Icariin prevents cytokine‑induced β‑cell death by inhibiting NF‑κB signaling

  • Authors:
    • Shao Zhong
    • Jing Ge
    • Jiang‑Yi Yu
  • View Affiliations

  • Published online on: July 20, 2018     https://doi.org/10.3892/etm.2018.6502
  • Pages: 2756-2762
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The loss of insulin secretion in type I diabetes mellitus (T1DM) is caused by autoimmune‑mediated destruction of insulin‑producing pancreatic β‑cells. Inflammatory cytokines and immune cell infiltration activate oxidative and endoplasmic reticulum (ER) stress, resulting in reduced β‑cell viability. The current pharmacological agents used to control blood glucose have a limited effective duration and are accompanied by strong side effects. Blocking the inflammatory and immune responses that cause the β‑cell damage has been investigated as a novel therapeutic approach to control T1DM. Icariin is a flavonoid component of Chinese medicinal herbs that has anti‑inflammatory effects in vitro and in vivo. The results of the present study revealed that icariin abrogates the pro‑apoptotic effect of inflammatory cytokines and significantly suppresses the activation of nuclear factor (NF)‑κB in rat pancreatic β‑cell lines. The present study may provide a basis for the potential use of icariin as a therapeutic agent for T1DM.

Introduction

The incidence of diabetes mellitus (DM), a complex metabolic disorder associated with defective insulin secretion and activity, has been increasing worldwide over the past 20 years (1,2). According to the World Health Organization estimation, ~7% of the world's adult population is diabetic and the diabetic population is likely to increase to ≥300 million by the year 2025 (3). Type 1 DM (T1DM) is an organ-specific autoimmune disease associated with failure to distinguish self- from non-self-antigens (4). It is caused by T cell-mediated destruction of insulin-producing pancreatic β-cells (4). The incidence of T1DM is increasing steadily by 3% annually and has a concordance rate of 40–60% for monozygotic twins (4). As such, environmental and genetic factors may contribute to disease onset (57). Currently, insulin therapy is the primary treatment for T1DM. However, tight glycemic control is difficult to achieve in a number of patients, leading to long-term vascular damage associated with kidney failure, heart disease, retinopathy and neuropathy (8). Recent advances in pancreatic islet transplantation and partial or whole pancreas transplantation represent alternate treatment options for T1DM (9). However, due to the limited number of organs available for transplant, this approach is not widely used (9). Since β-cell damage is crucial to the development of T1DM, treatments that are able to prevent β-cell damage may slow disease progression.

Pancreatic β-cell damage is known to be mediated by the immune response (1012). Previous studies have suggested that inflammatory cytokines and immune cell infiltration activate oxidative and endoplasmic reticulum (ER) stress and damage β-cell viability (1016). In the early stages of disease, the infiltration of inflammatory cells promotes the release of cytokines, including interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) (1516). IL-1β, alone or in combination with TNF-α or IFN-γ, upregulates the expression of inducible nitric oxide synthase (iNOS) and promotes the generation of nitric oxide (NO) in pancreatic islets (17,18). Excessive NO production leads to dysfunctions of mitochondrial metabolism, protein modification and DNA cleavage, which may contribute to the impairment of insulin secretion and triggering β-cell death (19).

Considering the inflammatory nature of T1DM, it is plausible that anti-inflammatory agents may have potential as anti-DM drugs. Icariin is a naturally occurring flavonoid isolated from traditional Chinese medicinal herbs of the Epimedium genus (20). The compound has been revealed to have anti-inflammatory, antidepressant, male reproductive, antineoplastic, bone-healing and neuroprotective effects (20). Early in vivo and in vitro studies revealed that icariin acts as a natural anti-inflammatory drug via multiple mechanisms targeting pro-inflammatory cytokines (TNF-α and IL-6), inflammatory mediators (NO) and adhesion molecules (CD11b) (21,22). Xu et al (23) reported that icariin activates the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway to ameliorate lipopolysaccharide (LPS)-induced acute inflammatory responses. The known anti-inflammatory effects of icariin suggest that it may inhibit inflammation-induced β-cell death. The aim of the present study was to use rat pancreatic β-cell lines as an in vitro model to investigate the role of icariin. The results suggest that icariin inhibits cytokine-induced NF-κB activation and prevents β-cell death.

Materials and methods

Cell culture

Rat pancreatic β-cell RINm5F cells were obtained from ATCC (Manassas, VA, USA). Cells were cultured in RPMI-1640 medium (Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) with 10% (v/v) heat-inactivated fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), 2 mM glutamine (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), 1% non-essential amino acids (Sigma-Aldrich; Merck KGaA), 100 U/ml streptomycin and 100 U/ml penicillin (Sigma-Aldrich; Merck KGaA) at 37°C in an atmosphere containing 5% CO2. Icariin was purchased from Sigma-Aldrich (Merck KGaA). Rat IL-1β and IFN-γ proteins were obtained from R&D Systems (Minneapolis, MN, USA).

MTT assay

MTT (Sigma-Aldrich; Merck KGaA) was used to determine cell viability according to the manufacturer's protocols. Briefly, 5 ml MTT solvent (Beyotime Institute of Biotechnology, Haimen, China) was used to dissolve 25 mg MTT to form an MTT solution at 5 mg/ml. A total of 10 µ1 MTT solution was added to each well and incubated for 4 h at 37°C in an incubator. Subsequently, 100 µl formazan solution (Beyotime Institute of Biotechnology) was added for 4 h at 37°C. The optical density of viable cells was measured using a microplate reader (BMG Labtech GmbH, Ortenburg, Germany) at a wavelength of 570 nm.

NO measurement

Biologically synthesized NO is quickly oxidized to form nitrite and nitrate in aqueous solutions (19). Therefore, detecting the nitrite concentration in cell-free culture supernatants using a colorimetric assay may be indicative of NO generation. In brief, RINm5F cells (5×106) or 30 islets were treated with the 5 or 10 µM concentrations of icariin for 3 h, prior to being treated with IL-1β (1 U/ml) and IFN-γ (100 U/ml) for 24 h. Subsequently, 100 µl aliquots of culture supernatant were incubated at room temperature for 5 min with 100 µl modified Griess reagent in a 1:1 mixture of 1% sulfanilamide in 30% acetic acid and 0.1% N-(1-naphthyl) ethylenediamine dihydrochloride in 60% acetic acid (Beyotime Institute of Biotechnology). The absorbance was measured at 540 nm. The NO concentration was calculated from the linear standard curve of serial dilutions of sodium nitrite in a working medium.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from cultured cells using TRIzol reagent (Thermo Fisher Scientific, Inc.). The primer for iNOS was synthesized based on the following previously published sequences (24): Forward, 5′-GAATCTTGGAGCGAGTTGTGG-3′ and reverse, 5′-AGGAAGTAGGTGAGGGCTTGG-3′. First-strand cDNA was obtained using Super M-MLV Reverse transcriptase (BioTeke Corporation, Beijing, China). Reverse transcription was performed at 42°C for 15 min and 72°C for 2 min according to the manufacturer's protocols. PCR was performed using SYBR-Green master mix (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China). The following thermocycling conditions were used: Predenaturation at 95°C for 30 sec followed by 40 cycles of amplification at 95°C for 5 sec and annealing and extension at 60°C for 30 sec. GADPH was used to normalize iNOS mRNA expression. GAPDH forward, 5′-GATGACCTTGCCCACAGCCT-3′ and reverse, 5′-ATCTCTGCCCCCTCTGCTGA-3′. The 2−∆∆Cq method was used to quantify data (24). ABI Prism 7000 software (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used to analyze data.

Western blotting

Following treatment, proteins were extracted from RINm5F cells using a Nuclear and Cytoplasmic Protein Extraction kit (cat. no. P0027; Beyotime Institute of Biotechnology). Protein concentrations were determined using an Enhanced BCA Protein Assay kit (cat. no. P0010S; Beyotime Institute of Biotechnology). A total of 20 µg/lane was separated by 12% SDS-PAGE and transferred to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked using Blocking Buffer (cat. no. P0023B; Beyotime Institute of Biotechnology) for 2 h at room temperature. Proteins were probed using specific primary antibodies at 4°C overnight, followed by incubation with secondary antibodies at room temperature for 1 h. Specific primary antibodies against pro-caspase-3 (ab44976; 1:500), cleaved caspase-3 (ab13847; 1:500) and cleaved poly ADP-ribose polymerase (PARP; ab32064; 1:2,000) were purchased from Abcam (Cambridge, UK). Secondary antibodies against β-actin (ab8227; 1:2,000) and Larmin A (ab26300; 1:1,000) used in this study were horseradish peroxidase (HRP) conjugated goat anti-rabbit IgG or anti-mouse IgG-HRP (Beyotime Institute of Biotechnology). β-actin and Larmin A were used as internal controls to normalize results. Signals were monitored using a chemiluminescent substrate (KPL, Inc., Gaithersburg, MD, USA). Following electrophoresis, gray values were analyzed using Quantity One v4.4.0.36 software (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Caspase-3 activity

The activity of caspase-3 was conducted using a commercial ELISA kit (cat. no. HC079; Shanghai Gefan Biotechnology Co., Ltd., Shanghai, China) according to the manufacturer's protocols. In brief, cells (1×106) were resuspended in 50 µl lysis buffer (Shanghai Gefan Biotechnology Co., Ltd.) and incubated for 1 h in an ice bath. The supernatant was collected following centrifugation for 10 min at 800 × g at room temperature, following which a colorimetric reagent was added and incubated for 4 h at 37°C. The colorimetric product was monitored using an ELISA reader at a wavelength of 405 nm.

Apoptosis detection using flow cytometry

A total of 1×106 cells were washed with PBS and resuspended in binding buffer containing Annexin V-APC and propidium iodide, and incubated at 20–25°C for 10–20 min (Beyotime Institute of Biotechnology). The samples were analyzed using a FACScan flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA). The percentage of apoptotic cells in a 10,000-cell cohort was determined using flow cytometry.

NF-κB P65 activity

Following treatment, nuclear extracts were isolated using the Nuclear Extract kit according to the manufacturer's protocols (Active Motif, Carlsbad, CA, USA; cat. no. 40010). The activity of NF-κB p65 was assessed using an ELISA kit (cat. no. 40596; Active Motif).

Statistical analysis

Values are presented as the mean ± standard deviation. Statistical comparisons between cell lines were performed using one-way analysis of variance, followed by Dunnett's t-test. GraphPad Prism 7.03 software (GraphPad Software Inc., La Jolla, CA, USA) was used to analyze experimental data and a P<0.05 was considered to indicate a statistically significant difference.

Results

Icariin prevents cytokine-induced loss of cell viability

To assess the therapeutic potential of icariin in rat pancreatic β cells, the viability of cultured RINm5F cells was initially examined. As presented in Fig. 1, treatment with icariin up to 10 µM did not result in a significant loss of cell viability. Next, whether icariin protected RINm5F cells from cytokine toxicity was investigated. Treatment with cytokines IL-1β and IFN-γ significantly reduced the cell viability to 49.9±5.2% of the control value (Fig. 1). Pretreatment with icariin significantly abrogated the cytotoxic effects of cytokines on RINm5F cells in a concentration-dependent manner.

Icariin prevents cytokine-induced NO production

NO production was significantly increased following 24 h treatment with cytokines (Fig. 2A). However, the cytokine-induced NO production was effectively inhibited by treatment with 10 µM icariin (Fig. 2A). To investigate the underlying mechanisms responsible for the effects of icariin, RT-qPCR and western blotting were performed to measure the expression of iNOS at the mRNA and protein level, respectively. Treatment with IL-1β and IFN-γ significantly increased the expression of iNOS, while icariin treatment significantly ameliorated this increase at the mRNA and protein level (Fig. 2).

Icariin prevents cytokine-induced apoptosis

Cytokines are able to promote β-cell death through apoptosis and necrosis (25). Caspase-3 serves a pivotal role in the apoptotic signaling pathway, and so the activation status of caspase-3 was assessed in the present study. Treatment with IL-1β and IFN-γ increased the activity of caspase-3 and cell apoptosis in RINm5F cells, while icariin effectively reversed these effects (Fig. 3). The activation of apoptotic signaling was also confirmed by western blotting (Fig. 3B). Cleaved caspase-3 is the main marker of cell apoptosis (26), and so its expression was assessed. As presented in Fig. 3B, IL-1β and IFN-γ were able to activate caspase-3 and increase the cleavage of PARP in RINm5F cells, while treatment with icariin reduced cleaved caspase-3 and cleaved PARP levels in cytokine-stimulated cells.

Icariin suppresses the cytokine-induced activation of NF-κB

NF-κB is a key transcription factor that induces iNOS and regulates subsequent NO production (27). The results of a previous study by our group demonstrated that NF-κB was activated by cytokines or oxidative stress (28). Based on this, it was investigated whether icariin affects the cytokine-induced activation and translocation of NF-κB from the cytosol to the nucleus in RINm5F cells. NF-κB and the nuclear translocation of p65, a key subunit of the NF-κB complex, were significantly promoted by treatment with IL-1β and IFN-γ compared with the control (Fig. 4). In contrast, icariin pretreatment markedly suppressed the cytokine-stimulated activation and nuclear translocation of NF-κB. In summary, these data suggest that icariin may downregulate iNOS expression via inhibiting the cytokine-stimulated activation of NF-κB.

Discussion

Icariin is a biologically active flavonoid with a favorable therapeutic profile in metabolic syndrome (29,30). Notably, icariin has been reported to ameliorate streptozocin-induced rat diabetic retinopathy and nephropathy (31,32). A previous study demonstrated that icariin could serve as a peroxisome proliferator-activated receptor α agonist, which activates gene expression associated with lipid metabolism in the liver to contribute towards diabetes management (33). In the present study, it was revealed that icariin is able to prevent cytokine-induced β-cell death, which is an important cause of T1DM.

Inflammation is the primary cause of T1DM as well as a direct cause of a number of diabetic complications (34). An acute, intense inflammatory response triggers T1DM through lymphocyte-mediated destruction of pancreatic β cells. A chronic state of low-grade inflammation persists within in the body, which is periodically exacerbated by hyperglycemic fluctuations (3435). Increased inflammation markers (35), immune activation (36) and oxidative stress have been recorded in patients with T1DM (37,38). It has therefore been hypothesized that anti-inflammatory agents may be an effective clinical treatment for patients with T1DM. A number of in vivo and in vitro studies have confirmed the anti-inflammatory effect of icariin, including in the brain, heart, bones and airways (22,23,3950). The present study demonstrated that icariin prevents viability loss in rat pancreatic β cells, as well as suppressing cytokine-induced NO production and apoptosis activation. These results suggest that icariin may interfere with the inflammatory response and resulting pancreatic β cell death during T1DM.

Furthermore, a key factor in cytokine-induced pancreatic β-cell damage is NF-κB. In vivo studies of transgenic mice revealed that NF-κB inhibition is a protective mechanism against cytokine-induced apoptosis in pancreatic β-cells (28). In addition, the use of A20-overexpressing islets to abrogate NF-κB signaling during islet transplantation reduces the number of islets required to achieve euglycemia in diabetic recipients (51). Therefore, suppression of the NF-κB pathway may also be a novel strategy for delaying the progression of T1DM. The regulatory role of icariin on NF-κB has been reported, however it may vary between different cell types (20). Xu and Huang (52) demonstrated that icariin could increase the expression of endothelial NOS in human endothelial cells, which was implicated in the activation of NF-κB (53). In contrast, icariin was able to abrogate the effects of LPS on neuroinflammation, lung inflammation, osteoclast differentiation and bone resorption via decreasing NF-κB activity (23,49,54,55). It has also been reported that icariin inhibits NF-κB activity in a wide range of cancerous cells (5659). The results of the present study demonstrated that icariin suppresses the cytokine-induced activation of NF-κB in rat pancreatic β cells. It is likely that icariin exerts cell-specific regulatory effects and only suppresses high levels of NF-κB activity in tumor cells or cells stimulated by inflammatory cytokines.

The results of the present study demonstrate that icariin abrogates the pro-apoptotic effect of cytokines and significantly suppresses NF-κB activation in rat pancreatic β-cells. Despite being used extensively as a model for the human pancreas, the physiology of rat pancreatic β cells does not perfectly mimic that of primary cells (60). The RINm5F cells used in the present study have limitations in terms of glucose sensitivity, transport and phosphorylation (60,61). Therefore, experiments utilizing human pancreatic cells and in vivo analysis are required to confirm these findings. Nevertheless, the results of the present study suggest that icariin may have potential as a therapeutic agent against T1DM.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81573911).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Authors' contributions

SZ and J-YY conceived the study, acquired data, interpreted the results and drafted the manuscript. JG made substantial contributions to the experiments and data analysis.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Philippe J and Raccah D: Treating type 2 diabetes: How safe are current therapeutic agents? Int J Clin Pract. 63:321–332. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Akkati S, Sam KG and Tungha G: Emergence of promising therapies in diabetes mellitus. J Clin Pharmacol. 51:796–804. 2011. View Article : Google Scholar : PubMed/NCBI

3 

King H, Aubert RE and Herman WH: Global burden of diabetes, 1995–2025: Prevalence, numerical estimates and projections. Diabetes Care. 21:1414–1431. 1998. View Article : Google Scholar : PubMed/NCBI

4 

American Diabetes Association, : Diagnosis and classification of diabetes mellitus. Diabetes Care. 31 Suppl 1:S55–S60. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Patterson CC, Gyurus E, Rosenbauer J, Cinek O, Neu A, Schober E, Parslow RC, Joner G, Svensson J, Castell C, et al: Trends in childhood type 1 diabetes incidence in Europe during 1989–2008: Evidence of non-uniformity over time in rates of increase. Diabetologia. 55:2142–2147. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Patterson CC, Dahlquist GG, Gyurus E, Green A and Soltész G; EURODIAB Study Group, : Incidence trends for childhood type 1 diabetes in Europe during 1989–2003 and predicted new cases 2005–20: a multicentre prospective registration study. Lancet. 373:2027–2033. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Redondo MJ, Jeffrey J, Fain PR, Eisenbarth GS and Orban T: Concordance for islet autoimmunity among monozygotic twins. N Engl J Med. 359:2849–2850. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Davidson MH: Cardiovascular risk factors in a patient with diabetes mellitus and coronary artery disease: Therapeutic approaches to improve outcomes: Perspectives of a preventive cardiologist. Am J Cardiol. 110 9 Suppl:43B–49B. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Robertson RP: Islet transplantation a decade later and strategies for filling a half-full glass. Diabetes. 59:1285–1291. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Ehses JA, Perren A, Eppler E, Ribaux P, Pospisilik JA, Maor-Cahn R, Gueripel X, Ellingsgaard H, Schneider MK, Biollaz G, et al: Increased number of islet-associated macrophages in type 2 diabetes. Diabetes. 56:2356–2370. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Eizirik DL, Sammeth M, Bouckenooghe T, Bottu G, Sisino G, Igoillo-Esteve M, Ortis F, Santin I, Colli ML, Barthson J, et al: The human pancreatic islet transcriptome: expression of candidate genes for type 1 diabetes and the impact of pro-inflammatory cytokines. PLoS Genet. 8:e10025522012. View Article : Google Scholar : PubMed/NCBI

12 

Wellen KE and Hotamisligil GS: Inflammation, stress and diabetes. J Clin Invest. 115:1111–1119. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Arif S, Moore F, Marks K, Bouckenooghe T, Dayan CM, Planas R, Vives-Pi M, Powrie J, Tree T, Marchetti P, et al: Peripheral and islet interleukin-17 pathway activation characterizes human autoimmune diabetes and promotes cytokine-mediated beta-cell death. Diabetes. 60:2112–2119. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Atkinson MA, Eisenbarth GS and Michels AW: Type 1 diabetes. Lancet. 383:69–82. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Baldwin AC, Green CD, Olson LK, Moxley MA and Corbett JA: A role for aberrant protein palmitoylation in FFA-induced ER stress and beta-cell death. Am J Physiol Endocrinol Metab. 302:E1390–E1398. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Imai Y, Dobrian AD, Weaver JR, Butcher MJ, Cole BK, Galkina EV, Morris MA, Taylor-Fishwick DA and Nadler JL: Interaction between cytokines and inflammatory cells in islet dysfunction, insulin resistance and vascular disease. Diabetes Obes Metab. 15 Suppl 3:S117–S129. 2013. View Article : Google Scholar

17 

Cnop M, Welsh N, Jonas JC, Jörns A, Lenzen S and Eizirik DL: Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes. 54 Suppl 2:S97–S107. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Eizirik DL, Colli ML and Ortis F: The role of inflammation in insulitis and beta-cell loss in type 1 diabetes. Nat Rev Endocrinol. 5:219–226. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Corbett JA and McDaniel ML: Does nitric oxide mediate autoimmune destruction of beta-cells? Possible therapeutic interventions in IDDM. Diabetes. 41:897–903. 1992. View Article : Google Scholar : PubMed/NCBI

20 

Chen Y, Huang JH, Ning Y and Shen ZY: Icariin and its pharmaceutical efficacy: research progress of molecular mechanism. Zhong Xi Yi Jie He Xue Bao. 9:1179–1184. 2011.(In Chinese). View Article : Google Scholar : PubMed/NCBI

21 

Wu JF, Dong JC and Xu CQ: Effects of icariin on inflammation model stimulated by lipopolysaccharide in vitro and in vivo. Zhongguo Zhong Xi Yi Jie He Za Zhi. 29:330–334. 2009.(In Chinese). PubMed/NCBI

22 

Liu MH, Sun JS, Tsai SW, Sheu SY and Chen MH: Icariin protects murine chondrocytes from lipopolysaccharide-induced inflammatory responses and extracellular matrix degradation. Nutr Res. 30:57–65. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Xu CQ, Liu BJ, Wu JF, Xu YC, Duan XH, Cao YX and Dong JC: Icariin attenuates LPS-induced acute inflammatory responses: involvement of PI3K/Akt and NF-kappaB signaling pathway. Eur J Pharmacol. 642:146–153. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Bae UJ, Lee da Y, Song MY, Lee SM, Park JW, Ryu JH and Park BH: A prenylated flavan from Broussonetia kazinoki prevents cytokine-induced beta-cell death through suppression of nuclear factor-kB activity. Biol Pharm Bull. 34:1026–1031. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Saldeen J: Cytokines induce both necrosis and apoptosis via a common Bcl-2-inhibitable pathway in rat insulin-producing cells. Endocrinology. 141:2003–2010. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Tewari M, Quan LT, O'Rourke K, Desnoyers S, Zeng Z, Beidler DR, Poirier GG, Salvesen GS and Dixit VM: Yama/CPP32 beta, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly (ADP-ribose) polymerase. Cell. 81:801–809. 1995. View Article : Google Scholar : PubMed/NCBI

27 

Heimberg H, Heremans Y, Jobin C, Leemans R, Cardozo AK, Darville M and Eizirik DL: Inhibition of cytokine-induced NF-kappaB activation by adenovirus-mediated expression of a NF-kappaB super-repressor prevents beta-cell apoptosis. Diabetes. 50:2219–2224. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Eldor R, Yeffet A, Baum K, Doviner V, Amar D, Ben-Neriah Y, Christofori G, Peled A, Carel JC, Boitard C, et al: Conditional and specific NF-kappaB blockade protects pancreatic beta cells from diabetogenic agents. Proc Natl Acad Sci USA. 103:5072–5077. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Gong Y, Shi J, Xie GY, Liu HR and Qi MY: Amelioration of icariin for the epididymis impairment induced by streptozocin (STZ) in rats. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 29:47–50. 2013.(In Chinese). PubMed/NCBI

30 

Zhang WP, Bai XJ, Zheng XP, Xie XL and Yuan ZY: Icariin attenuates the enhanced prothrombotic state in atherosclerotic rabbits independently of its lipid-lowering effects. Planta Med. 79:731–736. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Xin H, Zhou F, Liu T, Li GY, Liu J, Gao ZZ, Bai GY, Lu H and Xin ZC: Icariin ameliorates streptozotocin-induced diabetic retinopathy in vitro and in vivo. Int J Mol Sci. 13:866–878. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Qi MY, Kai C, Liu HR, Su YH and Yu SQ: Protective effect of Icariin on the early stage of experimental diabetic nephropathy induced by streptozotocin via modulating transforming growth factor beta1 and type IV collagen expression in rats. J Ethnopharmacol. 138:731–736. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Lu YF, Xu YY, Jin F, Wu Q, Shi JS and Liu J: Icariin is a PPARα activator inducing lipid metabolic gene expression in mice. Molecules. 19:18179–18191. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Tran B, Oliver S, Rosa J and Galassetti P: Aspects of inflammation and oxidative stress in pediatric obesity and type 1 diabetes: An overview of ten years of studies. Exp Diabetes Res. 2012:6836802012. View Article : Google Scholar : PubMed/NCBI

35 

Devaraj S, Dasu MR, Rockwood J, Winter W, Griffen SC and Jialal I: Increased toll-like receptor (TLR) 2 and TLR4 expression in monocytes from patients with type 1 diabetes: Further evidence of a proinflammatory state. J Clin Endocrinol Metab. 93:578–583. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Devaraj S, Glaser N, Griffen S, Wang-Polagruto J, Miguelino E and Jialal I: Increased monocytic activity and biomarkers of inflammation in patients with type 1 diabetes. Diabetes. 55:774–779. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Yamagishi S: Advanced glycation end products and receptor-oxidative stress system in diabetic vascular complications. Ther Apher Dial. 13:534–539. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Maritim AC, Sanders RA and Watkins JB III: Diabetes, oxidative stress and antioxidants: A review. J Biochem Mol Toxicol. 17:24–38. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Chen Y, Sun T, Wu J, Kalionis B, Zhang C, Yuan D, Huang J, Cai W, Fang H and Xia S: Icariin intervenes in cardiac inflammaging through upregulation of SIRT6 enzyme activity and inhibition of the NF-Kappa B pathway. Biomed Res Int. 2015:8959762015.PubMed/NCBI

40 

Zhou H, Yuan Y, Liu Y, Ni J, Deng W, Bian ZY, Dai J and Tang QZ: Icariin protects H9c2 cardiomyocytes from lipopolysaccharide-induced injury via inhibition of the reactive oxygen species-dependent c-Jun N-terminal kinases/nuclear factor-kB pathway. Mol Med Rep. 11:4327–4332. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Wei Y, Liu B, Sun J, Lv Y, Luo Q, Liu F and Dong J: Regulation of Th17/Treg function contributes to the attenuation of chronic airway inflammation by icariin in ovalbumin-induced murine asthma model. Immunobiology. 220:789–797. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Shen R, Deng W, Li C and Zeng G: A natural flavonoid glucoside icariin inhibits Th1 and Th17 cell differentiation and ameliorates experimental autoimmune encephalomyelitis. Int Immunopharmacol. 24:224–231. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Chi L, Gao W, Shu X and Lu X: A natural flavonoid glucoside, icariin, regulates Th17 and alleviates rheumatoid arthritis in a murine model. Mediators Inflamm. 2014:3920622014. View Article : Google Scholar : PubMed/NCBI

44 

Li L, Sun J, Xu C, Zhang H, Wu J, Liu B and Dong J: Icariin ameliorates cigarette smoke induced inflammatory responses via suppression of NF-kB and modulation of GR in vivo and in vitro. PLoS One. 9:e1023452014. View Article : Google Scholar : PubMed/NCBI

45 

Cui J, Zhu M, Zhu S, Wang G, Xu Y and Geng D: Inhibitory effect of icariin on Ti-induced inflammatory osteoclastogenesis. J Surg Res. 192:447–453. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Tao F, Qian C, Guo W, Luo Q, Xu Q and Sun Y: Inhibition of Th1/Th17 responses via suppression of STAT1 and STAT3 activation contributes to the amelioration of murine experimental colitis by a natural flavonoid glucoside icariin. Biochem Pharmacol. 85:798–807. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Zhou J, Wu J, Chen X, Fortenbery N, Eksioglu E, Kodumudi KN, Pk EB, Dong J, Djeu JY and Wei S: Icariin and its derivative, ICT, exert anti-inflammatory, anti-tumor effects and modulate myeloid derived suppressive cells (MDSCs) functions. Int Immunopharmacol. 11:890–898. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Chen SR, Xu XZ, Wang YH, Chen JW, Xu SW, Gu LQ and Liu PQ: Icariin derivative inhibits inflammation through suppression of p38 mitogen-activated protein kinase and nuclear factor-kappaB pathways. Biol Pharm Bull. 33:1307–1313. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Hsieh TP, Sheu SY, Sun JS and Chen MH: Icariin inhibits osteoclast differentiation and bone resorption by suppression of MAPKs/NF-kB regulated HIF-1α and PGE (2) synthesis. Phytomedicine. 18:176–185. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Guo J, Li F, Wu Q, Lu Y and Shi J: Protective effects of icariin on brain dysfunction induced by lipopolysaccharide in rats. Phytomedicine. 17:950–955. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Grey ST, Longo C, Shukri T, Patel VI, Csizmadia E, Daniel S, Arvelo MB, Tchipashvili V and Ferran C: Genetic engineering of a suboptimal islet graft with A20 preserves beta cell mass and function. J Immunol. 170:6250–6256. 2003. View Article : Google Scholar : PubMed/NCBI

52 

Xu HB and Huang ZQ: Icariin enhances endothelial nitric-oxide synthase expression on human endothelial cells in vitro. Vascul Pharmacol. 47:18–24. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Wo Y, Zhu D, Yu Y and Lou Y: Involvement of NF-kappaB and AP-1 activation in icariin promoted cardiac differentiation of mouse embryonic stem cells. Eur J Pharmacol. 586:59–66. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Zeng KW, Fu H, Liu GX and Wang XM: Icariin attenuates lipopolysaccharide-induced microglial activation and resultant death of neurons by inhibiting TAK1/IKK/NF-kappaB and JNK/p38 MAPK pathways. Int Immunopharmacol. 10:668–678. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Wu W, Pan C, Yu H, Gong H and Wang Y: Heparanase expression in gallbladder carcinoma and its correlation to prognosis. J Gastroenterol Hepatol. 23:491–497. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Yang L, Wang Y, Guo H and Guo M: Synergistic anti-cancer effects of icariin and temozolomide in glioblastoma. Cell Biochem Biophys. 71:1379–1385. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Han H, Xu B, Hou P, Jiang C, Liu L, Tang M, Yang X, Zhang Y and Liu Y: Icaritin sensitizes human glioblastoma cells to TRAIL-induced apoptosis. Cell Biochem Biophys. 72:533–542. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Zhang Y, Wei Y, Zhu Z, Gong W, Liu X, Hou Q, Sun Y, Chai J, Zou L and Zhou T: Icariin enhances radiosensitivity of colorectal cancer cells by suppressing NF-kB activity. Cell Biochem Biophys. 69:303–310. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Shi DB, Li XX, Zheng HT, Li DW, Cai GX, Peng JJ, Gu WL, Guan ZQ, Xu Y and Cai SJ: Icariin-mediated inhibition of NF-kB activity enhances the in vitro and in vivo antitumour effect of 5-fluorouracil in colorectal cancer. Cell Biochem Biophys. 69:523–530. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Gazdar AF, Chick WL, Oie HK, Sims HL, King DL, Weir GC and Lauris V: Continuous, clonal, insulin- and somatostatin-secreting cell lines established from a transplantable rat islet cell tumor. Proc Natl Acad Sci USA. 77:3519–3523. 1980. View Article : Google Scholar : PubMed/NCBI

61 

Praz GA, Halban PA, Wollheim CB, Blondel B, Strauss AJ and Renold AE: Regulation of immunoreactive-insulin release from a rat cell line (RINm5F). Biochem J;. 210:345–352. 1983. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 16 Issue 3

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
Zhong S, Ge J and Yu JY: Icariin prevents cytokine‑induced β‑cell death by inhibiting NF‑κB signaling. Exp Ther Med 16: 2756-2762, 2018
APA
Zhong, S., Ge, J., & Yu, J. (2018). Icariin prevents cytokine‑induced β‑cell death by inhibiting NF‑κB signaling. Experimental and Therapeutic Medicine, 16, 2756-2762. https://doi.org/10.3892/etm.2018.6502
MLA
Zhong, S., Ge, J., Yu, J."Icariin prevents cytokine‑induced β‑cell death by inhibiting NF‑κB signaling". Experimental and Therapeutic Medicine 16.3 (2018): 2756-2762.
Chicago
Zhong, S., Ge, J., Yu, J."Icariin prevents cytokine‑induced β‑cell death by inhibiting NF‑κB signaling". Experimental and Therapeutic Medicine 16, no. 3 (2018): 2756-2762. https://doi.org/10.3892/etm.2018.6502