Skip to main content
Erschienen in: BMC Complementary Medicine and Therapies 1/2017

Open Access 01.12.2017 | Research article

A putative Chondroprotective role for IL-1β and MPO in herbal treatment of experimental osteoarthritis

verfasst von: Nora M. Aborehab, Mahitab H. El Bishbishy, Abeer Refaiy, Nermien E. Waly

Erschienen in: BMC Complementary Medicine and Therapies | Ausgabe 1/2017

Abstract

Background

Herbal treatment may have a chondroprotective and therapeutic effect on Osteoarthritis (OA). We investigated the mechanism of action of ginger and curcumin rhizomes cultivated in Egypt in treatment of OA in rat model.

Methods

Thirty-five albino rats were intra-articularly injected with Monosodium Iodoacetate in the knee joint. Ginger and curcumin was orally administered at doses of 200 and 400 mg/kg (F200 and F400). Serum levels of cartilage oligomeric matrix protein (COMP), hyaluronic acid (HA), malondialdehyde (MDA), myeloperoxidase (MPO), Interleukin-1 beta (IL-1β) and superoxide dismutase activity (SOD) were measured using ELISA. The composition of the herbal formula hydro-ethanolic extract was characterized using UPLC-ESI-MS. Histopathological changes in injected joints was examined using routine histopathology. Statistical analysis was performed using one-way ANOVA.

Results

Serum levels of COMP, HA, MPO, MDA, and IL-1β were significantly decreased in F 200, F 400 and V groups when compared to OA group (P value <0.0001). On the other hand SOD levels were significantly elevated in treated groups compared to OA groups (P value <0.0001).

Conclusions

The ginger/curcumin at 1:1 had chondroprotective effect via anti-inflammatory and antioxidant effect in rat OA model. Further pharmacological and clinical studies are needed to evaluate this effect.
Abkürzungen
COMP
Cartilage oligomeric matrix protein
HA
Hyaluronic acid
IL-1β
Interleukin-1 beta
MDA
Malondialdehyde
MIA
Monosodium Iodoacetate
MPO
Myeloperoxidase
OA
Osteoarthritis
OARSI
Osteoarthritis Research Society International
ROS
Reactive oxygen species
SOD
Superoxide dismutase

Background

Osteoarthritis (OA) is a common degenerative joint disease in the elderly and a leading cause of physical disability worldwide [1]. It mainly affects the weight– bearing joints, such as hips and knees [2]. OA is a degenerative disease characterized by structural changes of the knee joint including articular cartilage erosion, synovitis and remodeling of subchondral bone [3]. Patients affected by OA experience pain that worsens with motion, exercise and, with pathology progression, also at rest [4]. It was proposed that OA is the result of both mechanical and biological changes, causing loss of the normal homeostatic mechanism occurring between catabolic and anabolic processes in the articular cartilage. Chondrocyte in the mature articular cartilage is the only type of cell that is capable of production and maintenance of the extracellular matrix. Therefore any reduction in chondrocyte density will contribute to the development of OA [5]. To date, there is no effective pharmacological treatment for OA and the end stage of disease usually requires joint replacement surgery [6, 7].
A monosodium Iodoacetate (MIA) induced OA model is well described in rats especially in terms of the pathological progression of the disease. It triggers changes similar to the histological and pathophysiological features of that in human OA. The mechanism of action of MIA has been attributed to inhibition of glyceraldehyde-3-phosphate dehydrogenase. This disrupts the glucose metabolism of chondrocytes, leading to reactive oxygen species (ROS) production and caspase activation, and further to the catabolism of cartilage matrix and cell death, that can be detected both in vivo and in vitro. It therefore targets the avascular cartilage and causes chondrocyte death, fragmentation of cartilage and exposure of subchondral tissue. In addition to evoking cartilage degradation, MIA induces an acute inflammation, which is associated with increased expression of pro-inflammatory factors such as IL-1β IL-6, IL-15, COX-2 and metalloproteinase [810].
Since the pharmacologic treatment has several limitations and is not always effective in the management of OA, researchers have been striving to find alternative therapeutic agents. Herbs and extracts present a potential hope for treatment of many diseases. The use of tomato and broccoli extracts has been shown to have beneficial effect in the treatment of obesity both in rats and humans [1113].
Ginger, Zingiber officinale Rosc., rhizomes have been shown to contain a large amount of zerurnbone [14]. Zerurnbone has been shown to have anticancer and antioxidant actions [5]. In addition several studies has shown that ginger possesses medicinal effects in animals. Its ethanol extract had hypoglycemic and anti-inflammatory effects in rats [15]. Another study has shown that ginger protects against ulcer development in albino rats [16]. Ginger also has been shown to exert chondroprotective effects in an experimental model of OA in rats [5]. In addition it is safe to use as the LD50 of ginger extract was is up to 1 g/kg [17].
Curcuma longa L., Zingiberaceae (turmeric) is a herb that is used in the Fareast folk medicine for the treatment of biliary disorders among other diseases [18]. Its extract has been shown to have beneficial effects in the treatment of OA through lowering oxidative stress and reducing inflammation [19, 20]. It has also been shown to have protective effects against rheumatoid arthritis in rat model [21]. Furthermore, ginger has no toxic effects when administered up to 5 g/kg in rats which provides wide safety margin of testing for its beneficial effects [22].
The aim of this study was to characterize the composition of the ginger and turmeric herbal formula hydro-ethanolic extract evaluate the possible chondro-protective effect of a mixed formula of both herbs in MIA induced OA rat model. In addition we investigated the possible mechanism of action of the herbal formula and examine its histopatholgical effect compared it to standard therapeutic agents.

Methods

Animals

Thirty-five male albino rats, weighing 200 ± 30 g at the start of the experiments were used. Prior to the initiation of the studies, the animals were randomized and assigned to treatment groups. Four rats were housed per cage (size 26 × 41 cm) and placed in the experimental room for acclimatization 24 h before any procedure was carried out. The animals were fed with standard laboratory diet and tap water ad libitum, and kept in an air-conditioned animal room at 23 ± 1 °C with a 12 h light/dark cycle.
Animal care and handling was performed in conformance with approved protocols of Cairo University and Egyptian Community guidelines for animal care. Research Ethics Committee at MSA School of Pharmacy approved the protocol of this study.

Monosodium iodoacetate injection

A single intra-articular (i.a.) injection of 2 mg of MIA (Sigma-Aldrich, Egypt) was performed through the infra-patellar ligament into the joint space of the right knee of lightly anesthetized rats (3% isoflurane in O2 at 1.5 l/min) in a total volume of 50 μl saline according to methodology of Nagase et al., 2012 [8].

Plant material

Fresh rhizomes of Z. officinale Rosc., and C. longa L., Zingiberaceae were purchased from the local market, Giza, Egypt during 2015/2016. The taxonomical identity was kindly verified by Dr. Mohamed El Gebaly, National Research Center, Giza, Egypt. The plant samples were air dried in the absence of direct sunlight and ground just before extraction.

Herbal formula preparation

The plant material was separately grinded then mixed together in a 1:1 ratio, macerated in 70% ethanol till exhaustion. The hydro-ethanolic extracts were concentrated under reduced pressure and kept in tightly closed amber glass containers for LC-ESI-MS and biological analysis. A voucher specimen (RS 0019) was deposited in the herbarium of faculty of Pharmacy, MSA University.

Experimental groups

Rats were randomly allocated into five groups of seven animals each.
Group 1: Normal control group injected (i.a) by 50 μl saline in the right knee.
Group 2 (OA): rats injected (i.a) of 2 mg of MIA in a total volume of 50 μl saline in the right knee.
Group 3 (F 200): Osteoarthritic rats treated with Herbal formula 200 mg/kg/day, orally for 1 month.
Group 4 (F 400): Osteoarthritic rats treated with Herbal formula 400 mg/kg/day, orally. For 1 month.
Group 5 (V): Osteoarthritic rats treated with Voltaren 30 mg/kg/day, orally for 1 month.
Doses were selected based on previous published results of toxicity studies in similar species [17, 22].

Drugs

Voltaren (Novartis, Egypt) were dissolved in distilled water and administered orally.

Chemicals

Ethanol was of HPLC grade and purchased from Sigma–Aldrich (Steinheim, Germany).

Blood samples and biochemical analysis

At the end of the study, rats were fasted overnight, anesthetized with thiopental sodium (50 mg/kg) [23] and blood samples were collected in the morning (5 ml per rat). Blood samples were centrifuged at 3000 rpm for 15 min after 30 min of collection and stored at −80 °C until analyzed. Serum cartilage oligomeric matrix protein (COMP), Hyaluronic acid (HA), Interleukin-1β (IL-1), Myeloperoxidase (MPO), Superoxide dismutase (SOD) were measured using the corresponding rat enzyme immunoassay kits (COMP: Cusabio Biotech Co, China), (HA: Uscn Life Science Inc. Wuhan, China), (IL-1: Koma Biotech Inc., Korea), (MPO: Hycult Biotech, Netherland), and (SOD: Cusabio Biotech Co, China) according to manufactured instructions. Serum Malondialdehyde (MDA): Cell Biolabs, Inc., USA was determined quantitatively using spectrophotometric method.

Histopathological examination

Anesthetized rats (thiopental sodium (50 mg/kg) [23])were rapidly decapitated; knee joints were dissected, rinsed in saline. Specimens were fixed in 10% formalin and then joints were decalcified in nitric oxide for 4 days, routinely processed and embedded in paraffin. Five microns sections were cut and stained with hematoxylin and Eosin (H&E). Histopathological evaluation were done according to Osteoarthritis Research Society International (OARSI) cartilage OA grading system [24].

UPLC-ESI–MS apparatus

The analysis was performed on a MS QQQ mass spectrometer coupled to Aglient 6420 series UPLC system (Agilent Technologies, Waldbronn, Germany), equipped with a 1290 ultra-performance auto sampler, 1290 infinity Quad pump, and 6420 triple quadrupole detector. Chromatographic separation was performed on a Acquity UPLC® HSS T3 (150 mm × 2.1 mm i.d.; 1.8 μm) column (Waters, USA).

Identification of the major compounds

Mobile phase consisted of two solvents, (A) 10% acetonitrile and (B) water. The separation was performed using gradient elution, from 15% to 90% A at 40 °C at a flow rate of 0.35 mL/min. Sample injection volume was 5 μL. The ionization technique was electrospray ionization. Spectra were recorded in negative ion mode between m/z 50 and 1000 with capillary voltage, 4500 V and heated dry nitrogen gas temperature, 325 °C and flow rate 9 l/min, the gas flow to the nebulizer was set at pressure 60 psi.

Statistical analyses

All data were expressed as mean ± SEM and analyzed using Prism program version 6. For all parameters, comparisons among groups (N = 7) were carried out using one-way analysis of variance (ANOVA) followed by Bonferroni’s multiple comparisons test. All P values reported are two-tailed and P ‹ 0.05 was set as the level of significance.

Results

Effect of herbal formula treatment on blood cartilage oligomeric matrix protein (COMP)

Mean serum level of COMP was significantly increased in OA group compared to the control group (P value <0.0001). On the other hand, the mean serum level of COMP was significantly decreased in F 200, F 400 and V groups when compared to OA group (P value <0.0001). The mean serum level of COMP was significantly decreased in F 400 group compared to F 200 group (P value <0.0001). Non-significant difference of COMP serum level was found between F 400 group and V group (Figure 1a).

Effect of herbal formula on blood hyaluronic acid (HA)

Mean serum level of HA was significantly increased in OA group compared to the control group (P value <0.0001). The mean serum level of HA was significantly decreased in F 200, F 400 and V groups when compared to OA group (P value <0.0001). The mean serum level of HA was significantly decreased in F 400 group compared to F 200 group (P value <0.0001). Also a significant difference was found between V and F 400 groups (P value <0.005) (Fig. 1b).

Effect of herbal formula on blood myeloperoxidase (MPO)

Mean serum level of MPO was significantly increased in OA group compared to the control group (P value <0.0001). The mean serum level of MPO was significantly decreased in F 200, F 400 and V groups when compared to OA group (P value <0.0001). The mean serum level of MPO was significantly decreased in F 400 group compared to F 200 group (P value <0.0001). A significant difference was found between V and F 400 groups (P value <0.01) (Fig. 2).

Effect of herbal formula on blood Interleukin-1 beta (IL-1β)

Mean serum level of IL-1β was significantly increased in OA group compared to the control group (P value <0.0001). The mean serum level of IL-1β was significantly decreased in F 200, F 400 and V groups when compared to OA group (P value <0.0001). The mean serum level of IL-1β was significantly decreased in F 400 group compared to F 200 group (P value <0.0001). A significant difference was found between V and F 400 groups (P value <0.002) (Fig. 3).

Effect of herbal formula on blood malondialdehyde (MDA) and superoxide dismutase (SOD)

Mean serum level of MDA was significantly increased in OA group compared to the control group (P value <0.0001). The mean serum level of MDA was significantly decreased in F 200, F 400 and V groups when compared to OA group (P value <0.0001). The mean serum level of MDA was significantly decreased in F 400 group compared to F 200 group (P value <0.0001). While non-significant difference was found between F 400 and V groups. (Fig. 4).
The mean serum level of SOD was significantly decreased in OA group compared to the control group, while it was increased in F 200, F 400 and V groups when compared to OA group (P value <0.0001). The mean serum level of SOD was significantly increased in F 400 group compared to F 200 group (P value <0.0001) and non-significant difference was found between F 400 and V groups. (Fig. 5).

UPLC-ESI-MS characterization of the herbal formula

UPLC-ESI-MS analysis of the hydro-ethanolic extract of the herbal formula demonstrated the presence of several gingerol derivatives in addition to curcumin and dihydro-curcumin as previously reported in different ginger and turmeric extracts [2529]. In total, 11 compounds belonging to different classes were tentatively identified by UPLC–MS. Data of peaks identification, retention times, and negative ion mode tentative identification of major compounds detected is presented in Table 1.
Table 1
Chromatographic, mass spectral characteristics and tentative identification of compounds in the herbal formula by UPLC-ESI(−ve)-MS
Compound
Tr (min)
Tentative Identification
Molecular Weight
(−)-ESI-MS
(m/z)
Reference(s)
1
2.07
10- gingerdiol
352
351 [M-H]-
Tanaka et al. 2015 [28]
2
7.76
Acetoxy-10-gingerol
392
391 [M-H]-
H. Jiang et al. 2005 [25]
3
9.43
13-Paradol
376
375 [M-H]-
Tanaka et al. 2015 [28]
4
14.91
1- Dehydro-12- gingerdione
374
373 [M-H]-
H. Jiang et al. 2005 [25]
5
16.09
Methyl-6-gingerol
308
307 [M-H]-
L. B., Ahui M. et al. 2013 [27]
6
19.24
7- Paradol
292
291 [M-H]-
Tanaka et al. 2015 [28]
7
19.86
Curcumin
368
367 [M-H]-
Herebian et al. 2009 [29]
8
22.17
Methyl diacetoxy-6-gingerdiol
394
393 [M-H]-
Tanaka et al. 2015 [28]
9
22.88
10-gingerol
350
349 [M-H]-
H. Jiang et al. 2005 [25]
TAO et al. 2009 [26]
L. B., Ahui M. et al. 2013 [27]
10
23.6
6-Paradol
278
277 [M-H]-
TAO et al. 2009 [26]
Tanaka et al. 2015 [25]
11
26.29
Dihydrocurumin
370
369 [M-H]-
Herebian et al. 2009 [29]

Histopathological changes associated with glucosamine treatment

Sections from the control group (C) showed normal histological appearance and proteoglycan content of the cartilage as judged by the preserved integrity of the articular surface, normal orientation and distribution of chondrocytes and the preservation of superficial layer. The OA group showed features of osteoarthritis grade 5 (ORASI grading system) recognized by denudation and erosion of the cartilage with reparative fibrotic changes (clefting, chondrocytes degeneration, calcification, fibrosis and matrix changes).
Other groups showed different grades of OA according to ORASI scale is illustrated in Fig. 6.

Discussion

Currently the only available pharmacological treatment for OA is non-steroid anti-inflammatory drugs (NSAIDs). Although the use of NSAIDs in osteoarthritis is highly controversial [30], many physicians and patients favor these agents for short and long-term use. However, the therapeutic utility of these agents is frequently limited by the development of side effects especially gastrointestinal ulceration and its complications [31]. Current treatment can control the pain of OA yet side effects could be serious, hence the need to find alternative therapeutic agents. Herbs and extracts present a potential candidate for treatment of OA [5].
In this study we report, for the first time, the beneficial effect of the use of a ginger/curcumin formula in the treatment of OA. Our results show that the ginger/curcumin herbal formula had chondroprotective effect in rat OA model possibly via a combined anti-inflammatory and antioxidant effect.
Our results come in agreement with many researchers who found beneficial effects of ginger and turmeric separately in OA and other related arthritic diseases. Ginger was effective as indomethacin in relieving symptoms of OA with negligible side effects [32]. To our knowledge this is the first report that combines ginger and turmeric extract in one formula as a treatment of OA.
Histopathological evaluation of the ginger/curcumin herbal formula treated groups showed different grades of healing according to Osteoarthritis Research Society International (OARSI) grading system (Fig. 6).
To explain the mechanism of action of ginger/curcumin formula, we measured serum level of COMP as well as other inflammatory and oxidative stress markers in our rat model of OA. Our results show that treatment with herbal formula at two different doses significantly reduced the serum level of COMP compared to OA group (P value <0.0001). The herbal formula at dose 400 mg/kg had similar effects on serum COMP levels as Voltaren® as there was a non-significant difference was found between F 400 and V groups (Fig. 1). These results on serum COMP could in part explain the effect of herbal formula observed in our study.
COMP is a large extracellular matrix protein, and a structural component of cartilage. Elevated levels of COMP are released during erosive joint disease such as rheumatoid arthritis and osteoarthritis [33]. The concentration of COMP in OA group was found to be increased over reference levels in the early stages of knee OA development. This is due to series of catabolic events undergoing in articular cartilage, which results in high turnover rate by the chondrocytes in order to repair the cartilage matrix. This process led first to dismantling of cartilage matrix and then a net loss of tissue [34]. There was a significant difference in the concentration of COMP biomarkers between patients with shorter and longer osteophytes on Kondyles of tibia and femur [14]. In another study patients with longer medial osteophytes and with great capsular distension had higher levels of HA and COMP in serum compared to patients with shorter osteophytes [35]. This suggests a role for COMP in the pathogenesis of OA [14].
HA is considered to be a good marker of synovitis in OA as it reflects local inflammation in synovial lining and, cartilage degradation [33]. Our results show that treatment with herbal formula at two-dose levels, significantly reduced serum level of HA compared to OA group (P value <0.0001); while its level was significantly increased in OA group compared to control group (Fig. 1).
MPO is released by activated neutrophils and used as a marker of leukocyte recruitment and function and subsequent inflammation. MPO concentration is related to inflammatory activity and could play an important role in the maintenance of oxidative stress in OA [36]. In this study MPO was significantly increased in OA group compared to control group (Fig. 2). On the other hand it was reduced after 1-month treatment with the herbal formula at two-dose levels (P value <0.0001).
Increased inflammation is the consequence of many factors, including mechanical overloading, joint injury, adipose tissue, and cartilage matrix fragments. IL-1β is considered one of the most prominent pro-inflammatory cytokines involved in OA. Elevated level of IL-1β is found in OA joint tissues, including the articular cartilage, subchondral bone, synovial fluid and synovium. IL-1β alters the homeostatic balance of chondrocytes by suppressing anabolic activity, stimulating catabolic breakdown of the articular cartilage, and increasing production of inflammatory mediators and reactive oxygen species (ROS) [37].
Ramadan et al., [21] found that the dried rhizomes of turmeric and ginger, separately, reduced inflammation by inhibiting the nuclear factor-κB activation. This lead to inhibition of gene expression of proinflammatory cytokines (TNF-α and IL-1β) in adjuvant-induced rat arthritis. This theory may explain why herbal formula was more effective in reducing inflammation and oxidative stress levels due to the synergistic effects of ginger and turmeric in herbal formula extract (Fig. 7).
Lipid peroxidation may lead to production of several toxic products as MDA. MDA showed a significant decrease in groups treated with herbal formula at two dose levels compared to OA group (P value <0.0001). This result comes in agreement with a study that found that there was significant decrease in lipid peroxidation groups treated with Zingiber officinale (ginger) when compared to animals treated with DMBA alone. The observed reduction in the level of lipid peroxidation in herbal formula treated animals was presumably due to the ability of ginger to scavenge the hydroxyl and peroxyl radicals [38, 39].
To evaluate the antioxidant effect of the formula we measured serum SOD activity. SOD showed significant elevation in herbal formula treated groups at two dose levels compared to OA group (P value <0.0001). This could indicate a direct or indirect (enhancing hepatic antioxidant activity) radical scavenging capability of the formula. Aqueous and ethanolic extract of Zingiber officinale was shown to have hepato-protective effect against acetaminophen-induced acute toxicity due to its antioxidant activity [39, 40].

Conclusion

The composition and characterization of the herbal formula hydro-ethanolic extract confirmed the identification of 11 curcumin and gingerol derivatives. The ginger/curcumin 1:1 herbal formula had chondroprotective effect in rat OA model in rats. Herbal formula treatment also reduced oxidative stress levels. Herbal formula at concentration of 400 mg/kg had more potent anti-inflammatory and anti oxidant effect compared with concentration of 200 mg/kg. Further pharmacological and clinical studies are still needed to evaluate this effect as well as its potential use in treatment of OA in human.

Acknowledgements

The authors would like to acknowledge Dr. Nashwa Waly, Professor of Small Animal Medicine Assiut University, Egypt, for her invaluable editorial assistance.

Funding

This research is not supported by any funding agency.

Availability of data and materials

The datasets supporting the conclusions of this article are included within the article and its additional files. Any further data will be available upon request.

Ethics approval

Animal care and handling was performed in conformance with approved protocols of Cairo University and Egyptian Community guidelines for animal care. Research Ethics Committee at MSA School of Pharmacy approved the protocol of this study.
Not applicable.

Competing interests

The authors declare that they have no competing interest.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
2.
Zurück zum Zitat Hootman JM, Helmick CG. Projections of US prevalence of arthritis and associated activity limitations. Arthritis Rheum. 2006;54(1):226–9.CrossRefPubMed Hootman JM, Helmick CG. Projections of US prevalence of arthritis and associated activity limitations. Arthritis Rheum. 2006;54(1):226–9.CrossRefPubMed
3.
Zurück zum Zitat Cialdai C, Giuliani S, Valenti C, Tramontana M, Maggi CA. Comparison between oral and intra-articular antinociceptive effect of dexketoprofen and tramadol combination in monosodium iodoacetate-induced osteoarthritis in rats. Eur J Pharmacol. 2013;714(1–3):346–51.CrossRefPubMed Cialdai C, Giuliani S, Valenti C, Tramontana M, Maggi CA. Comparison between oral and intra-articular antinociceptive effect of dexketoprofen and tramadol combination in monosodium iodoacetate-induced osteoarthritis in rats. Eur J Pharmacol. 2013;714(1–3):346–51.CrossRefPubMed
5.
Zurück zum Zitat Al-Saffar FJ, Ganabadi S, Fakurazi S, Yaakub H, Lip M. Chondroprotective effect of Zerumbone on monosodium Iodoacetate induced osteoarthritis in rats. J Appl Sci. 2010;10(4):248–60.CrossRef Al-Saffar FJ, Ganabadi S, Fakurazi S, Yaakub H, Lip M. Chondroprotective effect of Zerumbone on monosodium Iodoacetate induced osteoarthritis in rats. J Appl Sci. 2010;10(4):248–60.CrossRef
6.
Zurück zum Zitat Berenbaum F. Osteoarthritis year 2010 in review: pharmacological therapies. Osteoarthr Cartil. 2011;19(4):361–5.CrossRefPubMed Berenbaum F. Osteoarthritis year 2010 in review: pharmacological therapies. Osteoarthr Cartil. 2011;19(4):361–5.CrossRefPubMed
7.
Zurück zum Zitat Hawker GA, Mian S, Bednis K, Stanaitis I. Osteoarthritis year 2010 in review: non-pharmacologic therapy. Osteoarthr Cartil. 2011;19(4):366–74.CrossRefPubMed Hawker GA, Mian S, Bednis K, Stanaitis I. Osteoarthritis year 2010 in review: non-pharmacologic therapy. Osteoarthr Cartil. 2011;19(4):366–74.CrossRefPubMed
8.
Zurück zum Zitat Nagase H, Kumakura S, Shimada K. Establishment of a novel objective and quantitative method to assess pain-related behavior in monosodium iodoacetate-induced osteoarthritis in rat knee. J Pharmacol Toxicol Methods. 2012;65(1):29–36.CrossRefPubMed Nagase H, Kumakura S, Shimada K. Establishment of a novel objective and quantitative method to assess pain-related behavior in monosodium iodoacetate-induced osteoarthritis in rat knee. J Pharmacol Toxicol Methods. 2012;65(1):29–36.CrossRefPubMed
9.
Zurück zum Zitat Bianchi E, Di Cesare Mannelli L, Menicacci C, Lorenzoni P, Agliano M, Ghelardini C. Prophylactic role of acetyl-l-carnitine on knee lesions and associated pain in a rat model of osteoarthritis. Life Sci. 2014;106(1–2):32–9.CrossRefPubMed Bianchi E, Di Cesare Mannelli L, Menicacci C, Lorenzoni P, Agliano M, Ghelardini C. Prophylactic role of acetyl-l-carnitine on knee lesions and associated pain in a rat model of osteoarthritis. Life Sci. 2014;106(1–2):32–9.CrossRefPubMed
10.
Zurück zum Zitat Moilanen LJ, Hamalainen M, Nummenmaa E, Ilmarinen P, Vuolteenaho K, Nieminen RM, Lehtimaki L, Moilanen E. Monosodium iodoacetate-induced inflammation and joint pain are reduced in TRPA1 deficient mice--potential role of TRPA1 in osteoarthritis. Osteoarthr Cartil. 2015;23(11):2017–26.CrossRefPubMed Moilanen LJ, Hamalainen M, Nummenmaa E, Ilmarinen P, Vuolteenaho K, Nieminen RM, Lehtimaki L, Moilanen E. Monosodium iodoacetate-induced inflammation and joint pain are reduced in TRPA1 deficient mice--potential role of TRPA1 in osteoarthritis. Osteoarthr Cartil. 2015;23(11):2017–26.CrossRefPubMed
11.
Zurück zum Zitat Aborehab NM, El Bishbishy MH, Waly NE. Resistin mediates tomato and broccoli extract effects on glucose homeostasis in high fat diet-induced obesity in rats. BMC Complement Altern Med. 2016;16:225.CrossRefPubMedPubMedCentral Aborehab NM, El Bishbishy MH, Waly NE. Resistin mediates tomato and broccoli extract effects on glucose homeostasis in high fat diet-induced obesity in rats. BMC Complement Altern Med. 2016;16:225.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Ghavipour M, Sotoudeh G, Ghorbani M. Tomato juice consumption improves blood antioxidative biomarkers in overweight and obese females. Clin Nutr. 2015;34(5):805–9.CrossRefPubMed Ghavipour M, Sotoudeh G, Ghorbani M. Tomato juice consumption improves blood antioxidative biomarkers in overweight and obese females. Clin Nutr. 2015;34(5):805–9.CrossRefPubMed
13.
Zurück zum Zitat Ghavipour M, Saedisomeolia A, Djalali M, Sotoudeh G, Eshraghyan MR, Moghadam AM, Wood LG. Tomato juice consumption reduces systemic inflammation in overweight and obese females. Br J Nutr. 2013;109(11):2031–5.CrossRefPubMed Ghavipour M, Saedisomeolia A, Djalali M, Sotoudeh G, Eshraghyan MR, Moghadam AM, Wood LG. Tomato juice consumption reduces systemic inflammation in overweight and obese females. Br J Nutr. 2013;109(11):2031–5.CrossRefPubMed
14.
Zurück zum Zitat Zivanovic S, Rackov LP, Zivanovic A, Jevtic M, Nikolic S, Kocic S. Cartilage oligomeric matrix protein - inflammation biomarker in knee osteoarthritis. Bosn J Basic Med Sci. 2011;11(1):27–32.CrossRefPubMedPubMedCentral Zivanovic S, Rackov LP, Zivanovic A, Jevtic M, Nikolic S, Kocic S. Cartilage oligomeric matrix protein - inflammation biomarker in knee osteoarthritis. Bosn J Basic Med Sci. 2011;11(1):27–32.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Ojewole JA. Analgesic, antiinflammatory and hypoglycaemic effects of ethanol extract of Zingiber Officinale (roscoe) rhizomes (Zingiberaceae) in mice and rats. Phytother Res. 2006;20(9):764–72.CrossRefPubMed Ojewole JA. Analgesic, antiinflammatory and hypoglycaemic effects of ethanol extract of Zingiber Officinale (roscoe) rhizomes (Zingiberaceae) in mice and rats. Phytother Res. 2006;20(9):764–72.CrossRefPubMed
16.
Zurück zum Zitat al-Yahya MA, Rafatullah S, Mossa JS, Ageel AM, Parmar NS, Tariq M. Gastroprotective activity of ginger zingiber officinale rosc., in albino rats. Am J Chin Med. 1989;17(1–2):51–6.CrossRefPubMed al-Yahya MA, Rafatullah S, Mossa JS, Ageel AM, Parmar NS, Tariq M. Gastroprotective activity of ginger zingiber officinale rosc., in albino rats. Am J Chin Med. 1989;17(1–2):51–6.CrossRefPubMed
17.
Zurück zum Zitat Anosike CA, Obidoa O, Ezeanyika LUS, Nwuba MM. Anti-inflammatory and anti-ulcerogenic activity of the ethanol extract of ginger (Zingiber Officinale). Afr J Biochem Res. 2009;3(12):379–84. Anosike CA, Obidoa O, Ezeanyika LUS, Nwuba MM. Anti-inflammatory and anti-ulcerogenic activity of the ethanol extract of ginger (Zingiber Officinale). Afr J Biochem Res. 2009;3(12):379–84.
18.
Zurück zum Zitat Chin KY. The spice for joint inflammation: anti-inflammatory role of curcumin in treating osteoarthritis. Drug Des Devel Ther. 2016;10:3029–42.CrossRefPubMedPubMedCentral Chin KY. The spice for joint inflammation: anti-inflammatory role of curcumin in treating osteoarthritis. Drug Des Devel Ther. 2016;10:3029–42.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Rahimnia AR, Panahi Y, Alishiri G, Sharafi M, Sahebkar A. Impact of supplementation with Curcuminoids on systemic inflammation in patients with knee osteoarthritis: findings from a randomized double-blind placebo-controlled trial. Drug Res (Stuttg). 2015;65(10):521–5. Rahimnia AR, Panahi Y, Alishiri G, Sharafi M, Sahebkar A. Impact of supplementation with Curcuminoids on systemic inflammation in patients with knee osteoarthritis: findings from a randomized double-blind placebo-controlled trial. Drug Res (Stuttg). 2015;65(10):521–5.
20.
Zurück zum Zitat Panahi Y, Alishiri GH, Parvin S, Sahebkar A. Mitigation of systemic oxidative stress by Curcuminoids in osteoarthritis: results of a randomized controlled trial. J Diet Suppl. 2016;13(2):209–20.CrossRefPubMed Panahi Y, Alishiri GH, Parvin S, Sahebkar A. Mitigation of systemic oxidative stress by Curcuminoids in osteoarthritis: results of a randomized controlled trial. J Diet Suppl. 2016;13(2):209–20.CrossRefPubMed
21.
Zurück zum Zitat Ramadan G, Al-Kahtani MA, El-Sayed WM. Anti-inflammatory and anti-oxidant properties of Curcuma Longa (turmeric) versus Zingiber Officinale (ginger) rhizomes in rat adjuvant-induced arthritis. Inflammation. 2011;34(4):291–301.CrossRefPubMed Ramadan G, Al-Kahtani MA, El-Sayed WM. Anti-inflammatory and anti-oxidant properties of Curcuma Longa (turmeric) versus Zingiber Officinale (ginger) rhizomes in rat adjuvant-induced arthritis. Inflammation. 2011;34(4):291–301.CrossRefPubMed
22.
Zurück zum Zitat Shankar TN, Shantha NV, Ramesh HP, Murthy IA, Murthy VS. Toxicity studies on turmeric (Curcuma Longa): acute toxicity studies in rats, guineapigs & monkeys. Indian J Exp Biol. 1980;18(1):73–5.PubMed Shankar TN, Shantha NV, Ramesh HP, Murthy IA, Murthy VS. Toxicity studies on turmeric (Curcuma Longa): acute toxicity studies in rats, guineapigs & monkeys. Indian J Exp Biol. 1980;18(1):73–5.PubMed
23.
Zurück zum Zitat Vogler GA, Suckow MA, Weisbroth S, Franklin CL. Anesthesia and analgesia in the laboratory rat. New York: Elsevier Academic Press; 2006. p. 627–95. Vogler GA, Suckow MA, Weisbroth S, Franklin CL. Anesthesia and analgesia in the laboratory rat. New York: Elsevier Academic Press; 2006. p. 627–95.
24.
Zurück zum Zitat Pritzker KP, Gay S, Jimenez SA, Ostergaard K, Pelletier JP, Revell PA, Salter D, van den Berg WB. Osteoarthritis cartilage histopathology: grading and staging. Osteoarthr Cartil. 2006;14(1):13–29.CrossRefPubMed Pritzker KP, Gay S, Jimenez SA, Ostergaard K, Pelletier JP, Revell PA, Salter D, van den Berg WB. Osteoarthritis cartilage histopathology: grading and staging. Osteoarthr Cartil. 2006;14(1):13–29.CrossRefPubMed
25.
Zurück zum Zitat Jiang H, Solyom AM, Timmermann BN, Gang DR. Characterization of gingerol-related compounds in ginger rhizome (Zingiber Officinale Rosc.) by high-performance liquid chromatography/electrospray ionization mass spectrometry. Rapid Commun Mass Spectrom. 2005;19(20):2957–64.CrossRefPubMed Jiang H, Solyom AM, Timmermann BN, Gang DR. Characterization of gingerol-related compounds in ginger rhizome (Zingiber Officinale Rosc.) by high-performance liquid chromatography/electrospray ionization mass spectrometry. Rapid Commun Mass Spectrom. 2005;19(20):2957–64.CrossRefPubMed
26.
Zurück zum Zitat Tao Y, Li W, Liang W, Van Breemen RB. Identification and quantification of gingerols and related compounds in ginger dietary supplements using high-performance liquid chromatography-tandem mass spectrometry. J Agric Food Chem. 2009;57(21):10014–21.CrossRefPubMedPubMedCentral Tao Y, Li W, Liang W, Van Breemen RB. Identification and quantification of gingerols and related compounds in ginger dietary supplements using high-performance liquid chromatography-tandem mass spectrometry. J Agric Food Chem. 2009;57(21):10014–21.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Ahui MLB, Konan AB, Zannou-Tchoko VJ, Amonkan AK, Kati-Coulibaly S, Offoumou MA. Identification of Gingerols in ginger (Zingiber Officinale roscoe) by high performance liquid chromatography-tandem mass spectrometry and pharmacologic studies of its aqueous extract on the rabbit isolated duodenum contractility. J Phys Pharm Adv. 2013;3(2):16–26.CrossRef Ahui MLB, Konan AB, Zannou-Tchoko VJ, Amonkan AK, Kati-Coulibaly S, Offoumou MA. Identification of Gingerols in ginger (Zingiber Officinale roscoe) by high performance liquid chromatography-tandem mass spectrometry and pharmacologic studies of its aqueous extract on the rabbit isolated duodenum contractility. J Phys Pharm Adv. 2013;3(2):16–26.CrossRef
28.
Zurück zum Zitat Tanaka K, Arita M, Sakurai H, Ono N, Tezuka Y. Analysis of chemical properties of edible and medicinal ginger by metabolomics approach. Biomed Res Int. 2015;2015:671058.CrossRefPubMedPubMedCentral Tanaka K, Arita M, Sakurai H, Ono N, Tezuka Y. Analysis of chemical properties of edible and medicinal ginger by metabolomics approach. Biomed Res Int. 2015;2015:671058.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Herebian D, Choi JH, Abd El-Aty AM, Shim JH, Spiteller M. Metabolite analysis in Curcuma Domestica using various GC-MS and LC-MS separation and detection techniques. Biomed Chromatogr. 2009;23(9):951–65.CrossRefPubMed Herebian D, Choi JH, Abd El-Aty AM, Shim JH, Spiteller M. Metabolite analysis in Curcuma Domestica using various GC-MS and LC-MS separation and detection techniques. Biomed Chromatogr. 2009;23(9):951–65.CrossRefPubMed
30.
Zurück zum Zitat Nelson AE, Allen KD, Golightly YM, Goode AP, Jordan JM. A systematic review of recommendations and guidelines for the management of osteoarthritis: the chronic osteoarthritis management initiative of the U.S. bone and joint initiative. Semin Arthritis Rheum. 2014;43(6):701–12.CrossRefPubMed Nelson AE, Allen KD, Golightly YM, Goode AP, Jordan JM. A systematic review of recommendations and guidelines for the management of osteoarthritis: the chronic osteoarthritis management initiative of the U.S. bone and joint initiative. Semin Arthritis Rheum. 2014;43(6):701–12.CrossRefPubMed
31.
Zurück zum Zitat Bove SE, Calcaterra SL, Brooker RM, Huber CM, Guzman RE, Juneau PL, Schrier DJ, Kilgore KS. Weight bearing as a measure of disease progression and efficacy of anti-inflammatory compounds in a model of monosodium iodoacetate-induced osteoarthritis. Osteoarthr Cartil. 2003;11(11):821–30.CrossRefPubMed Bove SE, Calcaterra SL, Brooker RM, Huber CM, Guzman RE, Juneau PL, Schrier DJ, Kilgore KS. Weight bearing as a measure of disease progression and efficacy of anti-inflammatory compounds in a model of monosodium iodoacetate-induced osteoarthritis. Osteoarthr Cartil. 2003;11(11):821–30.CrossRefPubMed
32.
Zurück zum Zitat Haghighi ATN, Owlia MB. Effects of ginger on primary knee osteoarthritis. Indian J Rheumatol. 2006;1:3–7.CrossRef Haghighi ATN, Owlia MB. Effects of ginger on primary knee osteoarthritis. Indian J Rheumatol. 2006;1:3–7.CrossRef
33.
Zurück zum Zitat Ramonda R, Lorenzin M, Modesti V, Campana C, Ortolan A, Frallonardo P, Punzi L. Serological markers of erosive hand osteoarthritis. Eur J Intern Med. 2013;24(1):11–5.CrossRefPubMed Ramonda R, Lorenzin M, Modesti V, Campana C, Ortolan A, Frallonardo P, Punzi L. Serological markers of erosive hand osteoarthritis. Eur J Intern Med. 2013;24(1):11–5.CrossRefPubMed
34.
Zurück zum Zitat Verma P, Dalal K. Serum cartilage oligomeric matrix protein (COMP) in knee osteoarthritis: a novel diagnostic and prognostic biomarker. J Orthop Res. 2013;31(7):999–1006.CrossRefPubMed Verma P, Dalal K. Serum cartilage oligomeric matrix protein (COMP) in knee osteoarthritis: a novel diagnostic and prognostic biomarker. J Orthop Res. 2013;31(7):999–1006.CrossRefPubMed
35.
Zurück zum Zitat Jung YO, Do JH, Kang HJ, Yoo SA, Yoon CH, Kim HA, Cho CS, Kim WU. Correlation of sonographic severity with biochemical markers of synovium and cartilage in knee osteoarthritis patients. Clin Exp Rheumatol. 2006;24(3):253–9.PubMed Jung YO, Do JH, Kang HJ, Yoo SA, Yoon CH, Kim HA, Cho CS, Kim WU. Correlation of sonographic severity with biochemical markers of synovium and cartilage in knee osteoarthritis patients. Clin Exp Rheumatol. 2006;24(3):253–9.PubMed
36.
Zurück zum Zitat Punzi L, Ramonda R, Deberg M, Frallonardo P, Campana C, Musacchio E, Henrotin Y. Coll2-1, Coll2-1NO2 and myeloperoxidase serum levels in erosive and non-erosive osteoarthritis of the hands. Osteoarthr Cartil. 2012;20(6):557–61.CrossRefPubMed Punzi L, Ramonda R, Deberg M, Frallonardo P, Campana C, Musacchio E, Henrotin Y. Coll2-1, Coll2-1NO2 and myeloperoxidase serum levels in erosive and non-erosive osteoarthritis of the hands. Osteoarthr Cartil. 2012;20(6):557–61.CrossRefPubMed
37.
Zurück zum Zitat Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier JP, Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011;7(1):33–42.CrossRefPubMed Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier JP, Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011;7(1):33–42.CrossRefPubMed
38.
Zurück zum Zitat Blessy D, Suresh K, Manoharan S, Arokia A, Vijayaan MA and, Sugunadevi G. Evaluation of chemo preventive potential of Zingiber Officinale roscoe ethanolic root extract on 7, 12-dimethyl Benz[a] anthracene induced oral carcinogenesis. Res J Agric Biol Sci. 2009;5(5): 775 - 81. Blessy D, Suresh K, Manoharan S, Arokia A, Vijayaan MA and, Sugunadevi G. Evaluation of chemo preventive potential of Zingiber Officinale roscoe ethanolic root extract on 7, 12-dimethyl Benz[a] anthracene induced oral carcinogenesis. Res J Agric Biol Sci. 2009;5(5): 775 - 81.
39.
Zurück zum Zitat Al-Kushi AG, El-Boshy ME, ElSawy NA, Shaikh Omar OA, Header EA. Pathological comparative studies on aqueous and Ethanolic extracts of Zingiber Officinale on antioxidants and Hypolipidemic effects in rats. Life Science Journal. 2013;10(2):2393–403. Al-Kushi AG, El-Boshy ME, ElSawy NA, Shaikh Omar OA, Header EA. Pathological comparative studies on aqueous and Ethanolic extracts of Zingiber Officinale on antioxidants and Hypolipidemic effects in rats. Life Science Journal. 2013;10(2):2393–403.
40.
Zurück zum Zitat Ajith TA, Hema U, Aswathy MS. Zingiber Officinale roscoe prevents acetaminophen-induced acute hepatotoxicity by enhancing hepatic antioxidant status. Food Chem Toxicol. 2007;45(11):2267–72.CrossRefPubMed Ajith TA, Hema U, Aswathy MS. Zingiber Officinale roscoe prevents acetaminophen-induced acute hepatotoxicity by enhancing hepatic antioxidant status. Food Chem Toxicol. 2007;45(11):2267–72.CrossRefPubMed
41.
Zurück zum Zitat Leong DJ, Choudhury M, Hirsh DM, Hardin JA, Cobelli NJ, Sun HB. Nutraceuticals: potential for chondroprotection and molecular targeting of osteoarthritis. Int J Mol Sci. 2013;14(11):23063–85.CrossRefPubMedPubMedCentral Leong DJ, Choudhury M, Hirsh DM, Hardin JA, Cobelli NJ, Sun HB. Nutraceuticals: potential for chondroprotection and molecular targeting of osteoarthritis. Int J Mol Sci. 2013;14(11):23063–85.CrossRefPubMedPubMedCentral
Metadaten
Titel
A putative Chondroprotective role for IL-1β and MPO in herbal treatment of experimental osteoarthritis
verfasst von
Nora M. Aborehab
Mahitab H. El Bishbishy
Abeer Refaiy
Nermien E. Waly
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Complementary Medicine and Therapies / Ausgabe 1/2017
Elektronische ISSN: 2662-7671
DOI
https://doi.org/10.1186/s12906-017-2002-y

Weitere Artikel der Ausgabe 1/2017

BMC Complementary Medicine and Therapies 1/2017 Zur Ausgabe