Open Access

Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation

  • Authors:
    • Aydin Yavuz
    • Ayşegül Küçük
    • Aydan İremnur Ergörün
    • Ali Doğan Dursun
    • Zeynep Yiğman
    • Metin Alkan
    • Mustafa Arslan
  • View Affiliations

  • Published online on: April 3, 2024     https://doi.org/10.3892/etm.2024.12530
  • Article Number: 242
  • Copyright: © Yavuz et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sepsis is a systemic inflammatory response syndrome that develops in the host against microorganisms. This response develops away from the primary infection site and results in end‑organ damage. The present study aimed to investigate the protective and therapeutic effects on lung and kidney tissue of silymarin (S) and dexmedetomidine (DEX) applied 1 h before and after sepsis induced by the cecal ligation and puncture (CLP) method in rats. A total of 62 rats was randomly divided into eight groups: i) Control (n=6); ii) cecal perforation (CLP; n=8); iii) S + CLP (n=8; S + CLP; S administered 1 h before CPL); iv) CLP + S (n=8; S administered 1 h after CLP); v) DEX + CLP (n=8; D + CLP; DEX administered 1 h before CLP); vi) CLP + D (n=8; DEX administered 1 h after CLP); vii) SD + CLP (n=8; S and DEX administered 1 h before CLP) and viii) CLP + SD (n=8; S and DEX administered 1 h after CLP). After the cecum filled with stool, it was tied with 3/0 silk under the ileocecal valve and the anterior surface of the cecum was punctured twice with an 18‑gauge needle. A total of 100 mg/kg silymarin and 100 µg/kg DEX were administered intraperitoneally to the treatment groups. Lung and kidney tissue samples were collected to evaluate biochemical and histopathological parameters. In the histopathological examination, all parameters indicating kidney injury; interstitial edema, peritubular capillary dilatation, vacuolization, ablation of tubular epithelium from the basement membrane, loss of brush border in the proximal tubule epithelium, cell swelling and nuclear defragmentation; were increased in the CLP compared with the control group. Silymarin administration increased kidney damage, including ablation of tubular epithelium from the basement membrane, compared with that in the CLP group. DEX significantly reduced kidney damage compared with the CLP and silymarin groups. The co‑administration of DEX + silymarin decreased kidney damage, although it was not as effective as DEX‑alone. To conclude, intraperitoneal DEX ameliorated injury in CLP rats. DEX + silymarin partially ameliorated injury but silymarin administration increased damage. As a result, silymarin has a negative effects with this dosage and DEX has a protective effect. In the present study, it was determined that using the two drugs together had a greater therapeutic effect than silymarin and no differences in the effects were not observed any when the application times of the agents were changed.

Introduction

Sepsis is a systemic inflammatory response syndrome (SIRS) that develops in the host against microorganisms. This response develops away from the primary infection area and results in end-organ damage (1). The response that occurs during infection in healthy individuals continues with pathogen recognition, control and rapid tissue repair (2,3). Upon activation of the cell-mediated immune response, anti-/pro-inflammatory mediators are released (4,5). Overactivation by powerful pathogens leads to endothelial damage, tissue hypoperfusion, disseminated intravascular coagulation, treatment-resistant shock, multiple organ damage and death (6). Although a number of treatment methods have been developed such as antibiotics, corticosteroids, fluid and adjunctive therapies; SIRS and sepsis have high mortality and morbidity in intensive care units (7). In 2017, 48.9 million cases of sepsis were reported worldwide, of which 11 million resulted in death (8). Similar proportions of mortality and incidence have been reported in European countries (9-14). The current clinical approach to treatment starts with early diagnosis, identification of the source of infection and early antibiotic treatment, with corticosteroids also playing an important role (1). However, although there are studies showing that steroid treatment reduces mortality in sepsis, its effects on long-term mortality are controversial (15,16). Therefore, the effectiveness of novel drugs is being investigated in experimental and clinical studies (17-21).

Milk thistle (Silybum marianum) is a historical medicinal plant and its well-known flavonoid silymarin is an agent that has promising therapeutic efficacy in different clinical studies (22-24). S. marianum is a herbal product used in Ancient Greek medicine to treat gallbladder disorder and protect the liver from toxic agents (25). Furthermore, silymarin preparations have been used to treat liver and other gastrointestinal diseases due to hepatoprotectivity, neuroprotectivity, anti-fungal and anti-cancer activity (23,26,27). The anti-inflammatory activity of silymarin may underlie the positive effects of the agent (26,28). Several studies have demonstrated the anti-inflammatory activities of silymarin, which inhibits interferon-g, IL-4 and IL-10 in a dose-dependent manner (29-31). Silymarin suppresses NF-κB binding transporter gene transcription in a rat model of sepsis (32). In addition to its cell-protective effects via antioxidative and radical scavenging activity, silymarin also acts via specific receptor interactions such as P-glycoproteins and estrogen and nuclear receptors (29). Derivatives of silymarin could provide new avenues for therapeutic applications. However, although certain researchers have reported silymarin to be well-tolerated and safe clinically, there are also conflicting results (24,33-35). While gastrointestinal and neurological side effects were reported in the study by Schrieber et al (33); there are also studies in the literature, in which no adverse events were observed despite using similar or higher doses (34,35). Therefore, it is crucial that this agent be studied experimentally in organs and tissues before use in clinical practice. Furthermore, the origin of the milk thistle plant, from which silymarin is obtained, is along the Mediterranean coast of Europe and therefore, the fact that this herbal flavonoid is quite common in Anatolia (36) was also effective in its selection in the present study as it is possible to obtain pure raw materials from this plant in Turkey, where the present study was performed.

The activation of adrenergic α2 receptors causes hypotension, bradycardia, sedation, arterial and venous vasoconstriction, decreased presynaptic transmitter release, thrombus stabilization, hypothermia, decreased gastric acid secretion and motility and inhibition of lipolysis and pancreatic insulin release (37,38). A number of studies has shown that sepsis is associated with sympathetic overactivation, which may contribute to end-organ damage (39,40). In septic shock, increased endogenous sympathetic outflow plays a major role in maintaining vascular tone and tissue perfusion (41). Despite elevated concentrations of endogenous vasoconstrictors, such as noradrenaline, downregulation of adrenergic receptors and post-receptor signaling pathways leads to significant decline in vascular response (40,41). To prevent the negative consequences of excessive sympathetic flow, researchers have investigated the use of sympathetic blockade in the treatment of sepsis (42,43). According to Pichot et al, inhibiting sympathetic activity with an α2 agonist corrects vascular reactivity by upregulating α1 receptors in septic shock, thereby decreasing the need for vasopressors (44). Similarly, response to norepinephrine decreases following application of lipopolysaccharide and the administration of α2 agonists increases this response in rats (45). Dexmedetomidine (DEX), is one of the most commonly used sedation agents in intensive care (46-48). As a highly selective α2-adrenoreceptor agonist, DEX serves as an adjunctive therapy through pro-inflammatory downregulation and control of the anti-inflammatory response in patients with sepsis (49). DEX suppresses the release of TNF-α, IL-6, IL-8 and high mobility group box-1 (HMGB-1) in human whole blood cultured with lipopolysaccharide (50). The suppressive effect of DEX on proinflammatory mediator production occurs via α2 adrenergic receptors (49). There are numerous experimental and retrospective observational studies on the benefits of this agent in sepsis, which is the most common cause of mortality in intensive care units (8,51). To the best of our knowledge, however, there are still insufficient data on the specific protective benefits of this agent on tissue and organs. Various studies have shown that DEX, similar to silymarin, has potential benefits by inducing antioxidant pathways in different clinical situations such as ischemia-reperfusion, cancer and sepsis (52-55). Therefore, it was hypothesized these two agents together may show strong antioxidant activity and decrease tissue and organ damage.

There are three current approaches frequently used to construct sepsis models: Lipopolysaccharide administration, intravascular or intraperitoneal administration of live bacteria and the cecal ligation and puncture (CLP) method (56). The CLP method provides the closest results to sepsis in humans (57). Although the efficacy of experimental sepsis models in animals and their adaptability to human studies have been discussed for some time (58), the cecal ligation and puncture method still remains valid (59).

The aim of the present study was to investigate the protective and therapeutic effects of silymarin and DEX in CLP-induced sepsis in rat lung and kidney tissues.

Materials and methods

Animal studies

The present study was conducted at the Gazi University Animal Experiments Laboratory (Ankara, Turkey) in July 2021 in accordance with the ARRIVE guidelines (60). The present study was approved by The Local Ethics Committee of Gazi University Animal Experiments (approval no. G.Ü.E.T-20.022; Ankara, Turkey). Animal studies were performed in accordance with The Guide for the Care and Use of Laboratory Animals by the National Institutes of Health (61). A total 62 male Wistar Albino rats (Gazi University Animal Experiments Laboratory, Ankara, Turkey) weighing 225-300 g were used. Rats were kept in a temperature-controlled (21±1˚C) and humidity-controlled (45-55%) room and were maintained under a 12-h light/dark cycle. The animals were fed a standard pellet diet and allowed to drink water ad libitum. Rats were randomly divided into eight groups as follows: i) Control (n=6); ii) cecal perforation (CLP; n=8); iii) silymarin + CLP (n=8; S + CLP; silymarin administered 1 h before CPL); iv) CLP + S (n=8; silymarin administered 1 h after CLP); v) DEX + CLP (n=8; D + CLP; DEX administered 1 h before CLP); vi) CLP + D (n=8; DEX administered 1 h after CLP); vii) SD + CLP (n=8; silymarin and DEX administered 1 h before CLP) and viii) CLP + SD (n=8; silymarin and DEX administered 1 h after CLP).

Rats were anesthetized by 50 mg/kg intramuscular ketamine hydrochloride (Ketalar® vial; Parke-Davis; Pfizer, Inc.) and 10 mg/kg xylazine hydrochloride (Alfazyne; 2%; EGE VET) and placed on a heating pad to maintain their body temperature. Midline laparotomy was performed in rats whose skin was aseptically prepared. The intestines were removed using wet gauze. In the control group, the cecum was manipulated. However, drilling and ligation were not performed.

After the cecum filled with stool, it was tied with 3/0 silk under the ileocecal valve and the anterior surface of the cecum was punctured twice using an 18-gauge needle. No treatment (e.g., dexmedetomidine or slymarin) was applied to the sham or CLP group. Saline was applied to the peritoneal space to minimize heat and fluid loss. A total of 100 mg/kg silymarin (Sigma-Aldrich; Merck KGaA; cat. no. SO292-50G) and 100 µg/kg DEX (Sedodamid; 100 µg/2 ml; Koçak Farma®) was administered intraperitoneally to the treatment groups. All the rats were sacrificed 24 h after the operation; rats were anesthetized with ketamine (50 mg/kg) and xylazine (10 µg/kg) and sacrificed by collecting blood (5-10 ml) from the abdominal aorta. After heartbeat and respiration ceased, rats were monitored for a further 2 min to confirm death. Tissue samples were stored at -70˚C for biochemical analysis and immersed in 10% neutral buffered formalin for histopathological assessment.

In the present study, two rats were lost in the CLP and S + CLP groups and one rat in the CLP + S group. No losses were observed in any of the other groups. In the first 24 h, mortality rates in the CLP and S groups were similar to those reported by Kang et al (32), Al-Kadi et al (62) and Canikli Adıgüzel et al (63).

Histopathological evaluation

Lung and kidney tissue specimens were fixed in 10% neutral-buffered formalin for 48 h at room temperature and embedded in paraffin after routine tissue processing. Tissue specimens were dehydrated through an increasing-grade series of ethanol. Dehydrated specimens were cleared in xylene, infiltrated in liquid paraffin at 60˚C, and embedded in paraffin. Thereafter, 5 µm-thick tissue sections were cut from paraffin blocks using a microtome (Leica SM 2000; Leica Microsystems GmbH) and stained with hematoxylin and eosin (H&E) to analyze histopathological changes. Lung and kidney sections were incubated with hematoxylin and eosin stain solutions for 12 min each, at room temperature. The stained sections were assessed under a light microscope (Leica DM 4000 B; Leica Microsystems GmbH) equipped with a computer, and micrographs were captured using Leica LAS V4.9 software (Leica Microsystems GmbH).

H&E-stained kidney sections were examined under x400 magnification and renal injury was evaluated semi-quantitatively. Histopathological parameters, including interstitial edema, peritubular capillary dilatation, vacuolization, ablation of tubular epithelium from the basement membrane, loss of brush border in the proximal tubule epithelium, cell swelling and nuclear defragmentation, were scored 0-3 (0, none; 1, mild; 2, moderate; 3, severe) and the mean score was determined for each parameter in each group (64).

H&E-stained lung samples were examined under 200x and 400x magnification and lung injury was assessed semi-quantitatively. Alveolar wall thickening, capillary congestion, intra-alveolar hemorrhage and interstitial and intra-alveolar neutrophil infiltration were scored 0-3 (0, none; 1, mild; 2, moderate; 3, severe), and the mean score was determined for each parameter (65).

Biochemical determination

Total antioxidant status (TAS) and total oxidative status (TOS) were analyzed in blood samples. TAS and TOS were measured using test kits according to the manufacturer's instructions (Rel Assay Diagnostics®). TAS levels were calculated as follows: TAS=[(ΔAbsorbance (Abs) H2O-ΔAbs sample)/(ΔAbs H2O-ΔAbs standard)], and the results were expressed in mmol Trolox Eq/l. TOS levels were calculated as follows: TOS=(ΔAbs sample/ΔAbs standard) x standard concentration (10 µmol/l), and the results were expressed in µmol H2O2 Eq/l.

Statistical analysis

All data are expressed as the mean ± standard deviation (SD) or standard error of mean (SEM). The experiments was performed once. All statistical analyses were performed using SPSS (version 26.0; IBM Corp.). The distribution of data was analyzed using the Shapiro-Wilk test. Comparisons of >2 groups were performed using Kruskal-Wallis test followed by Dunn's post hoc test or one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference. The intention to treat analysis method was used (66-68).

Results

Kidney tissue histopathological results

The mean scores for histopathological changes in kidney specimens are summarized in Table I. The severity of interstitial edema in kidney was significantly different between the groups (P=0.003); it was more severe in the CLP, S + CLP, SD + CLP and CLP + SD groups than in the control group (P=0.008, P=0.001, P=0.016 and P=0.004, respectively). Interstitial edema was decreased in the D + CLP group compared with that in the CLP group (P=0.013). The interstitial edema score was significantly lower in the D + CLP and CLP + D groups than in the S + CLP group (P=0.001 and P=0.013, respectively). Peritubular capillary dilatation mean scores were also different (P=0.034), with a significantly higher score in the CLP, S + CLP and CLP + S groups than in the control group (P=0.047, P=0.012 and P=0.012, respectively), whereas it was lower in the CLP + D and CLP + SD groups than in both the S + CLP (P=0.020 and P=0.020, respectively) and CLP + S groups (P=0.020 and P=0.020, respectively; Table I; Fig. 1). Focal cystic formations along the more prominent tubular dilatation were observed in the cortex and medulla of the kidney from S + CLP and CLP + S groups (Fig. 1).

Table I

Histopathological findings in kidney tissue (mean ± SEM).

Table I

Histopathological findings in kidney tissue (mean ± SEM).

Histopathological findingControl, n=6CLP, n=8S + CLP, n=8CLP + S, n=8D + CLP, n=8CLP + D, n=8SD + CLP, n=8CLP + SD, n=8Kruskal Wallis test P-value
Interstitial edema0.33±0.21 1.63±0.26a 2.00±0.50a1.25±0.17 0.50±0.19a,b,c 0.88±0.30a,b 1.50±0.19a 1.75±0.25a0.003
Dilatation of peritubular capillaries0.83±0.31 1.75±0.45a 2.00±0.33a 2.00±0.27a1.63±0.33 1.00±0.19c,d1.25±0.16 1.00±0.27c,d0.034
Vacuolization0.17±0.17 1.75±0.45a 2.50±0.27a 2.25±0.49a 0.38±0.18b,c,d 0.63±0.26*a,c,d 0.38±0.18b,c,d 1.00±0.00*b,d<0.001
Ablation of tubular epithelium from the basement membrane0.50±0.221.00±0.50 1.88±0.40a 2.75±0.17a,b 1.13±0.44d 0.50±0.19c,d 0.38±0.19c,d 0.50±0.19c,d<0.001
Loss of brush border in proximal tubule epithelium1.16±0.31 2.88±0.13a 2.38±0.32a 2.75±0.16a 1.75±0.25b,d 1.75±0.16b,d 2.12±0.30a,b 1.88±0.23b,d<0.001
Cell swelling and nuclear defragmentation1.33±0.21 2.63±0.25a 2.50±0.27a 1.75±0.31b 1.50±0.19b,c 1.38±0.19b,c 1.88±0.30b 1.75±0.31b0.004

[i] P<0.05 vs.

[ii] acontrol,

[iii] bCLP,

[iv] cS + CLP and

[v] dCLP + S. CLP, cecal ligation and puncture; S, silymarin; D, dexmedetomidine; SD, S + D.

Lung tissue histopathological results

The histopathological changes in the lung samples are summarized in Table II. Alveolar wall thickening in lung scores were significantly different between the groups (P<0.0001). Alveolar wall thickening in the CLP, S + CLP, CLP + S, D + CLP, CLP + D and CLP + SD groups was greater than that in the control group (P<0.0001, P<0.0001, P=0.004, P=0.012, P=0.004 and P=0.012, respectively). However, it was significantly reduced in S + CLP, CLP + S, D + CLP, CLP + D, SD + CLP and CLP + SD groups compared with the CLP group (P=0.002, P<0.0001, P<0.0001, P<0.0001 and P<0.0001, respectively). Furthermore, this decrease was more prominent in the SD + CLP group than that of the S + CLP group (P=0.007). The difference in severity of interstitial neutrophil infiltration between the groups was also significant (P=0.015). It was significantly more severe in the CLP than in the control group (P<0.0001), whereas it was improved in the S + CLP, CLP + S, D + CLP, CLP + D, SD + CLP and CLP + SD groups compared with that in the CLP group (P=0.024, P=0.007, P=0.024, P=0.007, P<0.0001 and P=0.007, respectively). By contrast, intra-alveolar neutrophil infiltration scores of all the groups were similar (P=0.158; Table II; Fig. 2).

Table II

Histopathological findings of lung tissue (mean ± SEM).

Table II

Histopathological findings of lung tissue (mean ± SEM).

Histopathological findingControl, n=6CLP, n=8S + CLP, n=8CLP + S, n=8D + CLP, n=8CLP + D, n=8SD + CLP, n=8CLP + SD, n=8Kruskal Wallis test P-value
Thickening in the alveolar wall0.50±0.34 2.88±0.13a 1.88±0.30a,b 1.50±0.27a,b 1.38±0.27a,b,c 1.50±0.19a,b,c 1.00±0.00b,c 1.38±0.18a,b<0.001
Capillary congestion1.00±0.52 2.63±0.18a 2.00±0.19a 1.75±0.32b 1.25±0.16b,c 1.50±0.16b 1.38±0.26b 1.38±0.18b0.001
Intra-alveolar hemorrhage0.00±0.00 1.38±0.32a 1.00±0.36a 0.88±0.35a 0.38±0.26b 0.13±0.13b,c 0.38±0.26b 0.00±0.00b,c,d0.006
Interstitial neutrophil infiltration1.17±0.17 2.25±0.16a 1.62±0.18b 1.50±0.19b 1.63±0.18b 1.50±0.19b 1.25±0.25b 1.50±0.19b0.015
Intra-alveolar neutrophil infiltration0.17±0.170.50±0.190.25±0.160.50±0.190.38±0.260.00±0.000.13±0.130.00±0.000.158

[i] P<0.05 vs.

[ii] acontrol,

[iii] bCLP,

[iv] cS + CLP and

[v] dCLP + S. CLP, cecal ligation and puncture; S, silymarin; D, dexmedetomidine; SD, S + D.

Lung tissue biochemical results

There was a significant difference in lung TOS and TAS levels (P=0.001 and P=0.001, respectively). The TOS levels were significantly higher in the CLP, S + CLP, CLP + S, SD + CLP and CLP + SD groups than in the control group (P<0.0001, P<0.0001, P<0.0001, P=0.044 and P=0.005, respectively). TOS levels were significantly lower in the D + CLP, CLP + D and SD + CLP groups than in the CLP group (P=0.032, P=0.002 and P=0.043, respectively). TOS levels were significantly lower in the D + CLP and CLP + D groups than in the S + CLP group (P=0.041 and P=0.006, respectively; Fig. 3). Similarly, TOS levels were significantly lower in the D + CLP and CLP + D groups than in the CLP + S group (P=0.036 and P=0.003, respectively; Fig. 3).

TAS levels were significantly lower in the CLP, S + CLP and CLP + S groups than in the control group (P=0.002, P=0.039 and P=0.047, respectively). TAS levels were significantly higher in the D + CLP, CLP + D, SD + CLP and CLP + SD groups than in the CLP group (P<0.0001, P<0.0001, P=0.001 and P<0.0001, respectively). Similarly, TAS levels were significantly higher in the D + CLP, CLP + D, SD + CLP and CLP + SD groups than in the S + CLP group (P=0.014, P=0.008, P=0.035 and P=0.021, respectively; Fig. 4).

Kidney tissue biochemical results

There was a significant difference in kidney TOS and TAS levels (P<0.0001 and P<0.0001, respectively). TOS levels were significantly higher in the CLP, S + CLP and CLP + S groups than in the control group (P=0.004, P=0.010 and P=0.027, respectively). TOS levels were significantly lower in the D + CLP, CLP + D and SD + CLP groups than in the CLP group (P<0.0001, P<0.0001 and P=0.006, respectively). TOS levels were significantly lower in the D + CLP, CLP + D and SD + CLP groups than in the S + CLP group (P<0.0001, P=0.001 and P=0.015, respectively). Similarly, TOS levels were significantly lower in the D+ CLP, CLP + D, SD + CLP and CLP+ SD groups than in the CLP + S group (P=0.001, P=0.003, P=0.035 and P=0.042, respectively; Fig. 5).

TAS levels were significantly lower in the CLP, S + CLP and CLP + S groups than in the control group (P<0.0001, P=0.044 and P=0.035, respectively). TAS levels were significantly higher in the D + CLP, CLP + D, SD + CLP and CLP + SD groups than in the CLP group (all P<0.0001). The TAS levels were significantly higher in the D + CLP, CLP + D and CLP + SD groups than in the CLP group (P=0.005, P=0.003 and P=0.012, respectively). Similarly, TAS levels were significantly higher in the D + CLP, CLP + D, and CLP + SD groups than in the S + CLP group (P=0.044, P=0.002, and P=0.009, respectively; Fig. 6).

Discussion

In the clinical use of agents, prophylactic efficacy is as important as therapeutic efficacy. Therefore, the present study aimed to observe both the therapeutic and preventive effects of dexmedetomidine and silymarin. The present study observed differences following application of agents both before and after sepsis modeling.

Silymarin and DEX have been used in different doses in different studies and a definite effective dose has not been determined yet (24,69). In the present study, dose selection was based on similar studies (70-75). Treatment time was also determined based on previous studies, but since both preventive and therapeutic effects were investigated in the clinical sepsis model, separate groups were created for application times (62,76).

Since the polymicrobial peritonitis table created by the CLP model is termed sepsis in studies in the literature (77-84), it was assumed that the clinical picture created by the CLP method in the present study constitutes a sepsis model. CLP, which is an experimental technique, may not mimic sepsis in exactly the same way. Of course, due to the dynamic and developing nature of science, it may be possible to perform more accurate sepsis modeling in the coming years if different techniques are discovered.

In the present study, histopathological damage in lung and kidney tissue following sepsis modeling was observed. However, this damage was accompanied by decreased TAS and increased TOS. Tissue oxidant-antioxidant balance may result in organ damage, which is in line with the literature (85,86). Sepsis is a common clinical problem and silymarin and DEX have shown promising results in recent experimental studies (87-90).

Sepsis is the most common cause of mortality in intensive care units (91). Lung and kidney involvement is relatively common in sepsis, and dysfunction of these organs is associated with poor survival outcome (92,93). Therefore, it has become increasingly important to identify agents that have therapeutic or protective effects on the lungs and kidney during sepsis.

Silymarin is a herbal flavonoid obtained from the seeds or fruits of S. marianum (thistle) (25). Flavonoids, a class of secondary metabolites of plants and fungi, have both prooxidant and antioxidant activity due to their polyphenolic structure (94,95). These effects vary depending on the dose and cell or tissue types (72). For example, Malekinejad et al (96) determined that silymarin applied at the same dose and time had a protective effect on the liver, while increasing damage in the brain. Numerous studies have examined the curative and protective effects of silymarin on kidney and lung tissue through various mechanisms (94,95,97).

Al-Kadi et al showed that 1 h after CLP induction, 100 mg/kg silymarin has a protective effect on kidney tissue (62). Toklu et al (94) studied serum and plasma oxidation markers in lung tissue and concluded that 50 mg/kg per oral silymarin has potential therapeutic efficacy in a similar sepsis model and they found that silymarin may reduce sepsis-induced oxidative organ injury and that this can be attributed to its ability to balance oxidant-antioxidant status. By contrast with previous studies (32,88,94) in the present study, silymarin was administered 1 h before and after sepsis induction. In our study, a decrease in organ damage was observed in the kidney and lung tissues examined in histopathological samples, but no statistically significant difference was detected between the groups. In addition, TAS and TOS measurements did not improve in the silymarin-treated groups (S + CLP, CLP + S). This may indicate that the biochemical improvement reported in the literature (98,99) does not significantly contribute to tissue damage observed in sepsis.

Silymarin improves kidney tissue damage (62,71,95), however, this was not observed in the present study. This may be due to differences in the mechanisms that cause damage (ischemia reperfusion, sepsis, toxicity, malignancy) or changes in the selection of drug doses. Flavonoids have also been shown to have pro-oxidant activity and these pro-oxidant mechanisms are thought to provide anticarcinogenic activity by triggering cell death in malignant cells (100). Therefore, silymarin has different effects on different tissues at different doses (72,96,100). Although the antioxidant activity of silymarin is well-known (22,100,101), further studies are required to understand its protective effects against sepsis and associated organ damage. In the review of Soleimani et al (24), the side effects and doses used in the studies conducted with silymarin were examined and it was seen that it can be used safely at a number of different doses. However, present study suggested that it may have a prooxidant effect on the lung and kidney at the dose used in the experimental sepsis model (100 mg/kg, intraperitoneal). The continued use of silymarin, one of the oldest known plant-derived medicinal agents, in experimental studies may be due to novel effects, as demonstrated in the present study.

Although silymarin has demonstrated promising results in numerous clinical situations (31,35,72), it needs larger studies with different doses and drug combinations before it can be used clinically for its therapeutic or prophylactic effects. The present study evaluated both preventive and therapeutic efficacy, performed with the one of the highest intraperitoneal doses found in the literature (102) and also including interaction with a different agent. It was hypothesized that the dose of silymarin used had a pro-oxidant effect, as in other studies (72,96,100), and that this is why the animal losses occurred. Using the two drugs together had a greater therapeutic effect than silymarin.

DEX is a α2-adrenergic receptor agonist that exerts sympatholytic effects such as anxiolysis, sedation and analgesia in certain regions of the brain (103). Owing to the absence of side effects such as respiratory depression, it is a frequently preferred agent for sedation in intensive care units (104). The positive effects of DEX on in vitro experimental sepsis models have been reported in literature (105,106). For example, Koca et al (107) applied 50 µg/kg DEX to rats and observed improvements in both histomorphological and immunohistochemical findings in a sepsis model using CLP technique. Different hypotheses have been proposed for the similar organ-protective effects of DEX and positive results have been obtained. Li et al (108) suggested that the lung protective effect of dexmedetomidine in septic rats was achieved through increasing vagal tone; Wu et al (109), in the same experimental model, argued that the protective effect occurs through the TLR4/NF-κB pathway. Qiu et al (76) observed that DEX decreased acute renal failure and increased survival in a sepsis model. They also suggested that this effect occurred via the NF-KB pathway induced by lipoxin A4.

In the present study, it was hypothesized that DEX, which is known to regulate the oxidant-antioxidant balance in ischemia-reperfusion models (74,75), may show organ-protective effects in sepsis through oxidant-antioxidant balance pathway. Statistically significant positive effects were observed at both the histopathological and biochemical levels in the DEX treatment groups (D + CLP, CLP + D, SD + CLP, CLP + SD). DEX improved lung and kidney tissue damage in the treatment groups. The effects of DEX administration before and after sepsis were not significantly different.

In the present study, DEX application statistically significantly increased the total antioxidant score in tissues and decreased the oxidant score. In a study conducted by Şengel et al (110), TAS and TOS scores in kidney tissue after DEX application showed similar changes as in the present study and a statistically significant improvement in histopathological damage was observed. These data support the hypothesis that DEX may exert positive effects on the lung and kidneys during sepsis. The positive effects of DEX on both TAS and TOS levels and pathological examinations may be a guide for further studies on its mechanism of action.

In previous studies, silymarin and dexmedetomidine have been studied together with different combinations (98,111-116), but no study has been performed in which these two agents were used together. Thus, the present study aimed to investigate the effects of these two agents used together. However, a limitation of the present study was that the mechanism of action of these agents was not examined, and only tissue and organ results were studied. Further studies should study the mechanisms of the effects of these agents.

Although the present study aimed to observe the prophylactic effects of the agents before and after sepsis induction, no differences in the effects were observed when the application times of the agents were changed; however, it may be possible to obtain different results using larger sample sizes. The present study compared both the interactions and preventive and therapeutic effects of promising agents in an experimental sepsis model.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

MAr and AK designed the study and analyzed and interpreted the data. ADD, AY, MAl and ZY performed the experiments. MAl, AİE and ZY confirm the authenticity of all the raw data. AİE, AK, MArr and MAl critically revised the article for important intellectual content. AİE and collected samples. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Ethical approval for the study was obtained from The Gazi University Experimental Animals Ethics Committee (Ankara, Turkey; approval no: G.Ü.E.T-20.022).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Evans L, Rhodes A, Alhazzani W, Antonelli M, Coopersmith CM, French C, Machado FR, Mcintyre L, Ostermann M, Prescott HC, et al: Surviving sepsis campaign: International guidelines for management of sepsis and septic shock 2021. Intensive Care Med. 47:1181–1247. 2021.PubMed/NCBI View Article : Google Scholar

2 

Beisel WR: Metabolic response to infection. Annu Rev Med. 26:9–20. 1975.PubMed/NCBI View Article : Google Scholar

3 

Dyck B, Unterberg M, Adamzik M and Koos B: The impact of pathogens on sepsis prevalence and outcome. Pathogens. 13(89)2024.PubMed/NCBI View Article : Google Scholar

4 

Aziz M, Jacob A, Yang WL, Matsuda A and Wang P: Current trends in inflammatory and immunomodulatory mediators in sepsis. J Leukoc Biol. 93:329–342. 2013.PubMed/NCBI View Article : Google Scholar

5 

Chen XH, Yin YJ and Zhang JX: Sepsis and immune response. World J Emerg Med. 2:88–92. 2011.PubMed/NCBI

6 

Arina P and Singer M: Pathophysiology of sepsis. Curr Opin Anaesthesiol. 34:77–84. 2021.PubMed/NCBI View Article : Google Scholar

7 

Fleischmann-Struzek C, Mellhammar L, Rose N, Cassini A, Rudd KE, Schlattmann P, Allegranzi B and Reinhart K: Incidence and mortality of hospital- and ICU-treated sepsis: Results from an updated and expanded systematic review and meta-analysis. Intensive Care Med. 46:1552–1562. 2020.PubMed/NCBI View Article : Google Scholar

8 

Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al: Global, regional, and national sepsis incidence and mortality, 1990-2017: Analysis for the global burden of disease study. Lancet. 395:200–211. 2020.PubMed/NCBI View Article : Google Scholar

9 

Engel C, Brunkhorst FM, Bone HG, Brunkhorst R, Gerlach H, Grond S, Gruendling M, Huhle G, Jaschinski U, John S, et al: Epidemiology of sepsis in Germany: Results from a national prospective multicenter study. Intensive Care Med. 33:606–618. 2007.PubMed/NCBI View Article : Google Scholar

10 

Karlsson S, Varpula M, Ruokonen E, Pettilä V, Parviainen I, Ala-Kokko TI, Kolho E and Rintala EM: Incidence, treatment, and outcome of severe sepsis in ICU-treated adults in Finland: The Finnsepsis study. Intensive Care Med. 33:435–443. 2007.PubMed/NCBI View Article : Google Scholar

11 

Blanco J, Muriel-Bombín A, Sagredo V, Taboada F, Gandía F, Tamayo L, Collado J, García-Labattut A, Carriedo D, Valledor M, et al: Incidence, organ dysfunction and mortality in severe sepsis: A Spanish multicentre study. Crit Care. 12(R158)2008.PubMed/NCBI View Article : Google Scholar

12 

Sakr Y, Elia C, Mascia L, Barberis B, Cardellino S, Livigni S, Fiore G, Filippini C and Ranieri VM: Epidemiology and outcome of sepsis syndromes in Italian ICUs: A muticentre, observational cohort study in the region of Piedmont. Minerva Anestesiol. 79:993–1002. 2013.PubMed/NCBI

13 

Weng L, Xu Y, Yin P, Wang Y, Chen Y, Liu W, Li S, Peng JM, Dong R, Hu XY, et al: National incidence and mortality of hospitalized sepsis in China. Crit Care. 27(84)2023.PubMed/NCBI View Article : Google Scholar

14 

Rhee C, Dantes R, Epstein L, Murphy DJ, Seymour CW, Iwashyna TJ, Kadri SS, Angus DC, Danner RL, Fiore AE, et al: Incidence and trends of sepsis in US hospitals using clinical vs claims data, 2009-2014. JAMA. 318:1241–1249. 2017.PubMed/NCBI View Article : Google Scholar

15 

Lamontagne F, Rochwerg B, Lytvyn L, Guyatt GH, Møller MH, Annane D, Kho ME, Adhikari NKJ, Machado F, Vandvik PO, et al: Corticosteroid therapy for sepsis: A clinical practice guideline. BMJ. 362(k3284)2018.PubMed/NCBI View Article : Google Scholar

16 

Rochwerg B, Oczkowski SJ, Siemieniuk RAC, Agoritsas T, Belley-Cote E, D'Aragon F, Duan E, English S, Gossack-Keenan K, Alghuroba M, et al: Corticosteroids in sepsis: An updated systematic review and meta-analysis. Crit Care Med. 46:1411–1420. 2018.PubMed/NCBI View Article : Google Scholar

17 

Vignon P, Laterre PF, Daix T and François B: New agents in development for sepsis: Any reason for hope? Drugs. 80:1751–1761. 2020.PubMed/NCBI View Article : Google Scholar

18 

Üstündağ H, Doğanay S, Kalındemirtaş FD, Demir Ö, Huyut MT, Kurt N, Özgeriş FB and Akbaba Ö: A new treatment approach: Melatonin and ascorbic acid synergy shields against sepsis-induced heart and kidney damage in male rats. Life Sci. 329(121875)2023.PubMed/NCBI View Article : Google Scholar

19 

Usmani J, Khan T, Ahmad R and Sharma M: Potential role of herbal medicines as a novel approach in sepsis treatment. Biomed Pharmacother. 144(112337)2021.PubMed/NCBI View Article : Google Scholar

20 

Zhang W, Jiang H, Wu G, Huang P, Wang H, An H, Liu S and Zhang W: The pathogenesis and potential therapeutic targets in sepsis. MedComm (2020). 4(e418)2023.PubMed/NCBI View Article : Google Scholar

21 

Holubar M, Meng L, Alegria W and Deresinski S: Bacteremia due to methicillin-resistant staphylococcus aureus: An update on new therapeutic approaches. Infect Dis Clin North Am. 34:849–861. 2020.PubMed/NCBI View Article : Google Scholar

22 

Hadi A, Pourmasoumi M, Mohammadi H, Symonds M and Miraghajani M: The effects of silymarin supplementation on metabolic status and oxidative stress in patients with type 2 diabetes mellitus: A systematic review and meta-analysis of clinical trials. Complement Ther Med. 41:311–319. 2018.PubMed/NCBI View Article : Google Scholar

23 

Koltai T and Fliegel L: Role of silymarin in cancer treatment: Facts, hypotheses, and questions. J Evid Based Integr Med. 27(2515690X211068826)2022.PubMed/NCBI View Article : Google Scholar

24 

Soleimani V, Delghandi PS, Moallem SA and Karimi G: Safety and toxicity of silymarin, the major constituent of milk thistle extract: An updated review. Phytother Res. 33:1627–1638. 2019.PubMed/NCBI View Article : Google Scholar

25 

Abenavoli L, Izzo AA, Milić N, Cicala C, Santini A and Capasso R: Milk thistle (Silybum marianum): A concise overview on its chemistry, pharmacological, and nutraceutical uses in liver diseases. Phytother Res. 32:2202–2213. 2018.PubMed/NCBI View Article : Google Scholar

26 

Camini FC and Costa DC: Silymarin: Not just another antioxidant. J Basic Clin Physiol Pharmacol. 31(20190206)2020.PubMed/NCBI View Article : Google Scholar

27 

Tighe SP, Akhtar D, Iqbal U and Ahmed A: Chronic liver disease and silymarin: A biochemical and clinical review. J Clin Transl Hepatol. 8:454–458. 2020.PubMed/NCBI View Article : Google Scholar

28 

Aghazadeh S, Amini R, Yazdanparast R and Ghaffari SH: Anti-apoptotic and anti-inflammatory effects of Silybum marianum in treatment of experimental steatohepatitis. Exp Toxicol Pathol. 63:569–574. 2011.PubMed/NCBI View Article : Google Scholar

29 

Saller R, Melzer J, Reichling J, Brignoli R and Meier R: An updated systematic review of the pharmacology of silymarin. Forsch Komplementmed. 14:70–80. 2007.PubMed/NCBI View Article : Google Scholar

30 

Surai A and Surai PF: Chapter 10 Silymarin and inflammation: From understanding molecular mechanisms to practical applications. In: Silymarin Puzzle. Wageningen Academic, pp287-317, 2023.

31 

Sharma S, Kumar P, Ashawat MS, Pandit V, Verma CS and Sharma DK: Silymarin: A Phytoconstituent with Significant Therapeutic Potential-A Narrative Review. Curr Drug Ther. 18:89–97. 2023.

32 

Kang JS, Jeon YJ, Park SK, Yang KH and Kim HM: Protection against lipopolysaccharide-induced sepsis and inhibition of interleukin-1beta and prostaglandin E2 synthesis by silymarin. Biochem Pharmacol. 67:175–181. 2004.PubMed/NCBI View Article : Google Scholar

33 

Schrieber SJ, Hawke RL, Wen Z, Smith PC, Reddy KR, Wahed AS, Belle SH, Afdhal NH, Navarro VJ, Meyers CM, et al: Differences in the disposition of silymarin between patients with nonalcoholic fatty liver disease and chronic hepatitis C. Drug Metab Dispos. 39:2182–2190. 2011.PubMed/NCBI View Article : Google Scholar

34 

Shahbazi F, Sadighi S, Dashti-Khavidaki S, Shahi F, Mirzania M, Abdollahi A and Ghahremani MH: Effect of silymarin administration on cisplatin nephrotoxicity: Report from a pilot, randomized, double-blinded, placebo-controlled clinical trial. Phytother Res. 29:1046–1053. 2015.PubMed/NCBI View Article : Google Scholar

35 

Fried MW, Navarro VJ, Afdhal N, Belle SH, Wahed AS, Hawke RL, Doo E, Meyers CM and Reddy KR: Silymarin in NASH and C Hepatitis (SyNCH) Study Group. Effect of silymarin (milk thistle) on liver disease in patients with chronic hepatitis C unsuccessfully treated with interferon therapy: A randomized controlled trial. JAMA. 308:274–282. 2012.PubMed/NCBI View Article : Google Scholar

36 

Marmouzi I, Bouyahya A, Ezzat SM, El Jemli M and Kharbach M: The food plant Silybum marianum (L.) Gaertn.: Phytochemistry, Ethnopharmacology and clinical evidence. J Ethnopharmacol. 265(113303)2021.PubMed/NCBI View Article : Google Scholar

37 

Nguyen V, Tiemann D, Park E adz and Salehi A: Alpha-2 agonists. Anesthesiol Clin. 35:233–245. 2017.PubMed/NCBI View Article : Google Scholar

38 

Giovannitti JA Jr, Thoms SM and Crawford JJ: Alpha-2 adrenergic receptor agonists: A review of current clinical applications. Anesth Prog. 62:31–39. 2015.PubMed/NCBI View Article : Google Scholar

39 

Cioccari L, Luethi N, Bailey M, Shehabi Y, Howe B, Messmer AS, Proimos HK, Peck L, Young H, Eastwood GM, et al: The effect of dexmedetomidine on vasopressor requirements in patients with septic shock: A subgroup analysis of the Sedation Practice in Intensive care evaluation [SPICE III] trial. Crit Care. 24(441)2020.PubMed/NCBI View Article : Google Scholar

40 

Ferreira J: The Theory is out there: The use of ALPHA-2 agonists in treatment of septic shock. Shock. 49:358–363. 2018.PubMed/NCBI View Article : Google Scholar

41 

Morelli A, Sanfilippo F, Arnemann P, Hessler M, Kampmeier TG, D'Egidio A, Orecchioni A, Santonocito C, Frati G, Greco E, et al: The effect of propofol and dexmedetomidine sedation on norepinephrine requirements in septic shock patients: A crossover trial. Crit Care Med. 47:e89–e95. 2019.PubMed/NCBI View Article : Google Scholar

42 

Suzuki T, Suzuki Y, Okuda J, Kurazumi T, Suhara T, Ueda T, Nagata H and Morisaki H: Sepsis-induced cardiac dysfunction and β-adrenergic blockade therapy for sepsis. J Intensive Care. 5(22)2017.PubMed/NCBI View Article : Google Scholar

43 

Ferreira JA and Bissell BD: Misdirected sympathy: The role of sympatholysis in sepsis and septic shock. J Intensive Care Med. 33:74–86. 2018.PubMed/NCBI View Article : Google Scholar

44 

Pichot C, Géloën A, Ghignone M and Quintin L: Alpha-2 agonists to reduce vasopressor requirements in septic shock? Med Hypotheses. 75:652–656. 2010.PubMed/NCBI View Article : Google Scholar

45 

Geloen A, Chapelier K, Cividjian A, Dantony E, Rabilloud M, May CN and Quintin L: Clonidine and dexmedetomidine increase the pressor response to norepinephrine in experimental sepsis: A pilot study. Crit Care Med. 41:e431–e438. 2013.PubMed/NCBI View Article : Google Scholar

46 

Møller MH, Alhazzani W, Lewis K, Belley-Cote E, Granholm A, Centofanti J, McIntyre WB, Spence J, Al Duhailib Z, Needham DM, et al: Use of dexmedetomidine for sedation in mechanically ventilated adult ICU patients: A rapid practice guideline. Intensive Care Med. 48:801–810. 2022.PubMed/NCBI View Article : Google Scholar

47 

Wiegand A, Behal M, Robbins B, Bissell B, Pandya K and Mefford B: Niche roles for dexmedetomidine in the intensive care unit. Ann Pharmacother. 57:1207–1220. 2023.PubMed/NCBI View Article : Google Scholar

48 

Page V and McKenzie C: Sedation in the intensive care unit. Curr Anesthesiol Rep. 11:92–100. 2021.PubMed/NCBI View Article : Google Scholar

49 

Zi SF, Li JH, Liu L, Deng C, Ao X, Chen DD and Wu SZ: Dexmedetomidine-mediated protection against septic liver injury depends on TLR4/MyD88/NF-κB signaling downregulation partly via cholinergic anti-inflammatory mechanisms. Int Immunopharmacol. 76(105898)2019.PubMed/NCBI View Article : Google Scholar

50 

Chang Y, Huang X, Liu Z, Han G, Huang L, Xiong YC and Wang Z: Dexmedetomidine inhibits the secretion of high mobility group box 1 from lipopolysaccharide-activated macrophages in vitro. J Surg Res. 181:308–314. 2013.PubMed/NCBI View Article : Google Scholar

51 

Zhang T, Mei Q, Dai S, Liu Y and Zhu H: Use of dexmedetomidine in patients with sepsis: A systematic review and meta-analysis of randomized-controlled trials. Ann Intensive Care. 12(81)2022.PubMed/NCBI View Article : Google Scholar

52 

Zhang XY, Liu ZM, Wen SH, Li YS, Li Y, Yao X, Huang WQ and Liu KX: Dexmedetomidine administration before, but not after, ischemia attenuates intestinal injury induced by intestinal ischemia-reperfusion in rats. Anesthesiology. 116:1035–1046. 2012.PubMed/NCBI View Article : Google Scholar

53 

Kuyrukluyildiz U, Delen LA, Onk D, Yazici GN, Gulaboglu M and Suleyman H: The effect of dexmedetomidine on gastric ischemia reperfusion injury in rats. Biochemical and histopathological evaluation. Acta Cir Bras. 36(e360104)2021.PubMed/NCBI View Article : Google Scholar

54 

Kotanoğlu MS, Kadioğlu E, Emerce E, Kaymak C, Özcan A and Başar H: Antioxidant effects of dexmedetomidine against hydrogen peroxide-induced DNA damage in vitro by alkaline Comet assay. Turk J Med Sci. 50:1393–1398. 2020.PubMed/NCBI View Article : Google Scholar

55 

Li W, Chen M, Gong Y, Lin F and Sun C: Effects of dexmedetomidine on oxidative stress, programmed cell death, liver function, and expression of peripheral immune cells in patients with primary liver cancer undergoing hepatectomy. Front Physiol. 14(1159746)2023.PubMed/NCBI View Article : Google Scholar

56 

Poli-de-Figueiredo LF, Garrido AG, Nakagawa N and Sannomiya P: Experimental models of sepsis and their clinical relevance. Shock. 30 (Suppl 1):S53–S59. 2008.PubMed/NCBI View Article : Google Scholar

57 

Sjaastad FV, Jensen IJ, Berton RR, Badovinac VP and Griffith TS: Inducing experimental polymicrobial sepsis by cecal ligation and puncture. Curr Protoc Immunol. 131(e110)2020.PubMed/NCBI View Article : Google Scholar

58 

Alverdy JC, Keskey R and Thewissen R: Can the cecal ligation and puncture model be repurposed to better inform therapy in human sepsis? Infect Immun. 88:e00942–19. 2020.PubMed/NCBI View Article : Google Scholar

59 

Drechsler S and Osuchowski M: Cecal ligation and puncture. In: Sepsis: Methods and Protocols; Walker WE (ed). Springer: New York, NY, USA, pp1-8, 2021.

60 

Percie du Sert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, Browne WJ, Clark A, Cuthill IC, Dirnagl U, et al: The ARRIVE guidelines 2.0: Updated guidelines for reporting animal research. J Cereb Blood Flow Metab. 40:1769–1777. 2020.PubMed/NCBI View Article : Google Scholar

61 

Care IoLARCo, Animals UoL: Guide for the care and use of laboratory animals: US Department of Health and Human Services, Public Health Service, National Academies Press (US), 2011.

62 

Al-Kadi A, Ahmed AS, El-Tahawy NFG, Khalifa MMA and El-Daly M: Silymarin protects against sepsis-induced acute liver and kidney injury via anti-inflammatory and antioxidant mechanisms in the rat. J Adv Biomed Pharm Sci. 3:190–197. 2020.

63 

Canikli Adıgüzel Ş, Pirat A, Türkoğlu S, Bayraktar N, Özen Ö and Kaya M: A rat model of acute respiratory distress silymarin's antiinflamatory and antioxidant effect. J Turk Soc Intens Care. 14:18–27. 2016.

64 

Schick MA, Isbary TJ, Schlegel N, Brugger J, Waschke J, Muellenbach R, Roewer N and Wunder C: The impact of crystalloid and colloid infusion on the kidney in rodent sepsis. Intensive Care Med. 36:541–548. 2010.PubMed/NCBI View Article : Google Scholar

65 

Li XH, Gong X, Zhang L, Jiang R, Li HZ, Wu MJ and Wan JY: Protective effects of polydatin on septic lung injury in mice via upregulation of HO-1. Mediators Inflamm. 2013(354087)2013.PubMed/NCBI View Article : Google Scholar

66 

Abraha I, Cozzolino F, Orso M, Marchesi M, Germani A, Lombardo G, Eusebi P, De Florio R, Luchetta ML, Iorio A and Montedori A: A systematic review found that deviations from intention-to-treat are common in randomized trials and systematic reviews. J Clin Epidemiol. 84:37–46. 2017.PubMed/NCBI View Article : Google Scholar

67 

Gupta SK: Intention-to-treat concept: A review. Perspect Clin Res. 2:109–112. 2011.PubMed/NCBI View Article : Google Scholar

68 

Tripepi G, Chesnaye NC, Dekker FW, Zoccali C and Jager KJ: Intention to treat and per protocol analysis in clinical trials. Nephrology (Carlton). 25:513–517. 2020.PubMed/NCBI View Article : Google Scholar

69 

Dardalas I, Stamoula E, Rigopoulos P, Malliou F, Tsaousi G, Aidoni Z, Grosomanidis V, Milonas A, Papazisis G, Kouvelas D and Pourzitaki C: Dexmedetomidine effects in different experimental sepsis in vivo models. Eur J Pharmacol. 856(172401)2019.PubMed/NCBI View Article : Google Scholar

70 

Ni J, He J, Kang L, Zhong Z, Wang L and Yin S: Effects of dexmedetomidine pretreatment on rats with sepsis-induced acute kidney injury and miR-146a expression. Cell Mol Biol (Noisy-le-grand). 66:93–98. 2020.PubMed/NCBI

71 

Ustyol L, Demiroren K, Kandemir I, Erten R, Bulan K, Kaba S, Demir N and Basunlu MT: Comparative nephroprotective effects of silymarin, N-acetylcysteine, and thymoquinone against carbon tetrachloride-induced nephrotoxicity in rats. Iran Red Crescent Med J. 19(e37746)2017.

72 

Guzel S, Sahinogullari ZU, Canacankatan N, Antmen SE, Kibar D and Coskun Yilmaz B: Potential renoprotective effects of silymarin against vancomycin-induced nephrotoxicity in rats. Drug Chem Toxicol. 43:630–636. 2020.PubMed/NCBI View Article : Google Scholar

73 

Ozdemir A, Topçu A, Mercantepe T, Arpa M, Karakaş SM, Ozdemir A, Tümkaya L and Mercantepe F: The effects of dexmedetomidine on early acute kidney injury in severely burned rats. Eur Rev Med Pharmacol Sci. 27:1311–1321. 2023.PubMed/NCBI View Article : Google Scholar

74 

Gonullu E, Ozkardesler S, Kume T, Duru LS, Akan M, Guneli ME, Ergur BU, Meseri R and Dora O: Comparison of the effects of dexmedetomidine administered at two different times on renal ischemia/reperfusion injury in rats. Braz J Anesthesiol. 64:152–158. 2014.PubMed/NCBI View Article : Google Scholar

75 

Cakir M, Polat A, Tekin S, Vardi N, Taslidere E, Rumeysa Duran Z and Tanbek K: The effect of dexmedetomidine against oxidative and tubular damage induced by renal ischemia reperfusion in rats. Ren Fail. 37:704–708. 2015.PubMed/NCBI View Article : Google Scholar

76 

Qiu R, Yao W, Ji H, Yuan D, Gao X, Sha W, Wang F, Huang P and Hei Z: Dexmedetomidine restores septic renal function via promoting inflammation resolution in a rat sepsis model. Life Sci. 204:1–8. 2018.PubMed/NCBI View Article : Google Scholar

77 

Tanaka S, Genève C, Zappella N, Yong-Sang J, Planesse C, Louedec L, Viranaïcken W, Bringart M, Montravers P, Denamur E, et al: Reconstituted high-density lipoprotein therapy improves survival in mouse models of sepsis. Anesthesiology. 132:825–838. 2020.PubMed/NCBI View Article : Google Scholar

78 

Bedet A, Voiriot G, Ternacle J, Marcos E, Adnot S, Derumeaux G and Mekontso Dessap A: Heart rate control during experimental sepsis in mice: Comparison of ivabradine and β-blockers. Anesthesiology. 132:321–329. 2020.PubMed/NCBI View Article : Google Scholar

79 

Zhong M, Wu W, Wang Y, Mao H, Song J, Chen S and Zhu D: Inhibition of sphingosine kinase 1 attenuates sepsis-induced microvascular leakage via inhibiting macrophage NLRP3 inflammasome activation in mice. Anesthesiology. 132:1503–1515. 2020.PubMed/NCBI View Article : Google Scholar

80 

Park D, Ro M, Lee A-J, Kwak DW, Chung Y and Kim JH: Contributory role of BLT2 in the production of proinflammatory cytokines in cecal ligation and puncture-induced sepsis. Mol Cells. 44:893–899. 2021.PubMed/NCBI View Article : Google Scholar

81 

Abdelnaser M, Alaaeldin R, Attya ME and Fathy M: Hepatoprotective potential of gabapentin in cecal ligation and puncture-induced sepsis; targeting oxidative stress, apoptosis, and NF-kB/MAPK signaling pathways. Life Sci. 320(121562)2023.PubMed/NCBI View Article : Google Scholar

82 

Kim GO, Kim N, Song GY and Bae JS: Inhibitory activities of rare ginsenoside Rg4 on cecal ligation and puncture-induced sepsis. Int J Mol Sci. 23(10836)2022.PubMed/NCBI View Article : Google Scholar

83 

Li J, Li M, Li L, Ma J, Yao C and Yao S: Hydrogen sulfide attenuates ferroptosis and stimulates autophagy by blocking mTOR signaling in sepsis-induced acute lung injury. Mol Immunol. 141:318–327. 2022.PubMed/NCBI View Article : Google Scholar

84 

Tripathi AS, Awasthi S, Maurya RK, Yasir M, Mohapatra L and Srivastav V: Protective effect of vanillin on the management of cecal ligation and puncture induced sepsis rat model. Microb Pathog. 165(105493)2022.PubMed/NCBI View Article : Google Scholar

85 

Daenen K, Andries A, Mekahli D, Van Schepdael A, Jouret F and Bammens B: Oxidative stress in chronic kidney disease. Pediatr Nephrol. 34:975–991. 2019.PubMed/NCBI View Article : Google Scholar

86 

Krzemińska J, Wronka M, Młynarska E, Franczyk B and Rysz J: Arterial hypertension-oxidative stress and inflammation. Antioxidants (Basel). 11(172)2022.PubMed/NCBI View Article : Google Scholar

87 

Zhao X, Wang H, Yang Y, Gou Y, Wang Z, Yang D and Li C: Protective effects of silymarin against D-Gal/LPS-induced organ damage and inflammation in mice. Drug Des Devel Ther. 15:1903–1914. 2021.PubMed/NCBI View Article : Google Scholar

88 

Alikiaii B, Bagherniya M, Askari G, Johnston TP and Sahebkar A: The role of phytochemicals in sepsis: A mechanistic and therapeutic perspective. Biofactors. 47:19–40. 2021.PubMed/NCBI View Article : Google Scholar

89 

Mei B, Li J and Zuo Z: Dexmedetomidine attenuates sepsis-associated inflammation and encephalopathy via central α2A adrenoceptor. Brain Behav Immun. 91:296–314. 2021.PubMed/NCBI View Article : Google Scholar

90 

Hu H, An S, Sha T, Wu F, Jin Y, Li L, Zeng Z, Wu J and Chen Z: Association between dexmedetomidine administration and outcomes in critically ill patients with sepsis-associated acute kidney injury. J Clin Anesth. 83(110960)2022.PubMed/NCBI View Article : Google Scholar

91 

Esposito S, De Simone G, Boccia G, De Caro F and Pagliano P: Sepsis and septic shock: New definitions, new diagnostic and therapeutic approaches. J Glob Antimicrob Resist. 10:204–212. 2017.PubMed/NCBI View Article : Google Scholar

92 

Fujishima S: Organ dysfunction as a new standard for defining sepsis. Inflamm Regen. 36(24)2016.PubMed/NCBI View Article : Google Scholar

93 

Sun S, Chen R, Dou X, Dai M, Long J, Wu Y and Lin Y: Immunoregulatory mechanism of acute kidney injury in sepsis: A narrative review. Biomed Pharmacother. 159(114202)2023.PubMed/NCBI View Article : Google Scholar

94 

Toklu HZ, Akbay TT, Velioglu-Ogunc A, Ercan F, Gedik N, Keyer-Uysal M and Sener G: Silymarin, the antioxidant component of Silybum marianum, prevents sepsis-induced acute lung and brain injury. J Surg Res. 145:214–222. 2008.PubMed/NCBI View Article : Google Scholar

95 

Turgut F, Bayrak O, Catal F, Bayrak R, Atmaca AF, Koc A, Akbas A, Akcay A and Unal D: Antioxidant and protective effects of silymarin on ischemia and reperfusion injury in the kidney tissues of rats. Int Urol Nephrol. 40:453–460. 2008.PubMed/NCBI View Article : Google Scholar

96 

Malekinejad H, Rahmani F, Valivande-Azar S, Taheri-Broujerdi M and Bazargani-Gilani B: Long-term administration of Silymarin augments proinflammatory mediators in the hippocampus of rats: Evidence for antioxidant and pro-oxidant effects. Hum Exp Toxicol. 31:921–930. 2012.PubMed/NCBI View Article : Google Scholar

97 

Sajedianfard J, Nazifi S, Izadi A, Chahardahcherik M and Honarmand M: Effect of various doses of silymarin on the oxidative stress induced by busulfan administration in the different organs of rats. Turk J Pharm Sci. 13:233–240. 2016.

98 

Yardımcı M, Göz M, Aydın MS, Kankılıç N and Temiz E: Antioxidant actions of thymoquinone, silymarin, and curcumin on experimental aortic ischemia-reperfusion model in wistar albino rats. Braz J Cardiovasc Surg. 37:807–813. 2022.PubMed/NCBI View Article : Google Scholar

99 

Azizoğlu M, Arslan S, Gökalp Özkorkmaz E, Aşır F, Basuguy E, Okur MH, Aydoğdu B, Alagöz Karabel M and Kaplan I: Protective effects of Silymarin on testicular torsion/detorsion in rats. Eur Rev Med Pharmacol Sci. 27:10446–10453. 2023.PubMed/NCBI View Article : Google Scholar

100 

Surai PF: Silymarin as a natural antioxidant: An overview of the current evidence and perspectives. Antioxidants (Basel). 4:204–247. 2015.PubMed/NCBI View Article : Google Scholar

101 

Taleb A, Ahmad KA, Ihsan AU, Qu J, Lin N, Hezam K, Koju N, Hui L and Qilong D: Antioxidant effects and mechanism of silymarin in oxidative stress induced cardiovascular diseases. Biomed Pharmacother. 102:689–698. 2018.PubMed/NCBI View Article : Google Scholar

102 

Kim MJ, Kim DU, Choi JW, Kim DG, Song HJ, Bae GS and Park SJ: Silymarin attenuates the severity of cerulein-induced acute pancreatitis. Pancreas. 49:89–95. 2020.PubMed/NCBI View Article : Google Scholar

103 

Cormack JR, Orme RM and Costello TG: The role of alpha2-agonists in neurosurgery. J Clin Neurosci. 12:375–378. 2005.PubMed/NCBI View Article : Google Scholar

104 

Carollo DS, Nossaman BD and Ramadhyani U: Dexmedetomidine: A review of clinical applications. Curr Opin Anaesthesiol. 21:457–461. 2008.PubMed/NCBI View Article : Google Scholar

105 

Aidoni Z, Pourzitaki C, Stamoula E, Kotzampassi K, Tsaousi G, Kazakos G, Foroulis CN, Skourtis C, Vasilakos DG and Grosomanidis V: Circulatory effects of dexmedetomidine in early sepsis: A randomised controlled experimental study. Naunyn Schmiedebergs Arch Pharmacol. 393:89–97. 2020.PubMed/NCBI View Article : Google Scholar

106 

Wang C, Yuan W, Hu A, Lin J, Xia Z, Yang CF, Li Y and Zhang Z: Dexmedetomidine alleviated sepsis-induced myocardial ferroptosis and septic heart injury. Mol Med Rep. 22:175–184. 2020.PubMed/NCBI View Article : Google Scholar

107 

Koca U, Olguner ÇG, Ergür BU, Altekin E, Taşdöğen A, Duru S, Girgin P, Gündüz K, Cilaker Mıcılı S, Güzeldağ S and Akkuş M: The effects of dexmedetomidine on secondary acute lung and kidney injuries in the rat model of intra-abdominal sepsis. ScientificWorldJournal. 2013(292687)2013.PubMed/NCBI View Article : Google Scholar

108 

Li Y, Wu B, Hu C, Hu J, Lian Q, Li J and Ma D: The role of the vagus nerve on dexmedetomidine promoting survival and lung protection in a sepsis model in rats. Eur J Pharmacol. 914(174668)2022.PubMed/NCBI View Article : Google Scholar

109 

Wu Y, Liu Y, Huang H, Zhu Y, Zhang Y, Lu F and Zhou C, Huang L, Li X and Zhou C: Dexmedetomidine inhibits inflammatory reaction in lung tissues of septic rats by suppressing TLR4/NF-κB pathway. Mediators Inflamm. 2013(562154)2013.PubMed/NCBI View Article : Google Scholar

110 

Şengel N, Köksal Z, Dursun AD, Kurtipek Ö, Sezen ŞC, Arslan M and Kavutçu M: Effects of dexmedetomidine administered through different routes on kidney tissue in rats with spinal cord ischaemia-reperfusion injury. Drug Des Devel Ther. 16:2229–2239. 2022.PubMed/NCBI View Article : Google Scholar

111 

Hernández G, Tapia P, Alegría L, Soto D, Luengo C, Gomez J, Jarufe N, Achurra P, Rebolledo R, Bruhn A, et al: Effects of dexmedetomidine and esmolol on systemic hemodynamics and exogenous lactate clearance in early experimental septic shock. Crit Care. 20(234)2016.PubMed/NCBI View Article : Google Scholar

112 

Zhao W, Jia L, Yang HJ, Xue X, Xu WX, Cai JQ, Guo RJ and Cao CC: Taurine enhances the protective effect of dexmedetomidine on sepsis-induced acute lung injury via balancing the immunological system. Biomed Pharmacother. 103:1362–1368. 2018.PubMed/NCBI View Article : Google Scholar

113 

Yang CL, Chen CH, Tsai PS, Wang TY and Huang CJ: Protective effects of dexmedetomidine-ketamine combination against ventilator-induced lung injury in endotoxemia rats. J Surg Res. 167:e273–e281. 2011.PubMed/NCBI View Article : Google Scholar

114 

Özkan F, Yüksek A, Demirel A and Kantekin Ç: Effects of dexmetatomidine and midazolam on immunity in sepsis-induced rats. Med J Bakirkoy. 19:180–185. 2023.

115 

Choi MW, Ko DR, Kong T, Choa MH, You JS and Chung SP: Comparison of silymarin, penicillin, N-acetylcysteine in patient with amatoxin poisoning: A systematic review. J Korean Soc Clin Toxicol. 16:33–41. 2018.

116 

Abdel Salam OM, Sleem AA, Omara EA and Hassan NS: Effect of ribavirin alone or combined with silymarin on carbon tetrachloride induced hepatic damage in rats. Drug Target Insights. 2:19–27. 2007.PubMed/NCBI

Related Articles

Journal Cover

June-2024
Volume 27 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yavuz A, Küçük A, Ergörün AI, Dursun AD, Yiğman Z, Alkan M and Arslan M: Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation. Exp Ther Med 27: 242, 2024
APA
Yavuz, A., Küçük, A., Ergörün, A.I., Dursun, A.D., Yiğman, Z., Alkan, M., & Arslan, M. (2024). Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation. Experimental and Therapeutic Medicine, 27, 242. https://doi.org/10.3892/etm.2024.12530
MLA
Yavuz, A., Küçük, A., Ergörün, A. I., Dursun, A. D., Yiğman, Z., Alkan, M., Arslan, M."Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation". Experimental and Therapeutic Medicine 27.6 (2024): 242.
Chicago
Yavuz, A., Küçük, A., Ergörün, A. I., Dursun, A. D., Yiğman, Z., Alkan, M., Arslan, M."Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation". Experimental and Therapeutic Medicine 27, no. 6 (2024): 242. https://doi.org/10.3892/etm.2024.12530