Generic placeholder image

Current Molecular Medicine

Editor-in-Chief

ISSN (Print): 1566-5240
ISSN (Online): 1875-5666

Research Article

Crocetin Suppresses Uterine Ischemia/Reperfusion-induced Inflammation and Apoptosis through the Nrf-2/HO-1 Pathway

Author(s): Shu Peng, Yong Wu and Ying Wang*

Volume 23, Issue 6, 2023

Published on: 17 August, 2022

Page: [527 - 535] Pages: 9

DOI: 10.2174/1566524022666220520141943

Price: $65

Abstract

Background: Uterine ischemia/reperfusion (I/R) injury often occurs during many complex surgical procedures, such as uterus transplantation, cesarean, and myomectomy, which may lead to the loss of uterine function and failure of the operation. Crocetin (CRO), as one of the major active constituents from saffron extract, shows protective effects against reactive oxygen species, inflammation, and apoptosis. However, the role of CRO in protecting the uterus against I/R-induced injury has never been investigated. This study aims to clarify the protective role of CRO against I/R injury and the underlying mechanisms.

Materials and Methods: Sprague-Dawley rats were randomly divided into five groups: the control group, I/R group, 20 mg/kg CRO-treated I/R group, 40 mg/kg CRO-treated I/R group, and 80 mg/kg CRO-treated I/R group. Rats were given daily gavages with different doses of CRO or vehicle for five consecutive days. The rat uterine I/R model was created by routine method with 1h ischemia and 3h reperfusion. The serum and uterine tissues were collected, the changes in malondialdehyde (MDA) level and superoxide dismutase (SOD) activity, the mRNA and protein levels of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α and IL-10, the protein levels of B-cell chronic lymphocytic leukemia/lymphoma (Bcl)-2, Bcl-2-associated X protein (Bax), caspase-3, nuclear factor erythroid 2-related factor (Nrf)-2, and heme oxygenase (HO)-1, were measured. The histological changes were examined by HE staining. The number of apoptotic cells was analyzed by flow cytometry.

Results: Uterine I/R significantly induced MDA level, suppressed SOD activity, upregulated levels of pro-inflammatory cytokines, down-regulated level of the antiinflammatory cytokine, induced caspase-3-dependent apoptosis, activated the protein expression of Nrf-2 and HO-1, and caused uterine damage. However, pre-administration of CRO effectively reversed I/R-induced above changes and further enhanced Nrf-2/HO- 1 activation in a dose-dependent manner.

Conclusion: Pre-administration of CRO effectively alleviates I/R-induced oxidative stress, inflammation, apoptosis, and tissue injury probably through activating the Nrf- 2/HO-1 pathway, suggesting a protective role of CRO in I/R-induced uterus injury.

Keywords: Crocetin, ischemia/reperfusion, inflammatory response, apoptosis, uterus, UTx.

[1]
Brännström M, Dahm Kähler P, Greite R, Mölne J, Díaz-García C, Tullius SG. Uterus transplantation: A rapidly expanding field. Transplantation 2018; 102(4): 569-77.
[http://dx.doi.org/10.1097/TP.0000000000002035] [PMID: 29210893]
[2]
Brännström M. Womb transplants with live births: An update and the future. Expert Opin Biol Ther 2017; 17(9): 1105-12.
[http://dx.doi.org/10.1080/14712598.2017.1347633] [PMID: 28683576]
[3]
Brännström M. Uterus transplantation: Transition from experimental to clinical procedure. Minerva Ginecol 2019; 71(6): 460-6.
[PMID: 32064828]
[4]
Zitkute V, Kvietkauskas M, Leber B, Strupas K, Stiegler P, Schemmer P. Ischemia and reperfusion injury in uterus transplantation: A comprehensive review. Transplant Rev (Orlando) 2020; 34(3): 100550.
[http://dx.doi.org/10.1016/j.trre.2020.100550] [PMID: 32498979]
[5]
Yu H, Kalogeris T, Korthuis RJ. Reactive species-induced microvascular dysfunction in ischemia/reperfusion. Free Radic Biol Med 2019; 135: 182-97.
[http://dx.doi.org/10.1016/j.freeradbiomed.2019.02.031] [PMID: 30849489]
[6]
Hirao H, Dery KJ, Kageyama S, Nakamura K, Kupiec-Weglinski JW. Heme Oxygenase-1 in liver transplant ischemia-reperfusion injury: From bench-to-bedside. Free Radic Biol Med 2020; 157: 75-82.
[http://dx.doi.org/10.1016/j.freeradbiomed.2020.02.012] [PMID: 32084514]
[7]
Corona D, Ekser B, Gioco R, et al. Heme-oxygenase and kidney transplantation: A potential for target therapy? Biomolecules 2020; 10(6): 840.
[http://dx.doi.org/10.3390/biom10060840] [PMID: 32486245]
[8]
Shah HM, Jain AS, Joshi SV, Kharkar PS. Crocetin and related oxygen diffusion-enhancing compounds: Review of chemical synthesis, pharmacology, clinical development, and novel therapeutic applications. Drug Dev Res 2021; 82(7): 883-95.
[http://dx.doi.org/10.1002/ddr.21814] [PMID: 33817811]
[9]
Guo ZL, Li MX, Li XL, et al. Crocetin: A systematic review. Front Pharmacol 2022; 12: 745683.
[http://dx.doi.org/10.3389/fphar.2021.745683] [PMID: 35095483]
[10]
José Bagur M, Alonso Salinas GL, Jiménez-Monreal AM, et al. Saffron: An old medicinal plant and a potential novel functional food. Molecules 2017; 23(1): 30.
[http://dx.doi.org/10.3390/molecules23010030] [PMID: 29295497]
[11]
Boskabady MH, Farkhondeh T. Antiinflammatory, antioxidant, and immunomodulatory effects of Crocus sativus L. and its main constituents. Phytother Res 2016; 30(7): 1072-94.
[http://dx.doi.org/10.1002/ptr.5622] [PMID: 27098287]
[12]
Aydogan Kirmizi D, Baser E, Doganyigit Z. The activation of cannabinoid type-2 receptor with JWH-133 protects uterine ischemia/reperfusion-induced damage. Pharmacology 2021; 106(1-2): 106-13.
[http://dx.doi.org/10.1159/000511457] [PMID: 33105141]
[13]
Brännström M, Dahm-Kähler P. Uterus transplantation and fertility preservation. Best Pract Res Clin Obstet Gynaecol 2019; 55: 109-16.
[http://dx.doi.org/10.1016/j.bpobgyn.2018.12.006] [PMID: 30711374]
[14]
Lavoué V, Vigneau C, Duros S, et al. Which donor for uterus transplants: brain-dead donor or living donor? A systematic review. Transplantation 2017; 101(2): 267-73.
[http://dx.doi.org/10.1097/TP.0000000000001481] [PMID: 27607535]
[15]
Zhang R, Deng SL, Lian ZX, Yu K. Immunosuppression in uterine transplantation. Transpl Immunol 2020; 63: 101335.
[http://dx.doi.org/10.1016/j.trim.2020.101335] [PMID: 32927095]
[16]
Menke J, Sollinger D, Schamberger B, Heemann U, Lutz J. The effect of ischemia/reperfusion on the kidney graft. Curr Opin Organ Transplant 2014; 19(4): 395-400.
[http://dx.doi.org/10.1097/MOT.0000000000000090] [PMID: 24905021]
[17]
Yao W, Tai LW, Liu Y, Hei Z, Li H. Oxidative stress and inflammation interaction in ischemia reperfusion injury: Role of programmed cell death. Oxid Med Cell Longev 2019; 2019: 6780816.
[http://dx.doi.org/10.1155/2019/6780816] [PMID: 31089413]
[18]
Ghaffari S, Roshanravan N. Saffron; An updated review on biological properties with special focus on cardiovascular effects. Biomed Pharmacother 2019; 109: 21-7.
[http://dx.doi.org/10.1016/j.biopha.2018.10.031] [PMID: 30391705]
[19]
Yang M, Mao G, Ouyang L, Shi C, Hu P, Huang S. Crocetin alleviates myocardial ischemia/reperfusion injury by regulating inflammation and the unfolded protein response. Mol Med Rep 2020; 21(2): 641-8.
[PMID: 31974615]
[20]
Wang Y, Sun J, Liu C, Fang C. Protective effects of crocetin pretreatment on myocardial injury in an ischemia/reperfusion rat model. Eur J Pharmacol 2014; 741: 290-6.
[http://dx.doi.org/10.1016/j.ejphar.2014.07.052] [PMID: 25176181]
[21]
Michael CP, Derpapas M, Aravidou E, et al. The carotenoid compound of saffron crocetin alleviates effects of ischemia reperfusion injury via a mechanism possibly involving MiR-127. Cureus 2020; 12(2): e6979.
[http://dx.doi.org/10.7759/cureus.6979] [PMID: 32089976]
[22]
Ishizuka F, Shimazawa M, Umigai N, et al. Crocetin, a carotenoid derivative, inhibits retinal ischemic damage in mice. Eur J Pharmacol 2013; 703(1-3): 1-10.
[http://dx.doi.org/10.1016/j.ejphar.2013.02.007] [PMID: 23428630]
[23]
Chang G, Chen Y, Zhang H, Zhou W. Trans sodium crocetinate alleviates ischemia/reperfusion-induced myocardial oxidative stress and apoptosis via the SIRT3/FOXO3a/SOD2 signaling pathway. Int Immunopharmacol 2019; 71: 361-71.
[http://dx.doi.org/10.1016/j.intimp.2019.03.056] [PMID: 30952100]
[24]
Deng J, Xiong L, Zuo Z. Trans-sodium crocetinate provides neuroprotection against cerebral ischemia and reperfusion in obese mice. J Neurosci Res 2015; 93(4): 615-22.
[http://dx.doi.org/10.1002/jnr.23522] [PMID: 25491171]
[25]
Wang Y, Yu W, Shi C, Hu P. Crocetin attenuates sepsis-induced cardiac dysfunction via regulation of inflammatory response and mitochondrial function. Front Physiol 2020; 11: 514.
[http://dx.doi.org/10.3389/fphys.2020.00514] [PMID: 32581829]
[26]
Czerska M, Mikołajewska K, Zieliński M, Gromadzińska J, Wąsowicz W. Today’s oxidative stress markers. Med Pr 2015; 66(3): 393-405.
[http://dx.doi.org/10.13075/mp.5893.00137] [PMID: 26325052]
[27]
Ayala A, Muñoz MF, Argüelles S. Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev 2014; 2014: 360438.
[http://dx.doi.org/10.1155/2014/360438] [PMID: 24999379]
[28]
Nicco C, Batteux F. ROS modulator molecules with therapeutic potential in cancers treatments. Molecules 2017; 23(1): 84.
[http://dx.doi.org/10.3390/molecules23010084] [PMID: 29301225]
[29]
Jordan SC, Ammerman N, Choi J, et al. Interleukin-6: An important mediator of allograft injury. Transplantation 2020; 104(12): 2497-506.
[http://dx.doi.org/10.1097/TP.0000000000003249] [PMID: 32235253]
[30]
Barbier L, Ferhat M, Salamé E, et al. Interleukin-1 family cytokines: Keystones in liver inflammatory diseases. Front Immunol 2019; 10: 2014.
[http://dx.doi.org/10.3389/fimmu.2019.02014] [PMID: 31507607]
[31]
Esposito E, Cuzzocrea S. TNF-alpha as a therapeutic target in inflammatory diseases, ischemia-reperfusion injury and trauma. Curr Med Chem 2009; 16(24): 3152-67.
[http://dx.doi.org/10.2174/092986709788803024] [PMID: 19689289]
[32]
Wang L, Zhang Z, Wang H. Naringin attenuates cerebral ischemia-reperfusion injury in rats by inhibiting endoplasmic reticulum stress. Transl Neurosci 2021; 12(1): 190-7.
[http://dx.doi.org/10.1515/tnsci-2020-0170] [PMID: 34046215]
[33]
Hu Q, Jin J, Zhou H, et al. Crocetin attenuates DHT-induced polycystic ovary syndrome in mice via revising kisspeptin neurons. Biomed Pharmacother 2018; 107: 1363-9.
[http://dx.doi.org/10.1016/j.biopha.2018.08.135] [PMID: 30257351]
[34]
Di Emidio G, Rossi G, Bonomo I, et al. The natural carotenoid crocetin and the synthetic tellurium compound AS101 protect the ovary against cyclophosphamide by modulating SIRT1 and mitochondrial markers. Oxid Med Cell Longev 2017; 2017: 8928604.
[http://dx.doi.org/10.1155/2017/8928604] [PMID: 29270246]
[35]
Nakka VP, Gusain A, Mehta SL, Raghubir R. Molecular mechanisms of apoptosis in cerebral ischemia: Multiple neuroprotective opportunities. Mol Neurobiol 2008; 37(1): 7-38.
[http://dx.doi.org/10.1007/s12035-007-8013-9] [PMID: 18066503]
[36]
Lysiak JJ, Zheng S, Woodson R, Turner TT. Caspase-9-dependent pathway to murine germ cell apoptosis: Mediation by oxidative stress, BAX, and caspase 2. Cell Tissue Res 2007; 328(2): 411-9.
[http://dx.doi.org/10.1007/s00441-006-0341-y] [PMID: 17265069]
[37]
Garrido C, Galluzzi L, Brunet M, Puig PE, Didelot C, Kroemer G. Mechanisms of cytochrome c release from mitochondria. Cell Death Differ 2006; 13(9): 1423-33.
[http://dx.doi.org/10.1038/sj.cdd.4401950] [PMID: 16676004]
[38]
Skulachev VP. Cytochrome c in the apoptotic and antioxidant cascades. FEBS Lett 1998; 423(3): 275-80.
[http://dx.doi.org/10.1016/S0014-5793(98)00061-1] [PMID: 9515723]
[39]
Saha S, Buttari B, Panieri E, Profumo E, Saso L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules 2020; 25(22): 5474.
[http://dx.doi.org/10.3390/molecules25225474] [PMID: 33238435]
[40]
Loboda A, Damulewicz M, Pyza E, Jozkowicz A, Dulak J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: An evolutionarily conserved mechanism. Cell Mol Life Sci 2016; 73(17): 3221-47.
[http://dx.doi.org/10.1007/s00018-016-2223-0] [PMID: 27100828]
[41]
Chen L, Zhang WL, Xie DQ, Jia W. Sulforaphane alleviates hepatic ischemia-reperfusion injury through promoting the activation of Nrf-2/HO-1 signaling. Transpl Immunol 2021; 68: 101439.
[http://dx.doi.org/10.1016/j.trim.2021.101439] [PMID: 34320386]
[42]
Zhang HT, Wang XZ, Zhang QM, Zhao H. Neuroprotection of chromobox 7 knockout in the mouse after cerebral ischemiareperfusion injury via nuclear factor E2-related factor 2/hemeoxygenase-1 signaling pathway. Hum Exp Toxicol 2021; 40(12_suppl) (Suppl.): S178-86.
[http://dx.doi.org/10.1177/09603271211036122] [PMID: 34353139]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy