-signaling reveals potent antiviral and anti-inflammatory activity of 4-octyl-itaconate and dimethyl fumarate. Nat. Commun. 2020, 11, 4938. [Google Scholar] [CrossRef] [PubMed]
- Swindell, W.R.; Bojanowski, K.; Chaudhuri, R.K. A novel fumarate, isosorbide di-(methyl fumarate) (IDMF), replicates astrocyte transcriptome responses to dimethyl fumarate (DMF) but specifically down-regulates genes linked to a reactive phenotype. Biochem. Biophys. Res. Commun. 2020, 532, 475–481. [Google Scholar] [CrossRef] [PubMed]
- Sorrenti, V.; Vanella, L.; Platania, C.B.M.; Greish, K.; Bucolo, C.; Pittalà, V.; Salerno, L. Novel Heme Oxygenase-1 (HO-1) Inducers Based on Dimethyl Fumarate Structure. Int. J. Mol. Sci. 2020, 21, 9541. [Google Scholar] [CrossRef]
- Muri, J.; Wolleb, H.; Broz, P.; Carreira, E.M.; Kopf, M. Electrophilic Nrf2 activators and itaconate inhibit inflammation at low dose and promote IL-1β production and inflammatory apoptosis at high dose. Redox Biol. 2020, 36, 101647. [Google Scholar] [CrossRef]
- Bormann, M.; Alt, M.; Schipper, L.; van de Sand, L.; Le-Trilling, V.T.K.; Rink, L.; Heinen, N.; Madel, R.J.; Otte, M.; Wuensch, K.; et al. Turmeric Root and Its Bioactive Ingredient Curcumin Effectively Neutralize SARS-CoV-2 In Vitro. Viruses 2021, 13, 1914. [Google Scholar] [CrossRef]
- Marin-Palma, D.; Tabares-Guevara, J.H.; Zapata-Cardona, M.I.; Florez-alvarez, L.; Yepes, L.M.; Rugeles, M.T.; Zapata-Builes, W.; Hernandez, J.C.; Taborda, N.A. Curcumin Inhibits In Vitro SARS-CoV-2 Infection In Vero E6 Cells through Multiple Antiviral Mechanisms. Molecules 2021, 26, 6900. [Google Scholar] [CrossRef]
- Venkateswarlu, S.; Ramachandra, M.S.; Subbaraju, G.V. Synthesis and biological evaluation of polyhydroxycurcuminoids. Bioorg. Med. Chem. 2005, 13, 6374–6380. [Google Scholar] [CrossRef]
- Lee, K.-H.; Aziz, F.H.A.; Syahida, A.; Abas, F.; Shaari, K.; Israf, D.A.; Lajis, N.H. Synthesis and biological evaluation of curcumin-like diarylpentanoid analogues for anti-inflammatory, antioxidant and anti-tyrosinase activities. Eur. J. Med. Chem. 2009, 44, 3195–3200. [Google Scholar] [CrossRef]
- Su, I.-J.; Chang, H.-Y.; Wang, H.-C.; Tsai, K.-J. A Curcumin Analog Exhibits Multiple Biologic Effects on the Pathogenesis of Alzheimer’s Disease and Improves Behavior, Inflammation, and beta-Amyloid Accumulation in a Mouse Model. Int. J. Mol. Sci. 2020, 21, 5459. [Google Scholar] [CrossRef] [PubMed]
- Schloer, S.; Brunotte, L.; Mecate-Zambrano, A.; Zheng, S.; Tang, J.; Ludwig, S.; Rescher, U. Drug synergy of combinatory treatment with remdesivir and the repurposed drugs fluoxetine and itraconazole effectively impairs SARS-CoV-2 infection in vitro. Br. J. Pharmacol. 2021, 178, 2339–2350. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Gu, Y.-H.; Liu, M.; Bai, Y.; Wang, H.-L. Fluoxetine protects against methamphetamine-induced lung inflammation by suppressing oxidative stress through the SERT/p38 MAPK/Nrf2 pathway in rats. Mol. Med. Rep. 2017, 15, 673–680. [Google Scholar] [CrossRef] [PubMed]
- Zimniak, M.; Kirschner, L.; Hilpert, H.; Geiger, N.; Danov, O.; Oberwinkler, H.; Steinke, M.; Sewald, K.; Seibel, J.; Bodem, J. The serotonin reuptake inhibitor Fluoxetine inhibits SARS-CoV-2 in human lung tissue. Sci. Rep. 2021, 11, 5890. [Google Scholar] [CrossRef] [PubMed]
- Dechaumes, A.; Nekoua, M.P.; Belouzard, S.; Sane, F.; Engelmann, I.; Dubuisson, J.; Alidjinou, E.K.; Hober, D. Fluoxetine Can Inhibit SARS-CoV-2 In Vitro. Microorganisms 2021, 9, 339. [Google Scholar] [CrossRef] [PubMed]
- Park, J.-Y.; Kim, S.-W.; Lee, J.-K.; Im, W.B.; Jin, B.K.; Yoon, S.-H. Simplified Heterocyclic Analogues of Fluoxetine Inhibit Inducible Nitric Oxide Production in Lipopolysaccharide-Induced BV2 Cells. Biol. Pharm. Bull. 2011, 34, 538–544. [Google Scholar] [CrossRef]
- Yoon, S.-H.; Lee, E.; Cho, D.-Y.; Ko, H.M.; Baek, H.Y.; Choi, D.-K.; Kim, E.; Park, J.-Y. Synthesis of 4-(3-oxo-3-phenylpropyl)morpholin-4-ium chloride analogues and their inhibitory activities of nitric oxide production in lipopolysaccharide-induced BV2 cells. Bioorg. Med. Chem. Lett. 2021, 36, 127780. [Google Scholar] [CrossRef]
- Sun, Q.; Ye, F.; Liang, H.; Liu, H.; Li, C.; Lu, R.; Huang, B.; Zhao, L.; Tan, W.; Lai, L. Bardoxolone and bardoxolone methyl, two Nrf2 activators in clinical trials, inhibit SARS-CoV-2 replication and its 3C-like protease. Signal Transduct. Target. Ther. 2021, 6, 212. [Google Scholar] [CrossRef]
- Honda, T.; Rounds, B.V.; Bore, L.; Finlay, H.J.; Favaloro, F.G.; Suh, N.; Wang, Y.; Sporn, M.B.; Gribble, G.W. Synthetic Oleanane and Ursane Triterpenoids with Modified Rings A and C: A Series of Highly Active Inhibitors of Nitric Oxide Production in Mouse Macrophages. J. Med. Chem. 2000, 43, 4233–4246. [Google Scholar] [CrossRef]
- Turpaev, K.; Welsh, N. Aromatic malononitriles stimulate the resistance of insulin-producing beta-cells to oxidants and inflammatory cytokines. Eur. J. Pharmacol. 2016, 784, 69–80. [Google Scholar] [CrossRef]
- Zheng, S.; Santosh Laxmi, Y.R.; David, E.; Dinkova-Kostova, A.T.; Shiavoni, K.H.; Ren, Y.; Zheng, Y.; Trevino, I.; Bumeister, R.; Ojima, I.; et al. Synthesis, Chemical Reactivity as Michael Acceptors, and Biological Potency of Monocyclic Cyanoenones, Novel and Highly Potent Anti-inflammatory and Cytoprotective Agents. J. Med. Chem. 2012, 55, 4837–4846. [Google Scholar] [CrossRef]
- Fu, L.; Lin, Q.-x.; Onyango, E.O.; Liby, K.T.; Sporn, M.B.; Gribble, G.W. Design, synthesis, and biological activity of second-generation synthetic oleanane triterpenoids. Org. Biomol. Chem. 2017, 15, 6001–6005. [Google Scholar] [CrossRef] [PubMed]
- Fu, L.; Gribble, G.W. Efficient and Scalable Synthesis of Bardoxolone Methyl (CDDO-methyl Ester). Org. Lett. 2013, 15, 1622–1625. [Google Scholar] [CrossRef] [PubMed]
- Bellucci, E.; Terenzi, R.; La Paglia, G.M.C.; Gentileschi, S.; Tripoli, A.; Tani, C.; Alunno, A. One year in review 2016: Pathogenesis of rheumatoid arthritis. Clin. Exp. Rheumatol. 2016, 34, 793–801. [Google Scholar] [PubMed]
- Ito, H.; Moritoshi, F.; Hashimoto, M.; Tanaka, M.; Matsuda, S. Control of articular synovitis for bone and cartilage regeneration in rheumatoid arthritis. Inflamm. Regen. 2018, 38, 7. [Google Scholar] [CrossRef] [PubMed]
- Guo, Q.; Wang, Y.; Xu, D.; Nossent, J.; Pavlos Nathan, J.; Xu, J. Rheumatoid arthritis: Pathological mechanisms and modern pharmacologic therapies. Bone Res. 2018, 6, 107–120. [Google Scholar] [CrossRef] [PubMed]
- Maria Quinonez-Flores, C.; Aidee Gonzalez-Chavez, S.; Del Rio Najera, D.; Pacheco-Tena, C. Oxidative Stress Relevance in the Pathogenesis of the Rheumatoid Arthritis: A Systematic Review. BioMed Res. Int. 2016, 2016, 6097417. [Google Scholar] [CrossRef]
- Chadha, S.; Behl, T.; Kumar, A.; Khullar, G.; Arora, S. Role of Nrf2 in rheumatoid arthritis. Curr. Res. Transl. Med. 2020, 68, 171–181. [Google Scholar] [CrossRef] [PubMed]
- Holmstrom, K.M.; Kostov, R.V.; Dinkova-Kostova, A.T. The multifaceted role of Nrf2 in mitochondrial function. Curr. Opin. Toxicol. 2016, 1, 80–91. [Google Scholar] [CrossRef] [PubMed]
- Narapureddy, B.; Dubey, D. Clinical evaluation of dimethyl fumarate for the treatment of relapsing-remitting multiple sclerosis: Efficacy, safety, patient experience and adherence. Patient Prefer. Adherence 2019, 13, 1655–1666. [Google Scholar] [CrossRef] [PubMed]
- Nishioku, T.; Kawamoto, M.; Okizono, R.; Sakai, E.; Okamoto, K.; Tsukuba, T. Dimethyl fumarate prevents osteoclastogenesis by decreasing NFATc1 expression, inhibiting of erk and p38 MAPK phosphorylation, and suppressing of HMGB1 release. Biochem. Biophys. Res. Commun. 2020, 530, 455–461. [Google Scholar] [CrossRef]
- Yamaguchi, Y.; Kanzaki, H.; Katsumata, Y.; Itohiya, K.; Fukaya, S.; Miyamoto, Y.; Narimiya, T.; Wada, S.; Nakamura, Y. Dimethyl fumarate inhibits osteoclasts via attenuation of reactive oxygen species signalling by augmented antioxidation. J. Cell. Mol. Med. 2018, 22, 1138–1147. [Google Scholar] [CrossRef]
- Lal, R.; Dhaliwal, J.; Dhaliwal, N.; Dharavath, R.N.; Chopra, K. Activation of the Nrf2/HO-1 signaling pathway by dimethyl fumarate ameliorates complete Freund?s adjuvant-induced arthritis in rats. Eur. J. Pharmacol. 2021, 899, 174044. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Zhu, G.; Sun, Y.; Wu, Y.; Wu, B.; Zheng, W.; Ma, X.; Zheng, Y. 7-deacetyl-gedunin suppresses proliferation of Human rheumatoid arthritis synovial fibroblast through activation of Nrf2/ARE signaling. Int. Immunopharmacol. 2022, 107, 108557. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Ma, J.; Lacagnina, M.J.; Lorca, S.; Odem, M.A.; Walters, E.T.; Kavelaars, A.; Grace, P.M. Oral Dimethyl Fumarate Reduces Peripheral Neuropathic Pain in Rodents via NFE2L2 Antioxidant Signaling. Anesthesiology 2020, 132, 343–356. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.; Zhai, Y.; Heng, X.; Che, F.Y.; Chen, W.; Sun, D.; Zhai, G. Oral bioavailability of curcumin: Problems and advancements. J. Drug Target. 2016, 24, 694–702. [Google Scholar] [CrossRef]
- Gong, X.; Jiang, L.; Li, W.; Liang, Q.; Li, Z. Curcumin induces apoptosis and autophagy inhuman renal cell carcinoma cells via Akt/mTOR suppression. Bioengineered 2021, 12, 5017–5027. [Google Scholar] [CrossRef] [PubMed]
- Deng, H.; Wan, M.; Li, H.; Chen, Q.; Li, R.; Liang, B.; Zhu, H. Curcumin protection against ultraviolet-induced photo-damage in Hacat cells by regulating nuclear factor erythroid 2-related factor 2. Bioengineered 2021, 12, 9993–10006. [Google Scholar] [CrossRef]
- Xu, Z.; Shang, W.; Zhao, Z.; Zhang, B.; Liu, C.; Cai, H. Curcumin alleviates rheumatoid arthritis progression through the phosphatidylinositol 3-kinase/protein kinase B pathway: An in vitro and in vivo study. Bioengineered 2022, 13, 12899–12911. [Google Scholar] [CrossRef] [PubMed]
- Pourhabibi-Zarandi, F.; Rafraf, M.; Zayeni, H.; Asghari-Jafarabadi, M.; Ebrahimi, A.-A. Effects of curcumin supplementation on metabolic parameters, inflammatory factors and obesity values in women with rheumatoid arthritis: A randomized, double-blind, placebo-controlled clinical trial. Phytother. Res. 2022, 36, 1797–1806. [Google Scholar] [CrossRef] [PubMed]
- Bagherniya, M.; Darand, M.; Askari, G.; Guest, P.C.; Sathyapalan, T.; Sahebkar, A. The Clinical Use of Curcumin for the Treatment of Rheumatoid Arthritis: A Systematic Review of Clinical Trials. Adv. Exp. Med. Biol. 2021, 1291, 251–263. [Google Scholar] [CrossRef] [PubMed]
- Saha, S.; Buttari, B.; Profumo, E.; Tucci, P.; Saso, L. A Perspective on Nrf2 Signaling Pathway for Neuroinflammation: A Potential Therapeutic Target in Alzheimer’s and Parkinson’s Diseases. Front. Cell. Neurosci. 2022, 15, 787258. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.X.; Tian, Y.; Wang, Z.T.; Ma, Y.H.; Tan, L.; Yu, J.-T. The Epidemiology of Alzheimer’s Disease Modifiable Risk Factors and Prevention. J. Prev. Alzheimers Dis. 2021, 8, 313–321. [Google Scholar] [CrossRef]
- Chacko, A.; Delbaz, A.; Walkden, H.; Basu, S.; Armitage, C.W.; Eindorf, T.; Trim, L.K.; Miller, E.; West, N.P.; St John, J.A.; et al. Chlamydia pneumoniae can infect the central nervous system via the olfactory and trigeminal nerves and contributes to Alzheimer’s disease risk. Sci. Rep. 2022, 12, 2759. [Google Scholar] [CrossRef] [PubMed]
- Nazareth, L.; Chen, M.; Shelper, T.; Shah, M.; Velasquez, J.T.; Walkden, H.; Beacham, I.; Batzloff, M.; Rayfield, A.; Todorovic, M.; et al. Novel insights into the glia limitans of the olfactory nervous system. J. Comp. Neurol. 2019, 527, 1228–1244. [Google Scholar] [CrossRef]
- Jung, Y.-J.; Chung, W.-S. Phagocytic Roles of Glial Cells in Healthy and Diseased Brains. Biomol. Ther. 2018, 26, 350–357. [Google Scholar] [CrossRef] [PubMed]
- Al-Atrache, Z.; Lopez, D.B.; Hingley, S.T.; Appelt, D.M. Astrocytes infected with Chlamydia pneumoniae demonstrate altered expression and activity of secretases involved in the generation of β-amyloid found in Alzheimer disease. BMC Neurosci. 2019, 20, 6. [Google Scholar] [CrossRef] [PubMed]
- Rojo, A.I.; Pajares, M.; Rada, P.; Nunez, A.; Nevado-Holgado, A.J.; Killik, R.; Van Leuven, F.; Ribe, E.; Lovestone, S.; Yamamoto, M.; et al. NRF2 deficiency replicates transcriptomic changes in Alzheimer’s patients and worsens APP and TAU pathology. Redox Biol. 2017, 13, 444–451. [Google Scholar] [CrossRef] [PubMed]
- Bahn, G.; Park, J.S.; Yun, U.J.; Lee, Y.J.; Choi, Y.; Park, J.S.; Baek, S.H.; Choi, B.Y.; Cho, Y.S.; Kim, H.K.; et al. NRF2/ARE pathway negatively regulates BACE1 expression and ameliorates cognitive deficits in mouse Alzheimer’s models. Proc. Natl. Acad. Sci. USA 2019, 116, 12516–12523. [Google Scholar] [CrossRef]
- Jo, C.; Gundemir, S.; Pritchard, S.; Jin, Y.N.; Rahman, I.; Johnson, G.V. Nrf2 reduces levels of phosphorylated tau protein by inducing autophagy adaptor protein NDP52. Nat. Commun. 2014, 5, 3496. [Google Scholar] [CrossRef] [PubMed]
- Xi, X.; Feng, S. Neuroprotective Effect of EGCG on Alzheimer Model Rats and Expression of PPARgamma mRNA. Nat. Prod. Res. Dev. 2016, 28, 596–600. [Google Scholar]
- Cano, A.; Ettcheto, M.; Chang, J.-H.; Barroso, E.; Espina, M.; Kühne, B.A.; Barenys, M.; Auladell, C.; Folch, J.; Souto, E.B.; et al. Dual-drug loaded nanoparticles of Epigallocatechin-3-gallate (EGCG)/Ascorbic acid enhance therapeutic efficacy of EGCG in a APPswe/PS1dE9 Alzheimer’s disease mice model. J. Control. Release 2019, 301, 62–75. [Google Scholar] [CrossRef]
- El Khoury, A.; Seidler, P.M.; Eisenberg, D.S.; Harran, P.G. Catalytic Synthesis of PEGylated EGCG Conjugates that Disaggregate Alzheimer’s Tau. Synthesis 2021, 53, 4263–4271. [Google Scholar] [CrossRef]
- Guo, Y.; Zhao, Y.; Nan, Y.; Wang, X.; Chen, Y.; Wang, S. (-)-Epigallocatechin-3-gallate ameliorates memory impairment and rescues the abnormal synaptic protein levels in the frontal cortex and hippocampus in a mouse model of Alzheimer’s disease. Neuroreport 2017, 28, 590–597. [Google Scholar] [CrossRef]
- Lopez del Amo, J.M.; Fink, U.; Dasari, M.; Grelle, G.; Wanker, E.E.; Bieschke, J.; Reif, B. Structural Properties of EGCG-Induced, Nontoxic Alzheimer’s Disease Aβ Oligomers. J. Mol. Biol. 2012, 421, 517–524. [Google Scholar] [CrossRef] [PubMed]
- Hyung, S.J.; DeToma, A.S.; Brender, J.R.; Lee, S.; Vivekanandan, S.; Kochi, A.; Choi, J.S.; Ramamoorthy, A.; Ruotolo, B.T.; Lim, M.H. Insights into antiamyloidogenic properties of the green tea extract (-)-epigallocatechin-3-gallate toward metal-associated amyloid-β species. Proc. Natl. Acad. Sci. USA 2013, 110, 3743–3748. [Google Scholar] [CrossRef]
- Kim, H.J.; Jang, B.K.; Park, J.-H.; Choi, J.W.; Park, S.J.; Byeon, S.R.; Pae, A.N.; Lee, Y.S.; Cheong, E.; Park, K.D. A novel chalcone derivative as Nrf2 activator attenuates learning and memory impairment in a scopolamine-induced mouse model. Eur. J. Med. Chem. 2020, 185, 111777. [Google Scholar] [CrossRef]
- Zhang, R.; Zhang, J.; Fang, L.; Li, X.; Zhao, Y.; Shi, W.; An, L. Neuroprotective Effects of Sulforaphane on Cholinergic Neurons in Mice with Alzheimer’s Disease-Like Lesions. Int. J. Mol. Sci. 2014, 15, 14396–14410. [Google Scholar] [CrossRef]
- Zhang, J.; Zhang, R.; Zhan, Z.; Li, X.; Zhou, F.; Xing, A.; Jiang, C.; Chen, Y.; An, L. Beneficial Effects of Sulforaphane Treatment in Alzheimer’s Disease May Be Mediated through Reduced HDAC1/3 and Increased P75NTR Expression. Front. Aging Neurosci. 2017, 9, 121. [Google Scholar] [CrossRef]
- Lee, S.; Choi, B.-R.; Kim, J.; LaFerla, F.M.; Park, J.H.Y.; Han, J.-S.; Lee, K.W.; Kim, J. Sulforaphane Upregulates the Heat Shock Protein Co-Chaperone CHIP and Clears Amyloid-beta and Tau in a Mouse Model of Alzheimer’s Disease. Mol. Nutr. Food Res. 2018, 62, e1800240. [Google Scholar] [CrossRef]
- Zhao, H.; Wang, Q.; Cheng, X.; Li, X.; Li, N.; Liu, T.; Li, J.; Yang, Q.; Dong, R.; Zhang, Y.; et al. Inhibitive Effect of Resveratrol on the Inflammation in Cultured Astrocytes and Microglia Induced by Aβ1–42. Neuroscience 2018, 379, 390–404. [Google Scholar] [CrossRef] [PubMed]
- Gu, J.; Li, Z.; Chen, H.; Xu, X.; Li, Y.; Gui, Y. Neuroprotective Effect of Trans-Resveratrol in Mild to Moderate Alzheimer Disease: A Randomized, Double-Blind Trial. Neurol. Ther. 2021, 10, 905–917. [Google Scholar] [CrossRef] [PubMed]
- Alamro, A.A.; Alsulami, E.A.; Almutlaq, M.; Alghamedi, A.; Alokail, M.; Haq, S.H. Therapeutic Potential of Vitamin D and Curcumin in an In Vitro Model of Alzheimer Disease. J. Cent. Nerv. Syst. Dis. 2020, 12, 1179573520924311. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.J.; Li, Z.H.; Liu, L.; Tang, W.X.; Wang, Y.; Dong, M.R.; Xiao, C. Curcumin Attenuates Beta-Amyloid-Induced Neuroinflammation via Activation of Peroxisome Proliferator-Activated Receptor-Gamma Function in a Rat Model of Alzheimer’s Disease. Front. Pharmacol. 2016, 7, 261. [Google Scholar] [CrossRef]
- Alizade, A.; Ozbolat, G. Antioxidant activities of inula viscosa extract and curcumin on U87 cells induced by beta-amyloid. Cukurova Med. J. 2021, 46, 583–588. [Google Scholar] [CrossRef]
- Todorovic, M.; Wood, S.A.; Mellick, G.D. Nrf2: A modulator of Parkinson’s disease? J. Neural Transm. 2016, 123, 611–619. [Google Scholar] [CrossRef]
- Kasen, A.; Houck, C.; Burmeister, A.R.; Sha, Q.; Brundin, L.; Brundin, P. Upregulation of alpha-synuclein following immune activation: Possible trigger of Parkinson’s disease. Neurobiol. Dis. 2022, 166, 105654. [Google Scholar] [CrossRef]
- Latif, S.; Jahangeer, M.; Maknoon Razia, D.; Ashiq, M.; Ghaffar, A.; Akram, M.; El Allam, A.; Bouyahya, A.; Garipova, L.; Ali Shariati, M.; et al. Dopamine in Parkinson’s disease. Clin. Chim. Acta 2021, 522, 114–126. [Google Scholar] [CrossRef]
- Yi, G.; Din, J.U.; Zhao, F.; Liu, X. Effect of soybean peptides against hydrogen peroxide induced oxidative stress in HepG2 cells via Nrf2 signaling. Food Funct. 2020, 11, 2725–2737. [Google Scholar] [CrossRef]
- Paraidathathu, T.; Degroot, H.; Kehrer, J.P. PRODUCTION OF REACTIVE OXYGEN BY MITOCHONDRIA FROM NORMOXIC AND HYPOXIC RAT-HEART TISSUE. Free Radic. Biol. Med. 1992, 13, 289–297. [Google Scholar] [CrossRef]
- Wang, C.Y.; Zhang, Q.; Xun, Z.; Yuan, L.; Li, R.; Li, X.; Tian, S.Y.; Xin, N.; Xu, Y. Increases of iASPP-Keap1 interaction mediated by syringin enhance synaptic plasticity and rescue cognitive impairments via stabilizing Nrf2 in Alzheimer’s models. Redox Biol. 2020, 36, 101672. [Google Scholar] [CrossRef] [PubMed]
- Nesi, G.; Sestito, S.; Digiacomo, M.; Rapposelli, S. Oxidative Stress, Mitochondrial Abnormalities and Proteins Deposition: Multitarget Approaches in Alzheimer’s Disease. Curr. Top. Med. Chem. 2017, 17, 3062–3079. [Google Scholar] [CrossRef] [PubMed]
- Guo, C.J.; Zhang, Y.; Nie, Q.; Cao, D.D.; Wang, X.X.; Wan, X.K.; Liu, M.; Cui, J.; Sun, J.; Bai, Y.F.; et al. SQSTM1/ p62 oligomerization contributes to AO-induced inhibition of Nrf2 signaling. Neurobiol. Aging 2021, 98, 10–20. [Google Scholar] [CrossRef]
- Shah, S.Z.A.; Zhao, D.; Hussain, T.; Sabir, N.; Mangi, M.H.; Yang, L. p62-Keap1-NRF2-ARE Pathway: A Contentious Player for Selective Targeting of Autophagy, Oxidative Stress and Mitochondrial Dysfunction in Prion Diseases. Front. Mol. Neurosci. 2018, 11, 310. [Google Scholar] [CrossRef] [PubMed]
- Petrillo, S.; Schirinzi, T.; Di Lazzaro, G.; D’Amico, J.; Colona, V.L.; Bertini, E.; Pierantozzi, M.; Mari, L.; Mercuri, N.B.; Piemonte, F.; et al. Systemic activation of Nrf2 pathway in Parkinson’s disease. Mov. Disord. 2020, 35, 180–184. [Google Scholar] [CrossRef] [PubMed]
- Sivandzade, F.; Prasad, S.; Bhalerao, A.; Cucullo, L. NRF2 and NF-B interplay in cerebrovascular and neurodegenerative disorders: Molecular mechanisms and possible therapeutic approaches. Redox Biol. 2019, 21, 101059. [Google Scholar] [CrossRef]
- Buhlman, L.M. Parkin loss-of-function pathology: Premature neuronal senescence induced by high levels of reactive oxygen species? Mech. Ageing Dev. 2017, 161, 112–120. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.Y.; Xu, Y.; Wang, X.; Guo, C.; Wang, T.; Wang, Z.Y. Dl-3-n-Butylphthalide Inhibits NLRP3 Inflammasome and Mitigates Alzheimer’s-Like Pathology via Nrf2-TXNIP-TrX Axis. Antioxid. Redox Signal. 2019, 30, 1411–1431. [Google Scholar] [CrossRef]
- Schepici, G.; Bramanti, P.; Mazzon, E. Efficacy of Sulforaphane in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 8637. [Google Scholar] [CrossRef]
- DeBerardinis, A.M.; Banerjee, U.; Hadden, M.K. Identification of Vitamin D3-Based Hedgehog Pathway Inhibitors That Incorporate an Aromatic A-Ring Isostere. ACS Med. Chem. Lett. 2013, 4, 25–30. [Google Scholar] [CrossRef]
- Salman-Monte, T.C.; Torrente-Segarra, V.; Vega-Vidal, A.L.; Corzo, P.; Castro-Dominguez, F.; Ojeda, F.; Carbonell-Abello, J. Bone mineral density and vitamin D status in systemic lupus erythematosus (SLE): A systematic review. Autoimmun. Rev. 2017, 16, 1155–1159. [Google Scholar] [CrossRef] [PubMed]
- Tzilas, V.; Bouros, E.; Barbayianni, I.; Karampitsakos, T.; Kourtidou, S.; Ntassiou, M.; Ninou, I.; Aidinis, V.; Bouros, D.; Tzouvelekis, A. Vitamin D prevents experimental lung fibrosis and predicts survival in patients with idiopathic pulmonary fibrosis. Pulm. Pharmacol. Ther. 2019, 55, 17–24. [Google Scholar] [CrossRef] [PubMed]
- Hu, D.; Chen, W.; Wang, Y. Synthesis and Structure-Activity Relationship of Active Vitamin D3 Analogues. Prog. Chem. 2016, 28, 839–859. [Google Scholar]
- Choi, J.W.; Kim, S.; Yoo, J.S.; Kim, H.J.; Kim, H.J.; Kim, B.E.; Lee, E.H.; Lee, Y.S.; Park, J.H.; Park, K.D. Development and optimization of halogenated vinyl sulfones as Nrf2 activators for the treatment of Parkinson’s disease. Eur. J. Med. Chem. 2021, 212, 113103. [Google Scholar] [CrossRef]
- Pu, Y.Y.; Chang, L.J.; Qu, Y.G.; Wang, S.M.; Zhang, K.; Hashimoto, K. Antibiotic-induced microbiome depletion protects against MPTP-induced dopaminergic neurotoxicity in the brain. Aging 2019, 11, 6915–6929. [Google Scholar] [CrossRef]
- Liu, B.; Gao, Y.Q.; Zheng, Y.M.; Wang, X.M.; Wang, J.F. Protective Effect of Egcg on Rotenone-Induced Nerve Cell Damage and Oxidative Damage. Acta Med. Mediterr. 2022, 38, 215–219. [Google Scholar] [CrossRef]
- Doronicheva, N.; Yasui, H.; Sakurai, H. Chemical structure-dependent differential effects of flavonoids on the catalase activity as evaluated by a chemiluminescent method. Biol. Pharm. Bull. 2007, 30, 213–217. [Google Scholar] [CrossRef]
- Zhang, Z.; Hamada, H.; Gerk, P.M. Selectivity of Dietary Phenolics for Inhibition of Human Monoamine Oxidases A and B. BioMed Res. Int. 2019, 2019, 8361858. [Google Scholar] [CrossRef]
- Hamijoyo, L.; Candrianita, S.; Rahmadi, A.R.; Dewi, S.; Darmawan, G.; Suryajaya, B.S.; Rainy, N.R.; Hidayat, I.I.; Moenardi, V.N.; Wachjudi, R.G. The clinical characteristics of systemic lupus erythematosus patients in Indonesia: A cohort registry from an Indonesia-based tertiary referral hospital. Lupus 2019, 28, 1604–1609. [Google Scholar] [CrossRef]
- Zhao, M.; Chen, H.; Ding, Q.; Xu, X.; Yu, B.; Huang, Z. Nuclear Factor Erythroid 2-related Factor 2 Deficiency Exacerbates Lupus Nephritis in B6/lpr mice by Regulating Th17 Cell Function. Sci. Rep. 2016, 6, 38619. [Google Scholar] [CrossRef]
- Ye, Q.; Wang, G.; Huang, Y.; Lu, J.; Zhang, J.; Zhu, L.; Zhu, Y.; Li, X.; Lan, J.; Li, Z.; et al. Mycophenolic Acid Exposure Optimization Based on Vitamin D Status in Children with Systemic Lupus Erythematosus: A Single-Center Retrospective Study. Rheumatol. Ther. 2021, 8, 1143–1157. [Google Scholar] [CrossRef] [PubMed]
- Kasai, S.; Shimizu, S.; Tatara, Y.; Mimura, J.; Itoh, K. Regulation of Nrf2 by Mitochondrial Reactive Oxygen Species in Physiology and Pathology. Biomolecules 2020, 10, 320. [Google Scholar] [CrossRef] [PubMed]
- Ahmad, R.; Ahsan, H. Singlet oxygen species and systemic lupus erythematosus: A brief review. J. Immunoass. Immunochem. 2019, 40, 343–349. [Google Scholar] [CrossRef] [PubMed]
- Bai, Y.; Liu, Q.; Li, J.; Zhou, J. Protective mechanism of curcumin mediated HO-1 regulating Nrf2 signaling pathway on cadmium induced renal injury. J. Toxicol. 2020, 34, 291–295. [Google Scholar]
- Singh, U.; Barik, A.; Singh, B.G.; Priyadarsini, K.I. Reactions of reactive oxygen species (ROS) with curcumin analogues: Structure-activity relationship. Free Radic. Res. 2011, 45, 317–325. [Google Scholar] [CrossRef]
- Hou, L.F.; He, S.J.; Wang, J.X.; Yang, Y.; Zhu, F.H.; Zhou, Y.; He, P.L.; Zhang, Y.; Yang, Y.F.; Li, Y.; et al. SM934, a water-soluble derivative of arteminisin, exerts immunosuppressive functions in vitro and in vivo. Int. Immunopharmacol. 2009, 9, 1509–1517. [Google Scholar] [CrossRef]
- Li, Y.; Zhao, L.; Yang, X.; Chen, J.; Xu, W.; Gu, Q. Artemisinin derivative SM934, influences the activation, proliferation, differentiation and antibody-secreting capacity of β-cells in systemic lupus erythematosus mice via inhibition of TLR7/9 signaling pathway. Trop. J. Pharm. Res. 2021, 18, 1391–1396. [Google Scholar] [CrossRef]
- Deng, J.; Pan, J.; Li, H. Research progress of B cell-related targets in systemic lupus erythematosus. Chin. J. Cell. Mol. Immunol. 2016, 32, 1148–1150. [Google Scholar] [CrossRef]
- Lin, Z.M.; Liu, Y.T.; Chen, L.; Cao, S.Q.; Huang, Y.T.; Yang, X.Q.; Zhu, F.H.; Tang, W.; He, S.J.; Zuo, J.P. Artemisinin analogue SM934 protects against lupus-associated antiphospholipid syndrome via activation of Nrf2 and its targets. Sci. China-Life Sci. 2021, 64, 1702–1719. [Google Scholar] [CrossRef]
- Zheng, M.; Liu, G.; Tang, W.; Zuo, J.; Zhang, A.; Jiang, H. Structure-activity relationships of the antimalarial agent artemisinin and the research progress on the artemisinin analogues with novel pharmacological actions. Chin. Sci. Bull. 2017, 62, 1948–1963. [Google Scholar] [CrossRef]
- Yu, K.; Geng, X.; Chen, M.; Zhang, J.; Wang, B.; Ilic, K.; Tong, W. High Daily Dose and Being a Substrate of Cytochrome P450 Enzymes Are Two Important Predictors of Drug-Induced Liver Injury. Drug Metab. Dispos. 2014, 42, 744–750. [Google Scholar] [CrossRef]
- Lieber, C.S.; Rubin, E.; DeCarli, L.M. Hepatic microsomal ethanol oxidizing system (MEOS): Differentiation from alcohol dehydrogenase and NADPH oxidase. Biochem. Biophys. Res. Commun. 1970, 40, 858–865. [Google Scholar] [CrossRef]
- Bailey, S.M.; Cunningham, C.C. Contribution of mitochondria to oxidative stress associated with alcoholic liver disease. Free Radic. Biol. Med. 2002, 32, 11–16. [Google Scholar] [CrossRef]
- Ishida, K.; Kaji, K.; Sato, S.; Ogawa, H.; Takagi, H.; Takaya, H.; Kawaratani, H.; Moriya, K.; Namisaki, T.; Akahane, T.; et al. Sulforaphane ameliorates ethanol plus carbon tetrachloride-induced liver fibrosis in mice through the Nrf2-mediated antioxidant response and acetaldehyde metabolization with inhibition of the LPS/TLR4 signaling pathway. J. Nutr. Biochem. 2021, 89, 108573. [Google Scholar] [CrossRef] [PubMed]
- Abd El-Hameed, N.M.; Abd El-Aleem, S.A.; Khattab, M.A.; Ali, A.H.; Mohammed, H.H. Curcumin activation of nuclear factor E2-related factor 2 gene (Nrf2): Prophylactic and therapeutic effect in nonalcoholic steatohepatitis (NASH). Life Sci. 2021, 285, 119983. [Google Scholar] [CrossRef] [PubMed]
- Sunny, N.E.; Bril, F.; Cusi, K. NAFLD Mitochondrial Adaptation in Nonalcoholic Fatty Liver Disease: Novel Mechanisms and Treatment Strategies. Trends Endocrinol. Metab. 2017, 28, 250–260. [Google Scholar] [CrossRef] [PubMed]
- Begriche, K.; Igoudjil, A.; Pessayre, D.; Fromenty, B. Mitochondrial dysfunction in NASH: Causes, consequences and possible means to prevent it. Mitochondrion 2006, 6, 1–28. [Google Scholar] [CrossRef] [PubMed]
- Han, X.; Ding, C.H.; Zhang, G.D.; Pan, R.Y.; Liu, Y.P.; Huang, N.; Hou, N.N.; Han, F.; Xu, W.J.; Sun, X.D. Liraglutide ameliorates obesity-related nonalcoholic fatty liver disease by regulating Sestrin2-mediated Nrf2/HO-1 pathway. Biochem. Biophys. Res. Commun. 2020, 525, 895–901. [Google Scholar] [CrossRef] [PubMed]
- Hosseini, H.; Teimouri, M.; Shabani, M.; Koushki, M.; Khorzoughi, R.B.; Namvarjah, F.; Izadi, P.; Meshkani, R. Resveratrol alleviates non-alcoholic fatty liver disease through epigenetic modification of the Nrf2 signaling pathway. Int. J. Biochem. Cell Biol. 2020, 119, 105667. [Google Scholar] [CrossRef] [PubMed]
- Yan, C.; Zhang, Y.; Zhang, X.; Aa, J.; Wang, G.; Xie, Y. Curcumin regulates endogenous and exogenous metabolism via Nrf2-FXR-LXR pathway in NAFLD mice. Biomed. Pharmacother. 2018, 105, 274–281. [Google Scholar] [CrossRef] [PubMed]
- Shen, B.; Zhao, C.; Wang, Y.; Peng, Y.; Cheng, J.; Li, Z.; Wu, L.; Jin, M.; Feng, H. Aucubin inhibited lipid accumulation and oxidative stress via Nrf2/HO-1 and AMPK signalling pathways. J. Cell. Mol. Med. 2019, 23, 4063–4075. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Chen, J.; Gao, Q.; Shan, X.; Wang, J.; Lv, Z. Study on the attenuated effect of Ginkgolide B on ferroptosis in high fat diet induced nonalcoholic fatty liver disease. Toxicology 2020, 445, 152599. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Yang, M.; Lin, H.; Yan, W.; Deng, G.; Ye, H.; Shi, H.; Wu, C.; Ma, G.; Xu, S.; et al. Limonin Alleviates Non-alcoholic Fatty Liver Disease by Reducing Lipid Accumulation, Suppressing Inflammation and Oxidative Stress. Front. Pharmacol. 2022, 12, 801730. [Google Scholar] [CrossRef] [PubMed]
- Otani, K.; Korenaga, M.; Beard, M.R.; Li, K.; Qian, T.; Showalter, L.A.; Singh, A.K.; Wang, T.; Weinman, S.A. Hepatitis C virus core protein, cytochrome P450 2E1, and alcohol produce combined mitochondrial injury and cytotoxicity in hepatoma cells. Gastroenterology 2005, 128, 96–107. [Google Scholar] [CrossRef]
- Smirnova, O.A.; Ivanova, O.N.; Bartosch, B.; Valuev-Elliston, V.T.; Mukhtarov, F.; Kochetkov, S.N.; Ivanov, A.V. Hepatitis C Virus NS5A Protein Triggers Oxidative Stress by Inducing NADPH Oxidases 1 and 4 and Cytochrome P450 2E1. Oxidative Med. Cell. Longev. 2016, 201, 83419376. [Google Scholar] [CrossRef]
- Ivanov, A.V.; Smirnova, O.A.; Petrushanko, I.Y.; Ivanova, O.N.; Karpenko, I.L.; Alekseeva, E.; Sominskaya, I.; Makarov, A.A.; Bartosch, B.; Kochetkov, S.N.; et al. HCV Core Protein Uses Multiple Mechanisms to Induce Oxidative Stress in Human Hepatoma Huh7 Cells. Viruses 2015, 7, 2745–2770. [Google Scholar] [CrossRef]
- Carvajal-Yepes, M.; Himmelsbach, K.; Schaedler, S.; Ploen, D.; Krause, J.; Ludwig, L.; Weiss, T.; Klingel, K.; Hildt, E. Hepatitis C Virus Impairs the Induction of Cytoprotective Nrf2 Target Genes by Delocalization of Small Maf Proteins. J. Biol. Chem. 2011, 286, 8941–8951. [Google Scholar] [CrossRef]
- Yu, J.-S.; Chen, W.-C.; Tseng, C.-K.; Lin, C.-K.; Hsu, Y.-C.; Chen, Y.-H.; Lee, J.-C. Sulforaphane Suppresses Hepatitis C Virus Replication by Up-Regulating Heme Oxygenase-1 Expression through PI3K/Nrf2 Pathway. PLoS ONE 2016, 11, e0152236. [Google Scholar] [CrossRef]
- Shen, J.; Wang, G.; Zuo, J. Caffeic acid inhibits HCV replication via induction of IFN alpha antiviral response through p62-mediated Keap1/Nrf2 signaling pathway. Antivir. Res. 2018, 154, 166–173. [Google Scholar] [CrossRef] [PubMed]
- Mansouri, A.; Gattolliat, C.-H.; Asselah, T. Mitochondrial Dysfunction and Signaling in Chronic Liver Diseases. Gastroenterology 2018, 155, 629–647. [Google Scholar] [CrossRef] [PubMed]
- Liu, B.; Fang, M.; He, Z.; Cui, D.; Jia, S.; Lin, X.; Xu, X.; Zhou, T.; Liu, W. Hepatitis B virus stimulates G6PD expression through HBx-mediated Nrf2 activation. Cell Death Dis. 2015, 6, e1980. [Google Scholar] [CrossRef]
- Lee, C.K.; Choi, J.S. Effects of Silibinin, Inhibitor of CYP3A4 and P-Glycoprotein in vitro, on the Pharmacokinetics of Paclitaxel after Oral and Intravenous Administration in Rats. Pharmacology 2010, 85, 350–356. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.Y.; Kim, J.Y.; Jenis, J.; Li, Z.P.; Ban, Y.J.; Baiseitova, A.; Park, K.H. Tyrosinase inhibitory study of flavonolignans from the seeds of Silybum marianum (Milk thistle). Bioorg. Med. Chem. 2019, 27, 2499–2507. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, S.; Ullah, N.; Parveen, S.; Javed, I.; Jalil, N.A.C.; Murtey, M.D.; Sheikh, I.S.; Khan, S.; Ojha, S.C.; Chen, K. Effect of Silymarin as an Adjunct Therapy in Combination with Sofosbuvir and Ribavirin in Hepatitis C Patients: A Miniature Clinical Trial. Oxidative Med. Cell. Longev. 2022, 2022, 9199190. [Google Scholar] [CrossRef] [PubMed]
- Gong, J.X.; Weng, L.J.; Wang, F.; Bin Feng, Y.; Zhou, C.X.; Li, H.B.; Wu, Y.H.; Hao, X.J.; Wu, X.M.; Bai, H.; et al. Synthesis and antioxidant properties of novel silybin analogues. Chin. Chem. Lett. 2006, 17, 465–468. [Google Scholar]
- Kang, H.; Zheng, L.; Li, Z. Preparation and quality inspection of sodium 11-phosphate derivative of silymarin. Chin. J. Pharm. 2004, 35, 71–72. [Google Scholar]
- Wasik, U.; Milkiewicz, M.; Kempinska-Podhorodecka, A.; Milkiewicz, P. Protection against oxidative stress mediated by the Nrf2/Keap1 axis is impaired in Primary Biliary Cholangitis. Sci. Rep. 2017, 7, srep44769. [Google Scholar] [CrossRef]
- Czaja, A.J. Promising Pharmacological, Molecular and Cellular Treatments of Autoimmune Hepatitis. Curr. Pharm. Des. 2011, 17, 3120–3140. [Google Scholar] [CrossRef] [PubMed]
- Tam, J.; Icho, S.; Utama, E.; Orrell, K.E.; Gomez-Biagi, R.F.; Theriot, C.M.; Kroh, H.K.; Rutherford, S.A.; Lacy, D.B.; Melnyk, R.A. Intestinal bile acids directly modulate the structure and function of C. difficile TcdB toxin. Proc. Natl. Acad. Sci. USA 2020, 117, 6792–6800. [Google Scholar] [CrossRef]
- Kawata, K.; Kobayashi, Y.; Souda, K.; Kawamura, K.; Sumiyoshi, S.; Takahashi, Y.; Noritake, H.; Watanabe, S.; Suehiro, T.; Nakamura, H. Enhanced Hepatic Nrf2 Activation After Ursodeoxycholic Acid Treatment in Patients with Primary Biliary Cirrhosis. Antioxid. Redox Signal. 2010, 13, 259–268. [Google Scholar] [CrossRef] [PubMed]
- Brossard, D.; Lechevrel, M.; El Kihel, L.; Quesnelle, C.; Khalid, M.; Moslemi, S.; Reimund, J.-M. Synthesis and biological evaluation of bile carboxamide derivatives with pro-apoptotic effect on human colon adenocarcinoma cell lines. Eur. J. Med. Chem. 2014, 86, 279–290. [Google Scholar] [CrossRef]
- Silva, S.L.; Vaz, A.R.; Diogenes, M.J.; van Rooijen, N.; Sebastiao, A.M.; Fernandes, A.; Silva, R.F.M.; Brites, D. Neuritic growth impairment and cell death by unconjugated bilirubin is mediated by NO and glutamate, modulated by microglia, and prevented by glycoursodeoxycholic acid and interleukin-10. Neuropharmacology 2012, 62, 2398–2408. [Google Scholar] [CrossRef] [PubMed]
- Fiorucci, S.; Mencarelli, A.; Palazzetti, B.; Del Soldato, P.; Morelli, A.; Ignarro, L.J. An NO derivative of ursodeoxycholic acid protects against Fas-mediated liver injury by inhibiting caspase activity. Proc. Natl. Acad. Sci. USA 2001, 98, 2652–2657. [Google Scholar] [CrossRef]
- Ruart, M.; Chavarria, L.; Camprecios, G.; Suarez-Herrera, N.; Montironi, C.; Guixe-Muntet, S.; Bosch, J.; Friedman, S.L.; Carlos Garcia-Pagan, J.; Hernandez-Gea, V. Impaired endothelial autophagy promotes liver fibrosis by aggravating the oxidative stress response during acute liver injury. J. Hepatol. 2019, 70, 458–469. [Google Scholar] [CrossRef] [PubMed]
- Xiao, X.; Wang, C.H.; Chang, D.; Wang, Y.; Dong, X.J.; Jiao, T.; Zhao, Z.D.; Ren, L.L.; Dela Cruz, C.S.; Sharma, L.; et al. Identification of Potent and Safe Antiviral Therapeutic Candidates Against SARS-CoV-2. Front. Immunol. 2020, 11, 586572. [Google Scholar] [CrossRef] [PubMed]
- Kamel, E.O.; Gad-Elrab, W.M.; Ahmed, M.A.; Mohammedsaleh, Z.M.; Hassanein, E.H.M.; Ali, F.E.M. Candesartan Protects Against Cadmium-Induced Hepatorenal Syndrome by Affecting Nrf2, NF-kappa B, Bax/Bcl-2/Cyt-C, and Ang II/Ang 1-7 Signals. Biol. Trace Elem. Res. 2022, 1–18. [Google Scholar] [CrossRef]
- Sharawy, M.H.; El-Kashef, D.H.; Shaaban, A.A.; El-Agamy, D.S. Anti-fibrotic activity of sitagliptin against concanavalin A-induced hepatic fibrosis. Role of Nrf2 activation/NF-kappa B inhibition. Int. Immunopharmacol. 2021, 100, 108088. [Google Scholar] [CrossRef]
- Abo-Haded, H.M.; Elkablawy, M.A.; Al-Johani, Z.; Al-ahmadi, O.; El-Agamy, D.S. Hepatoprotective effect of sitagliptin against methotrexate induced liver toxicity. PLoS ONE 2017, 12, e0174295. [Google Scholar] [CrossRef]
- Song, J.-X.; An, J.-R.; Chen, Q.; Yang, X.-Y.; Jia, C.-L.; Xu, S.; Zhao, Y.-S.; Ji, E.-S. Liraglutide attenuates hepatic iron levels and ferroptosis in db/db mice. Bioengineered 2022, 13, 8334–8348. [Google Scholar] [CrossRef]
- Ge, C.; Tan, J.; Lou, D.; Zhu, L.; Zhong, Z.; Dai, X.; Sun, Y.; Kuang, Q.; Zhao, J.; Wang, L.; et al. Mulberrin confers protection against hepatic fibrosis by Trim31/Nrf2 signaling. Redox Biol. 2022, 51, 102274. [Google Scholar] [CrossRef] [PubMed]
- Wu, G.; Zhu, L.; Yuan, X.; Chen, H.; Xiong, R.; Zhang, S.; Cheng, H.; Shen, Y.; An, H.; Li, T.; et al. Britanin Ameliorates Cerebral Ischemia-Reperfusion Injury by Inducing the Nrf2 Protective Pathway. Antioxid. Redox Signal. 2017, 27, 754–768. [Google Scholar] [CrossRef] [PubMed]
- Rzepecka, J.; Pineda, M.A.; Al-Riyami, L.; Rodgers, D.T.; Huggan, J.K.; Lumb, F.E.; Khalaf, A.I.; Meakin, P.J.; Corbet, M.; Ashford, M.L.; et al. Prophylactic and therapeutic treatment with a synthetic analogue of a parasitic worm product prevents experimental arthritis and inhibits IL-1β production via NRF2-mediated counter-regulation of the inflammasome. J. Autoimmun. 2015, 60, 59–73. [Google Scholar] [CrossRef] [PubMed]
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
EGCG (1) | Previfenon® as Chemoprophylaxis of COVID-19 in Health Workers | 2/3 | Unknown | Event of clinical acute respiratory disease with a diagnosis of COVID-19 confirmed with rtPCR | 250 mg/8 h orally for 40–70 days | Sample size: 524; Gender: all; Ages: 25 years and older. | NCT04446065 |
Sulforaphane (2) | CO-Sprout: Broccoli sprout powder for COVID-19 positive pregnant women on the rate of hospital admission | 2 | Australia | Duration of COVID-19 associated symptoms (days) as self-reported by trial participants. | 21 mg, orally twice a day, morning and night (BD) | Sample size: 60; Gender: females; Ages: 18 years and older. | ACTRN12622000173796 |
SFX-01 treatment for Acute Respiratory Infections (STAR-Covid19) | 2 | United Kingdom | 1. Not hospitalized, no limitations on activities; 2. Not hospitalized, limitation on activities; 3. Hospitalized, not requiring supplemental oxygen; 4. Hospitalized, requiring supplemental oxygen; 5. Hospitalized, on non-invasive ventilation or high flow oxygen devices; 6. Hospitalized, on invasive mechanical ventilation or ECMO (Extracorporeal membrane oxygenation); 7. Death; timepoint(s) of evaluation of this end point day 15 (where day 1 is the first day of treatment). | 300 mg, orally | Sample size: 300; Gender: all; Ages: adults (18–64 years): 120, elderly (≥65 years): 180. | EUCTR2020-003486-19-GB | |
Resveratrol (3) | The Anti-fibrotic Therapeutic Effects of Resveratrol for Discharged COVID-19 Patients | N/A | Hong Kong, China | 1. The handheld basic spirometry; 2. PRO scores; 3. Borg Category Ratio 0–10 Scale; | 1.0 g, orally once a day for six months. | Sample size: 30; Gender: all; Ages: 18 years to 65 years | NCT04799743 |
Retrospective Study of ImmunoFormulation for COVID-19 | N/A | Spain | Clinical symptoms duration. Time Frame: 1 month, starting after start of treatment. | transfer factors (oligo- and polypeptides from porcine spleen, ultrafiltered at <10 kDa—Imuno TF®) 100 mg, 800 mg anti-inflammatory natural blend (Uncaria tomentosa, Endopleura uchi and Haematoccocus pluvialis—MiodesinTM), 60 mg zinc orotate, 48 mg selenium yeast (equivalent to 96 μg of Se), 20,000 IU cholecalciferol, 300 mg ascorbic acid, 480 mg ferulic acid, 90 mg resveratrol, 800 mg spirulina, 560 mg N-acetylcysteine, 610 mg glucosamine sulphate potassium chloride, and 400 mg maltodextrin-stabilized orthosilicic acid (equivalent to 6 mg of Si—SiliciuMax®). | Sample size: 40; Gender: all; Ages: 18 years and older | NCT04666753 | |
Evaluation of the combined effect of Hesperidin, Artemisinin-Artemisia annua, Noscapine, N-acetylcysteine, Resveratrol supplements and high dose of vitamin C on treatment, clinical symptoms of non-hospitalization and hospitalization patients with symptomatic COVID-19 | 3 | Iran (Islamic Republic of) | LDH, CBC diff, Na/K/Ca, CRP, ESR1, Weakness and nausea, respiratory quality | 1. Artemisia annua—Artemisinin (manufactured by Longlifenutri)150 mg every 12 h; 2. Dose of 1 g of vitamin C (manufactured by Daroupakhsh) intravenous vitamin (two 500 mg ampoules in 250 cc of sodium chloride serum for 30 min) every 12 h; 3. Dose of 5 cc of noscapine (manufactured by Faran Shimi) every eight h; 4. 500 mg dose of hesperidin (manufactured by Swanson) every 24 h; 5. resveratrol 500 mg (Manufactured by A Squard) every 24 h; 6. NAC 600 mg (Manufactured by Osvah) every 12 h. The duration of treatment is estimated to be ten days. Supplements are taken orally, and vitamin C is given to patients as an injection. | Sample size: 100; Gender: all; Ages: 12 years and older | IRCT20181030041504N1 | |
Can SARS-CoV-2 Viral Load and COVID-19 Disease Severity be Reduced by Resveratrol-assisted Zinc Therapy | 2; Terminated (Difficulty accruing patients) | United States | 1. Reduction in SARS-CoV-2 viral load; 2. Reduction in severity of COVID-19 Disease. | Zinc Picolinate (50 mg po TID × 5 days); resveratrol (2 g po BID × 5 days) | Sample size: 45; Gender: all; Ages: 18 years to 75 years | NCT04542993 | |
Randomized Controlled Trial of Resveretrol-Copper OR Sodium-Copper-Chlorophyllin Versus Standard Treatment In Severe COVID-19 Cancer Patients | 2 | India | The time to clinical improvement, defined as a two-point improvement on a seven-point ordinal scale. | Tablet of resveratrol-Cu containing 5.6 mg of resveratrol and 560 ng of copper, orally once every 6 h. | Sample size: 200; Gender: all; Ages: 18 years to 99 years | CTRI/2020/07/026514 | |
Randomized Controlled Trial of Resveretrol-Copper Or Sodium-Copper-Chlorophyllin Vs Standard Treatment In Mild COVID-19 infection with Cancer Patients | 3 | India | The proportion of patients who suffer clinical deterioration OR viral persistence at Day 10 from the date of randomization (excluding the date of randomization). | Sample size: 300; Gender: all; Ages: 18 years to 99 years | CTRI/2020/07/026515 | ||
Resveratrol and copper for the treatment of COVID-19 pnuemonia | N/A | India | To retrospectively access the clinical outcomes in the patients receiving R-Cu along with standard treatment versus those who received standard treatment. | N/A | Sample size: 230; Gender: all; Ages: 18 years to 99 years | CTRI/2020/06/026256 | |
Resveratrol in COVID-19 | 3 | Iran (Islamic Republic of) | 1. Time to clinical recovery; 2. Respiratory signs; 3. Intubation rate | 500 mg, orally once a day for 14 days. | Sample size: 50; Gender: all; Ages: no age limit. | IRCT20200112046089N1 | |
Randomized Controlled Trial of Resveretrol-Copper Or Sodium-Copper-Chlorophyllin Vs Standard Treatment In Mild COVID-19 infection | 3 | India | 1.Proportion of patients who suffer clinical deterioration OR viral infection; 2. Persistence at day 10 from the date of randomization (excluding the date of randomization); 3. Clinical deterioration will be defined as defined as a two-point or greater deterioration on a seven-point ordinal scale in every patient measured on each day, until day 10 from the date of randomization. | Tablet of resveratrol-Cu containing 5.6 mg of resveratrol and 560 ng of copper, orally once every 6 h. | Sample size: 300; Gender: all; Ages: 18 years to 99 years | CTRI/2020/05/025336 | |
Randomized Controlled Trial of Resveretrol-Copper OR Sodium-Copper-Chlorophyllin Versus Standard Treatment In Severe COVID-19 | 2 | India | The time to clinical improvement, defined as a two-point improvement on a seven-point ordinal scale. | Sample size: 200; Gender: all; Ages: 18 years to 99 years | CTRI/2020/05/025337 | ||
Evaluation efficacy of Curcumin and Resveratrol capsule in controlling symptoms in patients with COVID-19 | 3 | Iran (Islamic Republic of) | Clinical symptoms changes (dry cough, respiratory distress, fever). | “Curcumin and Resveratrol” capsule (each capsule contains 200 mg of curcumin, 200 mg of resveratrol as active ingredients and 100 mg of lactose as filler), 1 capsule every 12 h for 7 days | Sample size: 60; Gender: all; Ages: 18 years and older. | IRCT20080901001165N56 | |
Randomized Proof-of-Concept Trial to Evaluate the Safety and Explore the Effectiveness of Resveratrol, a Plant Polyphenol, for COVID-19 | 2 (Terminated (Feasibility)) | United States | Hospitalization rates for COVID-19: proportion of study participants admitted to the hospital within 21 days of randomization | Resveratrol 1000 mg 4 times/day for 15 days. Vitamin D3 100,000 IU on day 1 | Sample size: 100; Gender: all; Ages: 45 years and older. | NCT04400890 | |
Dimethyl Fumarate (4) | The Efficacy of dimethyl fumarate in the treatment of patients with COVID-19 | 2/3 | Iran (Islamic Republic of) | Death; need for mechanical ventilation; severe illness. | 240 mg capsules (CinnaGen, Tehran, Iran) daily for 5 days | Sample size: 30; Gender: all; Ages: 18 years and older. | IRCT20201024049134N4 |
Randomised Evaluation of COVID-19 Therapy | 2/3 | United Kingdom; Nepal; Sri Lanka; Ghana; Vietnam; Indonesia; India; South Africa | All-cause mortality: For each pairwise comparison with the “no additional treatment” arm, the primary objective is to provide reliable estimates of the effect of study treatments on all-cause mortality. | 120 mg every 12 h for 4 doses followed by 240 mg every 12 h by mouth for 8 days (10 days in total). | Sample size: 50,000; Gender: all; Ages: child, adult and older adult. | NCT04381936 | |
Curcumin (5) | The Effect of Micellized Food Supplements on Health-related Quality of Life in Patients with Post-acute COVID-19 Syndrome. | N/A | Unknown | Change in health-related quality of life. Health-related quality of life is measured with the “Short-Form 12” (SF-12) from 0 to 100. | The daily intake of 2 × 10 drops of a mixture of micellized curcumin (2%), Boswellia serrata (1.5%) and ascorbic acid (6%). | Sample size: 32; Gender: all; Ages: 18 years to 85 years | NCT05150782 |
Nutritional Supplementation of Flavonoids Quercetin and Curcumin for Early Mild Symptoms of COVID-19 | N/A | Pakistan | 1. Testing negative for SARS-CoV-2 using RT-PCR; 2. COVID-19 symptom improvement. | N/A | Sample size: 50; Gender: all; Ages: 18 years and older. | NCT05130671 | |
Determining the Safety and Effectiveness of ENDOR Oral Combination Drug in the Treatment of Patients with COVID-19 | 3 | Iran (Islamic Republic of) | Clinical symptoms, radiological findings, laboratory findings. | two oral capsules of Endor every 8 h for 7 days. This capsule contains beta-carotene 2.5 mg; curcumin 23.75 mg; DHA 30 mg; EPA 45 mg; vitamin C 50 mg; wheat germ oil 75 mg; zinc 10 mg. | Sample size: 200; Gender: all; Ages: 18 years and older. | IRCT20100601004076N26 | |
Nanocurcumin (6C & 30C) on incidence of ILI & COVID-19 type respiratory illness | 3 | India | Incidence of influenza-like illness and COVID-19-type respiratory illness. Timepoint weekly until completion of 1 year. | Children below 5 years: 2 pills of medicine Nanocurcumin 6C once a week for first 2 months, followed by once in 2 weeks. Individuals 5 years or above: 4 pills of medicine Nanocurcumin 6C once a week for first 2 months, followed by once in 2 weeks. | Sample size: 17,000; Gender: all; Ages: 1 years and over. | CTRI/2021/08/035906 | |
Effect of Bromelain, Curcumin and Epigallocatechin in the treatment of outpatient COVID-19 patients | 3 | Iran (Islamic Republic of) | Blood oxygen saturation, sense of smell, sense of taste, fever, lung involvement, cough, muscle pain, weakness, gastrointestinal symptoms, death, hospitalization. | Each capsule contained 150 mg Bromelain, 300 mg Curcumin and 50 mg epigallocatechin; orally twice a day for 5 days | Sample size: 300; Gender: all; Ages: 18 years and over. | IRCT20210724051971N1 | |
Study Designed to Evaluate the Effect of CimetrA in Patients Diagnosed With COVID-19 | 2 | Israel | 1. Change in WHO Ordinal Scale for Clinical Improvement; 2. Change in COVID-19-Related Symptoms score; 3. Safety endpoint: will be assessed through collection and analysis of adverse events, blood and urine laboratory test, blood pressure and saturation, body temperature. Time frame: up to 28 days | CimetrA-1 containing a combination of curcumin 40 mg, Boswellia 30 mg and Vitamin C 120 mg. CimetrA-2 containing a combination of Curcumin 28 mg, Boswellia 21 mg and vitamin C 84 mg. Spray administration twice a day on days 1 and 2. | Sample size: 240; Gender: all; Ages: 18 years and over. | NCT05037162 | |
Clinical Study Designed to Evaluate the Effect of CimetrA in Patients Diagnosed With COVID-19 | 3 | Israel | Clinical improvement in treatment groups. Time frame: up to 28 days. Time to sustained clinical improvement, defined as a national Early Warning Score 2 (NEWS2) of 2 maintained for 24 h in comparison to routine treatment (measured on days 7, 14, 28) | CimetrA-1 containing a combination of artemisinin 12 mg, curcumin 40 mg, Boswellia 30 mg, and vitamin C 120 mg. CimetrA-2 containing a combination of artemisinin 8.4 mg, curcumin 28 mg, Boswellia 21 mg, and Vitamin C 84 mg. Spray administration, twice a day on daays1 and 2. | Sample size: 252; Gender: all; Ages: 18 years and over. | NCT04802382 | |
A clinical study to see the effect of ArtemiC in patients with COVID-19 | 2 | India | 1.Time to clinical improvement, defined as a national Early Warning Score 2 (NEWS2); 2. Percentage of participants with definite or probable drug-related adverse events. | ArtemiC containing 12 mg artemisinin, 40 mg curcumin, 30 mg frankincense and 120 mg vitamin C as a maximum dose per 24 h, nasal spray, twice a day | Sample size: 20; Gender: all; Ages: 18 years to 65 years. | CTRI/2021/02/031520 | |
Oral Curcumin, Quercetin and Vitamin D3 Supplements for Mild to Moderate Symptoms of COVID-19 | N/A | Pakistan | 1.SARS-CoV-2 negativity determined by RT-PCR. Time frame: up to 14 days; 2. COVID-19 symptom improvement. Time frame: up to 7 days. | 168 mg curcumin, 260 mg quercetin, and 360 IU of vitamin D3 orally once a day for 14 days. | Sample size: 50; Gender: all; Ages: 18 years and older. | NCT04603690 | |
Assessment of the effect of nanocurcumin supplement in patients with COVID-19 | N/A | Iran (Islamic Republic of) | hs-CRP, recovery percentage, percentage of oxygen saturation, severity of infection symptoms of upper and lower respiratory tract, CBC. | 80 mg nanocurcumin orally once every 12 h for 6 days. | Sample size: 48; Gender: all; Ages: 30 years to 70 years old. | IRCT20131125015536N13 | |
Effect of curcumin in treatment of respiratory syndrome of corona | 2/3 | Iran (Islamic Republic of) | Body temperature; oxygen saturation; chest CT-scan at the beginning of the study and on the third and seventh days. | 150 mg curcumin orally every 8 h for 7 days. | Sample size: 42; Gender: all; Ages: no age limit. | IRCT20200418047119N1 | |
Evaluation of the effect of curcumin in improving patients with COVID-19 | 3 | Iran (Islamic Republic of) | CT-scan findings; Hospitalization duration; CBC; LDH; PT; PTT; D-DIMER; BUN/CR. | Patients are given 3 curcumin capsules (500 mg) daily after three meals. | Sample size: 60; Gender: all; Ages: 18 years to 70 years old. | IRCT20200514047445N1 | |
Curcumin for COVID-19 Pre Exposure Prophylaxis | 4 | India | SARS-CoV-2 infection rate Using RT-PCR. Time Frame: up to 12 weeks. | Oral curcumin capsule 500 mg twice daily (morning, evening) for 12 weeks. | Sample size: 200; Gender: all; Ages: 18 years to 70 years old. | CTRI/2020/07/026820 | |
A clinical study to see effect of ArtemiC in patients with COVID-19 | 2 | India | 1.Time to clinical improvement, defined as a national Early Warning Score 2 (NEWS2); 2. Percentage of participants with definite or probable drug-related adverse events. Time frame: up to 15 days. | ArtemiC is an oromucosal medical spray composed ofartemisinin (6 mg/mL), curcumin (20 mg/mL), frankincense (15 mg/mL) and vitamin C (60 mg/mL); spray administration two times a day on days 1 and 2. Each dose contains 1 mL (10 puffs/pushes on the spray bottle), total daily dose 2 mL (20 puffs/pushes on the spray bottle). The total treatment is 40 puffs over two days. | Sample size: 50; Gender: all; Ages: 18 years to 65 years old. | CTRI/2020/07/026789 | |
Evaluation of the effect of nano micelles containing curcumin (Sina Ccurcumin) as a therapeutic supplement in patients with COVID-19 | N/A | Iran (Islamic Republic of) | 1.COVID-19 symptoms improvement; 2. Changes in immune cell balance. Frame: up to 2 weeks. | Oral 40 mg nanocurcumin capsules four times a day for 2 weeks. | Sample size: 40; Gender: all; Ages: 18 years to 75 years old. | IRCT20200611047735N1 | |
Evaluation the anti-inflammatory effects of curcumin in the treatment of patients with COVID-19 | 3 | Iran (Islamic Republic of) | Cytokine gene expression, cytokine serum levels, clinical symptoms, laboratory findings. | 240 mg nanocurcumin for 7 days at the same time with common therapeutic protocol category | Sample size: 60; Gender: all; Ages: 18 years to 65 years old. | IRCT20200519047510N1 | |
Evaluation efficacy of Curcumin and Resveratrol capsule in controlling symptoms in patients with COVID-19 | 3 | Iran (Islamic Republic of) | Clinical symptoms changes (dry cough, respiratory distress, fever). | Each capsule contains 200 mg of curcumin, 200 mg of resveratrol as active ingredients, 1 capsule every 12 h for 7 days. | Sample size: 60; Gender: all; Ages: 18 years and over. | IRCT20080901001165N56 | |
Effect of curcumin-piperine in patients with coronavirus (COVID-19) | N/A | Iran (Islamic Republic of) | CT of the chest, body temperature, length of hospital stay, hs-CRP, ESR, ALT, AST, LDH, BUN, creatinine, CBC, blood oxidative stress indices (SOD, MDA, TAC), Albumin, Severity of the disease, severity and number of coughs. | Two curcumin-piperine capsules (500 mg curcumin + 5 mg piperine) will be given daily for 2 weeks after lunch and dinner. | Sample size: 100; Gender: all; Ages: 20 years to 75 years old. | IRCT20121216011763N46 | |
Evaluation of SinaCurcumin capsule efficacy as an supplement therapy for mild to moderate COVID-19 in Mashhad | 3 | Iran (Islamic Republic of) | Rates of treatment response and adverse drug reactions. | Nanocurcumin capsule 40 mg, two capsules twice daily for 2 weeks, then one capsule twice daily for 2 weeks. | Sample size: 60; Gender: all; Ages: 18 years to 65 years old. | IRCT20200408046990N1 | |
Effects of nano curcumin supplementation on the reduction of inflammation and mortality in patients with coronavirus 2019 admitted to ICU ward of imam Reza hospital in Tabriz | 2/3 | Iran (Islamic Republic of) | Gene expression rate; cytokine secretion rate; clinical observations; laboratory observations. | Oral 240 mg of nanocurcumin in 3 capsules of 80 mg daily. | Sample size: 86; Gender: all; Ages: 18 years to 80 years old. | IRCT20200324046851N1 | |
Fluoxetine (6) | Use of a Combined Regimen of Fluoxetine, Prednisolone and Ivermectin in the Treatment of Mild COVID-19 to Prevent Disease Progression in Papua New Guinea | 2/3 | Papua New Guinea | COVID-19 disease progression (time frame: up to 14 days); SARS-CoV-2 viral load (time frame: up to 7 days) | Fluoxetine 20 mg oral tablets daily for 9 days. Prednisolone 25 mg oral tablets daily for 4 days. Ivermectin 3 mg oral tablets daily for 5 days. | Sample size: 954; Gender: all; Ages: 18 years to 99 years old. | NCT05283954 |
efficacy and safety of adding fluoxetine to therapeutic regimen of patients with COVID-19 pneumonia | 3 | Iran (Islamic Republic of) | Blood oxygen saturation; number of days of hospitalization; need for intubation; ICU admission; death. | Oral fluoxetine capsules for 28 days, with 10 mg for the first 4 days followed by 20 mg for the rest of the 4-week period. | Sample size: 72; Gender: all; Ages: 16 years to 65 years old. | IRCT20200904048616N1 | |
Fluoxetine to Reduce Hospitalization From COVID-19 Infection (FloR COVID-19) | Early Phase 1; Withdrawn (study timeline is not feasible) | United States | Rate of hospitalization; physical symptoms assessed through daily checklist. Time frame: 8 weeks | Orally daily following: week 1: one pill (20 mg), week 2: two pills (40 mg), weeks 3–6: three pills (60 mg), week 7: two pills (40 mg), week 8: one pill (20 mg) | Sample size: 0. | NCT04570449 | |
Fluoxetine to Reduce Intubation and Death After COVID19 Infection | 4 | United States | Hospitalizations, intubation, death. Time frame: 2 months. | Oral 20 mg to 60 mg daily for 2 weeks to 2 months. | Sample size: 2000; Gender: all; Ages: 18 years and older. | NCT04377308 | |
Bardoxolone Methyl (7) | BARCONA: A Study of Effects of Bardoxolone Methyl in Participants With SARS-Corona Virus-2 (COVID-19) | 2 | United States | Number of serious adverse events. Time frame: 29 days. | Oral 20 mg once a day for the duration of hospitalization (until recovery) with a maximum treatment duration of 29 days. | Sample size: 40; Gender: all; Ages: 18 years and older. | NCT04494646 |
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
Dimethyl Fumarate (4) | Efficacy and Safety Study of BG00012 with Methotrexate in Patients With Active Rheumatoid Arthritis | 2 | Australia | The primary objective is the proportion of subjects with ACR20 response in their RA at Week 12. | 480 mg/day, oral and 720 mg/day, oral | Sample size: 153; Gender: all; Ages: 18 Years to 75 Years | NCT00810836 |
Curcumin (5) | Curcuma Longa L in Rheumatoid Arthritis | 1; terminated (insufficient enrollment) | United States | Number of participants with adverse events as a measure of safety and tolerability. | 4 250 mg curcumin capsules twice a day for one month | Sample size: 3; Gender: all; Ages: 18 Years and older. | NCT02543931 |
Curcumin in Rheumatoid Arthritis | Early phase 1 | United States | American College of Rheumatology 20%. Time frame: 4-month period. | 4 capsules once a day for 2 weeks, and then the dose will be increased to 4 capsules twice a day beginning at week 3. Subjects will remain at this dose for an additional 13 weeks for a total 16 weeks. After 16 weeks, the same procedures will be repeated for another 16 weeks | Sample size: 40; Gender: all; Ages: 18 Years to 75 Years. | NCT00752154 |
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
EGCG (1) | Prevention of Cognitive Decline in ApoE4 Carriers with Subjective Cognitive Decline After EGCG and a Multimodal Intervention | N/A | Spain | Preclinical Alzheimer Cognitive Composite Plus exe-like score (ADCS-PACC-like). | Oral 532 mg/day (weight > 50 kg). Oral 266 mg/day (weight < 50 kg). | Sample size: 200; Gender: all; Ages: 60 years to 80 years old. | NCT03978052 |
Sunphenon EGCg (Epigallocatechin-Gallate) in the Early Stage of Alzheimer’s Disease | 2/3 | Germany | ADAS-COG (Score 0–70) (baseline to treatment). Time frame: 18 months. | months 1–3: 200 mg/day (200-0-0 mg); months 4–6: 400 mg/day (200-0-200 mg); months 7–9: 600 mg/day (400-0–200 mg); months 10–18: 800 mg/day (400-0-400 mg) | Sample size: 21; Gender: all; Ages: 60 years and older. | NCT00951834 | |
Sunphenon EGCg (Epigallocatechin-Gallat) in the early stage of Alzheimer’s Disease—SUN-AK | 2 | Germany | Sample size: 50; Gender: all; Ages: 18 years and older. | EUCTR2009-009656-20-DE | |||
Sulforaphane (2) | Effects of Sulforaphane in Patients with Prodromal to Mild Alzheimer’s Disease | N/A | China | The Alzheimer’s Disease Assessment Scale. | Oral 2550 mg once a day for 24 weeks. | Sample size: 160; Gender: all; Ages: 50 years to 75 years old. | NCT04213391 |
Resveratrol (3) | BDPP Treatment for Mild Cognitive Impairment (MCI) and Prediabetes or Type 2 Diabetes Mellitus (T2DM) | 1 | United States | Assessment of AEs and SAEs. Brain penetrance of BDPP. Neuropsychiatric Inventory and Cornell Scale for Depression in Dementia. Memory, executive function, and attention measures (composite). | N/A | Sample size: 14; Gender: all; Ages: 50 years to 90 years old. | NCT02502253 |
Short Term Efficacy and Safety of Perispinal Administration of Etanercept in Mild to Moderate Alzheimer’s Disease | 1 | United States | Difference in effects of treatment for 6 weeks with etanercept + nutritional supplements versus nutritional supplements alone on the Mini-Mental Status Examination (MMSE) score. | N/A | Sample size: 12; Gender: all; Ages: 60 years to 85 years old. | NCT01716637 | |
Resveratrol for Alzheimer’s Disease | 2 | United States | Number of adverse events. Change from baseline in volumetric magnetic resonance imaging (MRI). | Begin at 500 mg taken once daily and increase after 13 weeks to 1 g taken by mouth twice daily. | Sample size: 119; Gender: all; Ages: 50 years and older. | NCT01504854 | |
Pilot Study of the Effects of Resveratrol Supplement in Mild-to-moderate Alzheimer’s Disease | 3; withdrawn (PI has left institution) | United States | Cognition. Time frame: 52 weeks. | Oral 215 mg once a day for 52 weeks. | Sample size: 0. | NCT00743743 | |
Randomized Trial of a Nutritional Supplement in Alzheimer’s Disease | 3 | United States | Alzheimer Disease Assessment Scale (ADAScog). Time frame: one year. | N/A | Sample size: 27; Gender: all; Ages: 50 years to 90 years old. | NCT00678431 | |
Curcumin (5) | KARVIAH_XTND: Longitudinal follow-up study examining the health and wellbeing of participants for identifying new biomarkers and the impact of lifestyle. (Following a 12 month intervention of curcumin for the prevention of Alzheimer’s disease.) | N/A | Australia | Blood biomarker compared with the brain amyloid levels. Blood biomarkers and PET imaging results. | N/A | Sample size: 100; Gender: all; Ages: 65 years and older. | ACTRN12620001325998 |
Curcumin and Yoga Therapy for Those at Risk for Alzheimer’s Disease | 2 | United States | Curcumin effects (first six-month period) or curcumin and aerobic yoga effects (second six-month period) on the changes in the levels of blood biomarkers for mild cognitive impairment relative to baseline or relative to placebo or non-aerobic yoga. | Oral 800 mg curcumin in 4 capsules BID per day prior to meals. | Sample size: 80; Gender: all; Ages: 50 years to 90 years old. | NCT01811381 | |
KARVIAH Sub-study: Examining the use of curcumin on cognition and mood in an older population | 2 | Australia | Attention tasks and working memory as measured using a computerized cognitive battery (CogState). | Oral 500 mg 3 times daily. | Sample size: 40; Gender: all; Ages: 65 years to 90 years old. | ACTRN12616001113448 | |
Effect of curcumin (tumeric) in Alzheimer’s disease | N/A | Iran (Islamic Republic of) | MMSE and quality of life questionnaires. Time frame: before and after intervention (12 weeks). | Oral 500 mg twice a day for 12 weeks. | Sample size: 70; Gender: all; Ages: no age limit. | IRCT201507271165N11 | |
The epigenetic effect of curcumin as measured in the blood and seen within lifestyle, for the prevention of Alzheimer’s disease | 2 | Australia | Measurement of blood biomarkers within healthy and MCI groups. | Oral 1.5 mg daily (×3 divided doses) for a period of 3 or 6 months. | Sample size: 60; Gender: all; Ages: 65 years to 90 years old. | ACTRN12614001024639 | |
McCusker KARVIAH: Curcumin in Alzheimer’s disease prevention | 2 | Australia | AD-related blood biomarker profiles. Pib PET imaging. Neuropsychological tests. Time frame: up to 12 months. | 500 mg daily for 2 weeks, progressing to 500 mg twice daily (1000 mg/daily) for 2 weeks, then 500 mg three times daily (1500 mg) for a period of 12 months in total. | Sample size: 134; Gender: all; Ages: 65 years to 90 years old. | ACTRN12613000681752 | |
Biocurcumax from curry spice turmeric in retaining cognitive function | N/A | Australia | Psychometric testing using Mini-Mental State Examination (MMSE), CAMDEX-R and (CAMCOG)-R, etc. | Oral 500 mg three times daily (total 1500 mg/day). | Sample size: 134; Gender: all; Ages: 65 years to 90 years old. | ACTRN12611000437965 | |
Efficacy and Safety of Curcumin Formulation in Alzheimer’s Disease | 2 | India | To determine if curcumin formulation affects mental capacity in Alzheimer’s patients based on mental exams. | Oral 2000 mg or 3000 mg daily BID. | Sample size: 26; Gender: all; Ages: 50 years to 80 years old. | NCT01001637 | |
A Pilot Study of Curcumin and Ginkgo for Treating Alzheimer’s Disease | 1/2 | Hong Kong, China | Change in isoprostane level in plasma. Change in A-beta level in serum. | Oral 1 g/4 g once daily. | Sample size: 36; Gender: all; Ages: 50 years and older. | NCT00164749 | |
Curcumin in Patients with Mild to Moderate Alzheimer’s Disease | 2 | United States | Side effect checklist. | N/A | Sample size: 33; Gender: all; Ages: 50 years and older. | NCT00099710 |
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
Vitamin D3 (8) | The Effects of Vitamin D and Bone Loss in Parkinson’s Disease | 2 | United States | Direct changes in bone formation and resorption will be investigated by measuring serum 25-hydroxyvitamin D [25(OH)D] level, serum parathyroid hormone (PTH) levels, serum osteocalcin, and serum n-telopeptides (N-Tx). Time frame: 12 months. | 1000 IU/day of vitamin D3. | Sample size: 23; Gender: all; Ages: 18 years and older. | NCT00907972 |
Clinical Effects of Vitamin D Repletion in Patients With Parkinson’s Disease | 4 | United State | Change from baseline visit to 3 months (treatment visit #1) in the TUG, timed walking task (8 m) and UPDRS III subscore. Time frame: 6 months. | 600 IU vitamin D3 capsule daily. | Sample size: 31; Gender: All; Ages: 18 years to 89 years. | NCT00571285 | |
12 Weeks Vitamin D Supplementation and Physical Activity in PD Patients With DBS | Not Applicable | Poland | The effects of vitamin D supplementation and physical activity on concentration of vitamin D3 in serum—the evaluation of changes before and after 12 weeks of supplementation and physical activity. Time frame: the outcome will be assessed up to 1 year after the last collection of blood. | Dosage based on the BMI as followed: for BMI under 25—4000 IU/day, for BMI between 25 and 30—5000 IU/day, and for BMI over 30—6000 IU/day. | Sample size: 72; Gender: all; Ages: 40 years to 90 years. | NCT04768023 | |
Effects of Vitamin D in Parkinson’s Disease (PD) | 2 | United States | Change in static balance as recorded using dynamic posturography with the sensory organization test (SOT 1–3). | Drug: vitamin D3 Vitamin D3 at 10,000 IU a day. Dietary supplement: calcium 1000 mg calcium daily. | Sample size: 101; Gender: all; Ages: 50 years to 99 years. | NCT01119131 | |
Resveratrol (3) | Tolerability, Safety and Pharmacokinetics of Four Single-doses of BIA 6-512 (Trans-resveratrol) and Their Effect on the Levodopa Pharmacokinetics | 1 | Portugal | 1.Maximum observed plasma drug concentration (Cmax) post-dose—levodopa. Time of occurrence of Cmax (tmax)—levodopa; 2. Area under the plasma concentration–time curve (AUC) from time zero to the last sampling time at which concentrations were at or above the limit of quantification (AUC0-t), calculated by the linear trapezoidal rule—–evodopa; 3. Area under the plasma concentration versus time curve from time zero to infinity (AUC0-∞), calculated from AUC0-t + (Clast/λz), where Clast is the last quantifiable concentration and λz is the apparent terminal rate constant—levodopa; 4. Apparent terminal half-life, calculated from ln 2/λz (t1/2)—levodopa; 5. Maximum observed plasma drug concentration (Cmax) post-dose—BIA 6-512; 6. Time of occurrence of Cmax (tmax)—BIA 6-512; 7. Area under the plasma concentration–time curve (AUC) from time zero to the last sampling time at which concentrations were at or above the limit of quantification (AUC0-t), calculated by the linear trapezoidal rule—BIA 6-512. 8; Area under the plasma concentration versus time curve from time zero to infinity (AUC0-∞), calculated from AUC0-t + (Clast/λz), where Clast is the last quantifiable concentration and λz the apparent terminal rate constant—BIA 6-512; 9. Apparent terminal half-life calculated from ln 2/λz (t1/2)—BIA 6-512. | One capsule of Madopar® HBS 125 (levodopa 100 mg/benserazide 25 mg) in an open label manner, concomitantly with BIA 6-512/Placebo. | Sample size: 20; Gender: all; Ages: 18 years to 45 years. | NCT03091543 |
Effect of BIA 6-512 at Steady-state on the Levodopa Pharmacokinetics With a Single-dose of Levodopa/Benserazide 200/50 mg or With a Single-dose of Levodopa/Benserazide 200/50 mg Plus a Single-dose of Nebicapone 150 mg | 1 | Portugal | 1.Day 4—Maximum observed plasma drug concentration (Cmax); 2. Day 4—Time of occurrence of Cmax (tmax); 3. Day 4—Area under the plasma concentration-time curve (AUC) from time zero to the last sampling time at which concentrations were at or above the limit of quantification (AUC0-t); 4. Day 4—AUC from time zero to 8 h post-dose (AUC0-τ); 5. Day 4—Area under the plasma concentration versus time curve from time zero to infinity (AUC0-∞); 6. Day 4—Apparent terminal elimination half-life, calculated from ln 2/λz (t1/2). 7; Day 5—Maximum observed plasma drug concentration (Cmax); 8. Day 5—Time of occurrence of Cmax (tmax); 9. Day 5—Area under the plasma concentration-time curve (AUC) from time zero to the last sampling time at which concentrations were at or above the limit of quantification (AUC0-t); 10. Day 5—AUC from time zero to 8 h post-dose (AUC0-τ); 11. Day 5—Area under the plasma concentration versus time curve from time zero to infinity (AUC0-∞); 12. Day 5—Apparent terminal elimination half-life, calculated from ln 2/λz (t1/2). | The investigational products consisted of capsules containing BIA 6-512 25 mg, 50 mg, 75 mg, 100 mg. Orally, with 240 mL of potable water. | Sample size: 38; Gender: all; Ages: 18 years to 45 years. | NCT03097211 | |
EGCG (1) | Efficacy and Safety of Green Tea Polyphenol in De Novo Parkinson’s Disease Patients | 2 | China | Delay of progression of motor dysfunction. | N/A | Sample size: 480; Gender: all; Ages: 30 years and older. | NCT00461942 |
Sulforaphane (2) | A 6-month Study to Evaluate Sulforaphane Effects in PD Patients | 2 | China | Cognitive improvement assessed using the MATRICS Consensus Cognitive Battery (MCCB) composite score. | N/A | Sample size: 100; Gender: all; Ages: 40 years to 75 years. | NCT05084365 |
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
Curcumin (5) | Effect of Curcumin on Systemic Lupus Erythematosus | 2 | California, United States | Change in SLEDAI. | Intervention is 2 g of curcumin supplement per day. | Sample size: 23; Gender: all; Ages: 18 years and older | NCT03953261 |
Vitamin D and Curcumin Piperine Attenuates Disease Activity and Cytokine Levels in Systemic Lupus Erythematosus Patients | 2 | Indonesia | 1.Disease activity from the SLE patients after the Treatments; 2. Fatigue assessment from the SLE patients after the treatments; 3. Comparison of cytokine levels before and after the treatments. | The third group received 400 IU cholecalciferol (Nature Plus) t.i.d and curcumin (600 mg)—piperine (15,800 mg) (Bioglan) one time daily. | Sample size: 45; Gender: all; Ages: 18 years to 45 years | NCT05430087 | |
Vitamin D3 (8) | Vitamin D3 Treatment in Pediatric Systemic Lupus Erythematosus | 2 | California | Change in average IFN module expression level Percentage of Subjects by treatment arm experiencing any adverse event (AE) ≥ grade 3. | 6000 IU of vitamin D3 by mouth daily until the subject’s serum 25 (OH) level is ≥ 40 ng/mL, at which point the supplementation dose is reduced to 4000 IU/day. Note: Subjects weighing < 40 kg (kg) at study entry will receive their dose five days a week and all other subjects seven days a week. | Sample size: 7; Gender: all; Ages: 5 years to 20 years. | NCT01709474 |
Vitamin D3 in Systemic Lupus Erythematosus | 2 | United States | Percent of s with an IFN alpha signature response at Week 12. | 8% vitamin D3 powder, 84% microcrystalline cellulose, 8% fumed silica by weight. | Sample size: 57; Gender: all; Ages: 18 years and older. | NCT00710021 | |
Vitamin D to Improve Endothelial Function in SLE | 2 | United States | Change at week 16 in % flow-mediated dilation in those who did and did not replete vitamin D. | 5000 International units versus 400 international units as an active comparator. | Sample size: 9; Gender: all; Ages: 18 years and older. | NCT01911169 | |
Vitamin D Therapy in Patients With Systemic Lupus Erythematosus (SLE) | 1 | United States | Hypercalcuria. | Cholecalciferol 800 IU oral daily. Cholecalciferol 2000 IU oral daily. Cholecalciferol 4000 IU oral daily. | Sample size: 18; Gender: all; Ages: 18 years to 85 years. | NCT00418587 | |
Vitamin D and Curcumin Piperine Attenuates Disease Activity and Cytokine Levels in Systemic Lupus Erythematosus Patients | 2 | Indonesia | 1. disease activity from the SLE patients after the treatments; 2. Fatigue assessment from the SLE patients after the treatments; 3. Comparison of cytokine levels before and after the treatment | The second group received a tablet containing curcumin (632 mg)—piperine (15,800 mg) (Bioglan) one time daily and a placebo (Saccharum lactis) t.i.d. | Sample size: 45; Gender: all; Ages:18 years to 45 years. | NCT05430087 | |
Effect of Vitamin D Supplement on Disease Activity in SLE | Not Applicable | Thailand | To examine the effect of vitamin D supplementation on SLE disease activity. | Add on vitamin D2 (calciferol) 40,000 IU/wk (2 cap) for 12 weeks. | Sample size: 100; Gender: all; Ages: 18 years and older. | NCT05260255 | |
The Effect of Vitamin D Supplementation on Disease Activity Markers in Systemic Lupus Erythematosus (SLE) | Not Applicable | Egypt | Decrease in SLE disease activity. | 2000 IU/day for 12 months. | Sample size: 248; Gender: all; Ages: 30 years and older. | NCT01425775 | |
SM934 (9) | Safety and Efficacy of SM934 Compared to Placebo in Adult Subjects With Active Systemic Lupus Erythematosus | 2 | China | 1.Percentage of subjects with lupus low disease activity score (LLDAS) in each group; 2. Percentage of subjects with systemic lupus erythematosus responder index—4 (SRI-4) response in each group; 3. Percentage of subjects with treatment-emergent adverse events (TEAEs) in each group. | SM934 10 mg (5 tablet) p.o. qd in combination with steroids. | Sample size: 48; Gender: all; Ages: 30 years and older. | NCT03951259 |
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
Liraglutide | Efficacy Study of Liraglutide vs. Sitagliptin vs. Glargine on Liver Fat in T2DM Subjects | 4 | China | To compare the change of intrahepatic lipids (IHL) in type 2 diabetic patients with nonalcoholic fatty liver disease after a 26-week treatment of liraglutide, sitagliptin, or insulin glargine per day combined with metformin. | Liraglutide, 0.6 mg per day for the first week, increased to 1.2 mg per day for the second week, and finally 1.8 mg per day from the third week. | Sample size: 75; Gender: all; Ages: 30 years to 75 years. | NCT02147925 |
Antidiabetic Effects on Intrahepatic Fat | 4 | China | Intrahepatic fat change from baseline by quantitative ultrasound. | 0.6 mg/day during the first week, 1.2 mg/day during the second week, and 1.8 mg/day from the third week. | Sample size: 87; Gender: all; Ages: 17 years to 80 years. | NCT03068065 | |
Liraglutide Efficacy and Action in Non-Alcoholic Steatohepatitis | 2 | England | Liver histological improvement. | 1.8 mg once daily, subcutaneous injection. | Sample size: 52; Gender: all; Ages: 18 years to 70 years. | NCT01237119 | |
Study of Liraglutide Versus Insulin on Liver Fat Fraction in Patients With Type 2 Diabetes | 2 | Canada | Improvement in liver steatosis defined by change in liver fat fraction as measured by MRI and MR spectroscopy at baseline and 12 weeks of treatment. | 0.6–1.8 mg subcutaneous per day. | Sample size: 35; Gender: all; Ages: 18 years and older. | NCT01399645 | |
Resveratrol (3) | Long-term Investigation of Resveratrol on Fat Metabolism in Obese Men With Nonalcoholic Fatty Liver Disease | N/A | Denmark | Hepatic VLDL-TG secretion and peripheral VLDL-TG clearance. Time frame: six months. - Changes from baseline after treatment with either resveratrol or placebo. | 500 mg 3 times daily for six months. | Sample size: 26; Gender: all; Ages: 25 years to 65 years. | NCT01446276 |
Resveratrol for the Treatment of Non Alcoholic Fatty Liver Disease and Insulin Resistance in Overweight Adolescents | 2/3 | Canada | Primary Side effect profile determined by interview and serum biochemistry. Side effect profile determined by serum biochemistry: AST, ALT, total and conjugated bilirubin, Creatinine, sodium, potassium, calcium, magnesium, chloride and TC02, haemoglobin, haematocrit, white blood cell and platelet counts, erythrocytes, and fasting lipid levels (total cholesterol, HDL-cholesterol, LDL-cholesterol and triglycerides). Fasting glucose and insulin levels. PT/INR and PTT levels. | Oral 75 mg twice daily (with breakfast and dinner) for a total daily dose of 150 mg for the duration of 30 days. | Sample size: 10; Gender: all; Ages: 13 years to 18 years. | NCT02216552 | |
Resveratrol in Patients With Non-alcoholic Fatty Liver Disease | 2/3 | Denmark | Changes in hepatic and inflammatory markers ind the blood such as ALT, hs-CRP, TNFa; changes in hepatic fat content, assessed by MR spectroscopy; changes in hepatic steatosis and inflammation, assessed histologically; changes in the expression of proteins in the relevant inflammatory pathways, assessed by gene expression studies. | 500 mg 3 times daily for 6 months. | Sample size: 28; Gender: all; Ages: 18 years to70 years. | NCT01464801 | |
The Effects of Resveratrol Supplement on Biochemical Factors and Hepatic Fibrosis in Patients With Nonalcoholic Steatohepatitis | 2/3 | America | Alaninaminotransferase (ALT). | One resveratrol capsule per day for 12 weeks. | Sample size: 50; Gender: all; Ages: 18 years to 80 years. | NCT02030977 | |
Potential Beneficial Effects of Resveratrol | N/A | Denmark | Metabolic parameters. Time frame: five weeks. Regarding glucose, protein, and fat metabolism. | 500 mg three times a day for five weeks. | Sample size: 24; Gender: male; Ages: 18 years and older. | NCT01150955 | |
Curcumin (5) | Curcumin for Pediatric Nonalcoholic Fatty Liver Disease | 2 | America | Change in serum alanine aminotransferase (ALT) from baseline. Time frame: 24 weeks. ALT value in U/L | 500 mg daily phosphatidylcholine–curcumin complex supplement orally for 24 weeks. | Sample size: 0; Gender: all; Ages: 8 years to 17 years. | NCT04109742 |
Curcumin Supplement in Nonalcoholic Fatty Liver Patients | 2/3 | America | Hepatic steatosis (time frame: 12 weeks) measured by CAP score using Fibroscan. | 1500 mg one capsule/day for 12 weeks. | Sample size: 50; Gender: all; Ages: 18 years and older. | NCT02908152 | |
The Effect of Curcumin on Liver Fat Content in Obese Subjects | N/A | Denmark | Curcumin’s effect on steatosis. Time frame: 42 days ± 3 days. Percentage of fat in the liver tissue measured by magnetic resonance spectroscopy. | 500 mg tablet (contains 100 mg curcumin); Dosage: 2 tablets twice daily for 42 days (± 3 days). | Sample size: 39; Gender: male; Ages: 20 years and older. | NCT03864783 | |
Efficacy of a Natural Components Mixture in the Treatment of Non Alcoholic Fatty Liver Disease (NAFLD) | N/A | Italy | Hematic levels of hepatic enzymes AST; hematic levels of hepatic enzymes ALT; hematic levels of hepatic enzymes GGT. Time frame: before and at the end of treatment (three months). | Nutraceutical mixture (two soft 800 mg gelatin capsules per day) for three months. | Sample size:126; Gender: male; Ages: 18 years to 80 years. | NCT02369536 |
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
Silymarin (10) | Effects of Silybum Marianum on Treatment of Patients With Chronic Hepatitis C | 2 | Iran | The investigators measured serum amino transferases using commercial AST kits and ALT kits (Bayer Diagnostics, Tarrytown, NY, USA) at six months after silymarin admission. | 210 mg tabs; 630 mg daily for six months. | Sample size: 55; Gender: all; Ages: child, adult, older adult. | NCT01292161 |
Clinical Study With Silymarin in the Patients With Chronic Hepatitis C Infection Who Failed Conventional Antiviral Therapy | 3 | Korea | The proportion of patient with serum ALT less than or equal to 40 IU/L or achieves at least 50% decline to less than 60 IU/L. | 700 mg thrice daily. | Sample size: 53; Gender: all; Ages: 18 years and older. | NCT01258686 | |
Phase II Trial of Silymarin for Patients With Chronic Hepatitis C Who Have Failed Conventional Antiviral Treatment | 2 | America | 1. Efficacy—whether or not serum ALT (mg/dl) is less than or equal to 45 IU/L (approximate normal range) or achieves at least 50% decline to less than 65 IU/L (approximately 1.5 times the upper limit of normal); 2. Safety—occurrence of a dose-limiting toxicity. Time frame: 24-week treatment period. | 1. 700 mg dose (5 pills, three times daily) for 24-week treatment period. 2. 420 mg dose (5 pills, three times daily) for 24-week treatment period. | Sample size: 154; Gender: all; Ages: 18 years and older. | NCT00680342 | |
Evaluating Silymarin for Chronic Hepatitis C | 2 | America | N/A | N/A | N/A | NCT00030030 | |
Randomized Placebo-controlled Trial Evaluating the Safety and Efficacy of Silymarin Treatment in Patients With Acute Viral Hepatitis | 2/3 | Egypt | 1. Incidence, severity, and duration of adverse events. Time frame: four weeks after enrollment. 2. Normalization of total (< 1.0 mg/dl) and direct bilirubin (< 0.3 mg/dl). Time frame: four weeks after enrollment. | 280 mg three times daily for four weeks. | Sample size: 199; Gender: all; Ages: 18 years and older. | NCT00755950 | |
Phase I Trial of Silymarin for Chronic Liver Diseases | 1 | America | Adverse events. Time frame: 10 days. | 280 mg every 8 h. | Sample size: 56; Gender: all; Ages: 18 years and older. | NCT00389376 | |
Effect of LEGALON SIL on Hepatitis C Virus Recurrence in Stable Liver Transplanted Patients | 2 | Italy | To determine the effect of post-transplant treatment with Legalon SIL on HCV viral load 30 days after the beginning of treatment. | 20 mg/kg silibinin, administered daily as a 2-h infusion for 14 days. | Sample size: 20; Gender: all; Ages: 18 years to 70 years. | NCT01518933 |
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
Ursodeoxycholic Acid (11) | Efficacy and Safety Study of TUDCA Compare UDCA to Treatment Chronic Cholestatic Liver Disease -PBC | 3 | China | Efficiency is defined as the proportion of patients whose ALP levels of serum decreased more than 25% compared to baseline at treatment for 24 weeks. | 250 mg/8 h orally for 24 weeks. | Sample size: 199; Gender: all; Ages: 18 years to 70 years. | NCT01829698 |
Clinical Research of UCDA Reducing Medication Regimen in Stable PBC | 4 | China | Liver biochemical markers (AST and ALP in U/L, BIL in umol/L) that restored to normal increase (bilirubin > 17 μmol/L, ALP > 3 ULM, AST > 2 ULN) again are considered to be PBC recurrence. The rate of recurrence will be described in percent. | 1. 250 mg orally twice a day; 2. 250 mg orally once a day; 3. 250 mg orally three times a day. | Sample size: 90; Gender: all; Ages: 18 years to 65 years. | NCT04650243 | |
Ursodeoxycholic Acid Combined With Low Dose Glucocorticoid in the Treatment of PBC With AIH Features II | 4 | China | The percentage of patients in biochemical remission, defined as normalization of serum ALT and IgG levels after treatment, per treatment group. | 13–15 mg/kg/d. | Sample size: 90; Gender: all; Ages: 18 years to 70 years. | NCT04617561 | |
Ursofalk Tablets (500 mg) Versus Ursofalk Capsules (250 mg) in the Treatment of Primary Biliary Cirrhosis | 4 | Germany | Change of liver enzymes between baseline and the end of the treatment period with 250 mg Ursofalk capsules and the end of treatment period with 500 mg Ursofalk tablets. | 500 mg orally for 24 weeks. | Sample size: 65; Gender: all; Ages: 18 years and older. | NCT01510860 | |
Development of Ursodeoxycholic Acid 300 mg at Hospital Das Clinicas of the University of São Paulo School of Medicine | N/A | America | Compare the liver enzyme parameters (alkaline phosphatase, alanine aminotranferase, aspartate. aminotransferase, gamma glutamyl transferase, and total bilirubin) in three different moments before the treatment, under the treatment, and at the end of treatment. | 300 mg 13–15 mg/kg day for 3 months. | Sample size: 30; Gender: all; Ages: 18 years and older. | NCT03489889 |
Intervention | Topic | Phase | Trial Country | Primary Endpoints | Dose | Subjects | Registration Number |
---|---|---|---|---|---|---|---|
Candesartan (12) | Effect of Some Drugs on Liver Fibrosis | 3 | Egypt | Change in Fibroscan or APRI score. | 8 mg/day for 6 months. | Sample size: 45; Gender: all; Ages: 20 years and older. | NCT03770936 |
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
Zhai, Z.; Huang, Y.; Zhang, Y.; Zhao, L.; Li, W. Clinical Research Progress of Small Molecule Compounds Targeting Nrf2 for Treating Inflammation-Related Diseases. Antioxidants 2022, 11, 1564. https://doi.org/10.3390/antiox11081564
Zhai Z, Huang Y, Zhang Y, Zhao L, Li W. Clinical Research Progress of Small Molecule Compounds Targeting Nrf2 for Treating Inflammation-Related Diseases. Antioxidants. 2022; 11(8):1564. https://doi.org/10.3390/antiox11081564
Chicago/Turabian StyleZhai, Zhenzhen, Yanxin Huang, Yawei Zhang, Lili Zhao, and Wen Li. 2022. "Clinical Research Progress of Small Molecule Compounds Targeting Nrf2 for Treating Inflammation-Related Diseases" Antioxidants 11, no. 8: 1564. https://doi.org/10.3390/antiox11081564
APA StyleZhai, Z., Huang, Y., Zhang, Y., Zhao, L., & Li, W. (2022). Clinical Research Progress of Small Molecule Compounds Targeting Nrf2 for Treating Inflammation-Related Diseases. Antioxidants, 11(8), 1564. https://doi.org/10.3390/antiox11081564