Butyrolactone I blocks the transition of acute kidney injury to chronic kidney disease in mice by targeting JAK1

Zijun Zhang , Ziming Zhao , Changxing Qi , Xiaotian Zhang , Yang Xiao , Chengjuan Chen , Yu Zou , Xia Chen , Lianghu Gu , Jianzheng Huang , Kun Huang , Ming Xiang , Tiantai Zhang , Qingyi Tong , Yonghui Zhang

MedComm ›› 2025, Vol. 6 ›› Issue (2) : e70064

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (2) : e70064 DOI: 10.1002/mco2.70064
ORIGINAL ARTICLE

Butyrolactone I blocks the transition of acute kidney injury to chronic kidney disease in mice by targeting JAK1

Author information +
History +
PDF

Abstract

Chronic kidney disease (CKD) is a disease that affects more than 850 million people. Acute kidney injury (AKI) is a common cause of CKD, and blocking the AKI–CKD transition shows promising therapeutic potential. Herein, we found that butyrolactone I (BLI), a natural product, exerts significant nephroprotective effects, including maintenance of kidney function, inhibition of inflammatory response, and prevention of fibrosis, in both folic acid- and ureteral obstruction-induced AKI–CKD transition mouse models. Notably, BLI showed greater blood urea nitrogen reduction and anti-inflammatory effects than telmisartan. Bioinformatics analysis and target confirmation assays suggested that BLI directly binds to JAK1, and kinase inhibition assay confirmed it is a potent JAK1inhibitor with an IC50 of 0.376 µM. Experiments in JAK1-knockdown mice also proved that BLI targets JAK1 to work. Furthermore, BLI demonstrated nephroprotective effects and safety comparable to ivarmacitinib, the well-known JAK1 inhibitor. Mechanistically, BLI targets JAK1 and inhibits its phosphorylation and JAK-STAT activation, subsequently regulating the downstream signaling pathways to inhibit reactive oxygen species production, inflammation, and ferroptosis, thereby preventing the occurrence of kidney fibrosis and blocking the AKI–CKD transition process. This study demonstrates for the first time that BLI is a JAK1 inhibitor and a promising candidate for delaying CKD progression, which warrants further investigation.

Keywords

AKI–CKD transition / butyrolactone I / ferroptosis / JAK1

Cite this article

Download citation ▾
Zijun Zhang, Ziming Zhao, Changxing Qi, Xiaotian Zhang, Yang Xiao, Chengjuan Chen, Yu Zou, Xia Chen, Lianghu Gu, Jianzheng Huang, Kun Huang, Ming Xiang, Tiantai Zhang, Qingyi Tong, Yonghui Zhang. Butyrolactone I blocks the transition of acute kidney injury to chronic kidney disease in mice by targeting JAK1. MedComm, 2025, 6(2): e70064 DOI:10.1002/mco2.70064

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Kalantar-Zadeh K, Jafar TH, Nitsch D, Neuen BL, Perkovic V. Chronic kidney disease. Lancet. 2021; 398(10302): 786-802.

[2]

Kellum JA, Romagnani P, Ashuntantang G, Ronco C, Zarbock A, Anders H-J. Acute kidney injury. Nat Rev Dis Primers. 2021; 7(1): 52.

[3]

Levin A, Ahmed SB, Carrero JJ, et al. Executive summary of the KDIGO 2024 clinical practice guideline for the evaluation and management of chronic kidney disease: known knowns and known unknowns. Kidney Int. 2024; 105(4): 684-701.

[4]

Sundström J, Bodegard J, Bollmann A, et al. Prevalence, outcomes, and cost of chronic kidney disease in a contemporary population of 2·4 million patients from 11 countries: the CaReMe CKD study. Lancet Regional Health—Europe. 2022; 20: 100438.

[5]

Lv J-C, Zhang L-X. Prevalence and disease burden of chronic kidney disease. In: Liu B-C, Lan H-Y, Lv L-L, eds. Renal Fibrosis: Mechanisms and Therapies. Springer; 2019: 3-15.

[6]

Bikbov B, Purcell C, Levey AS, et al. Global, regional, and national burden of chronic kidney disease, 1990–2017: a systematic analysis for the global burden of disease study 2017. Lancet (London, England). 2020; 395(10225): 709-733.

[7]

Kovesdy CP. Epidemiology of chronic kidney disease: an update 2022. Kidney Int Suppl. 2022; 12(1): 7-11.

[8]

Zhang X, Agborbesong E, Li X. The role of mitochondria in acute kidney injury and chronic kidney disease and its therapeutic potential. Int J Mol Sci. 2021; 22(20): 11253.

[9]

Guo RZ, Duan JY, Pan SK, et al. The road from AKI to CKD: molecular mechanisms and therapeutic targets of ferroptosis. Cell Death Dis. 2023; 14(7): 426.

[10]

Niu X, Dahse HM, Menzel KD, et al. Butyrolactone I derivatives from Aspergillus terreus carrying an unusual sulfate moiety. J Nat Prod. 2008; 71(4): 689-692.

[11]

Suzuki M, Hosaka Y, Matsushima H, Goto T, Kitamura T, Kawabe K. Butyrolactone I induces cyclin B1 and causes G2/M arrest and skipping of mitosis in human prostate cell lines. Cancer Lett. 1999; 138(1-2): 121-130.

[12]

Tsou YC, Wang HH, Hsieh CC, et al. Down-regulation of BNIP3 by olomoucine, a CDK inhibitor, reduces LPS-and NO-induced cell death in BV2 microglial cells. Neurosci Lett. 2016; 628: 186-193.

[13]

Nie Y, Yang J, Zhou L, et al. Marine fungal metabolite butyrolactone I prevents cognitive deficits by relieving inflammation and intestinal microbiota imbalance on aluminum trichloride-injured zebrafish. J Neuroinflammation. 2022; 19(1): 39.

[14]

Jhou RS, Sun KH, Sun GH, et al. Inhibition of cyclin-dependent kinases by olomoucine and roscovitine reduces lipopolysaccharide-induced inflammatory responses via down-regulation of nuclear factor kappaB. Cell Prolif. 2009; 42(2): 141-149.

[15]

Tian-Qi C, Yan-Fang D, Yan-Yan W, Yong-Hui Z. Butyrolactone I attenuates inflammation in murine NASH by inhibiting the NF-κB signaling pathway. Biochem Biophys Res Commun. 2022; 626: 167-174.

[16]

Wu W, Liu L, Zhu H, et al. Butyrolactone-I, an efficient α-glucosidase inhibitor, improves type 2 diabetes with potent TNF-α–lowering properties through modulating gut microbiota in db/db mice. FASEB J. 2019; 33(11): 12616-12629.

[17]

Hong BH, He JL, Fan CC, et al. Synthesis and biological evaluation of analogues of butyrolactone I as PTP1B Inhibitors. Mar Drugs. 2020; 18(11): 526.

[18]

Naeem A, Hu PY, Yang M, et al. Natural products as anticancer agents: current status and future perspectives. Molecules. 2022; 27(23): 8367.

[19]

Atanasov AG, Zotchev SB, Dirsch VM, Supuran CT, Taskforce INPS. Natural products in drug discovery: advances and opportunities. Nat Rev Drug Discovery. 2021; 20(3): 200-216.

[20]

Newman DJ, Cragg GM. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J Nat Prod. 2020; 83(3): 770-803.

[21]

Pavkovic M, Pantano L, Gerlach CV, et al. Multi omics analysis of fibrotic kidneys in two mouse models. Sci Data. 2019; 6(1): 92.

[22]

Kim EH, Lee CH, Hyun BH, et al. Quantitative trait loci for glomerulosclerosis, kidney weight and body weight in the focal glomerulosclerosis mouse model. Exp Anim. 2005; 54(4): 319-325.

[23]

Han X, Sun Z. Adult mouse kidney stem cells orchestrate the de novo assembly of a nephron via Sirt2-modulated canonical Wnt/β-catenin signaling. Adv Sci (Weinh). 2022; 9(15): e2104034.

[24]

Pan H-C, Yang S-Y, Chiou TT-Y, et al. Comparative accuracy of biomarkers for the prediction of hospital-acquired acute kidney injury: a systematic review and meta-analysis. Critical Care. 2022; 26(1): 349.

[25]

Kadatane SP, Satariano M, Massey M, Mongan K, Raina R. The role of inflammation in CKD. Cells. 2023; 12(12): 1581.

[26]

Xie Y, Hu X, Li S, et al. Pharmacological targeting macrophage phenotype via gut-kidney axis ameliorates renal fibrosis in mice. Pharmacol Res. 2022; 178:106161.

[27]

André C, Bodeau S, Kamel S, Bennis Y, Caillard P. The AKI-to-CKD transition: the role of uremic toxins. Int J Mol Sci. 2023; 24(22): 16152.

[28]

Kang HM, Ahn SH, Choi P, et al. Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development. Nat Med. 2014; 21(1): 37-46.

[29]

Li J, Cao F, Yin H-l, et al. Ferroptosis: past, present and future. Cell Death Dis. 2020; 11(2): 88.

[30]

Martin-Sanchez D, Ruiz-Andres O, Poveda J, et al. Ferroptosis, but not necroptosis, is important in nephrotoxic folic acid–induced AKI. J Am Soc Nephrol. 2017; 28(1): 218-229.

[31]

Zhang B, Chen X, Ru F, et al. Liproxstatin-1 attenuates unilateral ureteral obstruction-induced renal fibrosis by inhibiting renal tubular epithelial cells ferroptosis. Cell Death Dis. 2021; 12(9): 843.

[32]

Ta N, Qu C, Wu H, et al. Mitochondrial outer membrane protein FUNDC2 promotes ferroptosis and contributes to doxorubicin-induced cardiomyopathy. Proc Natl Acad Sci USA. 2022; 119(36): e2117396119.

[33]

Fuhrmann DC, Mondorf A, Beifuß J, Jung M, Brüne B. Hypoxia inhibits ferritinophagy, increases mitochondrial ferritin, and protects from ferroptosis. Redox Biol. 2020; 36:101670.

[34]

Shi P, Li M, Song C, et al. Neutrophil-like cell membrane-coated siRNA of lncRNA AABR07017145.1 therapy for cardiac hypertrophy via inhibiting ferroptosis of CMECs. Mol Therapy—Nucleic Acids. 2022; 27: 16-36. 10.1016/j.omtn.2021.10.024

[35]

Wang H, Yu X, Liu D, et al. VDR activation attenuates renal tubular epithelial cell ferroptosis by regulating Nrf2/HO-1 signaling pathway in diabetic nephropathy. Adv Sci (Weinh). 2024; 11(10): e2305563.

[36]

Wang L, Liu Y, Du T, et al. ATF3 promotes erastin-induced ferroptosis by suppressing system Xc–. Cell Death Differ. 2019; 27(2): 662-675.

[37]

Gao M, Yi J, Zhu J, et al. Role of mitochondria in ferroptosis. Mol Cell. 2019; 73(2): 354-363. e3.

[38]

Luo P, Liu D, Zhang Q, et al. Celastrol induces ferroptosis in activated HSCs to ameliorate hepatic fibrosis via targeting peroxiredoxins and HO-1. Acta Pharm Sin B. 2022; 12(5): 2300-2314.

[39]

Lomenick B, Hao R, Jonai N, et al. Target identification using drug affinity responsive target stability (DARTS). Proc Natl Acad Sci USA. 2009; 106(51): 21984-21989.

[40]

Chen C, Yin Y, Shi G, et al. A highly selective JAK3 inhibitor is developed for treating rheumatoid arthritis by suppressing γc cytokine–related JAK-STAT signal. Sci Adv. 2022; 8(33): eabo4363.

[41]

Chen Y, Fang Z-M, Yi X, Wei X, Jiang D-S. The interaction between ferroptosis and inflammatory signaling pathways. Cell Death Dis. 2023; 14(3): 205.

[42]

Nemeth E, Ganz T. Hepcidin and iron in health and disease. Annu Rev Med. 2023; 74: 261-277.

[43]

Saad HKM, Abd Rahman AA, Ab Ghani AS, et al. Activation of STAT and SMAD signaling induces hepcidin Re-expression as a therapeutic target for β-thalassemia patients. Biomedicines. 2022; 10(1): 189.

[44]

Rah B, Farhat NM, Hamad M, Muhammad JS. JAK/STAT signaling and cellular iron metabolism in hepatocellular carcinoma: therapeutic implications. Clin Exp Med. 2023; 23(7): 3147-3157.

[45]

Yu X, Zhu D, Luo B, Kou W, Cheng Y, Zhu Y. IFNγ enhances ferroptosis by increasing JAK-STAT pathway activation to suppress SLCA711 expression in adrenocortical carcinoma. Oncol Rep. 2022; 47(5): 97.

[46]

Wei TT, Zhang MY, Zheng XH, et al. Interferon-γ induces retinal pigment epithelial cell ferroptosis by a JAK1-2/STAT1/SLC7A11 signaling pathway in age-related macular degeneration. FEBS J. 2021; 289(7): 1968-1983.

[47]

Braña MF, García ML, López B, et al. Synthesis and biological evaluation of analogues of butyrolactone I and molecular model of its interaction with CDK2. Org Biomol Chem. 2004; 2(13): 1864-1871.

[48]

Ahn S, Jang DM, Park SC, et al. Cyclin-dependent kinase 5 inhibitor butyrolactone I elicits a partial agonist activity of peroxisome proliferator-activated receptor γ. Biomolecules. 2020; 10(2): 275.

[49]

Brosius FC, He JC. JAK inhibition and progressive kidney disease. Curr Opin Nephrol Hypertens. 2015; 24(1): 88-95.

[50]

Chen D, Liu Y, Chen J, et al. JAK/STAT pathway promotes the progression of diabetic kidney disease via autophagy in podocytes. Eur J Pharmacol. 2021; 902: 174121.

[51]

Liu Y, Wang W, Zhang J, Gao S, Xu T, Yin Y. JAK/STAT signaling in diabetic kidney disease. Front Cell Dev Biol. 2023; 11: 1233259.

[52]

Chen BL, Zhong J, Li XL, et al. Efficacy and safety of ivarmacitinib in patients with moderate-to-severe, active, ulcerative colitis: a phase II study. Gastroenterology. 2022; 163(6): 1555-1568.

[53]

Yao M, Qin S, Xiong J, et al. Oroxylin A ameliorates AKI-to-CKD transition through maintaining PPARα-BNIP3 signaling-mediated mitochondrial homeostasis. Front Pharmacol. 2022; 13: 935937.

[54]

Yan LJ. Folic acid-induced animal model of kidney disease. Anim Models Exp Med. 2021; 4(4): 329-342.

[55]

Fu Y, Tang CY, Cai J, Chen GC, Zhang DS, Dong Z. Rodent models of AKI-CKD transition. Am J Physiol Renal Physiol. 2018; 315(4): F1098-F1106.

[56]

Feng D, Ngov C, Henley N, Boufaied N, Gerarduzzi C. Characterization of matricellular protein expression signatures in mechanistically diverse mouse models of kidney injury. Sci Rep. 2019; 9(1): 16736.

[57]

Yan LJ. Folic acid-induced animal model of kidney disease. Animal Model Exp Med. 2021; 4(4): 329-342.

[58]

Ruiz-Ortega M, Rayego-Mateos S, Lamas S, Ortiz A, Rodrigues-Diez RR. Targeting the progression of chronic kidney disease. Nat Rev Nephrol. 2020; 16(5): 269-288.

[59]

Tuttle KR, Brosius FC, Adler SG, et al. JAK1/JAK2 inhibition by baricitinib in diabetic kidney disease: results from a Phase 2 randomized controlled clinical trial. Nephrol Dial Transplant. 2018; 33(11): 1950-1959.

[60]

Lee PL, Beutler E. Regulation of hepcidin and iron-overload disease. Annu Rev Pathol. 2009; 4: 489-515.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

194

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/