Open Access

Serum proteomic analysis of the anti‑arthritic effects of sinomenine on rats with collagen‑induced arthritis

  • Authors:
    • Xin Qian
    • Zhiming Zhao
    • Wei Shang
    • Zhihan Xu
    • Beibei Zhang
    • Hui Cai
  • View Affiliations

  • Published online on: May 3, 2018     https://doi.org/10.3892/mmr.2018.8959
  • Pages: 49-58
  • Copyright: © Qian 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

Rheumatoid arthritis (RA) is an autoimmune disease, which is characterized by inflammatory synovitis, and the subsequent destruction of articular cartilage and bone. Sinomenine is a traditional Chinese medicine, which has been employed as a clinical treatment for RA for several years in China. The present study investigated the anti‑arthritic effects of sinomenine on Sprague‑Dawley rats with collagen‑induced arthritis (CIA). The differentially expressed proteins in serum were measured by proteomic analysis in order to generate a differentially expressed protein network. A total of 320 differentially expressed proteins were detected in the drug‑treated group compared with in the control group. In the sinomenine‑treated group, 79 differentially expressed proteins were detected compared with in the model group, and among these, 46 proteins were upregulated. Gene ontology analysis revealed that five functions were affected by sinomenine treatment of CIA rats compared with in the model group. In addition, Ingenuity® Pathway Analysis was used to measure enriched signaling pathways, which revealed nuclear factor‑κB, histones, heat shock proteins and protein kinase B as core proteins, generating ~60 pair associations in the network. To the best of our knowledge, the present study is the first to perform proteomic analysis in sinomenine‑treated CIA rats, and the results revealed that numerous targets were involved in the process. In addition, the present study provided a novel approach and evidence for exploring the biological effects of sinomenine. Therefore, the findings of the present study may provide a novel insight into the anti‑RA mechanisms of sinomenine, and may justify further exploration into its function in other relevant diseases.

Introduction

Rheumatoid arthritis (RA) is primarily characterized by inflammatory synovitis, with subsequent destruction of articular cartilage and bone, joint swelling and space narrowing, and joint stiffness, deformity and dysfunction (1,2). RA is an autoimmune disease, and the pathological features mostly affect multiple small symmetrical joints of the hands and feet (1). The current therapeutic strategies available for RA include nonsteroidal anti-inflammatory drugs (NSAIDs), disease modifying anti-rheumatic drugs, glucocorticoids and surgery (3). Sinomenine is a bioactive alkaloid, which is extracted from the stems and roots of the Chinese medicine Sinomenium acutum. Sinomenine exerts various pharmacological activities, including anti-arthritic, immunosuppressive, neuroprotective, anti-inflammatory and anticancer effects (46). Sinomenine is widely used as a traditional Chinese medicine in the treatment of RA, as it exerts a biological effect on immunoregulation, anti-inflammation and bone protection (7,8). In addition, sinomenine is effective in ameliorating morning joint stiffness and painful joints, and exhibits fewer side effects on the digestive system compared with NSAIDs (9). Since sinomenine exerts immunosuppressive and anti-inflammatory effects, it is widely used to treat RA in Chinese clinical practice (10).

The proteome is an entire set of proteins encoded by the full genome produced by an organism or system. In addition, the proteome is the subject to sorting N-terminal peptides, which may be identified and quantified by mass spectrometrometry (MS) (1113). In recent years, proteome analysis has been employed to enhance understanding of malignant tumors and malaria (14,15). However, the association between RA and the serum proteome requires further analysis. The mechanisms underlying the development of RA are complex and are not fully understood. Few studies have focused on the association between RA and the serum proteome (1618); therefore, the present study performed proteomic analysis to detect the anti-RA mechanisms of sinomenine.

The pathological features of collagen-induced arthritis (CIA) in rats are consistent with typical pathological alterations in patients with RA; therefore, CIA is the most widely studied CIA model in preclinical studies (19). The present study demonstrated that sinomenine exerted anti-inflammatory effects, and alleviated the hyperplasia of fibrous tissue to exert an anti-arthritic effect. In addition, the present study investigated serum proteome profiles in the blank control group, model group, test group and positive control group, in order to generate a differentially expressed protein network map and evaluate the effectiveness of sinomenine in RA via multitarget methods.

Materials and methods

Animals

A total of 60 Sprague-Dawley female rats (aged 6–8 weeks old; 190–200 g) were purchased from the Laboratory Animal Research Center of Nanjing University of Chinese Medicine (Nanjing, China). All rats were housed at 26°C under pathogen-free conditions with a 12 h light/dark cycle and a 60% humidity, and fed with standard rat chow and water ad libitum. All experiments were conducted in compliance with the guidelines for the Care and Use of Laboratory Animals (20), and the present study was approved by the Institutional Animal Care and Use Committee of Nanjing University of Chinese Medicine. Rats were divided into six groups (10 rats per group), five of which underwent CIA.

Assessment of CIA in rats

The CIA model was established according to a previously described protocol (19). Briefly, 8 mg type II collagen (CII) (Chondrex, Redmond, WA, USA) was dissolved in 0.1 mol/l acetic acid and vortexed at 4°C; the concentration of CII reached 2 mg/ml. A total of 100 µg CII was emulsified thoroughly with the same volume of complete Freund's adjuvant (Chondrex) in an ice bath; the final concentration of CII reached 1 mg/ml. A total of 50 rats were injected subcutaneously at the tail base with 200 µl CII emulsion for the first immunization. After 7 days, 100 µg CII was dissolved and emulsified at the same concentration using incomplete Freund's adjuvant (Chondrex), and 100 µl emulsion was subcutaneously administered into the tail as a booster injection. Clinical arthritis was measured and the Arthritis Index (AI) was analyzed, as presented in Table I. The AI for each rat was expressed as the sum of the scores for all four limbs; therefore, the maximum AI was 16.

Table I.

Scoring system for the evaluation of arthritis severity.

Table I.

Scoring system for the evaluation of arthritis severity.

Severity scoreDegree of inflammation in the joints
0No evidence of erythema and swelling
1Erythema and mild swelling confined to the tarsals or ankle joint
2Erythema and mild swelling extending from the ankle to the tarsals
3Erythema and moderate swelling extending from the ankle to metatarsal joints
4Erythema and severe swelling encompass the ankle, foot and digits, or ankylosis of the limb
In vivo drug administration

CIA rats were randomly separated into the model control group, low dose group (sinomenine 30 mg/kg/day), middle dose group (sinomenine 60 mg/kg/day), high dose group (sinomenine 120 mg/kg/day) and positive control group (methotrexate 0.5 mg/kg/day). Sinomenine (cat no. Z20010174; Hunan Zhengqing Pharmaceutical, Hunan, China) was dissolved in normal saline at various concentrations and administered every day. Methotrexate (cat no. H31020644; Shanghai SINE Pharmaceutical Co., Ltd., Shanghai, China) was dissolved and 0.5 mg/kg/day methotrexate was administered every 3 days by gavage. The model group and blank control group were administered saline (1 ml/100 g) by gavage. The drug was administered continuously for 28 days.

Rheumatoid serum biochemical measurements

After treatment, rats were anesthetized with 5% isoflurane, and blood samples (3 ml) were obtained from the abdominal aorta, after which the rats were sacrificed. The blood samples were centrifuged at 3,000 × g for 10 min at 4°C. Serum was isolated for measurement of rheumatoid factor (RF) and C-reactive protein (CRP), according to the manufacturer's protocols (Abcam, Cambridge, UK; cat nos. ab178653 and ab108827).

Measurement of alanine aminotransferase (ALT) and aspartate aminotransferase (AST)

AST assay kit (GOT kit; cat no. C010-2) and ALT assay kit (GPT kit; cat no. C009-2) were purchased from Nanjing Jiancheng Bioengineering Institute (Nanjing, China). ALT and AST activity were measured after 6 weeks according to the manufacturer's protocols.

Histological scoring

Rats were euthanized using CO2 (displacement rate of the chamber volume/min, 10%), and the paw and knee joints of rats were fixed in 10% paraformaldehyde for 1 h at room temperature, decalcified in EDTA, embedded in paraffin and then sectioned (size, 4 µm). Tissue sections were mounted on slides for staining with hematoxylin and eosin for 1 h at room temperature. All sections were randomized and evaluated by two trained observers who were blinded to the treatment groups and the arthritis severity of each rat. The data were expressed as mean inflammation score and all scores were based on a scale of 0–5 (Table II). For each section, the number of positively stained cells was counted in 20 fields using a phase contrast microscope (magnification, ×200).

Table II.

Scoring system for the evaluation of histology.

Table II.

Scoring system for the evaluation of histology.

ScoreDegree of histological scoring
0No staining
1Few of the cells were positively stained
2Some (<50%) of the cells were stained
3~50% of the cells were stained
4>50% of the cells were stained
5All cells stained
Serum sample processing

After treatment, rats were anesthetized with 5% isoflurane, and blood samples were obtained from the abdominal aorta and transferred to a 1.5 ml Eppendorf protein tube (Eppendorf, Hamburg, Germany). The blood samples were briefly vortexed and incubated at 4°C for 6 h to precipitate serum proteins. Subsequently, the samples were centrifuged at 1,000 × g for 10 min at 4°C. The collected supernatants were then centrifuged at 16,000 × g for 10 min at 4°C to remove lipids and the clearest serum was collected and centrifuged 4,000 × g at 4°C for 10 min to remove any remaining cells. The extracted serum was transferred to a 200 µl Eppendorf tube and stored at −20°C.

Protein extraction

A total of 60 µl elution buffer [7 M urea, 1% (w/v) CHAPS] was added to 10 µl serum (n=6/group). Subsequently, dithiothreitol was added to reach a final concentration of 10 mM. The samples were boiled at 56°C in water for 1 h, followed by the addition of PBS to achieve a concentration of 55 mM, and were incubated for 1 h in the dark at room temperature. The addition of iced pure acetone resulted in the formation of precipitate, followed by centrifugation at 4,000 × g at 4°C for 10 min, and removal of the supernatant. The precipitate was dissolved in 300 µl physiological saline in a vortex tube for 3 min. The proteins were quantified using a Bradford assay.

Liquid chromatography and MS

Serum proteome was analyzed using LTQ-Orbitrap-Veces iFunnel (Thermo Fisher Scientific, Inc., Waltham, MA, USA) equipped with a reversed-phase capillary column and interfaced with the nanoflow LC system (1,100; Agilent Technologies, Inc., Santa Clara, CA, USA). The peptides (500 ml) were enriched on the C18 enrichment column and separated on a 75 µm ×43 mm analytical/separation column in the protein chip (Agilent HPLC-Chip: G4240-62001ZORBAX 300SB-C18; Agilent Technologies, Inc.) using a gradient mobile phase consisting of two different solvents, 0.1% formic acid solution (solvent A) and 90% acetonitrile (solvent B), at a flow rate of 200 nl/min. The following gradient method was used for the separation of peptides on the chip over a period of 60 min: From 0–60%. Nitrogen gas was maintained at 120°C with a 9 l/min flow rate and a nebulizer pressure of 207 kPa. Positive ions were generated via electrospray and MRM transitions were assessed using 350–1,750 m/z. The MS/MS data were further analyzed using MaxQuant (version 1.2.2.5; http://www.maxquant.org). MaxQuant is designed as a three-tiered application for the analysis of data, application logic and presentation. The spectra data were determined and subsequently searched using the UniProt database (http://www.uniprot.org/). Pathway analysis was performed with Ingenuity® Pathway Analysis (IPA) software version 1 (Ingenuity Systems; Qiagen, Inc., Valencia, CA, USA).

Statistical analysis

Data are expressed as the mean ± standard deviation of at least three independent experiments. Statistical analysis was performed using SPSS version 16.0 software (SPSS, Inc., Chicago, IL, USA). A Student's t-test was used to compare the discrepancy between two groups. One-way analysis of variance followed by Duncan's test was used to determine the difference between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Sinomenine improves clinical arthritic conditions in CIA rats

The present study investigated the in vivo efficacy of sinomenine in CIA rats. The CIA model was elicited in a genetically susceptible rat strain by immunization with CII emulsified in complete Freund's adjuvant. The body weights of the rats were monitored weekly, and the results demonstrated that beginning from day 21, CIA rats gained less weight compared with in the blank control group. Treatment with sinomenine (week 5, high dose) significantly reversed weight loss caused by RA compared with in the model group (Table III).

Table III.

Body weights (g) of rats with collagen-induced arthritis.

Table III.

Body weights (g) of rats with collagen-induced arthritis.

GroupWeek 2Week 3Week 4Week 5Week 6
Blank control244.0±15.4285.3±16.3324.3±22.9358.5±30.2380.5±30.7
Model244.8±23.6263.4±30.2 286.7±31.9a 312.0±31.7a 300.3±29.5b
Positive control246.0±24.2276.4±39.7299.8±41.2318.2±44.0 313.2±41.9b
High dose240.4±27.6267.7±32.7293.8±30.8 331.9±36.7a,d 319.6±33.2b,c
Middle dose234.1±26.2 251.0±34.0a 278.8±36.5a 309.1±42.5a 308.6±40.8b
Low dose246.8±27.2260.1±39.8 281.8±36.8a 310.4±34.4a 313.8±32.1b

a P<0.05

b P<0.01 vs. the blank control group.

c P<0.05

d P<0.01 vs. the model group.

Clinical score was used to measure the progression of arthritis development. The model group developed severe swelling, erythema and joint rigidity of the hind paws after 6 weeks (data not shown). Conversely, the experimental and positive control groups exhibited showed a lower AI (P<0.05) compared with in the model group. In addition, in the high dose sinomenine group, the AI was significantly attenuated compared with in the model group (P<0.01; Table IV). These results indicated that sinomenine and methotrexate may improve clinical arthritic conditions in CIA rats, and high dose sinomenine treatment exerted the optimal efficacy in CIA rats.

Table IV.

Arthritis index of rats with collagen-induced arthritis at week 6.

Table IV.

Arthritis index of rats with collagen-induced arthritis at week 6.

GroupArthritis index
Model13.5±0.9
Positive control 8.1±2.5a
High dose 6.4±2.3b
Middle dose 10.5±2.0a
Low dose 10.5±1.8a

a P<0.05

b P<0.01 vs. the model group.

Sinomenine improves histological parameters in CIA rats

The histology of tissues from CIA rats was analyzed, in order to determine whether sinomenine prevented articular destruction of talocrural joints. As shown in Fig. 1, infiltration of inflammatory cells, pannus invasion, cartilage damage and subchondral bone erosion were detected in CIA rats (Fig. 1). Histological scores of the individual groups are shown in Table V. The histological score of the model group was significantly higher than in the blank control group rats (P<0.01), and was reduced in the experimental groups, which indicated that sinomenine improved joint histological conditions, synovial swelling, congestion and hyperplasia in CIA rats (Table V). The joints of CIA rats treated with a high dose of sinomenine demonstrated less inflammatory cell infiltration and synovial hyperplasia than the positive control group. These results suggested that sinomenine improved the histological parameters in CIA rats.

Table V.

Histological score of joints in rats.

Table V.

Histological score of joints in rats.

GroupNumberScore
Blank control81.00±0.00
Model8 5.50±1.41a
Positive control8 4.00±1.07b
Low dose84.25±1.04
Middle dose8 4.00±1.07b
High dose8 3.75±1.04b

a P<0.01 vs. the blank control group.

b P<0.05 vs. the model group.

Sinomenine attenuates the inflammatory response in the joints of CIA rats

The present study next investigated the mechanisms underlying the decreased occurrence and severity of CIA following sinomenine treatment. The results of RF detection detected an apparent discrepancy between sinomenine-treated groups and the model group (P<0.05). Notably, high dose sinomenine exhibited an improved anti-arthritic effect compared with low dose sinometine (Table VI).

Table VI.

Expression levels of rheumatoid factor in serum at week 6.

Table VI.

Expression levels of rheumatoid factor in serum at week 6.

GroupRheumatoid factor (IU/ml)
Blank control 5,007.8±3,168.2
Model 9,176.5±3,757.2a
Positive control 5,048.2±3,132.2b
High dose 4,578.0±3,489.5b
Middle dose 4,750.8±4,764.3b
Low dose 5,756.3±4,963.4c

a P<0.05 vs. the blank control group.

b P<0.01

c P<0.05 vs. the model group.

The expression levels of CRP in the positive control group were significantly lower than in the model group (Table VII). Sinomenine treatment attenuated the secretion of CRP in CIA rats compared with in the model or positive control groups. These findings indicated that sinomenine exhibited an improved anti-arthritic effect compared with methotrexate.

Table VII.

Expression levels of C-reactive protein in serum at week 6.

Table VII.

Expression levels of C-reactive protein in serum at week 6.

GroupC-reactive protein (mg/ml)
Blank control261.0±50.2
Model 636.3±123.4a
Positive control 578.1±164.2a,b
High dose 340.0±85.4b,c
Middle dose 489.2±107.1d,e
Low dose679.9±21.2

a P<0.01 vs. the control group.

b P<0.01 vs. the model group.

c P<0.01 vs. the positive control group.

d P<0.05 vs. the model group.

e P<0.05 vs. the positive control group.

The results revealed that the secretion of ALT and AST was not significantly different between the sinomenine-treated groups and the blank control group; whereas ALT was significantly decreased in the high dose and positive control groups compared with the model group. Furthermore, a marked decrease of ALT and AST expression levels were observed in the high dose group compared with the middle and low dose groups (Table VIII). ALT and AST are the major markers of hepatic damage in the plasma. Taken together, the results suggest that a high dose of sinomenine exerted significant liver function improvement in CIA rats (Table VIII) compared with the model group. Furthermore, the positive control group demonstrated a significant decrease in leukocyte, erythrocyte and hemoglobin levels compared with the blank control group. Bone marrow suppression represents the decrease in the production of leukocytes, erythrocytes and/or platelets (21). Therefore, it can be suggested that methotrexate caused bone marrow suppression in CIA rats (Table IX). However, there were no significant alterations between the sinomenine-treated groups and the blank control group, which indicated that sinomenine did not induce bone marrow suppression and liver damage. In addition, the number of platelets in the low dose sinomenine group was significantly increased compared with in the blank control group, which indicated that low dose sinomenine treatment exerted a marked increase in platelet production or release. Therefore, sinomenine may exert an improved anti-arthritic effect, associated with no liver damage and bone marrow inhibition compared with methotrexate, which is associated with chronic liver damage (22). These results suggested that sinomenine may be used in the clinical treatment of RA.

Table VIII.

Expression levels of AST and ALT in serum.

Table VIII.

Expression levels of AST and ALT in serum.

GroupALT (U/l)AST (U/l)
Blank control7.99±3.3810.84±4.08
Model10.30±2.1114.39±1.98
Positive control 7.63±1.62a12.13±2.34
High dose 7.15±2.33a 11.77±3.69a
Middle dose7.96±2.1113.15±1.16
Low dose8.51±3.6913.13±5.48

a P<0.05 vs. the model group. AST, aspartate aminotransferase; ALT, alanine aminotransferase.

Table IX.

Blood analysis.

Table IX.

Blood analysis.

GroupLeukocytesErythrocytesHemoglobinPlatelets
Blank control6.57±1.547.64±0.71151.38±9.401,031.8±107.3
Model6.58±0.976.75±1.10130.75±18.30939.4±124.4
Positive control 5.60±1.22a,b 6.74±0.49a 134.25±8.96a 1,342.0±138.1a,b
High dose6.66±1.697.01±1.53133.88±34.661,115.3±270.9
Middle dose6.95±2.207.68±0.76151.57±18.441,063.1±172.8
Low dose6.93±1.757.57±0.53139.29±19.90 1,308.1±197.1a

a P<0.01 vs. the blank control group.

b P<0.01 vs. the model group.

Identification of differentially expressed proteins between sinomenine-treated groups and model group

To investigate the underlying mechanisms involved in sinomenine-treated CIA rats, proteomic analysis was performed. The data revealed that 320 differential proteins were expressed in the sinomenine-treated groups compared with in the model group. There were 79 differentially expressed proteins identified in the low dose group, and among them, 36 proteins were upregulated. The top 12 up- and downregulated proteins were presented in Table X. In addition, five highly relevant biological processes were identified from 16 relevant biological processes by gene ontology enrichment analysis (13). These biological processes were cell cycle (P-value 6.66×10−3-8.91×10−6; 14 associated genes); cell morphology (P-value, 6.43×10−3-2.82×10−4; 24 associated genes); cellular function and maintenance (P-value, 6.43×10−3-4.52×10−4; 22 associated genes); cellular assembly and organization (P-value, 6.43×10−3-7.78×10−4; 24 associated genes); and post-translational modification (P-value, 3.22×10−3-7.78×10−4; 6 associated genes) (Table XI). In addition, biological processes were associated with statistically relevant molecular pathways. IPA was used to investigate the involvement of signaling pathways. The results indicated that 213 signaling pathways were enriched, which were involved in the low dose sinomenine-treated group compared with in the model group. The 37 most enriched signaling pathways are listed in Fig. 2.

Table X.

Top 12 up- and downregulated differentially expressed proteins between the sinomenine-treated group and model group.

Table X.

Top 12 up- and downregulated differentially expressed proteins between the sinomenine-treated group and model group.

Gene nameAccession no.Fold change
AmotA0A067XG49−1.8538
Phf20l1Q6P7V2−1.6297
Myom1Q62234−1.5777
Rbl1Q64701−1.5508
Pla2g4dQ14CI2−1.5336
Slc4a3Q68EG4−1.5128
Dcaf5Q80T85−1.4726
Dido1Q8C9B9−1.4686
Speer1J3QMX3−1.4320
Hfm1F6XQ35−1.4088
Trcg1Q58Y74−1.4086
Tmem131O70472−1.3927
Dnah7cA0A087WR131.6626
Dnah7aE9Q0T81.6626
PtprfA2A8L51.7042
Ankrd27Q3UMR0-21.7052
Fgfr2A1YYM71.8464
Nup155Q6ZQ451.8588
Ctnna1Q545R01.8978
TprQ8BK712.0978
RictorQ6QI06-22.2287
Dnajc8F6TQL32.2831
Prokr2Q8K4583.6995
Antxr2Q6DFX26.0189

[i] Fold changes represent the model group vs. the sinomenine-treated group.

Table XI.

Functional analysis between the sinomenine-treated group and model group.

Table XI.

Functional analysis between the sinomenine-treated group and model group.

ComparisonNameP-valueMolecules
MVD (Model group vs. Sinomenine-treated group)Cell cycle 6.66×10−3-8.91×10−614
Cell morphology 6.43×10−3-2.82×10−424
Cellular function and maintenance 6.43×10−3-4.52×10−422
Cellular assembly and organization 6.43×10−3-7.78×10−424
Post-translational modification 3.22×10−3-7.78×10−46
Identification of proteins involved in the inflammation-associated pathway

The present study used IPA software to investigate the association between differentially expressed proteins by analyzing enrichment. A protein-protein interaction network was constructed by calculating the score, which indicated that 33 proteins were associated with other proteins and led to 60 paired relationships. These proteins were primarily involved in immunomodulatory and inflammatory reactions. In addition, nuclear factor (NF)-κB, histone H3, heat shock protein (Hsp)70 and protein kinase B (Akt) were the main proteins to regulate the network (Fig. 3). Among them, 14 proteins were upregulated and 7 proteins were downregulated. For example, if histone H3 was chosen, 14 paired relationships were connected (Fig. 3).

Identification of upstream regulators

To identify the key upstream regulators governing molecular status following sinomenine treatment, an upstream regulator analysis with IPA was applied. The top seven upstream regulators that were predicted to be activated or inhibited in the sinomenine treated group are presented in Table XII. E2F1, TCF7L2, NUPR1, TGFβ1 and lipopolysaccharide were most top upstream regulators predicted to be induced following treatment with sinomenine; whereas CST5 and AHR were the top upstream regulators predicted to be inhibited following treatment with sinomenine (Table XII). Fig. 4 showed a graphical representation of the molecular networks that may exert important roles following sinomenine treatment, which revealed that the most significant upstream regulators were transforming growth factor (TGF)-β1, aryl hydrocarbon receptor (AHR) and cystatin D (CST5).

Table XII.

Upstream analysis.

Table XII.

Upstream analysis.

Upstream regulatorMolecule typeActivation z-scoreP-value of overlapTarget molecules in dataset
CST5Other−0.8160.000331COL12A1, DDX21, DOCK9, NUP155, SSFA2, SSRP1
E2F3Transcription regulator 0.00154FGFR2, NCAPG2, PPP1R13B, RBL1
SP100Transcription regulator 0.00202BRCA1, HSPA8
SMARCE1Transcription regulator 0.00223BRCA1, CYLD
E2F2Transcription regulator 0.0201FGFR2, RBL1
E2F1Transcription regulator0.0770.0212BRCA1, FGFR2, HSPA8, PPP1R13B, RBL1
TCF7L2Transcription regulator10.0225CTNNA1, DOCK9, FGFR2, OTUD7B
MmpGroup 0.0232PLG
NUP107Transporter 0.0232TPR
TMPOOther 0.0232COL12A1
ZNF423Transcription regulator 0.0232BRCA1
PGK1Kinase 0.0232PLG
GW 5074Chemical-kinase inhibitor 0.0232FGFR2
Boc-D-FMKChemical reagent 0.0232RBL1
RbGroup 0.0243FGFR2, RBL1
AHRLigand-dependent nuclear Receptor−1.9820.0243COL12A1, FGFR2, FMO3, RBL1
TBX2Transcription regulator 0.0262NCAPG2, RBL1
PHF8Enzyme 0.0265RBL1
S100A10Other 0.0265PLG
LIMS1Other 0.0265CTNNA1
GRIP1Transcription regulator 0.0265FREM2
ALCAMOther 0.0265AMOT
TNRC6AOther 0.0265RBL1
NUPR1Transcription regulator0.4470.0271ANK1, BRCA1, DIDO1, SHROOM3, SYNE2
N-Ac-Leu-Leu-norleucinalChemical-protease inhibitor 0.0289BRCA1, RBL1
TIP60Complex 0.0298RBL1
SHOXTranscription regulator 0.0298RBL1
RBL2Other 0.0436BRCA1, RBL1
RRP1BOther 0.0444BRCA1, RBL1
COL9A1Other 0.0459COL12A1
PHBTranscription regulator 0.0459RBL1
Gamma-tocotrienolChemical drug 0.0459OTUD7B
CTGFGrowth factor 0.046ABCF1, MIA3
TGFB1Growth factor0.9560.0482AASS, ABCF1, COL12A1, DDX21, FGFR2, MIA3, PPP1R13B, RBL1, RICTOR, SSRP1
DactolisibChemical drug 0.0491RICTOR
TNFAIP2Other 0.0491RBL1
RGS1Other 0.0491RBL1
NDNTranscription regulator 0.0491RBL1
LipopolysaccharideChemical drug1.9691COL12A1, F2, HSPA8, PLG

Discussion

To gain an insight into the mechanism underlying the effects of sinomenine on RA, the present study used an animal experimental arthritis model. Rats were administered with sinomenine, methotrexate or vehicle. In the CIA study, the protective effects of sinomenine against arthritis were confirmed, as evidenced by the decreased incidence and severity of arthritis following CII immunization. Sinomenine also exerted an anti-inflammatory effect, as revealed by the suppression of CRP expression. These results are consistent with those of previous studies. Yang et al (23) reported that sinomenine exerts protective effects against lipopolysaccharide-induced inflammation in piglets. Additionally, Xu et al (9) reported that sinomenine and NSAID treatment regulates CRP and improved clinical conditions of RA. Furthermore, in the present study, sinomenine did not exert liver damage or bone marrow inhibition compared with in the positive control group. To the best of our knowledge, no previous studies have focused on proteomic analysis following treatment of CIA rats with sinomenine. The current proteomic analysis study revealed that NF-κB, histone, Hsp70 and Akt interacted with other proteins, leading to 60 relationship pairs. Taken together, these results indicated that the use of sinomenine has the potential to treat RA, and the present study identified pathways in the rats with CIA involved in the response to sinomenine. These results provide information to suggest that sinomenine may be used to treat RA.

In the present study, dual high-performance liquid chromatography and MS were performed to identify the protein profiles associated with sinomenine treatment in rats with CIA. A total of 320 proteins were differentially expressed. In response to treatment with low dose sinomenine, there were 79 differentially expression proteins, among which 36 proteins were downregulated. The differentially expressed proteins were involved in tumorigenesis, developmental disorder, inflammatory, cell morphology, lipid metabolism, cell cycle, amino acid metabolism, gene expression and drug metabolism. The present study focused on network analysis via IPA software to construct the protein network in the low dose sinomenine treatment group. NF-κB, histone H3, Hsp70 and Akt were involved in the enrichment networks as core proteins, which was determined using Uniprot (http://www.uniprot.org/). In addition, the key upstream regulators that governed molecular status following sinomenine treatment were predicted; the most significant upstream regulators were TGF-β1, AHR and CST5.

Akt is involved in the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Akt kinase activity is induced following activation of PI3K in growth factor receptor-mediated signaling cascades (24). Akt is involved in tumor formation, and is also involved in higher brain function, cell size matters, cell cycle regulation and metabolic functions, and serves various roles in diseases and biological functions (25). In the present study, the proteomic analysis results revealed that enriched proteins are associated with Akt expression, thus suggesting that Akt may function as an inducer of these proteins. Therefore, the present study provided evidence to suggest that Akt is a target of the far-reaching physiological effects regulated by sinomenine in RA, as in tumor and brain disease.

IPA upstream regulator analysis predicted that the most significant upstream regulators associated with sinomenine treatment are TGF-β1, AHR and CST5. Previously, Sugiura et al (26) reported that TGF-β1 is highly expressed in joints in RA, and it is considered an anti-inflammatory regulator in RA. AHR activation may induce the production of inflammatory cytokines and RA synoviocytes (27). The present findings suggested that TGF-β1 and AHR serve a key role in the anti-arthritic effects of sinomenine. Therefore, it was hypothesized that sinomenine exerted its anti-arthritic effects via inhibition of TGF-β1 and AHR. To the best of our knowledge, there are no published studies investigating the association between RA and the expression level of CST5. In the future, the authors of the present study aim to study the association between CST5 and RA. In the present study, Akt expression enrichment was inhibited by sinomenine in CIA rats. Therefore, it was hypothesized that TGF-β1 may mediate Akt activity in RA, and both were downregulated by sinomenine. It has previously been reported that TGF-β1 enhances Akt phosphorylation in MC3T3-E1 cells (28) and A549 cells (29). The present data are consistent with these studies; however, further investigations are required to confirm this hypothesis.

The present study explored the anti-arthritic and anti-inflammatory effects of sinomenine in vivo. To the best of our knowledge, the present study is the first to perform a proteomic analysis for analyzing the effects of sinomenine against RA using a CIA rat model. The present study aimed to elucidate the associated proteins involved in sinomenine-treated RA via proteomic analysis. The results of the present study revealed that sinomenine exerted anti-arthritic effects via numerous targets during CIA. In addition, the proteomic analysis provided a novel approach and evidence for exploring the other biological effects of sinomenine. Therefore, the findings may provide an insight into the anti-RA mechanisms of sinomenine and proteomic analysis may be used to explore its functions in other relevant diseases.

Acknowledgements

The authors would like to thank Jinjin Shang for proof reading the manuscript.

Funding

No funding was received.

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

XQ, ZZ and HC designed the study and performed the experiments. WS, ZX and BZ analyzed and interpreted the experimental data, and drafted the manuscript.

Ethics approval and consent to participate

The present study was approved by the Institutional Animal Care and Use Committee of Nanjing University of Chinese Medicine (Nanjing, China).

Consent for publication

Not applicable.

Conflicts of interest

The authors declare that they have no competing interests.

References

1 

Gu X, Gu B, Lv X, Yu Z, Wang R, Zhou X, Qiao W, Mao Z, Zuo G, Li Q, et al: 1, 25-dihydroxy-vitamin D3 with tumor necrosis factor-alpha protects against rheumatoid arthritis by promoting p53 acetylation-mediated apoptosis via Sirt1 in synoviocytes. Cell Death Dis. 7:e24232016. View Article : Google Scholar : PubMed/NCBI

2 

Boissier MC, Semerano L, Challal S, Saidenberg-Kermanac'h N and Falgarone G: Rheumatoid arthritis: From autoimmunity to synovitis and joint destruction. J Autoimmun. 39:222–228. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Burmester GR, Bijlsma JWJ, Cutolo M and McInnes IB: Managing rheumatic and musculoskeletal diseases - past, present and future. Nat Rev Rheumatol. 13:443–448. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Wang Y, Fang Y, Huang W, Zhou X, Wang M, Zhong B and Peng D: Effect of sinomenine on cytokine expression of macrophages and synoviocytes in adjuvant arthritis rats. J Ethnopharmacol. 98:37–43. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Qian L, Xu Z, Zhang W, Wilson B, Hong JS and Flood PM: Sinomenine, a natural dextrorotatory morphinan analog, is anti-inflammatory and neuroprotective through inhibition of microglial NADPH oxidase. J Neuroinflammation. 4:232007. View Article : Google Scholar : PubMed/NCBI

6 

Zhou B, Lu X, Tang Z, Liu D, Zhou Y, Zeng P and Xiong H: Influence of sinomenine upon mesenchymal stem cells in osteoclastogenesis. Biomed Pharmacother. 90:835–841. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Zhang HC, Liu MX, Wang EP, Lin Z, Lv GF and Chen X: Effect of sinomenine on the expression of rheumatoid arthritis fibroblast-like synoviocytes MyD88 and TRAF6. Genet Mol Res. 14:18928–18935. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Chen XM, Huang RY, Huang QC, Chu YL and Yan JY: Systemic review and meta-analysis of the clinical efficacy and adverse effects of zhengqing fengtongning combined with methotrexate in rheumatoid arthritis. Evid Based Complement Alternat Med. 2015:9103762015. View Article : Google Scholar : PubMed/NCBI

9 

Xu M, Liu L, Qi C, Deng B and Cai X: Sinomenine versus NSAIDs for the treatment of rheumatoid arthritis: A systematic review and meta-analysis. Planta Med. 74:1423–1429. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Wang Q and Li XK: Immunosuppressive and anti-inflammatory activities of sinomenine. Int Immunopharmacol. 11:373–376. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Kumar A and Snyder M: Protein complexes take the bait. Nature. 415:123–124. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Wepf A, Glatter T, Schmidt A, Aebersold R and Gstaiger M: Quantitative interaction proteomics using mass spectrometry. Nat Methods. 6:203–205. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Pflieger D, Gonnet F, de la Fuente van Bentem S, Hirt H and de la Fuente A: Linking the proteins-elucidation of proteome-scale networks using mass spectrometry. Mass Spectrom Rev. 30:268–297. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Gollapalli K, Ghantasala S, Kumar S, Srivastava R, Rapole S, Moiyadi A, Epari S and Srivastava S: Subventricular zone involvement in Glioblastoma-A proteomic evaluation and clinicoradiological correlation. Sci Rep. 7:14492017. View Article : Google Scholar : PubMed/NCBI

15 

Ray S, Patel SK, Venkatesh A, Chatterjee G, Ansari NN, Gogtay NJ, Thatte UM, Gandhe P, Varma SG, Patankar S and Srivastava S: Quantitative proteomics analysis of plasmodium vivax induced alterations in human serum during the acute and convalescent phases of infection. Sci Rep. 7:44002017. View Article : Google Scholar : PubMed/NCBI

16 

Noh R, Park SG, Ju JH, Chi SW, Kim S, Lee CK, Kim JH and Park BC: Comparative proteomic analyses of synovial fluids and serums from rheumatoid arthritis patients. J Microbiol Biotechnol. 24:119–126. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Cheng Y, Chen Y, Sun X, Li Y, Huang C, Deng H and Li Z: Identification of potential serum biomarkers for rheumatoid arthritis by high-resolution quantitative proteomic analysis. Inflammation. 37:1459–1467. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Yanagida M, Kawasaki M, Fujishiro M, Miura M, Ikeda K, Nozawa K, Kaneko H, Morimoto S, Takasaki Y, Ogawa H, et al: Serum proteome analysis in patients with rheumatoid arthritis receiving therapy with tocilizumab: An anti-interleukin-6 receptor antibody. Biomed Res Int. 2013:6071372013. View Article : Google Scholar : PubMed/NCBI

19 

Brand DD, Latham KA and Rosloniec EF: Collagen-induced arthritis. Nat Protoc. 2:1269–1275. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Jones-Bolin S: Guidelines for the care and use of laboratory animals in biomedical research. Curr Protoc Pharmacol Appendix 4: Appendix 4B. 2012. View Article : Google Scholar

21 

Feng L, Huang Q, Huang Z, Li H, Qi X, Wang Y, Liu Z, Liu X and Lu L: Optimized animal model of cyclophosphamide-induced bone marrow suppression. Basic Clin Pharmacol Toxicol. 119:428–435. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Safaei F, Mehrzadi S, Haghighian Khadem H, Hosseinzadeh A, Nesari A, Dolatshahi M, Esmaeilizadeh M and Goudarzi M: Protective effects of gallic acid against methotrexate-induced toxicity in rats. Acta Chir Belg. 25:1–9. 2017.

23 

Yang H, Jiang C, Chen X, He K and Hu Y: Protective effects of sinomenine against LPS-induced inflammation in piglets. Microb Pathog. 110:573–577. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Butler MG, Dasouki MJ, Zhou XP, Talebizadeh Z, Brown M, Takahashi TN, Miles JH, Wang CH, Stratton R, Pilarski R and Eng C: Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations. J Med Genet. 42:318–321. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Franke TF: PI3K/Akt: Getting it right matters. Oncogene. 27:6473–6488. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Sugiura Y, Niimi T, Sato S, Yoshinouchi T, Banno S, Naniwa T, Maeda H, Shimizu S and Ueda R: Transforming growth factor beta1 gene polymorphism in rheumatoid arthritis. Ann Rheum Dis. 61:826–828. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Nguyen NT, Nakahama T, Nguyen CH, Tran TT, Le VS, Chu HH and Kishimoto T: Aryl hydrocarbon receptor antagonism and its role in rheumatoid arthritis. J Exp Pharmacol. 7:29–35. 2015.PubMed/NCBI

28 

Suzuki E, Ochiai-Shino H, Aoki H, Onodera S, Saito A, Saito A and Azuma T: Akt activation is required for TGF-β1-induced Osteoblast differentiation of MC3T3-E1 pre-osteoblasts. PLoS One. 9:e1125662014. View Article : Google Scholar : PubMed/NCBI

29 

Jo E, Park SJ, Choi YS, Jeon WK and Kim BC: Kaempferol suppresses transforming growth factor-β1-induced epithelial-to-mesenchymal transition and migration of A549 lung cancer cells by inhibiting Akt1-mediated phosphorylation of Smad3 at Threonine-179. Neoplasia. 17:525–537. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 18 Issue 1

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
Qian X, Zhao Z, Shang W, Xu Z, Zhang B and Cai H: Serum proteomic analysis of the anti‑arthritic effects of sinomenine on rats with collagen‑induced arthritis. Mol Med Rep 18: 49-58, 2018
APA
Qian, X., Zhao, Z., Shang, W., Xu, Z., Zhang, B., & Cai, H. (2018). Serum proteomic analysis of the anti‑arthritic effects of sinomenine on rats with collagen‑induced arthritis. Molecular Medicine Reports, 18, 49-58. https://doi.org/10.3892/mmr.2018.8959
MLA
Qian, X., Zhao, Z., Shang, W., Xu, Z., Zhang, B., Cai, H."Serum proteomic analysis of the anti‑arthritic effects of sinomenine on rats with collagen‑induced arthritis". Molecular Medicine Reports 18.1 (2018): 49-58.
Chicago
Qian, X., Zhao, Z., Shang, W., Xu, Z., Zhang, B., Cai, H."Serum proteomic analysis of the anti‑arthritic effects of sinomenine on rats with collagen‑induced arthritis". Molecular Medicine Reports 18, no. 1 (2018): 49-58. https://doi.org/10.3892/mmr.2018.8959