Accès libre

Recent advances in the treatment of non-alcoholic fatty liver disease with astragaloside IV

, , , , ,  et   
16 juil. 2025
À propos de cet article

Citez
Télécharger la couverture

R. Loomba, S. L. Friedman and G. I. Shulman, Mechanisms and disease consequences of nonalcoholic fatty liver disease, Cell 184(10) (2021) 2537–2564; https://doi.org/10.1016/j.cell.2021.04.015 Search in Google Scholar

F. Bessone, M. V. Razori and M. G. Roma, Molecular pathways of nonalcoholic fatty liver disease development and progression, Cell. Mol. Life Sci. 76(1) (2019) 99–128; https://doi.org/10.1007/s00018-018-2942-5 Search in Google Scholar

M. Papatheodoridi and E. Cholongitas, Diagnosis of non-alcoholic fatty liver disease (NAFLD): Current concepts, Curr. Pharm. Des. 24(38) (2018) 4574–4586; https://doi.org/10.2174/1381612824666181002105543 Search in Google Scholar

C. D. Byrne and G. Targher, NAFLD: A multisystem disease, J. Hepatol. 62(1)(Suppl.) (2015) S47–S64; https://doi.org/10.1016/j.jhep.2014.12.012 Search in Google Scholar

A. Vadarlis, C. Antza, D. R. Bakaloudi, I. Doundoulakis, G. Kalopitas, M. Samara, T. Dardavessis, T. Maris and M. Chourdakis, Systematic review with meta-analysis: The effect of vitamin E supplementation in adult patients with non-alcoholic fatty liver disease, J. Gastroenterol. Hepatol. 36(2) (2021) 311–319; https://doi.org/10.1111/jgh.15221 Search in Google Scholar

M. Y. Wang, K. Prabahar, M. A. Găman and J. L. Zhang, Vitamin E supplementation in the treatment of nonalcoholic fatty liver disease (NAFLD): Evidence from an umbrella review of meta-analysis on randomized controlled trials, J. Dig. Dis. 24(6–7) (2023) 380–389; https://doi.org/10.1111/1751-2980.13210 Search in Google Scholar

C. Gofton, Y. Upendran, M. H. Zheng and J. George, MAFLD: How is it different from NAFLD?, Clin. Mol. Hepatol. 29(Suppl) (2023) S17–S31; https://doi.org/10.3350/cmh.2022.0367 Search in Google Scholar

S. L. Friedman, B. A. Neuschwander-Tetri, M. Rinella and A. J. Sanyal, Mechanisms of NAFLD development and therapeutic strategies, Nat. Med. 24(7) (2018) 908–922; https://doi.org/10.1038/s41591-018-0104-9 Search in Google Scholar

L. Rong, J. Zou, W. Ran, X. Qi, Y. Chen, H. Cui and J. Guo, Advancements in the treatment of non-alcoholic fatty liver disease (NAFLD), Front. Endocrinol. (Lausanne) 13 (2023) Article ID 1087260 (18 pages); https://doi.org/10.3389/fendo.2022.1087260 Search in Google Scholar

M. J. Watt, P. M. Miotto, W. De Nardo and M. K. Montgomery, The liver as an endocrine organ – Linking NAFLD and insulin resistance, Endocr. Rev. 40(5) (2019) 1367–1393; https://doi.org/10.1210/er.2018-00241 Search in Google Scholar

Y. Sakurai, N. Kubota, T. Yamauchi and T. Kadowaki, Role of insulin resistance in MAFLD, Int. J. Mol. Sci. 22(8) (2021) Article ID 4156 (26 pages); https://doi.org/10.3390/ijms22084156 Search in Google Scholar

G. Paradies, V. Paradies, F. M. Ruggiero and G. Petrosillo, Oxidative stress, cardiolipin and mitochondrial dysfunction in nonalcoholic fatty liver disease, World J. Gastroenterol. 20(39) (2014) 14205–14218; https://doi.org/10.3748/wjg.v20.i39.14205 Search in Google Scholar

J. Yang, M. Fernández-Galilea, L. Martínez-Fernández, P. González-Muniesa, A. Pérez-Chávez, J. A. Martínez and M. J. Moreno-Aliaga, Oxidative stress and non-alcoholic fatty liver disease: Effects of omega-3 fatty acid supplementation, Nutrients 11(4) (2019) Article ID 872 (37 pages); https://doi.org/10.3390/nu11040872 Search in Google Scholar

C. Luci, M. Bourinet, P. S. Leclère, R. Anty and P. Gual, Chronic inflammation in non-alcoholic steatohepatitis: Molecular mechanisms and therapeutic strategies, Front. Endocrinol. (Lausanne) 11 (2020) Article ID 597648 (14 pages); https://doi.org/10.3389/fendo.2020.597648 Search in Google Scholar

R. Forlano, B. H. Mullish, S. K. Mukherjee, R. Nathwani, C. Harlow, P. Crook, R. Judge, A. Soubieres, P. Middleton, A. Daunt, P. Perez-Guzman, N. Selvapatt, M. Lemoine, A. Dhar, M. R. Thursz, S. Nayagam and P. Manousou, In-hospital mortality is associated with inflammatory response in NAFLD patients admitted for COVID-19, PLoS One 15(10) (2020) e0240400; https://doi.org/10.1371/journal.pone.0240400. Search in Google Scholar

A. A. Kolodziejczyk, D. Zheng, O. Shibolet and E. Elinav, The role of the microbiome in NAFLD and NASH, EMBO Mol. Med. 11(2) (2019) e9302; https://doi.org/10.15252/emmm.201809302 Search in Google Scholar

A. Albillos, A. de Gottardi and M. Rescigno, The gut-liver axis in liver disease: Pathophysiological basis for therapy, J. Hepatol. 72(3) (2020) 558–577; https://doi.org/10.1016/j.jhep.2019.11.003 Search in Google Scholar

Z. Younossi, Q. M. Anstee, M. Marietti, T. Hardy, L. Henry, M. Eslam, J. George and E. Bugianesi, Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention, Nat. Rev. Gastroenterol. Hepatol. 15(1) (2018) 11–20; https://doi.org/10.1038/nrgastro.2017.109 Search in Google Scholar

S. Wei, L. Wang, P. C. Evans and S. Xu, NAFLD and NASH: Etiology, targets and emerging therapies, Drug Discov. Today 29(3) (2024) Article ID 103910; https://doi.org/10.1016/j.drudis.2024.103910 Search in Google Scholar

Y. Sun, Y. Ma, F. Sun, W. Feng, H. Ye, T. Tian and M. Lei, Astragaloside IV attenuates lipopolysaccharide induced liver injury by modulating Nrf2-mediated oxidative stress and NLRP3-mediated inflammation, Heliyon 9(4) (2023) e15436; https://doi.org/10.1016/j.heliyon.2023.e15436 Search in Google Scholar

S. Wu, F. Wen, X. Zhong, W. Du, M. Chen and J. Wang, Astragaloside IV ameliorate acute alcohol--induced liver injury in mice via modulating gut microbiota and regulating NLRP3/caspase-1 signaling pathway, Ann. Med. 55(1) (2023) Article ID 2216942 (16 pages); https://doi.org/10.1080/07853890.2023.2216942 Search in Google Scholar

Y. Li, X. Yang, X. Li, S. Wang, P. Chen, T. Ma and B. Zhang, Astragaloside IV and cycloastragenol promote liver regeneration through regulation of hepatic oxidative homeostasis and glucose/lipid metabolism, Phytomedicine 135 (2024) Article ID 156165; https://doi.org/10.1016/j.phymed.2024.156165 Search in Google Scholar

B. Zhou, D. L. Zhou, X. H. Wei, R. Y. Zhong, J. Xu and L. Sun, Astragaloside IV attenuates free fatty acid-induced ER stress and lipid accumulation in hepatocytes via AMPK activation, Acta Pharmacol. Sin. 38(7) (2017) 998–1008; https://doi.org/10.1038/aps.2016.175 Search in Google Scholar

Y. Zhai, W. Zhou, X. Yan, Y. Qiao, L. Guan, Z. Zhang, H. Liu, J. Jiang, J. Liu and L. Peng, Astragaloside IV ameliorates diet-induced hepatic steatosis in obese mice by inhibiting intestinal FXR via intestinal flora remodeling, Phytomedicine 107 (2022) Article ID 154444; https://doi.org/10.1016/j.phymed.2022.154444 Search in Google Scholar

Y.-L. Liu, Q.-Z. Zhang, Y.-R. Wang, L.-N. Fu, J.-S. Han, J. Zhang and B.-M. Wang, Astragaloside IV improves high-fat diet-induced hepatic steatosis in nonalcoholic fatty liver disease rats by regulating inflammatory factors level via TLR4/NF-κB signaling pathway, Front. Pharmacol. 11 (2021) Article ID 605064 (11 pages); https://doi.org/10.3389/fphar.2020.605064 Search in Google Scholar

J. M. Clark, Weight loss as a treatment for nonalcoholic fatty liver disease, J. Clin. Gastroenterol. 40(Suppl. 1) (2006) S39–S43; https://doi.org/10.1097/01.mcg.0000168641.31321.fa Search in Google Scholar

N. Hossain, P. Kanwar and S. R. Mohanty, A comprehensive updated review of pharmaceutical and nonpharmaceutical treatment for NAFLD, Gastroenterol. Res. Pract. 2016 (2016) Article ID 7109270 (17 pages); https://doi.org/10.1155/2016/7109270 Search in Google Scholar

S. Pouwels, N. Sakran, Y. Graham, A. Leal, T. Pintar, W. Yang, R. Kassir, R. Singhal, K. Mahawar and D. Ramnarain, Non-alcoholic fatty liver disease (NAFLD): A review of pathophysiology, clinical management and effects of weight loss, BMC Endocr. Disord. 22(1) (2022) Article ID 63 (9 pages); https://doi.org/10.1186/s12902-022-00980-1 Search in Google Scholar

M. F. Abdelmalek, Nonalcoholic fatty liver disease: Another leap forward, Nat. Rev. Gastroenterol. Hepatol. 18(2) (2021) 85–86; https://doi.org/10.1038/s41575-020-00406-0 Search in Google Scholar

G. Farrell, Insulin resistance, obesity, and liver cancer, Clin. Gastroenterol. Hepatol. 12(1) (2014) 117–119; https://doi.org/10.1016/j.cgh.2013.07.040 Search in Google Scholar

N. Sodum, G. Kumar, S. L. Bojja, N. Kumar and C. M. Rao, Epigenetics in NAFLD/NASH: Targets and therapy, Pharmacol. Res. 167 (2021) Article ID 105484; https://doi.org/10.1016/j.phrs.2021.105484 Search in Google Scholar

S. A. Harrison, A. M. Allen, J. Dubourg, M. Noureddin and N. Alkhouri, Challenges and opportunities in NASH drug development, Nat. Med. 29(3) (2023) 562–573; https://doi.org/10.1038/s41591-023-02242-6 Search in Google Scholar

S. J. Song, J. C. T. Lai, G. L. H. Wong, V. W. S. Wong and T. C. F. Yip, Can we use old NAFLD data under the new MASLD definition?, J. Hepatol. 80(2) (2024) e54–e56; https://doi.org/10.1016/j.jhep.2023.07.021 Search in Google Scholar

R. Younes and E. Bugianesi, The impact of metabolic syndrome on the outcome of NASH: Cirrhosis, hepatocellular carcinoma, and mortality, Curr. Hepatol. Rep. 17(4) (2018) 336–344; https://doi.org/10.1007/s11901-018-0422-x Search in Google Scholar

T. Puengel and F. Tacke, Efruxifermin, an investigational treatment for fibrotic or cirrhotic nonalcoholic steatohepatitis (NASH), Expert Opin. Investig. Drugs 32(6) (2023) 451–461; https://doi.org/10.1080/13543784.2023.2230115 Search in Google Scholar

K. Shree Harini and D. Ezhilarasan, Wnt/β-catenin signaling and its modulators in nonalcoholic fatty liver diseases, Hepatobiliary Pancreat. Dis. Int. 22(4) (2023) 333–345; https://doi.org/10.1016/j.hbpd.2022.10.003 Search in Google Scholar

N. Bhala, R. Younes and E. Bugianesi, Epidemiology and natural history of patients with NAFLD, Curr. Pharm. Des. 19(29) (2013) 5169–5176; https://doi.org/10.2174/13816128113199990336 Search in Google Scholar

H. Kojima, S. Sakurai, M. Uemura, H. Fukui, H. Morimoto and Y. Tamagawa, Mitochondrial abnormality and oxidative stress in nonalcoholic steatohepatitis, Alcohol Clin. Exp. Res. 31(s1) (2007) S61–S66; https://doi.org/10.1111/j.1530-0277.2006.00288.x Search in Google Scholar

N. Alkhouri, M. P. Berk, R. Lopez, T. Abu-Rajab Tamimi, L. Yerian, Y. Chung, R. Zhang, T. M. Mc-Intyre, S. L. Hazen and A. E. Feldstein, OxNASH score correlates with histologic features and severity of nonalcoholic fatty liver disease, Dig. Dis. Sci. 59(7) (2014) 1617–1624; https://doi.org/10.1007/s10620-014-3031-8 Search in Google Scholar

L. S. R. R. Okada, C. P. Oliveira, J. T. Stefano, M. A. Nogueira, I. D. C. G. da Silva, F. B. Cordeiro, V. A. F. Alves, R. S. Torrinhas, F. J. Carrilho, P. Puri and D. L. Waitzberg, Omega-3 PUFA modulate lipo-genesis, ER stress, and mitochondrial dysfunction markers in NASH – proteomic and lipidomic insight, Clin. Nutr. 37(5) (2017) 1474–1484; https://doi.org/10.1016/j.clnu.2017.08.031 Search in Google Scholar

J. S. Chang, J. H. Ahn, S. H. Kang, S. B. Koh, J. Y. Kim, S. K. Baik, J. H. Huh, S. S. Lee, M. Y. Kim and K. S. Park, Metabolic stress index including mitochondrial biomarker for noninvasive diagnosis of hepatic steatosis, Front. Endocrinol. 13 (2022) Article ID 896334 (8 pages); https://doi.org/10.3389/fendo.2022.896334 Search in Google Scholar

S. Seki, T. Kitada and H. Sakaguchi, Clinicopathological significance of oxidative cellular damage in non-alcoholic fatty liver diseases, Hepatol. Res. 33(2) (2005) 132–134; https://doi.org/10.1016/j.hepres.2005.09.020 Search in Google Scholar

J. Zhang, C. Wu, L. Gao, G. Du and X. Qin, Astragaloside IV derived from Astragalus membranaceus: A research review on the pharmacological effects, Adv. Pharmacol. 87 (2020) 89–112; https://doi.org/10.1016/bs.apha.2019.08.002 Search in Google Scholar

L. Li, X. Hou, R. Xu, C. Liu and M. Tu, Research review on the pharmacological effects of astragalo-side IV, Fundam. Clin. Pharmacol. 31(1) (2017) 17–36; https://doi.org/10.1111/fcp.12232 Search in Google Scholar

Z. Zhuang, Z.-H. Wang, L.-H. Deng, Q. Zheng, G.-Q. Zheng and Y. Wang, Astragaloside IV exerts cardioprotection in animal models of viral myocarditis: A preclinical systematic review and meta-analysis, Front. Pharmacol. 10 (2019) Article ID 1388 (11 pages); https://doi.org/10.3389/fphar.2019.01388 Search in Google Scholar

F. Xu, W. Q. Cui, Y. Wei, J. Cui, J. Qiu, L. L. Hu, W. Y. Gong, J. C. Dong and B. J. Liu, Astragaloside IV inhibits lung cancer progression and metastasis by modulating macrophage polarization through AMPK signaling, J. Exp. Clin. Cancer Res. 37(1) (2018) Article ID 207 (16 pages); https://doi.org/10.1186/s13046-018-0878-0 Search in Google Scholar

Z. Han, J. Zhu and Z. Han, Evaluation of astragaloside IV in hepatic fibrosis: A meta-analysis, Medicine 100(13) (2021) e25105; https://doi.org/10.1097/MD.0000000000025105 Search in Google Scholar

H. Wang, Z. Zhuang, Y.-Y. Huang, Y. Zhuang, J. Jin, X. Ye, Z. Lin, J. Zheng and H. Wang, Protective effect and possible mechanisms of astragaloside IV in animal models of diabetic nephropathy: A preclinical systematic review and meta-analysis, Front. Pharmacol. 11 (2020) Article ID 988 (17 pages); https://doi.org/10.3389/fphar.2020.00988 Search in Google Scholar

I. M. Costa, F. O. V. Lima, L. C. B. Fernandes, B. Norrara, F. I. Neta, R. D. Alves, J. R. L. P. Cavalcanti, E. E. S. Lucena, J. S. Cavalcante, A. C. M. Rego, I. A. Filho, D. B. Queiroz, M. A. M. Freire and F. P. Guzen, Astragaloside IV supplementation promotes a neuroprotective effect in experimental models of neurological disorders: A systematic review, Curr. Neuropharmacol. 17(7) (2019) 648–665; https://doi.org/10.2174/1570159X16666180911123341 Search in Google Scholar

M. Marušić, M. Paić, M. Knobloch and A.-M. Liberati Pršo, NAFLD, insulin resistance, and diabetes mellitus type 2, Can. J. Gastroenterol. Hepatol. 2021 (2021) Article ID 6613827 (9 pages); https://doi.org/10.1155/2021/6613827 Search in Google Scholar

D. Xia, W. Li, C. Tang and J. Jiang, Astragaloside IV, as a potential anticancer agent, Front. Pharmacol. 14 (2023) Article ID 1065505 (15 pages); https://doi.org/10.3389/fphar.2023.1065505 Search in Google Scholar

K. Mehta, D. H. Van Thiel, N. Shah and S. Mobarhan, Nonalcoholic fatty liver disease: Pathogenesis and the role of antioxidants, Nutr. Rev. 60(9) (2002) 289–293; https://doi.org/10.1301/002966402320387224 Search in Google Scholar

F. Item, S. Wueest, V. Lemos, S. Stein, F. C. Lucchini, R. Denzler, M. C. Fisser, T. D. Challa, E. Pirinen, Y. Kim, S. Hemmi, E. Gulbins, A. Gross, L. A. O’Reilly, M. Stoffel, J. Auwerx and D. Konrad, Fas cell surface death receptor controls hepatic lipid metabolism by regulating mitochondrial function, Nat. Commun. 8(1) (2017) Article ID 480 (10 pages); https://doi.org/10.1038/s41467-017-00566-9 Search in Google Scholar

L. Ding, W. Sun, M. Balaz, A. He, M. Klug, S. Wieland, R. Caiazzo, V. Raverdy, F. Pattou, P. Lefebvre, I. J. Lodhi, B. Staels, M. Heim and C. Wolfrum, Peroxisomal β-oxidation acts as a sensor for intracellular fatty acids and regulates lipolysis, Nat. Metab. 3(12) (2021) 1648–1661; https://doi.org/10.1038/s42255-021-00489-2 Search in Google Scholar

X. Zhou, L. L. Wang, W. J. Tang and B. Tang, Astragaloside IV inhibits protein tyrosine phosphatase 1B and improves insulin resistance in insulin-resistant HepG2 cells and triglyceride accumulation in oleic acid (OA)-treated HepG2 cells, J. Ethnopharmacol. 268(11) (2021) Article ID 113556; https://doi.org/10.1016/j.jep.2020.113556 Search in Google Scholar

J. L. Rains and S. K. Jain, Oxidative stress, insulin signaling, and diabetes, Free Radic. Biol. Med. 50(5) (2011) 567–575; https://doi.org/10.1016/j.freeradbiomed.2010.12.006 Search in Google Scholar

S. Vomund, A. Schäfer, M. J. Parnham, B. Brüne and A. Von Knethen, Nrf2, the master regulator of anti-oxidative responses, Int. J. Mol. Sci. 18(12) (2017) Article ID 2772 (19 pages); https://doi.org/10.3390/ijms18122772 Search in Google Scholar

S. Herzig and R. J. Shaw, AMPK: Guardian of metabolism and mitochondrial homeostasis, Nat. Rev. Mol. Cell Biol. 19(2) (2018) 121–135; https://doi.org/10.1038/nrm.2017.95 Search in Google Scholar

H. Yun, S. Park, M. J. Kim, W. K. Yang, D. U. Im, K. R. Yang, J. Hong, W. Choe, I. Kang, S. S. Kim and J. Ha, AMP-activated protein kinase mediates the antioxidant effects of resveratrol through regulation of the transcription factor FoxO1, FEBS J. 281(19) (2014) 4421–4438; https://doi.org/10.1111/febs.12949 Search in Google Scholar

Q. Lu, L. Yang, J.-J. Xiao, Q. Liu, L. Ni, J.-W. Hu, H. Yu, X. Wu and B.-F. Zhang, Empagliflozin attenuates the renal tubular ferroptosis in diabetic kidney disease through AMPK/NRF2 pathway, Free Radic. Biol. Med. 195 (2023) 89–102; https://doi.org/10.1016/j.freeradbiomed.2022.12.088 Search in Google Scholar

X. Li, D. Wu and T. Ye, Fibroblast growth factor 19 protects the heart from oxidative stress-induced diabetic cardiomyopathy via activation of AMPK/Nrf2/HO-1 pathway, Biochem. Biophys. Res. Commun. 502(1) (2018) 62–68; https://doi.org/10.1016/j.bbrc.2018.05.121 Search in Google Scholar

X. H. Zhou, L. Q. He, S. N. Zuo, Y. M. Zhang, D. Wan, C. M. Long, P. Huang, X. Wu, C. R. Wu, G. Liu and Y. Yin, Serine prevented high-fat diet–induced oxidative stress by activating AMPK and epigenetically modulating the expression of glutathione synthesis-related genes, Biochim. Biophys. Acta Mol. Basis Dis. 1864(2) (2018) 488–498; https://doi.org/10.1016/j.bbadis.2017.11.009 Search in Google Scholar

L. Li, W. X. Huang, S. K. Wang, K. Sun, W. Zhang, Y. Ding, L. Zhang, B. Tumen and L. Lang, Astragaloside IV attenuates acetaminophen-induced liver injuries in mice by activating the Nrf2 signaling pathway, Molecules 23(8) (2018) Article ID 2032 (9 pages); https://doi.org/10.3390/molecules23082032 Search in Google Scholar

N. Cheng, S. Chen, X. Liu, H. Zhao and W. Cao, Impact of Schisandra chinensis bee pollen on non-alcoholic fatty liver disease and gut microbiota in high-fat diet induced obese mice, Nutrients 11(2) (2019) Article ID 346 (16 pages); https://doi.org/10.3390/nu11020346 Search in Google Scholar

M. Martin, K. Rehani, R. S. Jope and S. M. Michalek, Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3, Nat. Immunol. 6(8) (2005) 777–784; https://doi.org/10.1038/ni1221 Search in Google Scholar

D. Sag, D. Carling, R. D. Stout and J. Suttles, Adenosine 5′-monophosphate-activated protein kinase promotes macrophage polarization to an anti-inflammatory functional phenotype, J. Immunol. 181(12) (2008) 8633–8641; https://doi.org/10.4049/jimmunol.181.12.8633 Search in Google Scholar

R. Wei, H. Liu, R. Chen, Y. Sheng and T. Liu, Astragaloside IV combating liver cirrhosis through the PI3K/Akt/mTOR signaling pathway, Exp. Ther. Med. 17(1) (2019) 393–397; https://doi.org/10.3892/etm.2018.6966 Spandidos Publications Search in Google Scholar

D. Qin, D. N. Ma, Z. G. Ren, X. D. Zhu, C. H. Wang, Y. C. Wang, B. Ye, M. Cao, D. Gao, Z. Tang and Z. Tang, Astragaloside IV inhibits metastasis in hepatoma cells through the suppression of epithelial-mesenchymal transition via the Akt/GSK-3β/β-catenin pathway, Oncol. Rep. 37(3) (2017) 1725–1735; https://doi.org/10.3892/or.2017.5389 Search in Google Scholar

X. Qu, H. Gao, L. Tao, Y. Zhang, J. Zhai, J. Sun, Y. Song and S. Zhang, Astragaloside IV protects against cisplatin-induced liver and kidney injury via autophagy-mediated inhibition of NLRP3 in rats, J. Toxicol. Sci. 44(3) (2019) 167–175; https://doi.org/10.2131/jts.44.167 Search in Google Scholar

J. R. Ham, H.-I. Lee, R.-Y. Choi, M.-O. Sim, K.-I. Seo and M.-K. Lee, Anti-steatotic and anti-inflammatory roles of syringic acid in high-fat diet-induced obese mice, Food Funct. 7(2) (2016) 689–697; https://doi.org/10.1039/C5FO01411A Search in Google Scholar

G. Baffy, Kupffer cells in non-alcoholic fatty liver disease: The emerging view, J. Hepatol. 51(1) (2009) 212–223; https://doi.org/10.1016/j.jhep.2009.03.008 Search in Google Scholar

S. Saravanan, V. I. Islam, N. P. Babu, P. Pandikumar, K. Thirugnanasambantham, M. Chellappandian, C. S. Raj, M. G. Paulraj and S. Ignacimuthu, Swertiamarin attenuates inflammation mediators via modulating NF-κB/IκB and JAK2/STAT3 transcription factors in adjuvant induced arthritis, Eur. J. Pharm. Sci. 56 (2014) 70–86; https://doi.org/10.1016/j.ejps.2014.02.005 Search in Google Scholar

Z. L. Bian, Y. Peng, Z. You, Q. Wang, Q. Miao, Y. Liu, X. Han, D. Qiu, Z. Li and X. Ma, CCN1 expression in hepatocytes contributes to macrophage infiltration in nonalcoholic fatty liver disease in mice, J. Lipid Res. 54(1) (2013) 44–54; https://doi.org/10.1194/jlr.M026013 Search in Google Scholar

E. Radi, P. Formichi, C. Battisti and A. Federico, Apoptosis and oxidative stress in neurodegenerative diseases, J. Alzheimer’s Dis. 42(Suppl. 3) (2014) S125–S152; https://doi.org/10.3233/JAD-132738 Search in Google Scholar

K. J. Campbell and S. W. G. Tait, Targeting BCL-2 regulated apoptosis in cancer, Open Biol. 8(5) (2018) Article ID 180002 (11 pages); https://doi.org/10.1098/rsob.180002 Search in Google Scholar

C. A. Gregory, H. Singh, A. S. Perry and D. J. Prockop, Deletion of the BH1 domain of Bcl-2 accelerates apoptosis by acting in a dominant negative fashion, J. Biol. Chem. 278(22) (2003) 19732–19742; https://doi.org/10.1074/jbc.M300373200 Search in Google Scholar

C. Cárdenas, R. A. Miller, I. Smith, T. Bui, J. Molgó, M. Müller, H. Vais, K.-H. Cheung, J. Yang, I. Parker, C. B. Thompson, M. J. Birnbaum, K. R. Hallows and J. K. Foskett, Essential regulation of cell bioenergetics by constitutive InsP₃ receptor Ca²⁺ transfer to mitochondria, Cell 142(2) (2010) 270–283; https://doi.org/10.1016/j.cell.2010.06.007 Search in Google Scholar

J. A. Martinez, Z. Zhang, S. I. Svetlov, R. L. Hayes, K. K. Wang and S. F. Larner, Calpain and caspase processing of caspase-12 contribute to the ER stress-induced cell death pathway in differentiated PC12 cells, Apoptosis 15(12) (2010) 1480–1493; https://doi.org/10.1007/s10495-010-0531-7 Search in Google Scholar

C. J. Hanson, M. D. Bootman, C. W. Distelhorst, R. J. H. Wojcikiewicz and H. L. Roderick, Bcl-2 suppresses Ca²⁺ release through inositol 1,4,5-trisphosphate receptors and inhibits Ca²⁺ uptake by mitochondria without affecting ER calcium store content, Cell Calcium 44(3) (2008) 324–338; https://doi.org/10.1016/j.ceca.2008.01.003 Search in Google Scholar

X.-Y. Liang, F.-F. Hong and S.-L. Yang, Astragaloside IV alleviates liver inflammation, oxidative stress and apoptosis to protect against experimental non-alcoholic fatty liver disease, Diabetes Metab. Syndr. Obes. 14 (2021) 1871–1883; https://doi.org/10.2147/DMSO.S304817 Search in Google Scholar

X. Liu, W. Chu, S. Shang, L. Ma, C. Jiang, Y. Ding, J. Wang, S. Zhang and B. Shao, Preliminary study on the anti-apoptotic mechanism of astragaloside IV on radiation-induced brain cells, Int. J. Immunopathol. Pharmacol. 34 (2020) Article ID 2058738420954594; https://doi.org/10.1177/2058738420954594 Search in Google Scholar

Z. Liu, Z. Zhou, P. Ai, C. Zhang, J. Chen and Y. Wang, Astragaloside IV attenuates ferroptosis after subarachnoid hemorrhage via Nrf2/HO-1 signaling pathway, Front. Pharmacol. 13 (2022) Article ID 924826 (13 pages); https://doi.org/10.3389/fphar.2022.924826 Search in Google Scholar

Z. Qi, P. Zhang, L. Yang, P. Song, J. Zhao, H. Wang, Y. Zhao and L. Cao, Astragaloside IV alleviates doxorubicin-induced cardiotoxicity by inhibiting cardiomyocyte pyroptosis through the SIRT1/NLRP3 pathway, Am. J. Chin. Med. 52(2) (2024) 453–469; https://doi.org/10.1142/S0192415X24500198 Search in Google Scholar

X. Meng, G. Zhang, H. Cao, D. Yu, X. Fang, W. M. de Vos and H. Wu, Gut dysbacteriosis and intestinal disease: Mechanism and treatment, J. Appl. Microbiol. 129(4) (2020) 787–805; https://doi.org/10.1111/jam.14661 Search in Google Scholar

L. L. Deng, Astragaloside IV as potential antioxidant against diabetic ketoacidosis in juvenile mice through activating JNK/Nrf2 signaling pathway, Arch. Med. Res. 51(7) (2020) 654–663; https://doi.org/10.1016/j.arcmed.2020.06.013 Search in Google Scholar

D. M. Chopyk and A. Grakoui, Contribution of the intestinal microbiome and gut barrier to hepatic disorders, Gastroenterology 159(3) (2020) 849–863; https://doi.org/10.1053/j.gastro.2020.04.077 Search in Google Scholar

J. W. Jung, F. Wang, A. Turk, J. S. Park, H. Ma, Y. Ma, H.-R. Noh, G. Sui, D. S. Shin, M. K. Lee and Y. S. Roh, Zaluzanin C alleviates inflammation and lipid accumulation in Kupffer cells and hepatocytes by regulating mitochondrial ROS, Molecules 28(22) (2023) Article ID 7484; https://doi.org/10.3390/molecules28227484 Search in Google Scholar

J. Fang, C.-H. Yu, X.-J. Li, J.-M. Yao, Z.-Y. Fang, S.-H. Yoon and W.-Y. Yu, Gut dysbiosis in nonalcoholic fatty liver disease: Pathogenesis, diagnosis, and therapeutic implications, Front. Cell. Infect. Microbiol. 12 (2022) Article ID 997018 (12 pages); https://doi.org/10.3389/fcimb.2022.997018 Search in Google Scholar

D. Zhang, X. Hao, L. Xu, J. Cui, L. Xue and Z. Tian, Intestinal flora imbalance promotes alcohol-induced liver fibrosis by the TGFβ/Smad signaling pathway in mice, Oncol. Lett. 14(4) (2017) 4511–4516; https://doi.org/10.3892/ol.2017.6762 Search in Google Scholar

A. Wieland, D. N. Frank, B. Harnke and K. Bambha, Systematic review: Microbial dysbiosis and nonalcoholic fatty liver disease, Aliment. Pharmacol. Ther. 42(9) (2015) 1051–1063; https://doi.org/10.1111/apt.13376 Search in Google Scholar

L. Xue, Z. Deng, W. Luo, X. He and Y. Chen, Effect of fecal microbiota transplantation on non-alcoholic fatty liver disease: A randomized clinical trial, Front. Cell. Infect. Microbiol. 12 (2022) Article ID 759306 (10 pages); https://doi.org/10.3389/fcimb.2022.759306 Search in Google Scholar

V. Kaden-Volynets, M. Basic, U. Neumann, D. Pretz, A. Rings, A. Bleich and S. C. Bischoff, Lack of liver steatosis in germ-free mice following hypercaloric diets, Eur. J. Nutr. 58 (2019) 1933–1945; https://doi.org/10.1007/s00394-018-1748-4 Search in Google Scholar

J. Fu, M. J. Bonder, M. C. Cenit, E. F. Tigchelaar, A. Maatman, J. A. M. Dekens, E. Brandsma, J. Marczynska, F. Imhann, R. K. Weersma, L. Franke, T. W. Poon, R. J. Xavier, D. Gevers, M. H. Hofker, C. Wijmenga and A. Zhernakova, The gut microbiome contributes to a substantial proportion of the variation in blood lipids, Circ. Res. 117(9) (2015) 817–824; https://doi.org/10.1161/CIRCRESAHA.115.306807 Search in Google Scholar

Z. Li, E. Hu, F. Zheng, S. Wang, W. Zhang, J. Luo, T. Tang, Q. Huang and Y. Wang, The effects of astragaloside IV on gut microbiota and serum metabolism in a mice model of intracerebral hemorrhage, Phytomedicine 121 (2023) Article ID 155086; https://doi.org/10.1016/j.phymed.2023.155086 Search in Google Scholar

X. Q. Du, L. P. Shi, Z. W. Chen, J. Y. Hu, B. Zuo, Y. Xiong and W. F. Cao, Astragaloside IV ameliorates isoprenaline-induced cardiac fibrosis in mice via modulating gut microbiota and fecal metabolites, Front. Cell. Infect. Microbiol. 12 (2022) Article ID 836150 (11 pages); https://doi.org/10.3389/fcimb.2022.836150 Search in Google Scholar

T. Yang, S. Xie, L. Cao, M. Li, L. Ding, L. Wang, S. Pang, Z. Wang and L. Geng, Astragaloside IV modulates gut macrophages M1/M2 polarization by reshaping gut microbiota and short chain fatty acids in sepsis, Shock 61(1) (2024) 120–131; https://doi.org/10.1097/SHK.0000000000002262 Search in Google Scholar

P. Golabi, H. Bush, M. Stepanova, C. T. Locklear, J. P. Weston, Z. Goodman, I. M. Younossi and Z. M. Younossi, Liver transplantation (LT) for cryptogenic cirrhosis (CC) and nonalcoholic steato-hepatitis (NASH) cirrhosis: Data from the Scientific Registry of Transplant Recipients (SRTR): 1994 to 2016, Medicine 97(31) (2018) e11518; https://doi.org/10.1097/MD.0000000000011518 Search in Google Scholar

S. A. Harrison, M. R. Bashir, C. D. Guy, R. Zhou, C. A. Moylan, J. P. Frias, N. Alkhouri, M. B. Bansal, S. Baum, B. A. Neuschwander-Tetri, R. Taub and S. E. Moussa, Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: A multicentre, randomised, double-blind, placebo- controlled, phase 2 trial, Lancet 394(10213) (2019) 2012–2024; https://doi.org/10.1016/S0140-6736(19)32517-6 Search in Google Scholar

A. R. Saran, S. Dave and A. Zarrinpar, Circadian rhythms in the pathogenesis and treatment of fatty liver disease, Gastroenterology 158(7) (2020) 1948–1966.e1; https://doi.org/10.1053/j.gastro.2020.01.050 Search in Google Scholar

S. M. Francque, P. Bedossa, V. Ratziu, Q. M. Anstee, E. Bugianesi, A. J. Sanyal, R. Loomba, S. A. Harrison, R. Balabanska, L. Mateva, N. Lanthier, N. Alkhouri, C. Moreno, J. M. Schattenberg, D. Stefanova-Petrova, L. Vonghia, R. Rouzier, M. Guillaume, A. Hodge, M. Romero-Gómez, P. Huot-Marchand, M. Baudin, M.-P. Richard, J.-L. Abitbol, P. Broqua, J.-L. Junien and M. F. Abdelmalek, A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH, N. Engl. J. Med. 385(17) (2021) 1547–1558; https://doi.org/10.1056/NEJMoa2036205 Search in Google Scholar

P. N. Newsome, K. Buchholtz, K. Cusi, M. Linder, T. Okanoue, V. Ratziu, A. J. Sanyal, A.-S. Sejling and S. A. Harrison, A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steato-hepatitis, N. Engl. J. Med. 384(12) (2021) 1113–1124; https://doi.org/10.1056/NEJMoa2028395 Search in Google Scholar

N. Alkhouri, R. Herring, H. Kabler, Z. Kayali, T. Hassanein, A. Kohli, R. S. Huss, Y. Zhu, A. N. Billin, L. H. Damgaard, K. Buchholtz, M. S. Kjær, C. Balendran, R. P. Myers, R. Loomba and M. Noureddin, Safety and efficacy of combination therapy with semaglutide, cilofexor and firsocostat in patients with non-alcoholic steatohepatitis: A randomised, open-label phase II trial, J. Hepatol. 77(3) (2022) 607–618; https://doi.org/10.1016/j.jhep.2022.04.003 Search in Google Scholar

R. Loomba, E. J. Lawitz, J. P. Frias, G. Ortiz-Lasanta, L. Johansson, B. B. Franey, L. Morrow, M. Rosenstock, C. L. Hartsfield, C.-Y. Chen, L. Tseng, R. W. Charlton, H. Mansbach and M. Margalit, Safety, pharmacokinetics, and pharmacodynamics of pegozafermin in patients with non-alcoholic steatohepatitis: A randomised, double-blind, placebo-controlled, phase 1b/2a multiple-ascending--dose study, Lancet Gastroenterol. Hepatol. 8(2) (2023) 120–132; https://doi.org/10.1016/S2468-1253(22)00347-8 Search in Google Scholar

S. A. Harrison, J. P. Frias, G. Neff, G. A. Abrams, K. J. Lucas, W. Sanchez, S. Gogia, M. Y. Sheikh, C. Behling, P. Bedossa, L. Shao, D. Chan, E. Fong, B. de Temple, R. Shringarpure, E. J. Tillman, T. Rolph, A. Cheng and K. Yale, Safety and efficacy of once-weekly efruxifermin versus placebo in non-alcoholic steatohepatitis (HARMONY): A multicentre, randomised, double-blind, placebo--controlled, phase 2b trial, Lancet Gastroenterol. Hepatol. 8(12) (2023) 1080–1093; https://doi.org/10.1016/S2468-1253(23)00272-8 Search in Google Scholar

J. Zhou, L. R. Waskowicz, A. Lim, X.-H. Liao, B. Lian, H. Masamune, S. Refetoff, B. Tran, D. D. Koeberl and P. M. Yen, A liver-specific thyromimetic, VK2809, decreases hepatosteatosis in glycogen storage disease type Ia, Thyroid 29(8) (2019) 1158–1167; https://doi.org/10.1089/thy.2019.0007 Search in Google Scholar

S. A. Harrison, R. Taub, G. W. Neff, K. J. Lucas, D. Labriola, S. E. Moussa, N. Alkhouri and M. R. Bashir, Resmetirom for nonalcoholic fatty liver disease: A randomized, double-blind, placebo-controlled phase 3 trial, Nat. Med. 29(11) (2023) 2919–2928; https://doi.org/10.1038/s41591-023-02603-1 Search in Google Scholar

Chinese Pharmacopoeia Commission, Pharmacopoeia of the People’s Republic of China, 11th ed., China Medical Science Press, Beijing 2020. Search in Google Scholar

Langue:
Anglais
Périodicité:
4 fois par an
Sujets de la revue:
Pharmacie, Pharmacie, autres