Inhibition of TLR4 protects rat islets against lipopolysaccharide-induced dysfunction

  • Authors:
    • Xiao Wang
    • Qin Min Ge
    • Fan Bian
    • Yan Dong
    • Chun Mei Huang
  • View Affiliations

  • Published online on: December 30, 2016     https://doi.org/10.3892/mmr.2016.6097
  • Pages: 805-812
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Oxidative stress leads to dysfunction in pancreatic cells, causing a reduction in insulin secretion following exposure to glucose. Toll-like receptor 4 (TLR4) may be activated by exposure to lipopolysaccharide (LPS) stress. TLR4 may mediate the initiation of inflammatory and immune defense responses; however, the importance of the LPS/TLR4 interaction in apoptosis induced by oxidative stress in pancreatic β cells remains to be elucidated. The present study aimed to investigate the importance of TLR4 during LPS‑induced oxidative stress, apoptosis and dysfunction of insulin secretion in isolated islets of rats. LPS‑induced stimulation of TLR4 increased the production of reactive oxygen species and promoted apoptosis by upregulating the expression levels of caspase‑3, poly ADP ribose polymerase and altering the expression ratio of B‑cell lymphoma‑2 (Bcl‑2)/Bcl‑2 associated X protein. Additionally, the insulin secretion of islets cells was reduced. Anti‑TLR4 antibody and a knockdown of TLR4 by TLR4‑short hairpin RNA were used to inhibit TLR4 activity, which may reverse LPS‑induced events. The present study determined that in islets exposed to LPS oxidative stress, dysfunction may be partly mediated via the TLR4 pathway. Inhibition of TLR4 may prevent dysfunction of rat islets due to oxidative stress. The present study revealed that targeting the LPS/TLR4 signaling pathway and antioxidant therapy may be a novel treatment for the severely septic patients with hyperglycemia stress.

Introduction

Sepsis remains an important challenge when treating patients in Intensive Care Units (ICUs). The pathology of sepsis is based on a systemic inflammatory response that is characterized by the upregulation of inflammatory cytokines, particularly in response to gram-negative bacteria (1,2). Lipopolysaccharides (LPS) are a major component of the outer membrane of gram-negative bacteria, which triggers inflammation and gram-negative sepsis. Patients that develop severe sepsis or septic shock often have a low survival rate, which may be associated with the host response to refractory hyperglycemic stress. However, the molecular mechanism underlying this remains to be elucidated. It is possible that LPS may directly limit the capacity of pancreatic β-cells to induce oxidative stress and apoptosis. It may be one of the mechanisms behind hyperglycemic stress in patients with sepsis.

The imbalance between the oxidation system and the antioxidant defense frequently results in oxidative stress. This imbalance may induce an inflammatory cascade and limit the function of pancreatic β-cells by the direct effect of free radicals (3). High levels of reactive oxygen species (ROS) in the pancreas and low levels of antioxidant defense mechanisms may lead to increased oxidative damage and reduced insulin secretion (4). Our previous study determined that LPS stress led to increased ROS production and reduced insulin secretion in Ins-1 pancreatic β-cells (5). Toll-like receptors (TLRs) are a family of pattern-recognition receptors that serve important functions in the innate immune system, by activating pro-inflammatory signaling pathways in response to the detection of microbial products (6). TLR4 is expressed in pancreatic β-cells and insulin-sensitive tissues, including adipocytes and muscle cells (79). TLR4 has been directly associated with proinflammatory signaling in β-cell dysfunction (10,11). The effect of TLR4 signaling in insulin resistance is a well-established concept; however, the specific function of TLR4 during LPS-induced oxidative stress on pancreatic β-cells remains to be elucidated. The present study blocked TLR4 using a TLR4 antibody and TLR4-short hairpin (shRNA) to observe its effect on LPS-induced dysfunction of pancreatic β-cells. It is possible that TLR4 activation may mediate oxidative stress within pancreatic β-cells, resulting in their apoptosis and dysfunction of insulin secretion. The current study aimed to reveal the possible mechanisms of infection-induced stress hyperglycemia.

Materials and methods

Animals

A total of six Male Sprague-Dawley rats were provided by SLAC Laboratory Animal Co., Ltd. (Shanghai, China). The rats were 6–8 weeks of age and weighed 250–300 g. They were maintained in a sterile environment prior to the start of the study. The rats were provided with standard chow and water, and were exposed to a 12 h light/dark cycles at 26°C in 50% humidity. The protocol was approved by the Ethics Committee of Shanghai Jiaotong University School of Medicine (approval no. XHEC-F-2012-003). The experiments were performed in accordance with the guidance for the Care and Use of Laboratory Animals of the Nationals Institutes of Health (Bethesda, MD, USA).

Reagents

Lipopolysacharide, collagenase V and Ficoll 400 were purchased from Sigma-Aldrich; Merck Millipore (Darmstadt, Germany). The β-actin antibody (cat. no. sc-81178) and mouse anti-TLR4 antibody (cat. no. sc-12511) and TLR4-short hairpin (sh)RNA were obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Fetal calf serum (FCS) and RPMI-1640 medium were obtained from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). The 2′,7′-dichlorofluorescin diacetate (DCFH-DA), annexin V-fluorescein isothiocyanate (FITC) kit and bicinchoninic acid protein assay kit were purchased from Beyotime Institute of Biotechnology (Haimen, China). The TRIzol reagent, PrimeScript RT reagent kit and SYBR Premix Ex Taq kit were obtained from Takara Biotechnology Co., Ltd. (Shiga, Japan).

Islet isolation and culture

Sprague-Dawley rats were fasted for 12 h and were subsequently sacrificed using an intraperitoneal injection of 2.5% sodium pentobarbital (Sinopharm Chemical Reagent Co., Ltd., Shanghai, China) (60 mg/kg body weight). The pancreatic and common bile ducts were exposed and 3 ml precooled Hank's Balanced Salt Solution (HBSS), combined with 6 ml of precooled collagenase V (Sigma-Aldrich; Merck Millipore) (1 mg/ml), were immediately injected through the pancreatic duct. The pancreas was subsequently removed and digested for 10 min in a water bath at 37°C. The tissues were then passed through a 600 µm mesh stainless steel filter. Precooled HBSS containing 10% FCS was used to terminate the digestion. The islets were purified using Ficoll 400 and density gradient centrifugation with 600 × g at 4°C. The isolated islets were cultured in RPMI-1640 media supplemented with 10% FCS at 37°C and an atmosphere of 5% CO2.

Knockdown of TLR4 by shRNA

The isolated islets were seeded into 24-well plates at a density ~0.5×103/ml and cultured overnight. TLR4-shRNA and negative control shRNA were transfected into cells using TLR4 shRNA (r) Lentiviral Particles (cat. no. sc-156001) and Control shRNA Lentiviral Particles (cat. no. sc-108080) (Santa Cruz Biotechnology, Inc.) according to the manufacturer's protocol. Subsequently, the cells were cultured by puromycin dihydrochloride (Sigma-Aldrich; Merck Millipore) for 3–4 days. Transfection efficiency was determined by western blot analysis.

ROS production determined using DCFH-DA probe

The islets were grouped into the following treatment groups: i) Control (CON) group treated with phosphate-buffered saline (PBS); ii) LPS group treated with 10 µg/ml LPS; iii) anti-TLR4 antibody intervention group pretreated with anti-TLR4 antibody (20 µg/ml) for 1 h, then treated with LPS (10 µg/ml); and iv) TLR4-shRNA intervention group pretreated with TLR4-shRNA (10 µmol/l) for 1 h, then treated with LPS (10 µg/ml). The islets were cultured for 24 h and were subsequently incubated in the presence of 10 µmol/l DCFH-DA for 1 h at 37°C in the dark, and then washed three times with PBS. The ROS production of islets was assessed by fluorescent microscopy (excitation 488 nm and emission 525 nm).

Apoptosis of rat islets detected by flow cytometry analysis

Following the incubation (the isolated islets were seeded into 24-well plates), the islets were harvested and centrifuged at 100 × g for 2 min at 4°C and then digested into single cells with trypsin and DNAse. The cells were subsequently centrifuged at 1,000 × g for 5 min at 4°C and washed with PBS twice. The cells were suspended at a density of 1.0×106 cells/well with annexin V-FITC composite liquid (195 µl), and the islets cells were incubated with annexin V-FITC (5 µl). Finally, the cells were stained with 10 µl propidium iodide for 15 min at room temperature in the dark. The apoptotic rate was detected by a flow cytometer (FACSCalibur; BD Biosciences, Franklin Lakes, NJ, USA) and analyzed by CellQuest software version 7.5.3 (BD Biosciences).

Western blotting

Isolated islets were cultured and treated as aforementioned, and were subsequently washed twice with PBS, placed immediately in precooled lysis buffer and centrifuged at 12,000 × g for 10 min at 4°C. The protein concentration of the extracts was determined using Enhanced BCA Protein assay kit (Beyotime Institute of Biotechnology). The samples (30–50 µg) were separated by electrophoresis on 8 or 12% sodium dodecyl sulphate-polyacrylamide gels and transferred onto nitrocellulose membranes. Non-specific binding sites of the samples were blocked using Tris-buffered saline and Tween-20 with 5% non-fat milk for 1 h at 4°C. Following blocking, the membraness were incubated overnight at 4°C with the anti-mouse TLR4 primary antibody (1:200), followed by incubation with the secondary goat anti-mouse horseradish peroxidase-conjugated antibody (1:2,000; Beyotime Institute of Biotechnology; cat. no. A0216) for 1 h at room temperature. β-actin (1:1,000) expression was used as an internal control. The blots were visualized using an enhanced chemiluminescence system (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The density of protein bands was analyzed using Image Lab 2.0.1 (Bio-Rad Laboratories, Inc.) and normalized against the intensity of β-actin.

mRNA expression levels of TLR4 and B-cell lymphoma (Bcl)-2/Bcl-2 associated X protein (Bax) detected by reverse transcription-quantitative PCR (RT-qPCR)

The total RNA was isolated using TRIzol reagent. The mRNA expression levels of TLR4 and Bax/Bcl-2 were determined using RT-qPCR. The total RNA concentration was 1,000 ng. The cDNA were synthesized using PrimeScript RT reagent kit and the samples were subsequently subjected to PCR amplification with primer sets (presented in Table I). RT-qPCR was performed using SYBR Premix Ex Taq kit. The thermocycling conditions were as follows: 95°C for 30 sec, 40 cycles of 5 sec at 95°C and 1 min at 60°C. To confirm that the specific PCR product was formed, a dissociation step of 15 sec at 95°C, 15 sec at 60°C and 15 sec at 95°C was added to confirm the melting temperature. Gene expression was determined by the 2−ΔΔCq method (12) and was normalized against GAPDH.

Table I.

Primer sequences used in reverse transcription-polymerase chain reaction.

Table I.

Primer sequences used in reverse transcription-polymerase chain reaction.

GeneSequence (5′-3′)
TLR4
  Forward AGTTGGCTCTGCCAAGTCTCAGAT
  Reverse TGGCACTCATCAGGATGACACCAT
Bcl-2
  Forward TTGTGGCCTTCTTTGAGTTCGGTG
  Reverse TCATCCACAGAGCGATGTTGTCCA
Bax
  Forward TTTGCAGACGGCAACTTCAACTGG
  Reverse TGTCCAGCCCATGATGGTTCTGAT
GAPDH
  Forward TGATGCTGGTGCTGAGTATGTCGT
  Reverse AGGTGGAAGAATGGGAGTTGCTGT

[i] TLR4, toll-like receptor 4; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2 associated X protein; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

Glucose-stimulated insulin secretion (GSIS) in islets

Isolated rat islets were seeded into 96-well plates at concentration about 0.5×103 cells/well. The cells were subjected to the indicated treatment for 24 h, and subsequently, the medium was removed and islets were balanced in Krebs Ringer Buffer [KRB; 115 mM NaCl; 4.7 mM KCl; 2.6 mM CaCl2; 1.2 mM KH2PO4; 1.2 mM MgSO4; 10 mM NaHCO3; 10 mM HEPES; 0.1% bovine serum albumen (pH 7.4)], containing 3.3 mM D-glucose, for 30 min. The islets were subsequently incubated in KRB containing 3.3 mM D-glucose for another 1 h and KRB containing 27.8 mM D-glucose for 1 h. The buffer was collected from the 3.3 and 27.8 mM incubations and stored at −80°C until analysis. Insulin concentration in the medium was determined using a Rat Insulin Radioimmunoassay kit (Diagnostic Systems Laboratories, Inc., Webster, TX, USA).

Statistical analysis

The data are presented as the mean ± standard deviation. The experiments were repeated at least three times for each group. Analysis was performed using SPSS version 19.0 software (IBM SPSS, Armonk, NY, USA). Statistical significance of differences between groups was determined using one-way analysis of variance. P<0.05 was were considered to indicate a statistically significant difference.

Results

TLR4 expression in rat islets increases with LPS treatment

The present study detected TLR4 expression levels in rat islets when stimulated by LPS for various durations. The protein and mRNA expression levels of TLR4 were upregulated subsequent to LPS stimulation. A significant increase in TLR4 protein expression at 24 and 48 h, which was 1.5-fold and 4-folds greater when compared with the control group (P<0.001; Fig. 1A and B). The TLR4 protein expression levels were significantly reduced when treated for >48 h (P<0.05; Fig. 1B). The mRNA expression levels of TLR4 were significantly increased at 12 h, and were 4-fold higher compared with the control group (P<0.001; Fig. 1C) and increased by 7-fold at 24 h (P<0.001; Fig. 1C). TLR4 expression levels were elevated following stimulation with LPS for 48 h (P<0.001; Fig. 1C) and 72 h (P<0.001; Fig. 1C). However, the expression levels fell with prolonged LPS treatment for >24 h (Fig. 1C).

Downregulation of TLR4 in LPS-treated rat isolated islets reduces ROS production

ROS production was observed as green fluorescence in the cells. The generation of ROS in rat islets was significantly increased following exposure to LPS (10 µg/ml) for 24 h when compared with the control (P<0.001; Fig. 2A and B). The mean fluorescence intensity of the LPS group increased by 10-fold compared with the control group (75.3±12.1 vs. 800±150.9; Fig. 2A). In order to investigate the function of TLR4 during oxidative stress in rat islets exposed to LPS, TLR4 expression was inhibited by using an anti-TLR4 antibody and TLR4 shRNA. The ROS production was significantly reduced by pretreatment of LPS-exposed cells with anti-TLR4 antibody or TLR4-shRNA when compared with the LPS only group (P<0.01; Fig. 2B). The efficiency of the TLR4-shRNA transfection was determined by western blot analysis. It was revealed that TLR4 expression was reduced by TLR4-shRNA (Fig. 2C). These findings suggested that TLR4 may contribute to the process of the oxidative stress induced by LPS.

Suppression of TLR4 decreases LPS-induced apoptosis in islet cells

Apoptosis is an important mechanism that may lead to the dysfunction of pancreatic β-cells. The present study revealed that the apoptotic rate of rat islets was higher following LPS stimulation for 24 h. The apoptotic rate was significantly higher (22.37±3.35%) in the LPS group compared with the control (9.33±1.97%) (P<0.001; Fig. 3). The activity of TLR4 was also suppressed to determine its involvement in the apoptosis of islet cells. A significant reduction on apoptotic rate was observed when cells were pretreated with anti-TLR4 antibody (P<0.001; Fig. 3) and TLR4-shRNA (P<0.001; Fig. 3) when compared with the LPS-treated group. The anti-TLR4 antibody and TLR4-shRNA decreased the LPS-induced apoptosis ratio to 10.60±1.04% and 12.10±0.79%, respectively. This revealed that the inhibition of TLR4 may protect rat islet cells from LPS-induced apoptosis.

TLR4 downregulation reduces the expression levels of apoptosis-associated genes and proteins

The dysregulation of apoptosis-associated genes and proteins may lead to apoptotic cell death. The present study examined the effects of TLR4 on the expression levels of caspase-3, poly ADP-ribose polymerase (PARP) and the ratio of Bax/Bcl-2. Treatment with 10 µg/ml LPS for 24 h led to significantly upregulated expression levels of caspase-3 and PARP increased by 2-fold compared with the control group (P<0.001; Fig. 4A and B), whereas the expression ratio of Bax/Bcl-2 was 1.5-fold higher when the LPS-treated group was compared with the control (P<0.05; Fig. 4A). Cells pretreated with anti-TLR4 antibody in order to inhibit TLR4 expression significantly reduced the LPS-induced upregulation of caspase-3 (P<0.05; Fig. 4A), and PARP (P<0.01; Fig. 4A). However, no significant difference was identified between cells pretreated with TLR4 antibodies and the LPS treatment group. Cells pretreated with TLR4-shRNA exhibited significantly reduced expression levels of caspase-3 (P<0.001; Fig. 4B), PARP (P<0.01; Fig. 4B) and Bac/Bcl-2 ratio (P<0.01; Fig. 4B) when compared with the LPS treatment group. The mRNA expression level ratio of Bax/Bcl-2 was significantly increased by 2.2-fold in the LPS treatment group compared with the control group (P<0.001; Fig. 4C). Pretreatment with TLR4 antibody and TLR4-shRNA resulted in significantly reduced expression ratio levels, when compared with the LPS treatment group (P<0.01 and P<0.001, respectively; Fig. 4C). Therefore, it is possible that TLR4 is involved in LPS-induced apoptosis in rat islet cells.

Downregulation of TLR4 expression levels in LPS-treated increases insulin secretion in islet cells

In order to determine the importance of TLR4 in LPS-induced rat pancreatic β-cell dysfunction GSIS assays were performed. Following the aforementioned treatments, the isolated islet cells were stimulated by 3.3 and 27.8 mmol/l glucose, and then insulin secretion was quantified. Isolated islet cells incubated with 10 µg/ml LPS for 24 h exhibited reduced insulin secretion. The insulin value was significantly reduced by 2.1-fold in the LPS-treated group compared with control group when stimulated with 27.8 mmol/l glucose (P<0.001; Fig. 5). The insulin secretion in isolated rat islets was significantly increased following pretreatment with anti-TLR4 antibody and TLR4-shRNA (P<0.01; Fig. 5). This suggested that inhibition of TLR4 may protect islet cells from LPS-induced damage.

Discussion

Stress hyperglycemia is common in ICU patients, particularly in patients with severe sepsis and septic shock. It directly increases the mortality of critically ill patients, despite the absence of pre-existing conditions, including diabetes mellitus. The inflammatory response has been regarded as a mechanism associated with insulin signal transduction, which may alter the normal structure of β-cells, induce insulin resistance and decrease insulin secretion (10,13). Acute severe hyperglycemia during a critical illness may excessively increase the existing systemic inflammatory response (14). However, no benefit of strict glucose control was observed in patients with severe sepsis in a previous study (15) and the glucose levels remained difficult to control when developed anti-inflammatory therapy was administered. Oxidative stress is a major contributing factor to the high mortality rates associated with several inflammatory diseases, including severe sepsis (16). ROS production is associated with the occurrence and deterioration of organ dysfunction. Islet β-cell damage is the direct cause of abnormal blood glucose. Pancreatic islet β-cells are sensitive to ROS levels due to their low expression levels of antioxidant enzymes. A previous study determined that acute hyperglycemia enhanced oxidative stress and the generation of ROS led to reduced glucose control (17). The effect of oxidative stress on patients with severe infection and stress hyperglycemia remains to be elucidated; however, it may be used as a novel strategy to treat refractory hyperglycemia in the future.

The generation of ROS is usually balanced by antioxidants. Oxidative stress results from the imbalance between the formation and sequestration of free radicals. Various pathological processes disrupt this balance by increasing ROS or decreasing the level of available antioxidants. Oxidative stress contributes to loss of islet mass and function and may trigger cell death via apoptosis and necrosis (18,19). Armann et al (20) determined that ROS production in islets was associated with the percentage of cells undergoing apoptosis and islet functional potency in vivo. Duprez et al (21) also demonstrated that oxidative stress resulted in the apoptosis of rat islet β-cells and dysfunction of insulin secretion. Our previous study revealed that LPS-induced oxidative stress and apoptosis results in the loss of GSIS in Ins-1 pancreatic β-cells (5). It is possible that oxidative stress may be the key mechanism contributing to the dysfunction of pancreatic β-cells. In order to investigate the importance of oxidative stress induced by LPS treatment in pancreatic β-cells, the present study was performed on isolated rat islet cells. ROS production in islet cells was significantly enhanced following LPS treatment for 24 h. This confirmed the increase in LPS-induced ROS production. Additionally, it was revealed that this process may partially depend on the TLR4 signaling pathway.

TLR4 is the signal-transducing molecule of the LPS receptor complex. The LPS-TLR4 signaling pathway has been identified as a critical upstream event in the pathogenesis of gram-negative sepsis (22). However, the function of TLR4 in pancreatic β-cells of patients with stress hyperglycemia remains to be elucidated. The protein and mRNA expression levels of TLR4 have previously been observed in human, mouse and rat pancreatic islets (9,23). Previous studies established that the dysfunction of pancreatic islet β-cells was triggered by LPS stimulation via the TLR4 signaling pathway (11,24). The TLR4 signaling pathway was involved in the early stage of pro-inflammatory and pro-oxidant of islet cells in vitro (3). TLR4 stimulation reduced the function of islet β-cells in mice by limiting GSIS (10). Shi et al (8) observed that the absence of TLR4 improved obesity-induced insulin resistance. It is possible that in the present study, TLR4 was involved in the process of LPS-induced oxidative stress, apoptosis and dysfunction in rat islet cells.

The present study revealed that the mRNA and protein expression levels of TLR4 significantly increased when isolated rat islet cells were cultured in the presence of LPS (10 µg/ml). LPS induced the upregulation of TLR4 expression, which may have led to increased ROS production. TLR4-shRNA and anti-TLR4 antibody pretreatments were used to suppress TLR4 activity, and the results demonstrated that inhibition of TLR4 reversed LPS-induced oxidative stress in rat isolated islets. It was confirmed that LPS-induced oxidative stress was associated with the expression levels of TLR4. Knockdown of TLR4 by shRNA demonstrated the involvement of TLR4 in LPS-induced subsequent effect.

A previous study determined that the permeability of the mitochondrial membrane was impaired in patients with sepsis and severe oxidative stress (25). Various apoptosis genes have been identified to be involved in mitochondrial apoptotic pathways associated with oxidative stress, including the caspase family and Bax/Bcl-2. Caspase-3 is crucial to the process of apoptosis. PARP, one of the substrates of caspase protein family, is associated with DNA repair and genetic integrity monitoring. The ratio of Bax/Bcl-2 has been established as an important regulator of apoptosis. A previous study reported that defective mitochondrial oxidative phosphorylation contributed to the dysfunction of pancreatic β-cells (26). The present study determined that LPS promoted apoptosis in rat islet cells by upregulation of the pro-apoptosis protein and gene expression levels. It was revealed that the expression levels of caspase-3, PARP and the ratio of Bax/Bcl-2 were increased following LPS stimulation. This effect was reversed by the inhibition of TLR4 expression. It is possible that LPS altered the permeability of the mitochondrial membrane resulting in an increased apoptotic rate.

The increase in ROS production and upregulation of apoptotic genes in pancreatic β-cells also resulted in dysfunction of insulin secretion. The present study clearly demonstrated dysfunction of insulin secretion in rat islet cells due to exposure to LPS. The reduction of insulin secretion was suggested to be associated with upregulation of TLR4 expression levels in pancreatic β-cells. Inhibition of TLR4 by anti-TLR4 antibody and TLR4-shRNA may increase the insulin secretion levels in LPS-induced islet cells.

It is of note that the observations of the present study are different to the results presented by Vivot et al (27). In this previous study, inhibition of TLR4 expression levels led to an increase in ROS production. They also determined that the generation of ROS and various inflammatory mediators, including cytochrome c oxidase subunit 2, interleukin-6 and C-C motif chemokine ligand 2 were two distinct pathways that depend on TLR4 expression (27). Additionally it is possible that the upstream regulator of TLR4 may be different under different conditions, such as apoptosis, oxidative stress or inflammatory reactions; however, to fully understand this, further investigation is required. This may be the molecular mechanism behind the poor clinical outcomes in patients with stress hyperglycemia. The observations of the present study highlighted that the control of ROS release must be taken into consideration, in order to improve the preservation of pancreatic islet cells in vitro.

The present study determined that the structure of islet cells sourced from Sprague-Dawley rats began to develop an irregular shape and exhibit reduced insulin secretion following treatment with LPS for 48 h, which led to adverse effects in follow-up experiments; therefore, islets were treated with LPS for 24 h instead of 48 h.

The results of the present study indicated that LPS may act directly on pancreatic islets via the TLR4 signaling pathway and induce oxidative stress and apoptosis, which may lead to dysfunction of pancreatic β-cells. Increase of ROS production may trigger an inflammatory cascade and promote severe hyperglycemia. Reducing ROS production via TLR4 inhibition may inhibit the positive-feedback of oxidative stress-induced damage to the pancreas in critically ill patients with stress hyperglycemia. This suggested that antioxidant therapy combined with anti-inflammatory therapy may be beneficial to patients with severe infections.

Acknowledgements

The present study was supported by the National Nature Science Foundation of China (no. 81000354) and the Shanghai Nature Science Foundation (no. 14ZR1427000).

References

1 

Beutler B and Rietschel ET: Innate immune sensing and its roots: The story of endotoxin. Nat Rev Immunol. 3:169–176. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Zahar JR, Timsit JF, Garrouste-Orgeas M, Français A, Vesin A, Descorps-Declere A, Dubois Y, Souweine B, Haouache H, Goldgran-Toledano D, et al: Outcomes in severe sepsis and patients with septic shock: Pathogen species and infection sites are not associated with mortality. Crit Care Med. 39:1886–1895. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Giribabu N, Kumar KE, Rekha SS, Muniandy S and Salleh N: Chlorophytum borivilianum root extract maintains near normal blood glucose, insulin and lipid profile levels and prevents oxidative stress in the pancreas of streptozotocin-induced adult male diabetic rats. Int J Med Sci. 11:1172–1184. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Coskun O, Kanter M, Korkmaz A and Oter S: Quercetin, a flavonoid antioxidant, prevents and protects streptozotocin-induced oxidative stress and beta-cell damage in rat pancreas. Pharmacol Res. 51:117–123. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Du SC, Ge QM, Lin N, Dong Y and Su Q: ROS-mediated lipopolysaccharide-induced apoptosis in INS-1 cells by modulation of Bcl-2 and Bax. Cell Mol Biol (Noisy-le-grand). 58 Suppl:OL1654–OL1659. 2012.PubMed/NCBI

6 

Medzhitov R: Toll-like receptors and innate immunity. Nat Rev Immunol. 1:135–145. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Reyna SM, Ghosh S, Tantiwong P, Meka CS, Eagan P, Jenkinson CP, Cersosimo E, Defronzo RA, Coletta DK, Sriwijitkamol A and Musi N: Elevated toll-like receptor 4 expression and signaling in muscle from insulin-resistant subjects. Diabetes. 57:2595–2602. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H and Flier JS: TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 116:3015–3025. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Li M, Song L, Gao X, Chang W and Qin X: Toll-like receptor 4 on islet beta cells senses expression changes in high-mobility group box 1 and contributes to the initiation of type 1 diabetes. Exp Mol Med. 44:260–267. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Cucak H, Mayer C, Tonnesen M, Thomsen LH, Grunnet LG and Rosendahl A: Macrophage contact dependent and independent TLR4 mechanisms induce β-cell dysfunction and apoptosis in a mouse model of type 2 diabetes. PLoS One. 9:e906852014. View Article : Google Scholar : PubMed/NCBI

11 

Wang Y: Attenuation of berberine on lipopolysaccharide-induced inflammatory and apoptosis responses in β-cells via TLR4-independent JNK/NF-κB pathway. Pharm Biol. Nov 5–2013.(Epub ahead of print).

12 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Hotamisligil GS: Inflammation and metabolic disorders. Nature. 444:860–867. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Yoneyama S, Terashima H, Yamaguchi R, Tadano S and Ohkohchi N: The manner of the inflammation-boosting effect caused by acute hyperglycemia secondary to overfeeding and the effects of insulin therapy in a rat model of sepsis. J Surg Res. 185:380–387. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Viana MV, Moraes RB, Fabbrin AR, Santos MF and Gerchman F: Assessment and treatment of hyperglycemia in critically ill patients. Rev Bras Ter Intensiva. 26:71–76. 2014.(In Portuguese). View Article : Google Scholar : PubMed/NCBI

16 

Muhl D, Woth G, Drenkovics L, Varga A, Ghosh S, Csontos C, Bogár L, Wéber G and Lantos J: Comparison of oxidative stress & leukocyte activation in patients with severe sepsis & burn injury. Indian J Med Res. 134:69–78. 2011.PubMed/NCBI

17 

Ling PR, Smith RJ and Bistrian BR: Acute effects of hyperglycemia and hyperinsulinemia on hepatic oxidative stress and the systemic inflammatory response in rats. Crit Care Med. 35:555–560. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Henriksnas J, Lau J, Zang G, Berggren PO, Kohler M and Carlsson PO: Markedly decreased blood perfusion of pancreatic islets transplanted intraportally into the liver: Disruption of islet integrity necessary for islet revascularization. Diabetes. 61:665–673. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Zhu HQ, Gao Y, Guo HR, Kong QZ, Ma Y, Wang JZ, Pan SH, Jiang HC and Dai WJ: Pretreatment with bilirubin protects islet against oxidative injury during isolation and purification. Transplant Proc. 43:1810–1814. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Armann B, Hanson MS, Hatch E, Steffen A and Fernandez LA: Quantification of basal and stimulated ROS levels as predictors of islet potency and function. Am J Transplant. 7:38–47. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Duprez J, Roma LP, Close AF and Jonas JC: Protective antioxidant and antiapoptotic effects of ZnCl2 in rat pancreatic islets cultured in low and high glucose concentrations. PLoS One. 7:e468312012. View Article : Google Scholar : PubMed/NCBI

22 

Roger T, Froidevaux C, Le Roy D, Reymond MK, Chanson AL, Mauri D, Burns K, Riederer BM, Akira S and Calandra T: Protection from lethal gram-negative bacterial sepsis by targeting Toll-like receptor 4. Proc Natl Acad Sci USA. 106:2348–2352. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Li Y, Zhou ZG, Xia QJ, Zhang J, Li HG, Cao GQ, Wang R, Lu YL and Hu TZ: Toll-like receptor 4 detected in exocrine pancreas and the change of expression in cerulein-induced pancreatitis. Pancreas. 30:375–381. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Amyot J, Semache M, Ferdaoussi M, Fontés G and Poitout V: Lipopolysaccharides impair insulin gene expression in isolated islets of langerhans via toll-like receptor-4 and NF-κB signalling. PLoS One. 7:e362002012. View Article : Google Scholar : PubMed/NCBI

25 

Apostolova N, Garcia-Bou R, Hernandez-Mijares A, Herance R, Rocha M and Victor VM: Mitochondrial antioxidants alleviate oxidative and nitrosative stress in a cellular model of sepsis. Pharm Res. 28:2910–2919. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Lu H, Koshkin V, Allister EM, Gyulkhandanyan AV and Wheeler MB: Molecular and metabolic evidence for mitochondrial defects associated with beta-cell dysfunction in a mouse model of type 2 diabetes. Diabetes. 59:448–459. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Vivot K, Langlois A, Bietiger W, Dal S, Seyfritz E, Pinget M, Jeandidier N, Maillard E, Gies JP and Sigrist S: Pro-inflammatory and pro-oxidant status of pancreatic islet in vitro is controlled by TLR-4 and HO-1 pathways. PLoS One. 9:e1076562014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2017
Volume 15 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang X, Ge QM, Bian F, Dong Y and Huang CM: Inhibition of TLR4 protects rat islets against lipopolysaccharide-induced dysfunction. Mol Med Rep 15: 805-812, 2017
APA
Wang, X., Ge, Q.M., Bian, F., Dong, Y., & Huang, C.M. (2017). Inhibition of TLR4 protects rat islets against lipopolysaccharide-induced dysfunction. Molecular Medicine Reports, 15, 805-812. https://doi.org/10.3892/mmr.2016.6097
MLA
Wang, X., Ge, Q. M., Bian, F., Dong, Y., Huang, C. M."Inhibition of TLR4 protects rat islets against lipopolysaccharide-induced dysfunction". Molecular Medicine Reports 15.2 (2017): 805-812.
Chicago
Wang, X., Ge, Q. M., Bian, F., Dong, Y., Huang, C. M."Inhibition of TLR4 protects rat islets against lipopolysaccharide-induced dysfunction". Molecular Medicine Reports 15, no. 2 (2017): 805-812. https://doi.org/10.3892/mmr.2016.6097