A narrative review of the protective effects of curcumin in treating ischemia-reperfusion injury
Review Article

A narrative review of the protective effects of curcumin in treating ischemia-reperfusion injury

Shiyong Teng1^, Mary Joseline Joseph2^, Huizhi Yu3^, Chunlan Hu3^, Xiaoshan Li4^, Chunxiao Hu5^

1Department of Anesthesiology, First Hospital of Jilin University, Changchun, China; 2Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, China; 3Department of Anesthesiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China; 4Wuxi Lung Transplant Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China; 5Department of Transplant Anesthesiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China

Contributions: (I) Conception and design: SY Teng; (II) Administrative support: CX Hu; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

^ORCID: Shiyong Teng, 0000-0003-2731-4459; Mary Joseline Joseph, 0000-0002-6817-8759; Huizhi Yu, 0000-0002-0408-362X; Chunlan Hu, 0000-0002-2202-0411; Xiaoshan Li, 0000-0003-1699-2545; Chunxiao Hu, 0000-0002-4825-0645.

Correspondence to: Prof. Chunxiao Hu. Department of Transplant Anesthesiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China. Email: huchunxiao91211@163.com.

Background and Objective: Ischemia-reperfusion (IR) injury is the cause of morbidity and mortality in a variety of diseases and surgical procedures including organ transplantation surgeries, acute coronary syndrome, strokes, and limb injuries. IR injury causes dysfunction of tissues and organs, and oxidative stress plays an important role in driving this process. Curcumin (CUR), a polyphenolic compound derived from turmeric, protects against IR injury by alleviating oxidative stress, reactive oxygen species (ROS) inflammation, apoptosis, and fibrosis. We review the protective effects of CUR against IR.

Methods: We searched PubMed, ScienceDirect, and Web of Science databases using the keywords: ischemic reperfusion, CUR and summarized the results.

Key Content and Findings: The effects of CUR during IR have been reported for animal models in vitro and in vivo and the compound has been shown in various organs by suppression of oxidative stress, prevention of inflammation, inhibition of apoptosis and autophagy. CUR with nanocarriers showed many advantages than free CUR in the treatment of IR injury, such as improved bioavailability, sustained-release, better water solubility, better target organ accumulation, improved permeability across the blood-brain-barrier and more effective.

Conclusions: Nanotechnology offers significant improvements and promising strategies to improve drug delivery to IR-injured tissues and achieve the desired protective effects. Thus, it is necessary to promote further clinical trials to promote the clinical application of CUR with nanocarriers.

Keywords: Ischemia; reperfusion; curcumin (CUR); review


Submitted May 25, 2022. Accepted for publication Jul 08, 2022.

doi: 10.21037/atm-22-3178


Introduction

Tissue hypoxia and IR injury are the underlying pathophysiological mechanisms culminating in tissue injury in a wide range of clinical conditions, including myocardial infarction, stroke, and acute limb ischemia. They are also a common manifestation in a variety of routine surgical procedures, such as organ transplantation and cardiac and vascular surgeries. IR injury is a major cause of organ malfunction, which can result in patient mortality (1).

IR injury is associated with a variety of pathophysiological features, including energy depletion, oxidative stress, calcium overload, endothelial dysfunction, increased membrane permeability, mitochondrial dysfunction, increased proinflammatory cytokines, and immune responses, resulting in apoptosis and autophagy. Oxidative stress plays an important role in the mechanism of IR injury (2).

Many medicinal plants contain active ingredients such as flavonoids, which are free radical scavengers that can reduce oxidative stress (3). An important member of the flavonoids family is curcumin (CUR), the orange-yellow and water-insoluble ingredient extracted from the rhizome of turmeric (Curcuma longa). CUR is considered relatively safe and is a commonly used household spice in certain dishes. Numerous animal studies and human trials have demonstrated CUR to be safe, with good anti-inflammatory properties without obvious side effects (4,5). Various pharmacological properties of CUR including its anti-inflammatory, antioxidant, immunomodulatory, anticarcinogenic, anticoagulant, hepatoprotective, analgesic, antidiabetic, lipid-lowering, and antidepressant properties have been of great interest to the scientific community (6).

Converging evidence suggests CUR has a protective effect on tissue with IR injury. A better understanding of this protection may shed light on the mechanisms underlying IR injury and provide solid evidence of the clinical therapeutic strategies that may be employed for protection against it.

To better understand the development of the protective effects of CUR in treating IR injury, we review the works to date on its role in tissue protection in a variety of models of IR in different organs. We present the following article in accordance with the Narrative Review reporting checklist (available at https://atm.amegroups.com/article/view/10.21037/atm-22-3178/rc).


Methods

A comprehensive literature search of all published studies was conducted using PubMed, ScienceDirect, and Web of Science databases [1993–2021] with the keywords: ischemic reperfusion, CUR. Inclusion criteria included studies focus on CUR and IR injury. Articles were searched independently by two reviewers (Shiyong Teng and Mary Joseline Joseph) using predefined standardized data extraction forms, and then references were evaluated by a third reviewer (Xiaoshan Li) independently (see Table 1). The detailed search strategy was shown in Table 2.

Table 1

The search strategy summary

Items Specification
Date of Search January 1, 2022 to January 10, 2022
Databases and other sources searched PubMed, ScienceDirect, and Web of Science
Search terms used (including Mesh and free text search terms and filters), See Table 2 for details “curcumin”, “demethoxycurcumin”, “curcuminoids”, “tetrahydrocurcumin”, “ischemia”, “reperfusion”
Timeframe 1993–2021
Inclusion and exclusion criteria (study type, language restrictions etc.) Inclusion criteria included studies focus on curcumin and ischemia-reperfusion injury. Exclusion criteria included studies did not focus on curcumin and ischemia-reperfusion injury
Selection process (who conducted the selection, whether it was conducted independently, how consensus was obtained, etc.) Articles retrieved from the searches were evaluated independently by 2 reviewers (Shiyong Teng and Mary Joseline Joseph) using predefined standardized data extraction forms, and then data were evaluated by a third reviewer (Xiaoshan Li) independently

Table 2

The detailed search strategy (take Web of Science for example)

Items Specification
MeSH 260 references were found and after carefully checked the abstract and full text, 191 references were included in this study
Free text Curcumin; demethoxycurcumin; curcuminoids; tetrahydrocurcumin; ischemia; reperfusion
Filters Studies did not focus on curcumin and ischemia-reperfusion injury

Hepatic IR

Liver IR injury is commonly seen in patients undergoing liver transplantation, hepatectomy, or hemorrhagic shock and can lead to a high level of morbidity and mortality (7,8). The increasing application of cadaveric or steatotic grafts in liver transplantation results in a higher susceptibility to IR injury and a much higher risk of primary non-function and mortality. Therefore, minimizing the adverse effects of hepatic IR injury could increase the number of successful outcomes after liver transplantation surgery (9,10).

Chen et al. (11) investigated the effects of CUR on hepatic IR in a rat liver isolated perfusion model. The investigators flushed rat livers with different preservation solutions with or without CUR (25–200 µM) and stored them at 4 degrees C for 24–48 h, followed by 2 h of reperfusion. In the CUR treated groups, portal flow rates and bile production were significantly higher, while the levels of liver enzymes (which serve as markers of cellular damage) were significantly lower. This indicated the use of CUR enhanced the preservation quality, thereby extending the preservation time while maintaining organ quality.

Shen et al. (12) observed the protective effect of CUR on liver thermal IR injury in a rat liver thermal IR model and found CUR (50 mg/kg) administered intravenously through the mesentery 30 min before ischemia significantly attenuated the extent of liver injury, suggesting its protective mechanism may be related to the overexpression of Hsp70 and antioxidant enzymes. Lin et al. (13) reported treatment with 25 mg/kg CUR orally (1 day before IR) in the rat significantly attenuated the extent of reperfusion injury to the liver. CUR reversed ATP content and decreased methyl guanidine (MG), tumor necrosis factor α (TNF-α), and nitric oxide (NO) release during hepatic ischemia. Inokuma et al. (14) treated rats with 340 mg/kg/day CUR orally for 7 days before a 90% hepatectomy and showed it improved the survival rate after a massive hepatectomy by maintaining the hepatic lobular structure in a relatively stable state without necrosis and increasing the heme oxygenase-1 (HO-1) protein level. Wu et al. (15) reported treatment with CUR in rats attenuated hepatic IR induced combined restrictive and obstructive lung disease by reducing lung inflammation and matrix metalloprotease 9 (MMP-9) activity, while Liu et al. (16) investigated the effects of CUR on orthotopic liver transplantation and Kupffer cells (KCs) polarization. CUR significantly alleviated liver injury while improving liver function and overall post-transplantation survival through activating PPAR gamma by inhibiting the activation of the nuclear factor kappa-B (NF-κB) pathway and remodeling the polarization of KCs. Ibrahim et al. (17) investigated the effects of Dimethyl fumarate (DMF) with CUR against hepatic IR injury in rats and found the combination of DMF and CUR offered significant protection via the antioxidant and anti-inflammatory properties mediated by the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) pathway. Wang et al. (18) showed that administration of CUR (100 mg/kg) 0.5 h before IR significantly inhibited apoptosis and reduced LDH levels and TNF-α, IL-1b, and IL-6 production by inhibiting the TLR4/NF-κB pathway, while Kosharskyy et al. (19) and Chi et al. (20) showed similar hepato-protective effects of CUR analogs.

Leong et al. (21) compared pentosidine with CUR for the pro-oxidant-induced glutathione antioxidant response, and in vitro studies (AML12 cells) showed Sch B (15 µm) and CUR (7.5 µm) protected against oxidant-induced damage. Kheradpezhouh et al. (22) investigated the effects of CUR on Transient Receptor Potential Melastatin 2 (TRPM2) channels in rat hepatocytes, and 5 µM CUR in the incubation medium prevented the H2O2 and paracetamol-induced (Ca2+) rise and inhibited activation of TRPM2 current.


Intestinal IR

The intestine is particularly sensitive to ischemia, and intestinal IR frequently occurs during abdominal surgery (23). Intestinal IR increases the production of reactive oxygen species (ROS), reactive nitrogen species (RNS), and polymorphonuclear neutrophil activity, which leads to oxidative stress (24).

Karatepe et al. (25) induced intestinal ischemia in rats for 1 hour by superior mesenteric artery ligation followed by reperfusion for 3 hours. These investigators pretreated rats with 40 mg/kg CUR for 15 days before ischemia and found administration of CUR before intestinal ischemia significantly increased GSH levels and decreased the intestinal mucosal injury scores, myeloperoxidase (MPO) activity, malondialdehyde (MDA), and NO levels.

CUR treatment reduced the severity of IR-induced histopathological damage of the intestine, including mucosal erosion, villi congestion, and hemorrhage (26,27), increased intestinal and gastric superoxide dismutase activity (28,29), and reduced ROS production in mesenteric vessels (30). Many signaling pathways may be involved in the protective role of CUR against intestinal IR injury, such as the NF-κB pathway (31), leptin and Ob-Rb-dependent ERK and p38 MAPK signaling pathways (32), TNF-alpha pathway (33), and the induction of Parkin dependent mitophagy through AMPK activation and subsequent TFEB nuclear translocation (34).

CUR treatment may also protect against damage to other organs caused by intestinal IR, such as intestinal IR injury associated esophageal injury (35), liver injury (36), and lung injury (37). Sozen et al. (38) studied the effect of CUR on an animal intestinal IR model for bacterial translocation and inflammatory response and showed it reduced bacterial translocation, hepatocyte damage, and plasma cytokine levels in the blood. These results suggest CUR would be clinically useful in the treatment of intestinal IR injury.


Lung IR

IR-associated lung injury occurs in various clinical circumstances, including lung transplantation, in which it is a severe complication in the early postoperative period affecting up to 20% of transplants, resulting in primary graft failure and a 60% death rate (39). It is important to develop an effective therapeutic strategy to attenuate IR associated lung injury, reduce the incidence of PGD, and improve perioperative survival.

Sun et al. (40) investigated the effects of CUR or dexamethasone on lung transplantation-associated lung injury in Sprague-Dawley rats. Rats received unilateral in situ lung transplantation with 4 h of cold ischemia followed by 2 h (or 24 h) of reperfusion. CUR was administered intraperitoneally to both donors and recipients at a dose of 200 mg/kg 3 h before anesthesia, while dexamethasone was administered intraperitoneally at a dose of 5 mg/kg 30 min before anesthesia. CUR and dexamethasone pretreatment significantly attenuated alveolar barrier disruption and PaO2 reduction, preventing post-transplant pulmonary edema and tissue inflammation through mediation of the NF-κB pathway.

Sun et al. (41) also observed the protective effects of CUR or dexamethasone in a rat lung IR model by performing clamped ischemia in the left lung hilar of rats for 90 min followed by 4 h of reperfusion. CUR or dexamethasone pretreatment significantly attenuated IR-induced barrier disruption, pulmonary edema, tissue inflammation, and hypoxemia. CUR attenuated NF-κB-mediated inflammatory cytokine expression, improved oxidative stress, and suppressed inflammatory responses in acute lung injury. Zhou et al. (42) established a unilateral in situ lung IR injury model in C57BL/6J mice, and CUR (150–200 mg/kg) administered intraperitoneally had a good protective effect, the mechanism of which may be related to the inhibition for pneumocyte apoptosis associated with Caspase-12 in an excessive unfolded protein response.

In keeping with these studies, Luo et al. (43) found CUR exerted significant protective effects on lung IR injury in C57BL/6J mice and suggested this may relate to inhibited overexpression of CHOP, JNK, and Caspase-12, reduced stress on the endoplasmic reticulum (ER), and reduction of pulmonary IR injury-induced apoptosis. Jiang et al. (44) reported CUR (50–200 mg/kg) intraperitoneally protected against lung IR injury in rats, resulting in significantly decreased carbon dioxide partial pressure, the ratio of lung wet weight to dry weight, and the lung cell apoptosis index, and significantly increased arterial partial pressure of oxygen. Shi et al. (45) investigated the effect of CUR on myocardium mediated by pulmonary IR injury in rats and found 50 mg/kg intraperitoneally effectively protected against myocardial injury.


Myocardial IR

Cardiac IR can injure the myocardium and cause acute infarction (46). The mitochondrial respiratory chain and NADPH oxidase generate ROS, which increases during cardiac IR injury (47), increasing myocardial injury and leading to apoptosis, arrhythmias, and functional impairment (6).

Cheng et al. (48) first reported the effect of CUR on the myocardial IR injury model by occluding the left anterior descending branch of the coronary artery for 60 min then removing the ligation to allow reperfusion for 60 min. The results showed administering CUR (20, 40 mg/kg via the sublingual vein) reduced the myocardial infarct size as well as the serum CK, LDH activity, and myocardial MDA and FFA content, and increased super oxide dismutase and glutathione peroxidase activity. Yeh et al. (49) investigated the effect of CUR on myocardial ischemia/reperfusion injury with cardioplegia during cardiopulmonary bypass (CPB), and found CUR (70 µm/kg, 100 µm/kg) ameliorated the pro-inflammatory cytokine surge during CPB and reduced cardiomyocyte apoptosis after total myocardial IR injury by inhibiting NF-κB activation. The same authors also proved CUR attenuates IR-induced contractile injury by inhibiting NF-κB activation, decreasing pro-inflammatory genes in cardiomyocytes, and attenuating matrix metalloproteinase activation (50).

A double-blind randomized controlled trial from Wongcharoen evaluated the effects of CUR on the incidence of acute myocardial infarction after coronary artery bypass grafting (51). A total of 121 consecutive patients undergoing CABG were randomly allocated to two groups; placebo or CUR (4 g/day). CUR was administered orally beginning 3 days before the scheduled surgery and continued until 5 days after surgery, and the incidence of in-hospital myocardial infarction was decreased to 13.1% in the CUR group from 30.0% in the placebo group. Postoperative C-reactive protein, MDA, and N-terminal pro-B-type natriuretic peptide levels were also lower in the CUR than in the placebo group. These studies suggest beneficial effects of CUR in decreasing myocardial infarction associated with CABG, possibly through its antioxidant and anti-inflammatory effects.

Duan et al. (52) evaluated the effects of CUR on myocardial ischemia in a rat isolated perfused heart model. They exposed the heart to 1 µM CUR 10 min before myocardial reperfusion and showed treatment significantly improved the recovery of cardiac function after ischemia, reduced myocardial infarct size, decreased lactate dehydrogenase release, improved coronary blood flow, and reduced the number of apoptotic cardiomyocytes. The protective effects of CUR may be mediated by upregulation of the anti-apoptotic protein Bcl2, downregulation of the pro-apoptotic protein Caspase3, and activation of the JAK2/STAT3 signaling pathway. Similarly, CUR (53) significantly decreased the expressions of the inflammation-related pathway in both rats and isolated hearts. In keeping with these studies, Ilyas et al. (54) found CUR exerted protective effects against myocardial IR injury in isolated perfused working guinea pig hearts and suggested these cardioprotective effects may be related to inhibited glutathione peroxidase expression. Wang et al. (55) showed CUR significantly improved cardiac function, reduced the infarct size, and decreased the lactate dehydrogenase levels in isolated rat hearts, in part through activation of the SIRT3 pathway. Broskova et al. (30) studied the effects of plant polyphenols on IR injury in isolated rat hearts and intestines and showed CUR and peppermint extracts were most effective in reducing reperfusion-induced arrhythmias.

In myocardial IR in vitro experiments, many investigators have selected H9C2 cells under oxygen-glucose deprivation/reoxygenation (OGD/R) conditions to observe the protective effects of CUR. Fiorillo et al. (56) found the protective effects of CUR (10 µM) given before ischemia (pre-treatment) or at reperfusion (post-treatment) were similar, with an equal antioxidant capacity as the antioxidant Trolox.

Briefly, CUR treatment reduced the severity of IR induced histopathological damage of the heart, lessened the severity of cardiac mechanical dysfunction, improved heart function, diminished infarct size, anti-fibrotic, improved left ventricular end-diastolic volume, stroke volume and ejection fraction, promoted neovascularization, increased the wall thickness of the infarcted middle anterior septum, and showed antiarrhythmic effects (for details see Table S1). CUR treatment could reduce apoptosis of IR injured myocardial cells by inhibiting GSK-3 (57,58), and by preventing apoptosis and autophagy through Bcl-2/Bax/beclin-1/BNIP3/SIRT1 signaling pathways (59), activation of caspase 3 enzyme and bax/bax3 signaling pathways (60), activation of SIRT3 (55), down regulation of the Notch pathway (61), and diminishing ER stress (62,63) (Table S1).


Neural IR

Brain IR

Stroke is a common cause of disability or death worldwide (64), and the most common causes include carotid pathology, hypoxic-ischemic brain injury, and shock (65). Although the pathophysiological mechanisms of IR are complex, apoptosis, inflammation, and intra-neuronal calcium excess are the main causes of IR injury (64).

In 2002, Ghoneim et al. (66) first reported the protective effects of CUR against brain IR injury in a rat bilateral common carotid artery occlusion (BCCAO) model. The results showed CUR (50, 100, or 200 mg/kg i.p.) administered 30 minutes after the onset of ischemia protected the rat forebrain against IR injury, and at the highest dosage (200 mg/kg), decreased the IR-induced elevated xanthine oxidase activity, superoxide anion production, MDA level, glutathione peroxidase, superoxide dismutase, and lactate dehydrogenase activity.

Many researchers have investigated the protective effects of CUR on brain IR in various models, such as BCCAO, middle cerebral artery occlusion (MCAO), global cerebral IR, retinal IR, and PC12 cell lines (Table S2). CUR treatment reduced the severity of IR-induced histopathological damage of the brain, including diminishing infarct volume, improving neurological deficit, decreasing mortality, reducing the water content of the brain, reducing hippocampal neuronal apoptosis, improving memory function, protecting against damage to the blood brain barrier, improving neuro-motor functions and ameliorating cerebrovascular permeability, and increasing the proliferation of neural stem cells (Table S2). The mechanisms of the neuroprotective effects of CUR are mainly through its antioxidant and anti-apoptotic functions and its inhibition of autophagy. Many signaling pathways may be involved in the protective effects of CUR on brain IR, such as the ONOO− donor SIN-1, Fos/Jun/NF-κB, iNOS, MDA, cytochrome c, caspase 3, Bcl-2, HMGB1, and MEK/ERK/cREB pathways (Table S2).

Spine IR

Spine IR injury is commonly seen in trauma and abdominal aorta occlusion. After trauma occurs, the spinal cord undergoes an initial physical injury (primary injury) followed by a progressive injury process (secondary injury) (67). It is believed one of the most important factors precipitating secondary injury of the spinal cord is lipid peroxidation caused by oxygen-free radicals (68), and CUR, as an antioxidant, has been extensively studied to evaluate its effect on spine IR (Table S2).

In 2010, Cemil et al. investigated the effects of CUR in rat traumatic spinal cord injury models, and the results showed CUR (200 mg/kg i.p.) improved early functional, biochemical, and pathological results by increasing tissue levels of GSH-Px, superoxide dismutase (SOD) and catalase (CAT) (69). Similarly, Kavakli’s study indicated CUR (200 mg/kg orally) effectively protects the spinal cord tissues against oxidative damage in a rat weight-drop spinal cord injury model (70). In keeping with these results, Ormond et al. showed CUR in combination with stem cell therapy, induced profound recovery from severe spinal cord injury by regulation of stem cell proliferation (71). CUR also showed good protective effects on transient spinal cord ischemia by aortic occlusion (72-74).


Pancreas IR

The pancreas is an organ highly susceptible to ischemia, and pancreas IR injury can be a consequence of pancreatic surgery, pancreas transplantation, pancreatitis, and shock. Pancreas IR could induce systemic inflammatory responses by increasing oxygen radical production, white blood cell count, and cytokine release (75,76).

Chen et al. investigated the effect of CUR on airway hyper-reactivity induced by pancreatic IR in rat models (77). Ischemia of the pancreas was induced by clamping the gastro-duodenal and the splenic artery for 2 hours followed by reperfusion for 6 hours, and the results showed CUR (20 mg/kg i.p. 2 hours before pancreatic I/R) significantly attenuated the inflammatory, oxidative, and nitrosative responses as well as the lung tissue iNOS and TNF-α expressions during IR and attenuated airway reactivity to methacholine challenge. These results suggest CUR has promising applications for the treatment of airway hyperreactivity and systemic inflammatory responses caused by pancreatic IR.


Renal IR

Renal IR contributes to the development of acute kidney injury (AKI), which directly influences patient survival. Renal IR occurs in a variety of medical and surgical settings and the mechanism consists of activation of neutrophils and release of ROS and inflammatory mediators (78,79).

Shoskes et al. first reported the renoprotective effect of CUR and quercetin in the left renal pedicle occlusion model in rats (80), and the results indicated CUR or quercetin (30 mg/kg i.p. 2 h before surgery) reduced IR injury and the inflammatory outcomes. They also examined the effects of CUR and quercetin on early graft function of dialysis-dependent cadaveric kidney recipients in a clinical RCT study (79). CUR (480, 960 mg) and quercetin (20, 40 mg) were given orally for 30 days after surgery, and the results showed two patients in the control group exhibited delayed graft function, while the treatment groups did not exhibit this outcome. Incidence rates of early function were 43% (control group), vs. 71% (low-dose group), and 93% (high-dose group). Serum creatinine (2 and 30 days) and incidences of acute rejection within 6 months were significantly lower in the treatment group. The authors of this study concluded CUR and quercetin can improve early outcomes in cadaveric renal transplantation, possibly by inducing HO-1 expression.

Epigenetic regulation, including histone acetylation, has been implicated in the pathogenesis of renal IRI (81). The acetylation of histone H3 by ischemia and reperfusion would damage renal function (82). Yang et al. investigated the effects of CUR on the regulation of histone acetylation on IRI-induced renal apoptosis and the molecular mechanisms in rats (83). The results showed that CUR significantly decreased renal apoptosis and caspase-3/-9 expression and enhanced renal function in IRI rats, and the protective mechanism of CUR involves suppression of activation of the c-Jun N-terminal kinase (JNK) pathway via epigenetic regulation of p300/CREB-binding protein (CBP)-mediated histone acetylation. The studies of the effects of CUR in renal IR injury are summarized in Table S3.


Reproductive system IR

Ovarian IR

Ovarian IR injury can be a consequence of ovarian torsion, often termed adnexal torsion, which is a common gynecological emergency. Ovarian IR injury is mediated by ROS generated via lipid peroxidation, promoting the release of inflammatory agents (84).

Sak et al. evaluated the protective effects of CUR in a rat ovarian torsion model (85), and found CUR (100 mg/kg, i.p. 30 minutes before IR) significantly decreased the mean levels of oxidant markers and histopathologic scores of the ovarian tissues, and reversed tissue damage induced by IR injury. Similarly, Eser et al. reported CUR (200 mg/kg i.p.) maintained and protected ovarian functions in an IR rat model (86).

Testicular IR

Testicular IR injury can be a consequence of testicular torsion which disrupts blood flow of the testis and causes ischemia. It is an emergency in newborns, children, adolescents, and adults and can lead to infertility (87).

Wei et al. studied the effect of CUR on testicular IR in a rat torsion–detorsion model (88) and found CUR (200 mg/kg iv via the tail vein) significantly decreased xanthine oxidase activity and MDA level, and showed a significant increase in HO-1 protein expression level and testicular spermatogenesis. Similarly, studies conducted by Takhtfooladi and Shahedi also demonstrated the protective effects of CUR on testicular IR injury (89,90).

It is worth mentioning that several studies found CUR had protective effects against drug-induced testicular toxicity, such as cisplatin (91,92) and dexamethasone toxicity (93).

Priapism IR

Priapism IR can be seen in patients undergoing ischemic priapism which lasts longer than four hours, leading to hypoxia, acidosis, and fibrosis, resulting in erectile dysfunction (94). Yilmaz et al. investigated the biochemical and histopathological effects of CUR in a rat ischemic priapism model (95), and the results indicated that CUR (200 mg/kg/day orally for 7 days) had preventive effects against oxidative stress parameters in priapism IR.


Skin and skeletal muscle IR

In 1994, Ashoori et al. first reported the protective effects of CUR and ellagic acid on skin IR in a rat skin flaps model (96), while Shoskes et al. found quercetin and CUR (30 mg subcutaneous injection) prolonged skin graft survival in a rat full-thickness skin allograft model (97). Jia et al. tested the efficacy of CUR on rabbit ear wounds (98), and found intravenous CUR produced accelerated wound healing and promoted non-ischemic wound healing in a dose-dependent manner which was associated with significant decreases in interleukin (IL) levels, namely IL-1, IL-6, and IL-8. Yen et al. created back wounds in mice and treated them with topical CUR (0.2 mg/mL) in Pluronic F127 gel (99), and the results showed topical CUR accelerated wound healing by regulating the levels of various cytokines, such as TNF-α, MMP-9, and αSMA.

Several studies showed CUR could not only protect against skeletal muscle IR (100-102) but also protect against renal injury (103) and lung injury (104-106) induced by skeletal muscle IR.


Molecular mechanisms

Because of its anti-oxidant, anti-inflammatory, and excellent safety profile, CUR is useful in the prevention and treatment of some diseases thanks to the control of inflammation, cell growth, and apoptosis (107). Many chronic inflammatory, degenerative disorders are caused by oxidative stress and oxidative damage, leading to a decline in health and an increased incidence of chronic diseases. CUR is a highly pleiotropic molecule that interacts with a wide range of inflammatory molecules. In the field of treating IR, numerous studies have revealed CUR modulates a variety of molecules in cell signal pathways including PI3K, Akt, mTOR, ERK5, AP-1, TGF-beta, Wnt, beta-catenin, Shh, PAK1, Rac1, STAT3, PPAR gamma, EBP alpha, NLRP3 inflammasome, p38MAPK, Nrf2, Notch-1, AMPK, TLR-4, and MyD-88 (108). Many studies both in vitro and in vivo have revealed CUR exerts a potent protective effect on IR injury mainly through the reduction of oxidative stress (41), prevention of inflammation, inhibition of apoptosis (109), and inhibition of autophagy (110). It is worth mentioning that evidence indicates both CUR pretreatment and post-treatment protect against IR, which is possible through endogenous antioxidant defense systems (111). So that the pleiotropic molecule, CUR, might be a promising therapeutic strategy via multiple pathways during the whole process of IR. Take lung transplantation as an example, CUR might take effect during donor organ preconditioning, organ preservation, and transplantation in the recipient. The detailed molecules in cell signal pathways of CUR protecting against IR injury are summarized in Figure 1.

Figure 1 Molecular mechanism demonstrating the protective function of CUR against ischemia-reperfusion injury. TNF-R, tumor necrosis factor receptor; PPAR, peroxisome proliferators-activated receptors; TGF, transforming growth factor; IL-6R, interleukin 6 receptor; CUR, curcumin; ATP, adenosine triphosphate; ER, endoplasmic reticulum; NF-κB, nuclear factor κB; I-κB, inhibitor of NF-κB; Casp, caspase; ROS, reactive oxygen species; JNK, c-Jun N-terminal kinase; Cyto-c, cytochrome c; ERK, extracellular signal-regulated kinase; BCL2, b-cell lymphoma-2; BAX, BCL2 associated X; AKT (PKB), protein kinase B; HSP, heat shock protein; PI3K, phosphoinositide 3-kinase; JAK, janus kinase; STAT, signal transducer and activator of transcription; RAS, rat sarcoma; MAPK, mitogen activated protein kinase; MTOR, mammalian target of rapamycin.

Nano CUR

Although CUR shows great protective effects against IR in many organs, its poor bioavailability and poor solubility hinder its clinical application. For example, Leong et al. reported CUR showed equivalent protective effects with Schisandrin B in AML12 cells but a much smaller effect than Schisandrin B in vivo. The authors of this study attributed this to the low bioavailability of CUR in vivo (21). In most studies in vivo (animals), CUR was dissolved in oil or DMSO and administrated by intraperitoneal injection, which is not acceptable in clinical practice.

In recent years, researchers have employed a variety of nanocarriers to address the poor bioavailability and water solubility of CUR, such as liposomes, solid lipid nanoparticles, exosomes, hydrogel, and nanofibres (Table S4). Compared with free CUR, CUR with nanocarriers showed many advantages in the treatment of IR injury, such as improved bioavailability (112), sustained-release (113), better water solubility, better target organ accumulation (114), and improved permeability across the blood-brain-barrier, and was found to be far more effective than free CUR (115). Nanotechnology offers significant improvements and promising strategies to improve drug delivery to IR-injured tissues and achieve the desired protective effects (116).


Narrative

Although many studies have shown the protective effects of CUR on various organs, some have claimed it has no significant protective effect on renal IR (117,118), hepatic IR (119), ovarian IR (120), and testicular IR (121).

It is worth mentioning CUR was administered orally in these studies, so the efficacy might have been diminished by poor bioavailability through gavage as the treatment was administered too late to take effect in these studies, for example, CUR 200 mg/kg p.o. 15 minutes before IR (119) and CUR 150 mg/kg p.o. 30 minutes before IR (121). Conversely, several studies mentioned earlier in this review used an oral route of administration for days before IR and had successful outcomes (14,51,122-128).


Summary

CUR possesses a wide-range of anti-inflammatory and antioxidant properties. Many studies showed the great protective effects of CUR against IR injury in various organs by suppression of oxidative stress, prevention of inflammation, inhibition of apoptosis, and autophagy. Although the low systemic bioavailability after oral administration seems to limit its ability to reach sufficient concentrations in tissues to exert beneficial effects, nanotechnology offers a solution to this problem and promises strategies that could enable the widespread clinical employment of CUR in treating IR injury.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://atm.amegroups.com/article/view/10.21037/atm-22-3178/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-22-3178/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Aragno M, Cutrin JC, Mastrocola R, et al. Oxidative stress and kidney dysfunction due to ischemia/reperfusion in rat: attenuation by dehydroepiandrosterone. Kidney Int 2003;64:836-43. [Crossref] [PubMed]
  2. Dorweiler B, Pruefer D, Andrasi TB, et al. Ischemia-Reperfusion Injury: Pathophysiology and Clinical Implications. Eur J Trauma Emerg Surg 2007;33:600-12. [Crossref] [PubMed]
  3. Orsolic N, Balta V, Odeh D, et al. Oxidative stress and inflammation caused by n-hexyl salicylate in mouse skin: the effectiveness of flavonoids. In J Phytomed 2017;9:279-88.
  4. Chainani-Wu N. Safety and anti-inflammatory activity of curcumin: a component of tumeric (Curcuma longa). J Altern Complement Med 2003;9:161-8. [Crossref] [PubMed]
  5. Sharma RA, Gescher AJ, Steward WP. Curcumin: the story so far. Eur J Cancer 2005;41:1955-68. [Crossref] [PubMed]
  6. Mokhtari-Zaer A, Marefati N, Atkin SL, et al. The protective role of curcumin in myocardial ischemia-reperfusion injury. J Cell Physiol 2018;234:214-22. [Crossref] [PubMed]
  7. Cursio R, Colosetti P, Gugenheim J. Autophagy and liver ischemia-reperfusion injury. Biomed Res Int 2015;2015:417590. [Crossref] [PubMed]
  8. Kim JY, Lee DY, Kang S, et al. Bilirubin nanoparticle preconditioning protects against hepatic ischemia-reperfusion injury. Biomaterials 2017;133:1-10. [Crossref] [PubMed]
  9. Serracino-Inglott F, Habib NA, Mathie RT. Hepatic ischemia-reperfusion injury. Am J Surg 2001;181:160-6. [Crossref] [PubMed]
  10. Jha S, Calvert JW, Duranski MR, et al. Hydrogen sulfide attenuates hepatic ischemia-reperfusion injury: role of antioxidant and antiapoptotic signaling. Am J Physiol Heart Circ Physiol 2008;295:H801-6. [Crossref] [PubMed]
  11. Chen C, Johnston TD, Wu G, et al. Curcumin has potent liver preservation properties in an isolated perfusion model. Transplantation 2006;82:931-7. [Crossref] [PubMed]
  12. Shen SQ, Zhang Y, Xiang JJ, et al. Protective effect of curcumin against liver warm ischemia/reperfusion injury in rat model is associated with regulation of heat shock protein and antioxidant enzymes. World J Gastroenterol 2007;13:1953-61. [Crossref] [PubMed]
  13. Lin CM, Lee JF, Chiang LL, et al. The protective effect of curcumin on ischemia-reperfusion-induced liver injury. Transplant Proc 2012;44:974-7. [Crossref] [PubMed]
  14. Inokuma T, Yamanouchi K, Tomonaga T, et al. Curcumin improves the survival rate after a massive hepatectomy in rats. Hepatogastroenterology 2012;59:2243-7. [PubMed]
  15. Wu NC, Wang JJ. Curcumin attenuates liver warm ischemia and reperfusion-induced combined restrictive and obstructive lung disease by reducing matrix metalloprotease 9 activity. Transplant Proc 2014;46:1135-8. [Crossref] [PubMed]
  16. Liu Y, Zhang W, Cheng Y, et al. Activation of PPARγ by Curcumin protects mice from ischemia/reperfusion injury induced by orthotopic liver transplantation via modulating polarization of Kupffer cells. Int Immunopharmacol 2018;62:270-6. [Crossref] [PubMed]
  17. Ibrahim SG, El-Emam SZ, Mohamed EA, et al. Dimethyl fumarate and curcumin attenuate hepatic ischemia/reperfusion injury via Nrf2/HO-1 activation and anti-inflammatory properties. Int Immunopharmacol 2020;80:106131. [Crossref] [PubMed]
  18. Wang L, Li N, Lin D, Zang Y. Curcumin protects against hepatic ischemia/reperfusion induced injury through inhibiting TLR4/NF-κB pathway. Oncotarget 2017;8:65414-20. [Crossref] [PubMed]
  19. Kosharskyy B, Vydyanathan A, Zhang L, et al. 2-Acetylcyclopentanone, an enolate-forming 1,3-dicarbonyl compound, is cytoprotective in warm ischemia-reperfusion injury of rat liver. J Pharmacol Exp Ther 2015;353:150-8. [Crossref] [PubMed]
  20. Chi X, Yu D, Li P, et al. The protection effects of (1E,6E)-1,7-diphenylhepta-1,6-diene-3,5-dione, a curcumin analogue, against operative liver injury in rats. Eur J Pharm Sci 2017;100:94-101. [Crossref] [PubMed]
  21. Leong PK, Chiu PY, Ko KM. Prooxidant-induced glutathione antioxidant response in vitro and in vivo: a comparative study between schisandrin B and curcumin. Biol Pharm Bull 2012;35:464-72. [Crossref] [PubMed]
  22. Kheradpezhouh E, Barritt GJ, Rychkov GY. Curcumin inhibits activation of TRPM2 channels in rat hepatocytes. Redox Biol 2016;7:1-7. [Crossref] [PubMed]
  23. Mallick IH, Yang W, Winslet MC, et al. Ischemia-reperfusion injury of the intestine and protective strategies against injury. Dig Dis Sci 2004;49:1359-77. [Crossref] [PubMed]
  24. Impellizzeri D, Cordaro M, Campolo M, et al. Anti-inflammatory and Antioxidant Effects of Flavonoid-Rich Fraction of Bergamot Juice (BJe) in a Mouse Model of Intestinal Ischemia/Reperfusion Injury. Front Pharmacol 2016;7:203. [Crossref] [PubMed]
  25. Karatepe O, Gulcicek OB, Ugurlucan M, et al. Curcumin nutrition for the prevention of mesenteric ischemia-reperfusion injury: an experimental rodent model. Transplant Proc 2009;41:3611-6. [Crossref] [PubMed]
  26. Yucel AF, Kanter M, Pergel A, et al. The role of curcumin on intestinal oxidative stress, cell proliferation and apoptosis after ischemia/reperfusion injury in rats. J Mol Histol 2011;42:579-87. [Crossref] [PubMed]
  27. Onder A, Kapan M, Gümüş M, et al. The protective effects of curcumin on intestine and remote organs against mesenteric ischemia/reperfusion injury. Turk J Gastroenterol 2012;23:141-7. [Crossref] [PubMed]
  28. Okudan N, Belviranlı M, Gökbel H, et al. Protective effects of curcumin supplementation on intestinal ischemia reperfusion injury. Phytomedicine 2013;20:844-8. [Crossref] [PubMed]
  29. Guzel A, Kanter M, Guzel A, et al. Protective effect of curcumin on acute lung injury induced by intestinal ischemia/reperfusion. Toxicol Ind Health 2013;29:633-42. [Crossref] [PubMed]
  30. Brosková Z, Drábiková K, Sotníková R, et al. Effect of plant polyphenols on ischemia-reperfusion injury of the isolated rat heart and vessels. Phytother Res 2013;27:1018-22. [Crossref] [PubMed]
  31. Cucolas C, Daneasa AI, Olteanu D, et al. Resveratrol and curcumin as protective agents in an experimental rat model of intestinal ischemia and reperfusion. Can J Physiol Pharmacol 2016;94:1151-8. [Crossref] [PubMed]
  32. Deng ZH, Liao J, Zhang JY, et al. Localized leptin release may be an important mechanism of curcumin action after acute ischemic injuries. J Trauma Acute Care Surg 2013;74:1044-51. [Crossref] [PubMed]
  33. Tian S, Guo R, Wei S, et al. Curcumin protects against the intestinal ischemia-reperfusion injury: involvement of the tight junction protein ZO-1 and TNF-α related mechanism. Korean J Physiol Pharmacol 2016;20:147-52. [Crossref] [PubMed]
  34. Cao S, Wang C, Yan J, et al. Curcumin ameliorates oxidative stress-induced intestinal barrier injury and mitochondrial damage by promoting Parkin dependent mitophagy through AMPK-TFEB signal pathway. Free Radic Biol Med 2020;147:8-22. [Crossref] [PubMed]
  35. Nurullahoglu-Atalik KE, Okudan N, Belviranli M, et al. Role of curcumin in mesenteric ischemia - reperfusion injury in rats. Bratisl Lek Listy 2012;113:465-70. [Crossref] [PubMed]
  36. Fan Z, Jing H, Yao J, et al. The protective effects of curcumin on experimental acute liver lesion induced by intestinal ischemia-reperfusion through inhibiting the pathway of NF-κB in a rat model. Oxid Med Cell Longev 2014;2014:191624. [Crossref] [PubMed]
  37. Fan Z, Yao J, Li Y, et al. Anti-inflammatory and antioxidant effects of curcumin on acute lung injury in a rodent model of intestinal ischemia reperfusion by inhibiting the pathway of NF-Kb. Int J Clin Exp Pathol 2015;8:3451-9. [PubMed]
  38. Sozen S, Aziret M, Bali I, et al. The Effect of Curcumin on an Animal Intestinal Ischemia/Reperfusion Model for Bacterial Translocation and Inflammatory Response. Int Surg 2015;100:1352-9. [Crossref]
  39. King RC, Binns OA, Rodriguez F, et al. Reperfusion injury significantly impacts clinical outcome after pulmonary transplantation. Ann Thorac Surg 2000;69:1681-5. [Crossref] [PubMed]
  40. Sun J, Guo W, Ben Y, et al. Preventive effects of curcumin and dexamethasone on lung transplantation-associated lung injury in rats. Crit Care Med 2008;36:1205-13. [Crossref] [PubMed]
  41. Sun J, Yang D, Li S, et al. Effects of curcumin or dexamethasone on lung ischaemia-reperfusion injury in rats. Eur Respir J 2009;33:398-404. [Crossref] [PubMed]
  42. Zhou JH, Zhao S, Chen HE, et al. Effect of curcumin on caspase-12 and apoptosis in pulmonary ischemia/reperfusion injury mice. Zhongguo Zhong Xi Yi Jie He Za Zhi 2014;34:1118-24. [PubMed]
  43. Luo ZY, Zhou JH, Xiang BQ, et al. Effects of curcumin on cell apoptosis in pulmonary ischemia/reperfusion injury in mice. Int J Clin Exp Pathol 2016;9:6633-44.
  44. Jiang W, Mei AH, Zhang SQ, et al. Curcumin Prevents Lung Ischemia-Reperfusion Injury in Rats through Reducing Oxidative Stress, Apoptosis and NF-kappa B Expression. Lat Am J Pharm 2020;39:2127-32.
  45. Shi M, Huang J, Pang L, et al. The influence of lung ischemia-reperfusion injury on myocardium. Int J Clin Exp Med 2014;7:4780-6. [PubMed]
  46. Ibáñez B, Heusch G, Ovize M, et al. Evolving therapies for myocardial ischemia/reperfusion injury. J Am Coll Cardiol 2015;65:1454-71. [Crossref] [PubMed]
  47. Cadenas S. ROS and redox signaling in myocardial ischemia-reperfusion injury and cardioprotection. Free Radic Biol Med 2018;117:76-89. [Crossref] [PubMed]
  48. Cheng H, Liu W, Ai X. Protective effect of curcumin on myocardial ischemia reperfusion injury in rats. Zhong Yao Cai 2005;28:920-2. [PubMed]
  49. Yeh CH, Chen TP, Wu YC, et al. Inhibition of NFkappaB activation with curcumin attenuates plasma inflammatory cytokines surge and cardiomyocytic apoptosis following cardiac ischemia/reperfusion. J Surg Res 2005;125:109-16. [Crossref] [PubMed]
  50. Yeh CH, Lin YM, Wu YC, et al. Inhibition of NF-kappa B activation can attenuate ischemia/reperfusion-induced contractility impairment via decreasing cardiomyocytic proinflammatory gene up-regulation and matrix metalloproteinase expression. J Cardiovasc Pharmacol 2005;45:301-9. [Crossref] [PubMed]
  51. Wongcharoen W, Jai-Aue S, Phrommintikul A, et al. Effects of curcuminoids on frequency of acute myocardial infarction after coronary artery bypass grafting. Am J Cardiol 2012;110:40-4. [Crossref] [PubMed]
  52. Duan W, Yang Y, Yan J, et al. The effects of curcumin post-treatment against myocardial ischemia and reperfusion by activation of the JAK2/STAT3 signaling pathway. Basic Res Cardiol 2012;107:263. [Crossref] [PubMed]
  53. Liu K, Chen H, You QS, et al. Curcumin attenuates myocardial ischemia-reperfusion injury. Oncotarget 2017;8:112051-9. [Crossref] [PubMed]
  54. Ilyas EI, Nur BM, Laksono SP, et al. Effects of Curcumin on Parameters of Myocardial Oxidative Stress and of Mitochondrial Glutathione Turnover in Reoxygenation after 60 Minutes of Hypoxia in Isolated Perfused Working Guinea Pig Hearts. Adv Pharmacol Sci 2016;2016:6173648. [Crossref] [PubMed]
  55. Wang R, Zhang JY, Zhang M, et al. Curcumin attenuates IR-induced myocardial injury by activating SIRT3. Eur Rev Med Pharmacol Sci 2018;22:1150-60. [PubMed]
  56. Fiorillo C, Becatti M, Pensalfini A, et al. Curcumin protects cardiac cells against ischemia-reperfusion injury: effects on oxidative stress, NF-kappaB, and JNK pathways. Free Radic Biol Med 2008;45:839-46. [Crossref] [PubMed]
  57. Yu YP, Zhou CI, Fu YF, et al. Effects of curcumin on ischemia/reperfusion induced apoptosis of H9c2 myocardial cells and the expression of glycogen synthase kinase-3 and its phosphorylation. Zhongguo Zhong Xi Yi Jie He Za Zhi 2013;33:240-3. [PubMed]
  58. Yu YP, Huang XM, Fu YF. Curcumin protects H9c2 cardiomyocyte against ischemia/reperfusion injury through inactivation of glycogen synthase kinase-3. Int J Clin Exp Pathol 2016;9:3226-32.
  59. Huang Z, Ye B, Dai Z, et al. Curcumin inhibits autophagy and apoptosis in hypoxia/reoxygenation-induced myocytes. Mol Med Rep 2015;11:4678-84. [Crossref] [PubMed]
  60. Chen XL, Liu XR, Fang YX, et al. Cardioprotective role of curcumin in myocardial ischemia-reperfusion of male albino rats. Int J Clin Exp Med 2016;9:7846-54.
  61. Zhu P, Yang M, He H, et al. Curcumin attenuates hypoxia/reoxygenation-induced cardiomyocyte injury by downregulating Notch signaling. Mol Med Rep 2019;20:1541-50. [Crossref] [PubMed]
  62. Wei W, Peng J, Li J. Curcumin attenuates hypoxia/reoxygenation-induced myocardial injury. Mol Med Rep 2019;20:4821-30. [PubMed]
  63. Guan G, Lei L, Lv Q, et al. Curcumin attenuates palmitic acid-induced cell apoptosis by inhibiting endoplasmic reticulum stress in H9C2 cardiomyocytes. Hum Exp Toxicol 2019;38:655-64. [Crossref] [PubMed]
  64. Luo SY, Li R, Le ZY, et al. Anfibatide protects against rat cerebral ischemia/reperfusion injury via TLR4/JNK/caspase-3 pathway. Eur J Pharmacol 2017;807:127-37. [Crossref] [PubMed]
  65. Ahmad N, Khan Z, Basit A, et al. Neuroprotective agents, natural plant herbs & drugs in ischemic stroke: a review. PharmaTutor 2017;5:29-36.
  66. Ghoneim AI, Abdel-Naim AB, Khalifa AE, et al. Protective effects of curcumin against ischaemia/reperfusion insult in rat forebrain. Pharmacol Res 2002;46:273-9. [Crossref] [PubMed]
  67. Kaptanoglu E, Okutan O, Akbiyik F, et al. Correlation of injury severity and tissue Evans blue content, lipid peroxidation and clinical evaluation in acute spinal cord injury in rats. J Clin Neurosci 2004;11:879-85. [Crossref] [PubMed]
  68. Hall ED. Lipid antioxidants in acute central nervous system injury. Ann Emerg Med 1993;22:1022-7. [Crossref] [PubMed]
  69. Cemil B, Topuz K, Demircan MN, et al. Curcumin improves early functional results after experimental spinal cord injury. Acta Neurochir (Wien) 2010;152:1583-90; discussion 1590. [Crossref] [PubMed]
  70. Sahin Kavaklı H, Koca C, Alıcı O. Antioxidant effects of curcumin in spinal cord injury in rats. Ulus Travma Acil Cerrahi Derg 2011;17:14-8. [Crossref] [PubMed]
  71. Ormond DR, Shannon C, Oppenheim J, et al. Stem cell therapy and curcumin synergistically enhance recovery from spinal cord injury. PLoS One 2014;9:e88916. [Crossref] [PubMed]
  72. Liu ZQ, Xing SS, Zhang W. Neuroprotective effect of curcumin on spinal cord in rabbit model with ischemia/reperfusion. J Spinal Cord Med 2013;36:147-52. [Crossref] [PubMed]
  73. Kurt G, Yildirim Z, Cemil B, et al. Effects of curcumin on acute spinal cord ischemia-reperfusion injury in rabbits Laboratory investigation. J Neurosurg Spine 2014;20:464-70. [Crossref] [PubMed]
  74. Akar İ, İnce İ, Arici A, et al. The Protective Effect of Curcumin on a Spinal Cord Ischemia-Reperfusion Injury Model. Ann Vasc Surg 2017;42:285-92. [Crossref] [PubMed]
  75. Hoffmann TF, Leiderer R, Harris AG, et al. Ischemia and reperfusion in pancreas. Microsc Res Tech 1997;37:557-71. [Crossref] [PubMed]
  76. Sakorafas GH, Tsiotos GG, Sarr MG. Ischemia/Reperfusion-Induced pancreatitis. Dig Surg 2000;17:3-14. [Crossref] [PubMed]
  77. Chen KH, Chao D, Liu CF, et al. Curcumin attenuates airway hyperreactivity induced by ischemia-reperfusion of the pancreas in rats. Transplant Proc 2010;42:744-7. [Crossref] [PubMed]
  78. Malek M, Nematbakhsh M. Renal ischemia/reperfusion injury; from pathophysiology to treatment. J Renal Inj Prev 2015;4:20-7. [PubMed]
  79. Shoskes D, Lapierre C, Cruz-Correa M, et al. Beneficial effects of the bioflavonoids curcumin and quercetin on early function in cadaveric renal transplantation: a randomized placebo controlled trial. Transplantation 2005;80:1556-9. [Crossref] [PubMed]
  80. Shoskes DA. Effect of bioflavonoids quercetin and curcumin on ischemic renal injury: a new class of renoprotective agents. Transplantation 1998;66:147-52. [Crossref] [PubMed]
  81. Zhao Y, Ding C, Xue W, et al. Genome-wide DNA methylation analysis in renal ischemia reperfusion injury. Gene 2017;610:32-43. [Crossref] [PubMed]
  82. Bomsztyk K, Denisenko O. Epigenetic alterations in acute kidney injury. Semin Nephrol 2013;33:327-40. [Crossref] [PubMed]
  83. Yang L, Chen X, Bi Z, et al. Curcumin attenuates renal ischemia reperfusion injury via JNK pathway with the involvement of p300/CBP-mediated histone acetylation. Korean J Physiol Pharmacol 2021;25:413-23. [Crossref] [PubMed]
  84. Ozkisacik S, Yazici M, Gursoy H, et al. Does gradual detorsion protect the ovary against ischemia-reperfusion injury in rats? Pediatr Surg Int 2014;30:437-40. [Crossref] [PubMed]
  85. Sak ME, Soydinc HE, Sak S, et al. The protective effect of curcumin on ischemia-reperfusion injury in rat ovary. Int J Surg 2013;11:967-70. [Crossref] [PubMed]
  86. Eser A, Hizli D, Namuslu M, et al. Protective effect of curcumin on ovarian reserve in a rat ischemia model: an experimental study. Clin Exp Obstet Gynecol 2017;44:453-7. [Crossref] [PubMed]
  87. Bozlu M, Acar D, Cayan S, et al. Protective effect of trapidil on long-term histologic damage in a rat model of testicular ischemia-reperfusion injury. World J Urol 2009;27:117-22. [Crossref] [PubMed]
  88. Wei SM, Yan ZZ, Zhou J. Curcumin attenuates ischemia-reperfusion injury in rat testis. Fertil Steril 2009;91:271-7. [Crossref] [PubMed]
  89. Takhtfooladi MA, Asghari A, Takhtfooladi HA, et al. The protective role of curcumin on testicular tissue after hindlimb ischemia reperfusion in rats. Int Urol Nephrol 2015;47:1605-10. [Crossref] [PubMed]
  90. Shahedi A, Talebi AR, Mirjalili A, et al. Protective effects of curcumin on chromatin quality, sperm parameters, and apoptosis following testicular torsion-detorsion in mice. Clin Exp Reprod Med 2021;48:27-33. [Crossref] [PubMed]
  91. Ilbey YO, Ozbek E, Cekmen M, et al. Protective effect of curcumin in cisplatin-induced oxidative injury in rat testis: mitogen-activated protein kinase and nuclear factor-kappa B signaling pathways. Hum Reprod 2009;24:1717-25. [Crossref] [PubMed]
  92. Mercantepe T, Unal D, Tümkaya L, et al. Protective effects of amifostine, curcumin and caffeic acid phenethyl ester against cisplatin-induced testis tissue damage in rats. Exp Ther Med 2018;15:3404-12. [Crossref] [PubMed]
  93. Khorsandi L, Mirhoseini M, Mohamadpour M, et al. Effect of curcumin on dexamethasone-induced testicular toxicity in mice. Pharm Biol 2013;51:206-12. [Crossref] [PubMed]
  94. Avisrror MU, Fernandez IA, Sánchez AS, et al. Doxazosin and priapism. J Urol 2000;163:238. [Crossref] [PubMed]
  95. Yılmaz Y, Taken K, Atar M, et al. Protective effect of curcumin on priapism and ischemia-reperfusion injury in rats. Eur Rev Med Pharmacol Sci 2015;19:4664-70. [PubMed]
  96. Ashoori F, Suzuki S, Zhou JH, et al. Involvement of lipid peroxidation in necrosis of skin flaps and its suppression by ellagic acid. Plast Reconstr Surg 1994;94:1027-37. [Crossref] [PubMed]
  97. Shoskes DA, Jones EA, Shahed A. Synergy of mycophenolate mofetil and bioflavonoids in prevention of immune and ischemic injury. Transplant Proc 2000;32:798-9. [Crossref] [PubMed]
  98. Jia S, Xie P, Hong SJ, et al. Intravenous curcumin efficacy on healing and scar formation in rabbit ear wounds under nonischemic, ischemic, and ischemia-reperfusion conditions. Wound Repair Regen 2014;22:730-9. [Crossref] [PubMed]
  99. Yen YH, Pu CM, Liu CW, et al. Curcumin accelerates cutaneous wound healing via multiple biological actions: The involvement of TNF-α, MMP-9, α-SMA, and collagen. Int Wound J 2018;15:605-17. [Crossref] [PubMed]
  100. Avci G, Kadioglu H, Sehirli AO, et al. Curcumin protects against ischemia/reperfusion injury in rat skeletal muscle. J Surg Res 2012;172:e39-46. [Crossref] [PubMed]
  101. Liu Y, Chen L, Shen Y, et al. Curcumin Ameliorates Ischemia-Induced Limb Injury Through Immunomodulation. Med Sci Monit 2016;22:2035-42. [Crossref] [PubMed]
  102. Receno CN, Liang C, Korol DL, et al. Effects of Prolonged Dietary Curcumin Exposure on Skeletal Muscle Biochemical and Functional Responses of Aged Male Rats. Int J Mol Sci 2019;20:1178. [Crossref] [PubMed]
  103. Karahan MA, Yalcin S, Aydogan H, et al. Curcumin and dexmedetomidine prevents oxidative stress and renal injury in hind limb ischemia/reperfusion injury in a rat model. Ren Fail 2016;38:693-8. [Crossref] [PubMed]
  104. Takhtfooladi HA, Takhtfooladi MA. Effect of curcumin on lung injury induced by skeletal muscle ischemia/reperfusion in rats. Ulus Travma Acil Cerrahi Derg 2019;25:7-11. [PubMed]
  105. Bo H, Feng X. Post-treatment curcumin reduced ischemia-reperfusion-induced pulmonary injury via the Notch2/Hes-1 pathway. J Int Med Res 2020;48:300060519892432. [Crossref] [PubMed]
  106. Zou HB, Sun XF. The mechanism of curcumin post-treatment relieving lung injuries by regulating miR-21/TLR4/NF-κB signalling pathway. J Int Med Res 2020;48:300060520965809. [Crossref] [PubMed]
  107. He Y, Yue Y, Zheng X, et al. Curcumin, inflammation, and chronic diseases: how are they linked? Molecules 2015;20:9183-213. [Crossref] [PubMed]
  108. Patel SS, Acharya A, Ray RS, et al. Cellular and molecular mechanisms of curcumin in prevention and treatment of disease. Crit Rev Food Sci Nutr 2020;60:887-939. [Crossref] [PubMed]
  109. Moulin S, Arnaud C, Bouyon S, et al. Curcumin prevents chronic intermittent hypoxia-induced myocardial injury. Ther Adv Chronic Dis 2020;11:2040622320922104. [Crossref] [PubMed]
  110. Tyagi N, Qipshidze N, Munjal C, et al. Tetrahydrocurcumin ameliorates homocysteinylated cytochrome-c mediated autophagy in hyperhomocysteinemia mice after cerebral ischemia. J Mol Neurosci 2012;47:128-38. [Crossref] [PubMed]
  111. Wu JX, Zhang LY, Chen YL, et al. Curcumin pretreatment and post-treatment both improve the antioxidative ability of neurons with oxygen-glucose deprivation. Neural Regen Res 2015;10:481-9. [Crossref] [PubMed]
  112. Kakkar V, Muppu SK, Chopra K, et al. Curcumin loaded solid lipid nanoparticles: an efficient formulation approach for cerebral ischemic reperfusion injury in rats. Eur J Pharm Biopharm 2013;85:339-45. [Crossref] [PubMed]
  113. Hardy N, Viola HM, Johnstone VP, et al. Nanoparticle-mediated dual delivery of an antioxidant and a peptide against the L-Type Ca2+ channel enables simultaneous reduction of cardiac ischemia-reperfusion injury. ACS Nano 2015;9:279-89. [Crossref] [PubMed]
  114. Hu JB, Li SJ, Kang XQ, et al. CD44-targeted hyaluronic acid-curcumin prodrug protects renal tubular epithelial cell survival from oxidative stress damage. Carbohydr Polym 2018;193:268-80. [Crossref] [PubMed]
  115. Rathore P, Arora I, Rastogi S, et al. Collagen-curcumin nanocomposites showing an enhanced neuroprotective effect against short term focal cerebral ischemia. RSC Adv 2020;10:2241-53. [Crossref] [PubMed]
  116. Zang X, Zhou J, Zhang X, et al. Ischemia Reperfusion Injury: Opportunities for Nanoparticles. ACS Biomater Sci Eng 2020;6:6528-39. [Crossref] [PubMed]
  117. Hammad FT, Lubbad L. Does curcumin protect against renal dysfunction following reversible unilateral ureteric obstruction in the rat? Eur Surg Res 2011;46:188-93. [Crossref] [PubMed]
  118. Garg AX, Devereaux PJ, Hill A, et al. Oral curcumin in elective abdominal aortic aneurysm repair: a multicentre randomized controlled trial. CMAJ 2018;190:E1273-80. [Crossref] [PubMed]
  119. Oguz A, Kapan M, Onder A, et al. The effects of curcumin on the liver and remote organs after hepatic ischemia reperfusion injury formed with Pringle manoeuvre in rats. Eur Rev Med Pharmacol Sci 2013;17:457-66. [PubMed]
  120. Eser A, Hizli D, Haltas H, et al. Effects of curcumin on ovarian ischemia-reperfusion injury in a rat model. Biomed Rep 2015;3:807-13. [Crossref] [PubMed]
  121. Basaran UN, Dokmeci D, Yalcin O, et al. Effect of curcumin on ipsilateral and contralateral testes after unilateral testicular torsion in a rat model. Urol Int 2008;80:201-7. [Crossref] [PubMed]
  122. Tanwar V, Sachdeva J, Golechha M, et al. Curcumin protects rat myocardium against isoproterenol-induced ischemic injury: attenuation of ventricular dysfunction through increased expression of Hsp27 along with strengthening antioxidant defense system. J Cardiovasc Pharmacol 2010;55:377-84. [Crossref] [PubMed]
  123. Hong D, Zeng X, Xu W, et al. Altered profiles of gene expression in curcumin-treated rats with experimentally induced myocardial infarction. Pharmacol Res 2010;61:142-8. [Crossref] [PubMed]
  124. González-Salazar A, Molina-Jijón E, Correa F, et al. Curcumin protects from cardiac reperfusion damage by attenuation of oxidant stress and mitochondrial dysfunction. Cardiovasc Toxicol 2011;11:357-64. [Crossref] [PubMed]
  125. Prakash P, Misra A, Surin WR, et al. Anti-platelet effects of Curcuma oil in experimental models of myocardial ischemia-reperfusion and thrombosis. Thromb Res 2011;127:111-8. [Crossref] [PubMed]
  126. Kim YS, Kwon JS, Cho YK, et al. Curcumin reduces the cardiac ischemia-reperfusion injury: involvement of the toll-like receptor 2 in cardiomyocytes. J Nutr Biochem 2012;23:1514-23. [Crossref] [PubMed]
  127. Wang NP, Wang ZF, Tootle S, et al. Curcumin promotes cardiac repair and ameliorates cardiac dysfunction following myocardial infarction. Br J Pharmacol 2012;167:1550-62. [Crossref] [PubMed]
  128. Yang Y, Duan W, Lin Y, et al. SIRT1 activation by curcumin pretreatment attenuates mitochondrial oxidative damage induced by myocardial ischemia reperfusion injury. Free Radic Biol Med 2013;65:667-79. [Crossref] [PubMed]
Cite this article as: Teng S, Joseph MJ, Yu H, Hu C, Li X, Hu C. A narrative review of the protective effects of curcumin in treating ischemia-reperfusion injury. Ann Transl Med 2022;10(14):807. doi: 10.21037/atm-22-3178

Download Citation