Skip to main content
Erschienen in: BMC Complementary Medicine and Therapies 1/2022

Open Access 01.12.2022 | Research

RETRACTED ARTICLE: Suppression of NF-κB signaling by ECN in an arthritic model of inflammation

verfasst von: Amna Khan, Li Zhang, Chang Hu Li, Ashraf Ullah Khan, Bushra Shal, Adnan Khan, Sajjad Ahmad, Fakhar ud Din, Zia ur rehman, Feng Wang, Salman Khan

Erschienen in: BMC Complementary Medicine and Therapies | Ausgabe 1/2022

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

The 7β-(3-ethyl-cis-crotonoyloxy)-1α-(2-methylbutyryloxy)-3,14-dehydro-Z-notonipetranone (ECN), a sesquiterpenoid isolated from the Tussilago farfara Linneaus (Asteraceae), was evaluated against acute Carrageenan and chronic complete Freund’s adjuvant (CFA)-induced arthritis in mice.

Methods

Acute and chronic arthritis were induced by administering Carrageenan and CFA to the intraplantar surface of the mouse paw. Edema, mechanical allodynia, mechanical hyperalgesia, and thermal hyperalgesia were assessed in the paw. Similarly, histological and immunohistological parameters were assessed following arthritis induced by CFA. Antioxidants, inflammatory cytokines, and oxidative stress markers were also studied in all the treated groups.

Results

The ECN treatment significantly attenuated edema in the paw and elevated the nocifensive threshold following induction of this inflammatory model. Furthermore, ECN treatment markedly improved the arthritis index and distress symptoms, while attenuating the CFA-induced edema in the paw. ECN treatment also improved the histological parameters in the paw tissue compared to the control. At the same time, there was a significant reduction in edema and erosion in the ECN-treated group, as measured by radiographic analysis. Using the Comet’s assay, we showed that ECN treatment protected the DNA from chronic CFA-induced arthritis. Immunohistochemistry analysis showed a marked decrease in the expression level of p-JNK (phosphorylated C-Jun N-terminal kinase), NF-κB (Nuclear factor-kappa B), COX-2 (Cyclooxygenase-2), and TNF-α (Tumour necrosis factor-alpha) compared to the CFA-treated group. Biophysical analysis involving molecular docking, molecular dynamics simulations, and binding free energies of ECN were performed to explore the underlying mechanism.

Conclusion

ECN exhibited significant anti-inflammatory and anti-arthritic activity against Carrageenan and CFA-induced models.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12906-022-03629-7.
Amna Khan, Li Zhang and Chang Hu Li contributed equally to this work.
This article has been retracted. Please see the retraction notice for more detail: https://doi.org/10.1186/s12906-023-03954-5"
A retraction note to this article is available online at https://​doi.​org/​10.​1186/​s12906-023-03954-5.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CAT
Catalase
p-JNK
Phosphorylated C-Jun N-terminal kinase
CFA
Complete Freund’s adjuvant
COX-2
Cyclooxygenase-2
DMSO
Dimethyl sulfoxide
ECN
7β-(3-ethyl-cis-crotonoyloxy)-1α-(2-methylbutyryloxy)-3, 14-dehydro-Z-notonipetranone
GSH
Glutathione
GST
Glutathione sulfotransferase
LPS
Lipopolysaccharide
MDA
Malonaldehyde
NO
Nitric oxide
NSAID’s
Non-steroidal anti-inflammatory drugs
NF-κB
Nuclear factor-kappa B
ROS
Reactive oxygen species
RA
Rheumatoid arthritis
SOD
Superoxide dismutase
TLR-4
Toll-like receptor 4

Background

Rheumatoid arthritis (RA) is an autoimmune disorder characterized by synovial inflammation leading to articular destruction, joint stiffness, and pain [1]. It is one of the most prevalent disorders worldwide, affecting nearly 1–2% of the population [1]. The incidence of RA appears to be higher in people over 40 years of age and in women due to the loss of bone integrity after menopause [2]. The ultimate target of RA treatment includes alleviating inflammation, protecting articular morphology, maintaining physiology, and systemic control of mediators [3]. Treatment strategies for RA include non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids, and immunomodulators [4]. These agents help to lower arthritis-associated inflammation, but 30% of the individuals with RA fail to respond to the treatment [5]. Cost-effectiveness and reduced side effects are the primary parameters considered in the treatment of RA [2].
Complete Freund’s adjuvant (CFA) has been widely used to model RA in animals. It establishes autoimmune arthritis by increasing the inflammatory cytokines such as TNF-α, COX-2, and interleukin-1β (IL-1β) via NF-κB signaling [68]. CFA administration also induces activation of NF-κB by facilitating the degradation of IκB (inhibitory kappa B) [1, 9]. Following degradation of IκB, the NF-κB translocates to the nucleus and induces the expression of pro-inflammatory cytokines and oxidative stress markers [68]. Numerous experimental findings associated with the pathogenesis of arthritis reported that activation of NF-κB, along with conversion of superoxide anions to nitric oxide (NO) and reactive oxygen species (ROS) productionplay a major role in exaggerating arthritis [68]. These reactive oxygen species may also be involved in the paw tissue’s oxidative DNA damage, resulting in apoptosis [10]. In addition, some of the intracellular enzymes such as catalase (CAT), superoxide dismutase (SOD), glutathione (GSH), and glutathione sulfotransferase (GST) are detoxifying enzymes that may attenuate the pathological symptoms [1113].
Phytochemicals have effectively alleviated or ameliorated numerous inflammatory disorders [14]. Tussilago farfara L. (Asteraceae), commonly called coltsfoot, is a perennial herb widely spread in Korea, China, North Africa, Siberia, and Europe [15]. The flowering buds of T. farfara have been used as an herbal medicine to treat asthma, bronchitis, and cough [15]. Several active sesquiterpenoids, such as faradiol, phytosterol, rutin, and tussilagone isolated from T. farfara possess anti-microbial and stimulate the cardiovascular system [16, 17].
One of these is 7β-(3-ethylcis-crotonoyloxy)-1α-(2-methylbutyryloxy)-3,14-dehydro-Z-notonipetranone (ECN) known to have antioxidant and neuroprotective effects [15]. This study assessed the anti-arthritic activity of ECNagainst Carrageenan and CFA (augmented with LPS)-induced arthritis in mice to understand its possible molecular mechanism of anti-rheumatism.

Methods

Chemicals

ECN (the dried flower buds of Tussilago farfara) were purchased from an Oriental Market in Seoul, Korea, and identified by Prof. Je-Hyun Lee. Carrageenan, Complete Freund’s adjuvant (CFA), and Griess reagent were purchased from Sigma Aldrich (USA). ELISA kits for assessing the inflammatory mediators TNF-α and IL-1β were purchased from Thermo Fisher Scientific (USA). Primary and secondary antibodies were purchased from Santa Cruz. A Neubauer hemocytometer (Feinoptik, Germany) was used for blood analysis, while Sahli’s hemoglobin meter was used to measure the hemoglobin concentration (Hb). The AMP diagnostic kits (Graz, Austria) were utilized for the liver function tests. All the compounds were dissolved in 2% DMSO and then diluted with normal saline. All experiments were performed in triplicate to assess reproducibility.

Plant material

The dried flower buds of T. farfara were purchased from Oriental Market in Seoul, Korea, and identified by Prof. Je-Hyun Lee (Dongkkuk University, Kyungju, South Korea). A voucher specimen (no. EA333) was deposited at Natural Product Chemistry Lab, College of Pharmacy, Ewha Womans University, Seoul, Korea) as reported [18].

Animals

Male albino BALB/c mice (4–5 weeks of age; 25–30 g) were procured from the National Institute of Health (NIH), Islamabad, Pakistan. Animals were caged in a controlled laboratory environment with a room temperature of 23 ± 1 °C, 50 ± 10% humidity, and a 12 h light-dark cycle. The animals were provided unrestricted access to water and food. Procedures were carried out between 8 a.m. and 6 p.m. in the pathogens-free zone of the Pharmacology Laboratory, Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan. All experiments were performed following the guidelines of the ethical committee of Quaid-i-Azam University of care and experimental animals (Approval No: BEC-FBS-QAU2018–125) and conformed to the ethical instructions for the analysis of investigational pain in live animals developed by the International Association of Pain [19]. Great care was taken to avoid any unnecessary harm to the animals. New animals were used for each experiment and used once.

Randomization and sample size selection

Animals were randomly divided into various groups [20]. Double blindness was maintained during the whole experiment to avoid experimental biases. The sample size (n = 7) selection was based on previous reports [20].

Animal models

Carrageenan-induced model: Mice were divided into six groups (n = 7 per group). The animals were divided randomly and equally into various groups to avoid scientific bias.
Group I: Normal control.
Group II: Carrageenan treated group (1% solution of Carrageenan, intraplantar).
Group III: Dexa 5 mg/kg, i.p. (60 min before Carrageenan administration).
Group IV: ECN 1 mg/kg, i.p. (60 min before Carrageenan administration).
Group V: ECN 5 mg/kg, i.p. (60 min before Carrageenan administration).
Group VI: ECN 10 mg/kg, i.p. (60 min before Carrageenan administration).
CFA-induced arthritis model: All the mice were allocated into four groups (n = 7 per group).
Group I: Normal control.
Group II: CFA treated group (20 μl, intraplantar).
Group III: Dexa 5 mg/kg, i.p. (forty min before CFA administration).
Group IV: ECN 10 mg/kg, i.p. (forty min before CFA injection).
Control animals received no treatment, the positive control received dexamethasone, and the treatment control regularly received ECN for 21 days. LPS (25 μg/kg, intraplantar) was also administered to all groups (except the control) on day 14 to augment the inflammatory response. The study design is depicted in Fig. 1.

Animal sampling

Blood was obtained from the heart directly following anesthesia using xylazine and ketamine injection (16 mg + 60 mg, i.p). The animals were sacrificed by cervical dislocation [21].

Assessment of distress symptoms and survival rate in CFA -induced mice

Animals were observed throughout the experiment to assess the distress symptoms and survival rate in CFA- induced mice [22]. As reported previously, the assessed distress symptoms included general health, gait weakness, and unwillingness to move [22]. The distress score was calculated at different time intervals, i.e., 0, 3, 6, 9, 12, 15, 18, and 21st day [22]. Furthermore, the mortality rate was assessed in each group from day 0 to the end of the experiment.

Assessment of arthritic score in CFA-induced mice

The structural characteristics of arthritis, such as redness, edema, and erythema, were assessed by the previously mentioned visual criteria [23]. A score of 0 indicatesno signs of redness and edema in the paw, 0.5 represents redness and swelling in just one finger, 1 indicates redness and light swelling of the ankle or 2–5 fingers, 2 indicates redness and medium swelling of the ankle and 2–5 fingers, 3 indicates redness and extreme swelling all fingers and 4 indicates decreased swelling and deformation resulting in an incapacitated limb.

Assessment of paw edema in Carrageenan and CFA-induced mice

The effect of ECN was assessed on Carrageenan and CFA-induced changes in paw thickness; the paw thickness was determined via dial thickness gauge [24, 25]. Paw edema was determined at four different time intervals in the case of Carrageenan and acute CFA-induced arthritis, i.e., 0, 2, 4, and 6 h. In the case of chronic CFA-induced arthritis, paw edema was determined on a daily basis for 21 days post-CFA administration .

Assessment of mechanical hyperalgesia in Carrageenan and CFA-induced mice

Paw withdrawal threshold in Carrageenan and CFA -induced mice were determined using the Randall Selitto apparatus (Digital Randall Selitto with Pressure Applicator) following published methods [5, 24]. The paw withdrawal threshold was determined on a daily basis for 21 days post-CFA.

Assessment of thermal hyperalgesia in Carrageenan and CFA-induced mice

Thermal pain threshold in Carrageenan and CFA-induced mice were determined using a unilateral hot plate apparatus [26]. The plate’s temperature was set at 50 ± 0.5 °C, and 35 seconds was set as a cut-off value to avoid the animal’s harm. The paw withdrawal latency was determined on a daily basis for 21 days post-CFA.

Assessment of mechanical allodynia in Carrageenan and CFA -induced mice

Mechanical allodynia was assessed using Von Frey filaments [11, 27, 28]. The paw withdrawal threshold was determined on a daily basis for 21 days post-CFA.

Estimation of GSH, GST, CAT, and SOD in CFA-induced mice paw tissue

The effect of ECN (10 mg/kg, i.p) on the antioxidants GSH, GST, CAT, and SOD was determined as reported previously [2931]. Briefly, the reduced glutathione was assessed by mixing tissue homogenate (0.1 ml), PBS solution (2.4 ml), and DTNB (0.5 ml) to make the final volume up to 3 ml. The absorbance was read by a microplate reader at 412 nm wavelength. The GST level was quantified following the previously mentioned method [2830]. The GST level in paw tissue was assessed by mixing tissue homogenate (0.1 ml), CDNB (0.1 ml), and by adding 0.1 M PBS (pH 6.5) to make the final volume of 3 ml. The absorbance was read by a microplate reader at a wavelength of 314 nm. Catalase level was analyzed following the already mentioned protocol with minor adjustments [22]. The catalase level was assessed by taking H2O2-phosphate buffer (3 ml) in a cuvette and by rapid addition of enzyme extract (40 μl) [31]. The absorbance was read by a microplate reader at 240 nm wavelength. The SOD activity was measured by mixing 50 mM (pH 8.5) Tris-EDTA buffer, 24 mM pyrogallol, and 10 μl of the sample [31]. The absorbance was read at 420 nm wavelength.

Estimation of NO and MDA in CFA-induced plasma

Nitric oxide (NO) levels in CFA -induced mice paw tissue were assessed via the Griess reagent method, as reported [24, 32]. Similarly, the rate of lipid peroxidation was analyzed by assessing the concentration of malonaldehyde (MDA) [29, 30]. Briefly, tissue homogenate (0.25 ml) in 10% PBS (pH 7.4) was incubated at 37 °C in a water bath. After 1 h incubation, 5% TCA (0.25 ml) and 0.67% TBA (0.5 ml) were mixed with the tissue homogenate (0.25 ml) and centrifuged at 1300 rpm for 10 min. The clear supernatant was transferred from the above mixture to a different tube, placed for 10 min in a water bath, cooled, and the absorbance read at 535 nm wavelength.

Detection of inflammatory cytokines

Production of TNF-α and IL-1β following CFA-induced arthritis was measured using ELISA kits. Briefly, the paw tissue samples from all the animal groups were prepared according to method described above. Protein was extracted from the tissue using PBS \ with 0.4 M NaCl, 0.05% Tween 20, and protein inhibitors. The samples were homogenized and centrifuged (at 3000 g for 10 min) to obtain the supernatant for the assessment of the TNF-α and IL-1β using commercially available kits [33].

Estimation of DNA damage in CFA-induced mice paw tissue

DNA damage was assessed by comet assay [34, 35]. A small part of the paw tissue was suspended in cold lysing buffer (1 ml) in an ultracentrifuge tube, and then the samples were homogenized. The cell suspension (5–10 μl) was mixed in 0.5% low melting point agarose, layered on the pre-coated slides with normal agarose solution (1%), and incubated for 10 min on an ice pack. After performing this step twice, the slides were placed in lysing mixture for 2 h at 4 °C. Post-electrophoresis, slides were marked with ethidium bromide (1%) and were analyzed under a fluorescent microscope. The content of DNA disruption was quantified by CASP software. Length of tail and % DNA in the tail was used to check DNA disruption’s content [34, 35].

Renal and hepatotoxicity testing

The effect of ECN (10 mg/kg, i.p) treatment on the vital organs such as the liver and kidneys was assessed by measuring the aspartate aminotransferase (ALT), alanine aminotransferase (AST), and creatinine concentration [21]. The collected blood was centrifuged at 5000 rpm for 5 min to separate the plasma from the blood. The plasma was used to estimate the concentration of AST, ALT, and creatinine [21].

Radiological and histological analysis

Radiological analysis was carried out to assess ECN (10 mg/kg) treatment on synovial hyperplasia, tissue and joint destruction following CFA-induced arthritis, as mentioned [21]. Radiographic images of all the groups were taken and quantified using the digital radiographic system, as reported [6]. H and E staining assessed the underlying histopathological changes following CFA-induced arthritis. The paw tissue was placed in formalin solution (10%), mounted in paraffin blocks, sliced into 4 μm sections, and observed under a microscope (100X) [6].

Hematological analysis

Blood was analyzed for total leukocyte count, different leukocytes, and erythrocytes in all the animal groups [6].

Immunohistochemical expression of TNF-α, COX-2, p-JNK, and p-NF-κB in CFA-induced mice paw tissue

Immunohistochemical analysis of TNF-α, COX-2, p-JNK, and p-NF-κB was performed according to the published method [22]. Briefly, paw tissues (paraffin-fixed) were deparaffinized and rehydrated through xylene and alcohol. The intracellular peroxidase was quenched by H2O2 (3%) in menthol and then incubated for 20 min with goat serum. The slides were incubated overnight with mouse anti-TNF-α, anti-COX-2, anti-p-JNK, and anti-NF-κB antibodies [36]. After washing, the slides were incubated in secondary antibodies (goat-antimouse), then incubated with ABC reagents for 1 h,washed with phosphate buffer saline and marked with diaminobenzidine solution. Images were taken under a light microscope. The relative expression of TNF-α, COX-2, p-JNK, and NF-κB was measured using Image_J software [37].

Molecular docking analysis

Computational analysis is commonly employed to assess the interaction of the ligand with the protein targets and explore the mechanism of binding of the ligand with the protein [37]. In the present study, the molecular docking analysis of ECN against the various targets was performed using AutoDock Vina 4.2. The protein targets were downloaded from the RCSB PDB site (RCSB.PDB) and saved in PDB format. The water molecules and co-crystallized ligands were removed during protein preparation, and the energy was minimized [37]. The ligand was prepared using chembiodraw_16 and saved as an SDF file. The SDF file was converted to PDB format, and the ligand’s energy was minimized [37]. The binding energies and interacting amino acids with the ligands were represented in 3D and 2D structures.

Molecular dynamics simulation

The stable energy ECN-JNK and ECN-p65 complexes were further investigated for structural stability and estimation of binding free energies. Molecular Dynamic simulation was performed on NVIDIA GPU RTX1080 TI using Particle Mesh Ewald Molecular Dynamics (PMEMD.CUDA) from AMBER20. Initially, the complex was processed by employing Antechamber and the FF14SB force field [37, 38]. The latter was considered to define molecular characteristics and generate coordinate and topology files. Each complex was then solvated in a TIP3P water box, and subsequently, counter ions were added to obtain neutral systems. A production run was performed for 200 ns under periodic boundary conditions for each system. SHAKE algorithm was used to allow constraints on bonds containing hydrogen while long-range electrostatic interactions were processed through Particle Mesh Ewald (PME) method [37, 38]. Prior, systems energy was equilibrated and gradually increased to 300 K for 100 ps with additional 500 ps constant number of particles, pressure, temperature (NPT) equilibration, and pressure 1 atm. The production run of MD via steepest descent and conjugate gradient methods. In the heating step, systems temperature simulation was performed without any constraints for the timescale of 200 ns. The MD simulation trajectories were analyzed through the CPPTRAJ program of AMBER. The MMPBSA.py was consequently used to estimate binding free energy based on Molecular Mechanics-Poisson Boltzmann Surface Area (MM-PBSA) method [37, 38].

Pharmacokinetic analysis

The pharmacokinetic parameter of the ECN was assessed using computational analysis as reported previously [37, 38]. The parameters that were assessed during the current studies include absorption, metabolism, distribution, and excretion using Swiss target predictions (www.​swisstargetpredi​ction.​com), pkCSM (biosig.unimelb .edu.au/pkcsm/prediction), and PASS online (www.​pharmaexpert.​ru/​passonline/​) [38]. Furthermore, the physico-chemicals properties such as water solubility, lipid solubility, number of rotational bonds, hydrogen donor/acceptor, and drug-likeness behavior were assessed [38]. The possible metabolites of ECN were predicted using GLORY metabolites software (https://​nerdd.​zbh.​uni-hamburg.​de/​glory/​about/​) and were ranked according to the possibility of their occurrence [38].

Statistical analysis

The values are expressed as mean ± S. D (n = 7) and evaluated using a one-way analysis of variance (ANOVA) followed by the post hoc test to assess the inter-group comparisons, while two-way repeated analysis of variance (ANOVA) was used where appropriate. P-values such as p < 0.05, p < 0.01, and p < 0.001 were considered significant. GraphPad Prism version_5 was used for the statistical analysis.

Results

Effect of ECN on Carrageenan-induced paw edema, thermal hyperalgesia, mechanical hyperalgesia, and mechanical allodynia

Carrageenan-induced paw edema was used as a preliminary model to investigate the effect of ECN and optimize the dose-response. The ECN treatment was evaluated in different concentrations (1 mg/kg, 5 mg/kg, and 10 mg/kg) against paw edema, mechanical hyperalgesia, mechanical allodynia, and thermal hyperalgesia induced by Carrageenan. ECN markedly reduced paw edema compared to the Carrageenan-induced group (p < 0.05). The dose of 10 mg/kg exhibited the maximum response. Similarly, ECN attenuated the mechanical hyperalgesia (p < 0.01) and allodynia (p < 0.05) while significantly increasing the pain threshold compared to the Carrageenan-induced group. Furthermore, ECN administration significantly reduced the acute Carrageenan-induced thermal hyperalgesia (p < 0.05) and increased the pain threshold compared to the Carrageenan-induced group, as shown in Fig. 2. The ECN dose of 10 mg/kg was optimal and used in subsequent experiments.

Effect of ECN on CFA-induced survival rate, distress symptoms, and arthritic index

CFA-induced arthritis is associated with the development of distress symptoms in animals [33]. CFA showed a marked increase in the distress symptoms. However, ECN administration showed remarkable improvement in the distress symptoms. Similarly, CFA showed a marked increase in the arthritic index (inflammation of the joints, number of inflamed digits, and erythema). However, the ECN exhibited a marked reduction in the arthritic index compared to the CFA-induced group (p < 0.01). Furthermore, survival analysis showed no significant changes in the mortality rate of any treated group (Fig. 3).

Effect of ECN on CFA-induced paw edema and thermal hyperalgesia

The CFA -induced group showed an increase in paw edema in both acute and chronic models. However, ECN administration reduced paw swelling compared to the CFA-induced group (p < 0.05) (Fig. 4). The thermal hyperalgesia threshold in acute and chronic studies was significantly decreased in the CFA-induced group. However, ECN increased the thermal hyperalgesia threshold compared to the CFA-induced group (p < 0.01) (Fig. 4).

Effect of ECN on CFA-induced mechanical hyperalgesia and allodynia

The CFA administration significantly reduced the mechanical hyperalgesia threshold in acute and chronic studies, while this was significantly increased following treatment with ECN (p < 0.05) (Fig. 5). Similarly, the CFA-induced group showed a marked decrease in the mechanical allodynic threshold in acute and chronic studies which was significantly enhanced ECN (Fig. 5).

Effect of ECN on antioxidant proteins and enzymes in CFA-induced mice paw tissue

CFA-induced arthritis is associated with increased oxidative stress (MDA, NO) and reduction in the antioxidants GSH, GST, Catalase, and SOD. In the present study, there was a reduction in the antioxidants GSH, GST, Catalase, and SOD and a marked increase in the oxidative stress markers MDA and NO. ECN treatment resulted in an increase (p < 0.001) in the antioxidantsand a significant reduction (p < 0.001) in the MDA and NO levels compared to the CFA-induced group (Fig. 6).

Effect of ECN on pro-inflammatory cytokines

CFA administration elevated the levels of IL-1β and TNF-α in the paw tissue measured by ELISA; this effect was reversed by ECN (Fig. 7). Likewise, CFA significantly increased the expression of TNF-α, COX-2, p-JNK, and p-NF-κB measured by immunohistochemistry (Fig. 8X). ECN reduced the expression level of these proteins compared to the CFA treated group.

Effect of ECN on kidney and liver functions

Changes in liver and kidney functions were assessed by determining ALT, AST, and creatinine levels in all the animal groups following CFA-induced arthritis. Daily ECN treatment for 21 days had no effect on the liver and kidney markers (AST, ALT, and creatinine) (Table 1).
Table 1
Effect of the ECN (10 mg/kg) on hematological parameters such as total WBCs count, differential WBCs count, and RBCs level following CFA-induced arthritis
Parameters
WBC
(109/L)
LYM
(109/L)
NEU
(109/L)
MON
(109/L)
RBC
(1012/L)
PLT
(109/L)
Normal control
3.8 ± 0.18
2.02 ± 0.06
0.40 ± 0.029
0.32 ± 0.015
5.42 ± 0.3
257 ± 12
CFA
5.33 ± 0.12###
3.6 ± 0.17###
0.71 ± 0.18###
0.54 ± 0.16###
5.63 ± 0.29###
113 ± 22###
ECN (10 mg/kg)
3.96 ± 0.24
2.26 ± 0.11
0.54 ± 0.056
0.38 ± 0.045
5.84 ± 0.12
205 ± 15
The results showed no significant changes in the hematological parameters in all the treated groups. The data were represented as the mean ± SD (n = 7). (*) p < 0.05, (**) p < 0.01 and (***) p < 0.001 indicates significant differences from the CFA control group, while (###) means comparison with the CFA treated group

Effect of ECN on radiological and histological examination

Radiological analysis was performed to assess the changes in the paw tissue and subsequently assess the effect of ECN following CFA-induced arthritis [39]. CFA administration resulted in an increase in soft tissue swelling, cartilage destruction, synovial hyperplasia, and bone erosion [39]. In contrast, ECN reduced paw swelling, synovial hyperplasia, and bone erosion (Fig. 9). By H&E staining, CFA induced a significant increase in immune cell infiltration, paw edema, and changes in histological architecture. ECN (10 mg/kg) treatment showed marked improvement in the histological features compared to the CFA-induced group (p < 0.01) (Fig. 9).

Effect of ECN on DNA damage in paw tissue

The Comet assay was performed to assess changes to DNA in the paw tissue [39]. We observed marked changes in the DNA integrity, including DNA tailing and % DNA in the tail in response to CFA. ECN preserved the DNA architecture (DNA tailing and % DNA in tail) compared to the CFA-induced group (p < 0.05) (Fig. 10).

Effect of ECN on hematological changes

Changes in the levels of RBCs, total leukocytes count, and differential leukocytes count (neutrophils, eosinophils, platelets, and monocytes) were measured in all the recruited groups. The hematological studies revealed no significant changes in the hematological parameters (Table 2).
Table 2
Effect of ECN (10 mg/kg) on the liver and renal function test following CFA-induced arthritis
Parameters
ALT (IU/L)
AST (IU/L)
Creatinine (mg/dl)
Normal control
25.33 ± 0.57
21.33 ± 0.57
1.09 ± 1.73
CFA
37.33 ± 1.52###
35.66 ± 3.51###
1.50 ± 4.04###
ECN (10 mg/kg)
30.81 ± 4.35
25.5 ± 2.64
1.20 ± 0.57
The results revealed no marked changes in the LFTs and RFTs in any group. The data were represented as the mean ± SD (n = 7). (*) p < 0.05, (**) p < 0.01 and (***) p < 0.001 indicates significant differences from the CFA control group, while (###) means comparison with the CFA treated group

Molecular docking analysis

Molecular docking analysis was performed to assess the interaction of ECN with the protein targets JNK (PDB ID = 1uki), NF-κB (PDB ID = 1vkx), COX-2 (PDB ID = 1pxx), and TNF-α (PDB ID = 2az5). ECN showed variable interaction with the protein targets via multiple hydrophobic and hydrophilic bonds. The grid dimension center and size was maintained at (x = − 18.625, y = 74.475, z = 42.058) and (x = 100, y = 100, z = 100), respectively. The interaction of the ligand-receptor complex was visualized using Discovery studio visualizer_16, and the 3D and 2D images are shown in Fig. 11. ECN showed multiple hydrogen bonds and hydrophobic bonds with p65 (His 58A, Ser 112A, Thr 60A). Similarly, ENC also showed multiple hydrogen bonds with JNK (Arg 125A), COX-2 (His 2382C, Asn 2214C), and TNF-α (Tyr 151A). The negative binding energies of ECN with p65, JNK, COX-2, and TNF-α, as well as the interacting amino acids involved, are described in Table 3.
Table 3
The interaction of the ECN with various protein targets using molecular docking analysis
Ligand-protein interaction
Binding energy
(Kcal/mol)
Hydrogen bonds
Hydrogen bond amino acids
Hydrophobic interactions
ECN-JNK
−6.0
1
Arg25
Leu363, Leu23, Lys24, Asn9, Ala510
ECN-NF-κB
−6.0
3
His58, Ser112, Thr60
Ile110
ECN-COX-2
−7.4
2
His2212, Asn2382
Phe2210, His2207, Val2442, Ala2450, His2388, His2386, Ala2446
ECN-TNF-α
−8.1
1
Tyr151
Leu57, Tyr59, Tyr119
The molecular docking analysis showed multiple H-bonds and hydrophobic bonds of the ECN with various protein targets such as JNK, NF-κB, COX-2, and TNF-α. Additionally, the molecular docking analysis also showed the binding energies of the ECN with various protein targets and the amino acid involved in the interaction

Molecular dynamics simulation

The binding stability of the ECN-JNK complex and ECN-p65 complex was explored by a run of molecular dynamics simulation for 200 ns. To investigate the intermolecular conformation and stability of interactions between the compounds and protein, root means square deviation (RMSD) analysis was performed using molecular dynamics simulation trajectories. RMSD measures the mean distance of superimposed protein atoms for a given simulation frame relative to a selected reference snapshot. Considering the RMSD plot for the ECN-JNK complex and ECN-p65 complex, both complexes attain sustainable structure conformational stability as the simulation proceeds (Fig. 12). The RMSD value of complexes throughout simulation time is within an acceptable range and depicts a highly stable nature. The ECN-p65 system, in particular, was reported to be more structurally stable due to strong hydrogen bonding with residues like His58 and Thr60. These interactions are seen regularly in interactions with the ECN compound and play a critical role in holding the ligand at the docked site. The initial RMSD deviations of the ECN-JNK complex correspond to ECN conformational moves. This suggests that the compound binding pose predicted by docking studies is not real, and the binding mode is subjected to conformational changes to make its interaction strength with active residues at the active site of JNK protein. Equilibrium of this complex can also be visualized after 100 ns, and this stability is stronger towards the end of the simulation. The mean RMSD of ENC-p65 and ECN-JNK complex is 1.40 Å and 1.69 Å, respectively.

MMPBSA binding free energies

The MMPBSA binding free energy was estimated to affirm the docking and simulation results. From the data in Table 4, both systems are highly stable by acquiring highly negative total binding energy in kcal/mol. The overall binding free energy of ECN-JNK complex and ECN-p65 is − 44.38 kcal/mol and − 53.27 kcal/mol, respectively. The latter system, by binding energy, is more stable than the formal one. This net binding energy suggests the protein-ligand interactions are dominated by electrostatic energy and is equally supported by van der Waals energy. On the other hand, the non-polar biding energy like gas-phase energy favors complex formation, whereas negative contribution was reported from polar solvation energy.
Table 4
MMPBSA binding free energy of complexes in kcal/mol using a molecular dynamics simulation approach
Complex
Net Binding Free Energy
Net Electrostatic Binding Energy
Net van der Waals Binding Free Energy
Net Gas Phase Binding Energy
Net Polar Solvation Binding Free Energy
Net Non- polar Solvation Binding Free Energy
Net Solvation Binding Free Energy
ECN-JNK
−44.38
−25.99
−21.04
−47.03
11.78
−9.13
2.65
ECN-p65
−53.27
−35.64
−18.66
−54.3
8.00
−6.97
1.03
The MD simulation was performed for the NF-kB (p65) and JNK using AMBER20. The MD simulation showed various parameters such as net binding free energies, net electrostatic binding free energy, net van der Walls free energy, net gas phase binding energy, net polar solvation binding free energy, and net non-polar solvation binding free energy, and net solvation binding free energy

Pharmacokinetic analysis

Pharmacokinetic parameters such as ADME (absorption, distribution, metabolism, and excretion), toxicokinetics, and physicochemical properties of the ECN were studied using an in silico approach. The absorption parameters included water-solubility, Caco2 cell permeability, intestinal absorption, P-glycoprotein inhibition, and P-glycoprotein substrate. Similarly, the distribution includes a volume of distribution (VDss), blood-brain barrier permeability, and fraction of unbound drug. The metabolism includes cytochrome p450 inhibition, while the excretion includes total clearance. The toxicokinetic parameters include Ames toxicity, hepatotoxicity, hERG I and II inhibition, and minnow toxicity. The drug-likeness behavior of ECN and various rules of the drug-likeness such as Lipinski, Ghose, Veber, Egan, and Muegge were assessed (Fig. 13). Similarly, the metabolites of ECN were predicted using a computational tool and were ranked according to the possibility of their occurrence (Fig. 14).

Discussion

Inflammation is a multidimensional biochemical and defensive process comprised of vascular permeability and nonspecific responses stimulated by the innate immune system to infection, injured cells, and irritants [40]. Histamine, serotonin, bradykinin, and prostaglandin are the important endogenous mediators critically involved in inflammation [41]. The Carrageenan-induced inflammatory event takes place in two phases. During the first phase, the various mediators of inflammation such as bradykinin, complement, and tachykinin are released; while in the second phase, there is a release of prostaglandins and other enzymes [42].
The CFA-induced model is commonly used to assess the chronic inflammatory microenvironment within arthritic inflammation and assess the mediators involved in the ongoing inflammatory process [43]. The pro-inflammatory mediators unleashed during the inflammatory event sensitize the pain receptors that cause inflammatory pain. All types of inflammatory pain are due to sensitization of primary nociceptive neurons, which results in hyperalgesia and allodynia [44, 45].
Currently, various strategies are used in clinical practice to deal with rheumatoid arthritis, including the NSAIDs (non-steroidal anti-inflammatory drugs), DMARDs (disease-modifying anti-rheumatic drugs), and biological drugs [46, 47]. However, these drugs are associated with various unavoidable adverse outcomes that necessitate drug abstinence. The NSAIDs are associated with gastric ulceration and cardiovascular adverse events [46, 47], while the biological and DMARDs are associated with immunosuppression and increased chance of infection. Given this, there is an urgent need to develop drugs that are safe and effective against rheumatoid arthritis. In the current study, ECN was assessed against Carrageenan and CFA-induced arthritis. ECN administration significantly increased paw withdrawal threshold and paw withdrawal latency in Carrageenan and CFA-induced mice.
The histological studies play a key role in assessing the pathological changes within the inflammatory environment, such as infiltration of the inflammatory cells, edema, and fibrotic changes [48, 49]. Similarly, the radiological analysis assesses joint damage, stiffness, and inflammation [50, 51]. In the present study, ECN treatment significantly improved the histological and radiological features compared to the control. Other studies also reported that improvement in histological parameters of the paw tissue reduces inflammatory arthritis [46, 52].
Oxidative stress exacerbates the inflammatory process and is critically involved in the pathogenesis of rheumatoid arthritis [9]. The free radicals generated during the inflammatory process interact with cellular proteins, DNA, and lipids [9]. Free radicals such as superoxide and peroxides lead to the production of nitric oxide and MDA, which induce the production of pro-inflammatory mediators [53]. The body possesses the endogenous antioxidants system such as GST, GSH, Catalase, and SOD, which trigger the neutralization of the free radicals [25, 53]. CFA administration is commonly associated with alleviating the antioxidants and elevating oxidative stress markers [54]. In the current study, ECN significantly attenuated the oxidative stress markers and induced the antioxidants compared to the CFA treated group. The result of the current study is consistent with previously reported studies in which the reduction in oxidative stress and increase in the antioxidant showed amelioration of rheumatoid arthritis [52].
The increased activity of NF-κB signaling induces the production of numerous pro-inflammatory cytokines such as TNF-α, COX-2, and other biologically active substances [21]. COX-2, a critical pro-inflammatory mediator, triggers prostaglandin synthesis and accelerates the inflammatory process [13]. Similarly, MAPK (JNK) also regulates the synthesis of pro-inflammatory mediators, and its activation augments the inflammatory process [55, 56]. In the present study, ECN treatment markedly attenuated the expression levels of the pro-inflammatory proteins TNF-α, p-JNK, COX-2 and NF-κB compared to the CFA-treated group. Several other studies reported that inhibition of inflammatory cytokines such as TNF-α and COX-2 attenuates rheumatoid arthritis and improves the patient’s quality of life [57]. Additionally, activation of the MAPKs (JNK) and NF-κB signaling aggravate the underlying condition, while attenuation of NF-κB and MAPKs (JNK) reduce the signs and symptoms associated with rheumatoid arthritis [57].
Quantitative DNA damage in paw tissue was identified by DNA relocation out of the nucleus and into the Comet’s tail. DNA strand breakage can be due to the enhanced generation of ROS, leading to free radical formation, which might be the reason for the alteration and strand breakage of double-helical strands causing cell death [34, 58]. Treatment with ECN remarkably reduced DNA damage compared to the CFA treated group.
Computational analysis has played a key role in developing and screening new drugs for the ailment of various diseases. Virtual screening is now being widely used to elucidate the possible mechanism of the drug against various targets [34]. In the present study, molecular docking analysis, molecular dynamics simulation, and MMPBSA binding free energy were performed to assess the binding affinity of ECN and its mechanism of interaction with the protein targets TNF-α p-JNK, COX-2, and NF-κB [34]. ECN had different binding energies and affinities for the protein targets; however, ECN exhibited a strong affinity for NF-κB and MAPKs (JNK). Based on the molecular docking binding energies, the ECN-NF-κB and ECN-JNK complexes were subjected to molecular dynamics simulation to assess the stability of the ligand-protein complexes and binding free energies. The ECN-NF-κB and ECN-JNK complexes showed RMSD values in the acceptable range, and the complexes remained stable during the simulation process. Similarly, the present computational analysis results are consistent with the previously reported studies [37, 38, 59, 60, 61].

Conclusion

The present study investigated the anti-rheumatic potential of the natural compound ECN against Carrageenan and CFA-induced arthritis. ECN administration improved the signs and symptoms associated with acute and chronic inflammatory arthritis, including paw edema and pain. ECN administration restores the antioxidant and oxidative stress imbalance while significantly attenuating the inflammatory cytokines involved in the pathogenesis of rheumatoid arthritis. Furthermore, ECN treatment improved radiological and histological parameters by attenuating NF-kB and MAPKs (JNK) signaling and preserving DNA integrity. Similarly, ECN portrayed various physico-chemical properties and showed no toxicity against vital organs using computational and in vivo approaches. Overall, this study provides scientific evidence of ECN activity against inflammation and rheumatic arthritis. However, additional studies should be carried out to understand the mechanism of actionof ECN against Rheumatoid Arthritis.

Acknowledgments

The authors are grateful to Prof. Yeong Shik Kim, College of Pharmacy, Natural Products Research Institute, Seoul National University, South Korea, for providing the natural compound, ECN.

Declarations

The study was approved by the animal ethical committee of Quaid-i-Azam University, Islamabad (Approval No: BEC-FBS-QAU2018–125). All the animal experiments were conducted in accordance with the guidelines of the institutional bioethical committee. The dried flower buds of T. farfara were purchased from an Oriental Market in Seoul, Korea, and identified by Prof. Je-Hyun Lee (Dongkkuk University, Kyungju, South Korea). A voucher specimen (no. EA333) was deposited at Natural Product Chemistry Lab, College of Pharmacy, Ewha Womans University, Seoul, Korea). The collection of plant materials complies with the Ehwa, Womans, University, Seoul, Korea) guidelines. The department reviewed the procedure for sample collection and laboratory analysis to ensure compliance. The study was reported in accordance to ARRIVE guidelines (https://​arriveguidelines​.​org).
Not applicable.

Competing interests

The author declares no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Mahdi HJ, Khan NAK, Asmawi MZB, Mahmud R, Vikneswaran A, Murugaiyah L. In vivo anti-arthritic and anti-nociceptive effects of ethanol extract of Moringa oleifera leaves on complete Freund's adjuvant (CFA)-induced arthritis in rats. Integr Med Res. 2018;7(1):85–94.PubMedCrossRef Mahdi HJ, Khan NAK, Asmawi MZB, Mahmud R, Vikneswaran A, Murugaiyah L. In vivo anti-arthritic and anti-nociceptive effects of ethanol extract of Moringa oleifera leaves on complete Freund's adjuvant (CFA)-induced arthritis in rats. Integr Med Res. 2018;7(1):85–94.PubMedCrossRef
2.
Zurück zum Zitat Kapoor B, Singh SK, Gulati M, Gupta R, Vaidya Y. Application of liposomes in treatment of rheumatoid arthritis: quo vadis. Sci World J. 2014;2014. Kapoor B, Singh SK, Gulati M, Gupta R, Vaidya Y. Application of liposomes in treatment of rheumatoid arthritis: quo vadis. Sci World J. 2014;2014.
4.
Zurück zum Zitat Liu Q, Xiao X-H, Hu L-B, Jie H-Y, Wang Y, Ye W-C, et al. Anhuienoside C ameliorates collagen-induced arthritis through inhibition of MAPK and NF-κB signaling pathways. Front Pharmacol. 2017;8:299.PubMedPubMedCentralCrossRef Liu Q, Xiao X-H, Hu L-B, Jie H-Y, Wang Y, Ye W-C, et al. Anhuienoside C ameliorates collagen-induced arthritis through inhibition of MAPK and NF-κB signaling pathways. Front Pharmacol. 2017;8:299.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Khalid S, Ullah MZ, Khan AU, Afridi R, Rasheed H, Khan A, et al. Antihyperalgesic properties of honokiol in inflammatory pain models by targeting of NF-κB and Nrf2 signaling. Front Pharmacol. 2018;9:140.PubMedPubMedCentralCrossRef Khalid S, Ullah MZ, Khan AU, Afridi R, Rasheed H, Khan A, et al. Antihyperalgesic properties of honokiol in inflammatory pain models by targeting of NF-κB and Nrf2 signaling. Front Pharmacol. 2018;9:140.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Khan A, Ullah MZ, Afridi R, Rasheed H, Khalid S, Ullah H, et al. Antinociceptive properties of 25-methoxy hispidol A, a triterpinoid isolated from Poncirus trifoliata (Rutaceae) through inhibition of NF-κB signalling in mice. Phytother Res. 2019;33(2):327–41.PubMedCrossRef Khan A, Ullah MZ, Afridi R, Rasheed H, Khalid S, Ullah H, et al. Antinociceptive properties of 25-methoxy hispidol A, a triterpinoid isolated from Poncirus trifoliata (Rutaceae) through inhibition of NF-κB signalling in mice. Phytother Res. 2019;33(2):327–41.PubMedCrossRef
7.
Zurück zum Zitat Ali H, Khan A, Ali J, Ullah H, Khan A, Ali H, et al. Attenuation of LPS-induced acute lung injury by continentalic acid in rodents through inhibition of inflammatory mediators correlates with increased Nrf2 protein expression. BMC Pharmacol Toxicol. 2020;21(1):1–14.CrossRef Ali H, Khan A, Ali J, Ullah H, Khan A, Ali H, et al. Attenuation of LPS-induced acute lung injury by continentalic acid in rodents through inhibition of inflammatory mediators correlates with increased Nrf2 protein expression. BMC Pharmacol Toxicol. 2020;21(1):1–14.CrossRef
8.
Zurück zum Zitat Zhang Y, Chen J, Fu H, Kuang S, He F, Zhang M, et al. Exosomes derived from 3D-cultured MSCs improve therapeutic effects in periodontitis and experimental colitis and restore the Th17 cell/Treg balance in inflamed periodontium. Int J Oral Sci. 2021;13(1):1–15.CrossRef Zhang Y, Chen J, Fu H, Kuang S, He F, Zhang M, et al. Exosomes derived from 3D-cultured MSCs improve therapeutic effects in periodontitis and experimental colitis and restore the Th17 cell/Treg balance in inflamed periodontium. Int J Oral Sci. 2021;13(1):1–15.CrossRef
9.
Zurück zum Zitat Ahmad SF, Ansari MA, Zoheir KM, Bakheet SA, Korashy HM, Nadeem A, et al. Regulation of TNF-α and NF-κB activation through the JAK/STAT signaling pathway downstream of histamine 4 receptor in a rat model of LPS-induced joint inflammation. Immunobiology. 2015;220(7):889–98.PubMedCrossRef Ahmad SF, Ansari MA, Zoheir KM, Bakheet SA, Korashy HM, Nadeem A, et al. Regulation of TNF-α and NF-κB activation through the JAK/STAT signaling pathway downstream of histamine 4 receptor in a rat model of LPS-induced joint inflammation. Immunobiology. 2015;220(7):889–98.PubMedCrossRef
10.
Zurück zum Zitat De S, Manna A, Kundu S, De Sarkar S, Chatterjee U, Sen T, et al. Allylpyrocatechol attenuates collagen-induced arthritis via attenuation of oxidative stress secondary to modulation of the MAPK, JAK/STAT, and Nrf2/HO-1 pathways. J Pharmacol Exp Ther. 2017;360(2):249–59.PubMedCrossRef De S, Manna A, Kundu S, De Sarkar S, Chatterjee U, Sen T, et al. Allylpyrocatechol attenuates collagen-induced arthritis via attenuation of oxidative stress secondary to modulation of the MAPK, JAK/STAT, and Nrf2/HO-1 pathways. J Pharmacol Exp Ther. 2017;360(2):249–59.PubMedCrossRef
11.
Zurück zum Zitat Rasheed H, Afridi R, Khan AU, Ullah MZ, Khalid S, Atiq A, et al. Anti-inflammatory, anti-rheumatic and analgesic activities of 2-(5-mercapto-1, 3, 4-oxadiazol-2-yl)-N-propylbenzenesulphonamide (MOPBS) in rodents. Inflammopharmacology. 2018;26(4):1037–49.PubMedCrossRef Rasheed H, Afridi R, Khan AU, Ullah MZ, Khalid S, Atiq A, et al. Anti-inflammatory, anti-rheumatic and analgesic activities of 2-(5-mercapto-1, 3, 4-oxadiazol-2-yl)-N-propylbenzenesulphonamide (MOPBS) in rodents. Inflammopharmacology. 2018;26(4):1037–49.PubMedCrossRef
12.
Zurück zum Zitat Ali J, Khan AU, Shah FA, Ali H, Islam SU, Kim YS, et al. Mucoprotective effects of Saikosaponin-A in 5-fluorouracil-induced intestinal mucositis in mice model. Life Sci. 2019;239:116888.PubMedCrossRef Ali J, Khan AU, Shah FA, Ali H, Islam SU, Kim YS, et al. Mucoprotective effects of Saikosaponin-A in 5-fluorouracil-induced intestinal mucositis in mice model. Life Sci. 2019;239:116888.PubMedCrossRef
13.
Zurück zum Zitat Chu S-J, Tang S-E, Pao H-P, Wu S-Y, Liao W-I. Protease-Activated Receptor-1 Antagonist Protects Against Lung Ischemia/Reperfusion Injury. Front Pharmacol. 2021:2717. Chu S-J, Tang S-E, Pao H-P, Wu S-Y, Liao W-I. Protease-Activated Receptor-1 Antagonist Protects Against Lung Ischemia/Reperfusion Injury. Front Pharmacol. 2021:2717.
14.
Zurück zum Zitat Rahman M, Beg S, Verma A, Al Abbasi FA, Anwar F, Saini S, et al. Phytoconstituents as pharmacotherapeutics in rheumatoid arthritis: challenges and scope of nano/submicromedicine in its effective delivery. J Pharm Pharmacol. 2017;69(1):1–14.PubMedCrossRef Rahman M, Beg S, Verma A, Al Abbasi FA, Anwar F, Saini S, et al. Phytoconstituents as pharmacotherapeutics in rheumatoid arthritis: challenges and scope of nano/submicromedicine in its effective delivery. J Pharm Pharmacol. 2017;69(1):1–14.PubMedCrossRef
15.
Zurück zum Zitat Lee J, Song K, Huh E, Oh MS, Kim YS. Neuroprotection against 6-OHDA toxicity in PC12 cells and mice through the Nrf2 pathway by a sesquiterpenoid from Tussilago farfara. Redox Biol. 2018;18:6–15.PubMedPubMedCentralCrossRef Lee J, Song K, Huh E, Oh MS, Kim YS. Neuroprotection against 6-OHDA toxicity in PC12 cells and mice through the Nrf2 pathway by a sesquiterpenoid from Tussilago farfara. Redox Biol. 2018;18:6–15.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Wang D, Fang L, Wang X, Qiu J, Huang L. Preparative separation and purification of sesquiterpenoids from Tussilago farfara L. by high-speed counter-current chromatography. Quim Nova. 2011;34(5):804–7. Wang D, Fang L, Wang X, Qiu J, Huang L. Preparative separation and purification of sesquiterpenoids from Tussilago farfara L. by high-speed counter-current chromatography. Quim Nova. 2011;34(5):804–7.
17.
Zurück zum Zitat Liu L-L, Yang J-L, Shi Y-P. Sesquiterpenoids and other constituents from the flower buds of Tussilago farfara. J Asian Nat Prod Res. 2011;13(10):920–9.PubMedCrossRef Liu L-L, Yang J-L, Shi Y-P. Sesquiterpenoids and other constituents from the flower buds of Tussilago farfara. J Asian Nat Prod Res. 2011;13(10):920–9.PubMedCrossRef
18.
Zurück zum Zitat Song K, Lee KJ, Kim YS. Development of an efficient fractionation method for the preparative separation of sesquiterpenoids from Tussilago farfara by counter-current chromatography. J Chromatogr A. 2017;1489:107–14.PubMedCrossRef Song K, Lee KJ, Kim YS. Development of an efficient fractionation method for the preparative separation of sesquiterpenoids from Tussilago farfara by counter-current chromatography. J Chromatogr A. 2017;1489:107–14.PubMedCrossRef
19.
Zurück zum Zitat Zimmermann M. Ethical guidelines for investigations of experimental pain in conscious animals. Pain. 1983;16(2):109–10.PubMedCrossRef Zimmermann M. Ethical guidelines for investigations of experimental pain in conscious animals. Pain. 1983;16(2):109–10.PubMedCrossRef
20.
Zurück zum Zitat Afridi R, Khan AU, Khalid S, Shal B, Rasheed H, Ullah MZ, et al. Toxicology: Anti-hyperalgesic properties of a flavanone derivative Poncirin in acute and chronic inflammatory pain models in mice. 2019;20(1):1–16. Afridi R, Khan AU, Khalid S, Shal B, Rasheed H, Ullah MZ, et al. Toxicology: Anti-hyperalgesic properties of a flavanone derivative Poncirin in acute and chronic inflammatory pain models in mice. 2019;20(1):1–16.
21.
Zurück zum Zitat Naveed M, Khan SZ, Zeeshan S, Khan A, Shal B, Atiq A, et al. A new cationic palladium (II) dithiocarbamate exhibits anti-inflammatory, analgesic, and antipyretic activities through inhibition of inflammatory mediators in in vivo models. Naunyn Schmiedeberg’s Arch Pharmacol. 2019:1–17. Naveed M, Khan SZ, Zeeshan S, Khan A, Shal B, Atiq A, et al. A new cationic palladium (II) dithiocarbamate exhibits anti-inflammatory, analgesic, and antipyretic activities through inhibition of inflammatory mediators in in vivo models. Naunyn Schmiedeberg’s Arch Pharmacol. 2019:1–17.
22.
Zurück zum Zitat Atiq A, Shal B, Naveed M, Khan A, Ali J, Zeeshan S, et al. Diadzein ameliorates 5-fluorouracil-induced intestinal mucositis by suppressing oxidative stress and inflammatory mediators in rodents. Eur J Pharmacol. 2019;843:292–306.PubMedCrossRef Atiq A, Shal B, Naveed M, Khan A, Ali J, Zeeshan S, et al. Diadzein ameliorates 5-fluorouracil-induced intestinal mucositis by suppressing oxidative stress and inflammatory mediators in rodents. Eur J Pharmacol. 2019;843:292–306.PubMedCrossRef
23.
Zurück zum Zitat Seeuws S, Jacques P, Van Praet J, Drennan M, Coudenys J, Decruy T, et al. A multiparameter approach to monitor disease activity in collagen-induced arthritis. Arthritis Res Ther. 2010;12(4):R160.PubMedPubMedCentralCrossRef Seeuws S, Jacques P, Van Praet J, Drennan M, Coudenys J, Decruy T, et al. A multiparameter approach to monitor disease activity in collagen-induced arthritis. Arthritis Res Ther. 2010;12(4):R160.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Khan S, Shehzad O, Chun J, Choi RJ, Park S, Islam MN, et al. Anti-hyperalgesic and anti-allodynic activities of capillarisin via suppression of inflammatory signaling in animal model. J Ethnopharmacol. 2014;152(3):478–86.PubMedCrossRef Khan S, Shehzad O, Chun J, Choi RJ, Park S, Islam MN, et al. Anti-hyperalgesic and anti-allodynic activities of capillarisin via suppression of inflammatory signaling in animal model. J Ethnopharmacol. 2014;152(3):478–86.PubMedCrossRef
25.
Zurück zum Zitat Shal B, Khan A, Naveed M, Ali H, Seo EK, Choi H, et al. Neuroprotective effect of 25-Methoxyhispidol A against CCl4-induced behavioral alterations by targeting VEGF/BDNF and caspase-3 in mice. Life Sci. 2020;117684. Shal B, Khan A, Naveed M, Ali H, Seo EK, Choi H, et al. Neuroprotective effect of 25-Methoxyhispidol A against CCl4-induced behavioral alterations by targeting VEGF/BDNF and caspase-3 in mice. Life Sci. 2020;117684.
26.
Zurück zum Zitat Arome D, Sunday AI, Onalike EI, Amarachi A. Pain and inflammation: Management by conventional and herbal therapy. Indian J Pain. 2014;28(1):5.CrossRef Arome D, Sunday AI, Onalike EI, Amarachi A. Pain and inflammation: Management by conventional and herbal therapy. Indian J Pain. 2014;28(1):5.CrossRef
27.
Zurück zum Zitat Cho H, Yang YD, Lee J, Lee B, Kim T, Jang Y, et al. The calcium-activated chloride channel anoctamin 1 acts as a heat sensor in nociceptive neurons. Nat Neurosci. 2012;15(7):1015–21.PubMedCrossRef Cho H, Yang YD, Lee J, Lee B, Kim T, Jang Y, et al. The calcium-activated chloride channel anoctamin 1 acts as a heat sensor in nociceptive neurons. Nat Neurosci. 2012;15(7):1015–21.PubMedCrossRef
28.
Zurück zum Zitat Khan A, Khalid S, Khan A, Shal B, Kang E, Lee H, et al. ECN from Tussilago Farfara Attenuates Neuropathic Pain by Suppressing Oxidative Stress, Inflammatory and Pro-Apoptotic Proteins Expression in Surgical Model; 2020.CrossRef Khan A, Khalid S, Khan A, Shal B, Kang E, Lee H, et al. ECN from Tussilago Farfara Attenuates Neuropathic Pain by Suppressing Oxidative Stress, Inflammatory and Pro-Apoptotic Proteins Expression in Surgical Model; 2020.CrossRef
29.
Zurück zum Zitat Doherty V, Ogunkuade O, Kanife U. Biomarkers of oxidative stress and heavy metal levels as indicators of environmental pollution in some selected fishes in Lagos, Nigeria. Am Eurasian J Agric Environ Sci. 2010;7(3):359–65. Doherty V, Ogunkuade O, Kanife U. Biomarkers of oxidative stress and heavy metal levels as indicators of environmental pollution in some selected fishes in Lagos, Nigeria. Am Eurasian J Agric Environ Sci. 2010;7(3):359–65.
30.
Zurück zum Zitat Farombi E, Adelowo O, Ajimoko Y. Biomarkers of oxidative stress and heavy metal levels as indicators of environmental pollution in African cat fish (Clarias gariepinus) from Nigeria Ogun River. Int J Environ Res Public Health. 2007;4(2):158–65.PubMedPubMedCentralCrossRef Farombi E, Adelowo O, Ajimoko Y. Biomarkers of oxidative stress and heavy metal levels as indicators of environmental pollution in African cat fish (Clarias gariepinus) from Nigeria Ogun River. Int J Environ Res Public Health. 2007;4(2):158–65.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Moulahoum H, Boumaza BMA, Ferrat M, Nagy A-L, Olteanu DE, Bounaama A, et al. Aberrant crypt foci are regionally affected by zinc treatment in a 1, 2-dimethylhydrazine induced colon carcinogenesis model. J Trace Elem Med Biol. 2018;47:21–30.PubMedCrossRef Moulahoum H, Boumaza BMA, Ferrat M, Nagy A-L, Olteanu DE, Bounaama A, et al. Aberrant crypt foci are regionally affected by zinc treatment in a 1, 2-dimethylhydrazine induced colon carcinogenesis model. J Trace Elem Med Biol. 2018;47:21–30.PubMedCrossRef
32.
Zurück zum Zitat Ullah H, Khan A, Baig MW, Ullah N, Ahmed N, Tipu MK, et al. Poncirin attenuates CCL4-induced liver injury through inhibition of oxidative stress and inflammatory cytokines in mice. BMC Complement Med Ther. 2020;20:1–14.CrossRef Ullah H, Khan A, Baig MW, Ullah N, Ahmed N, Tipu MK, et al. Poncirin attenuates CCL4-induced liver injury through inhibition of oxidative stress and inflammatory cytokines in mice. BMC Complement Med Ther. 2020;20:1–14.CrossRef
33.
Zurück zum Zitat Naveed M, Ullah R, Khan A, Shal B, Khan AU, Khan SZ, et al. Anti-Neuropathic Pain Activity of A Cationic Palladium (II) Dithiocarbamate By Suppressing The Inflammatory Mediators in Paclitaxel-Induced Neuropathic Pain Model; 2021. Naveed M, Ullah R, Khan A, Shal B, Khan AU, Khan SZ, et al. Anti-Neuropathic Pain Activity of A Cationic Palladium (II) Dithiocarbamate By Suppressing The Inflammatory Mediators in Paclitaxel-Induced Neuropathic Pain Model; 2021.
34.
Zurück zum Zitat Shal B, Khan A, Naveed M, Khan NU, AlSharari SD, Kim YS, et al. Effect of 25-methoxy hispidol A isolated from Poncirus trifoliate against bacteria-induced anxiety and depression by targeting neuroinflammation, oxidative stress and apoptosis in mice. Biomed Pharmacother. 2019;111:209–23.PubMedCrossRef Shal B, Khan A, Naveed M, Khan NU, AlSharari SD, Kim YS, et al. Effect of 25-methoxy hispidol A isolated from Poncirus trifoliate against bacteria-induced anxiety and depression by targeting neuroinflammation, oxidative stress and apoptosis in mice. Biomed Pharmacother. 2019;111:209–23.PubMedCrossRef
35.
Zurück zum Zitat Khan A, Shal B, Naveed M, Shah FA, Atiq A, Khan NU, et al. Matrine ameliorates anxiety and depression-like behaviour by targeting hyperammonemia-induced neuroinflammation and oxidative stress in CCl4 model of liver injury. Neurotoxicology. 2019;72:38–50.PubMedCrossRef Khan A, Shal B, Naveed M, Shah FA, Atiq A, Khan NU, et al. Matrine ameliorates anxiety and depression-like behaviour by targeting hyperammonemia-induced neuroinflammation and oxidative stress in CCl4 model of liver injury. Neurotoxicology. 2019;72:38–50.PubMedCrossRef
36.
Zurück zum Zitat Khan AM, Khan AU, Ali H, Islam SU, Seo EK, Khan S. Continentalic acid exhibited nephroprotective activity against the LPS and E coli-induced kidney injury through inhibition of the oxidative stress and inflammation. Int Immunopharmacol. 2020;80:106209.PubMedCrossRef Khan AM, Khan AU, Ali H, Islam SU, Seo EK, Khan S. Continentalic acid exhibited nephroprotective activity against the LPS and E coli-induced kidney injury through inhibition of the oxidative stress and inflammation. Int Immunopharmacol. 2020;80:106209.PubMedCrossRef
37.
Zurück zum Zitat Khan AU, Khan AM, Khan A, Shal B, Aziz A, Ahmed MN, et al. The newly synthesized compounds (NCHDH and NTHDH) attenuates LPS-induced septicemia and multi-organ failure via Nrf2/HO1 and HSP/TRVP1 signaling in mice. Chem Biol Interact. 2020;329:109220.PubMedCrossRef Khan AU, Khan AM, Khan A, Shal B, Aziz A, Ahmed MN, et al. The newly synthesized compounds (NCHDH and NTHDH) attenuates LPS-induced septicemia and multi-organ failure via Nrf2/HO1 and HSP/TRVP1 signaling in mice. Chem Biol Interact. 2020;329:109220.PubMedCrossRef
38.
Zurück zum Zitat Keum J, Yoo S, Lee D, Nam H. Prediction of compound-target interactions of natural products using large-scale drug and protein information. BMC Bioinformatics. 2016;17(6):219.PubMedPubMedCentralCrossRef Keum J, Yoo S, Lee D, Nam H. Prediction of compound-target interactions of natural products using large-scale drug and protein information. BMC Bioinformatics. 2016;17(6):219.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Khan A, Shehzad O, Seo EK, Onder A, Khan S. Therapies: Anti-allergic activities of Umbelliferone against histamine-and Picryl chloride-induced ear edema by targeting Nrf2/iNOS signaling in mice. BMC Complement Med Ther. 2021;21(1):1–17. Khan A, Shehzad O, Seo EK, Onder A, Khan S. Therapies: Anti-allergic activities of Umbelliferone against histamine-and Picryl chloride-induced ear edema by targeting Nrf2/iNOS signaling in mice. BMC Complement Med Ther. 2021;21(1):1–17.
40.
Zurück zum Zitat Antonisamy P, Dhanasekaran M, Kim H-R, Jo S-G, Agastian P, Kwon K-B. Anti-inflammatory and analgesic activity of ononitol monohydrate isolated from Cassia tora L. in animal models. Saudi J Biol Sci. 2017;24(8):1933–8.PubMedPubMedCentralCrossRef Antonisamy P, Dhanasekaran M, Kim H-R, Jo S-G, Agastian P, Kwon K-B. Anti-inflammatory and analgesic activity of ononitol monohydrate isolated from Cassia tora L. in animal models. Saudi J Biol Sci. 2017;24(8):1933–8.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Tatiya AU, Saluja AK, Kalaskar MG, Surana SJ, Patil PH. Evaluation of analgesic and anti-inflammatory activity of Bridelia retusa (Spreng) bark. J Tradit Complement Med. 2017;7(4):441–51.PubMedPubMedCentralCrossRef Tatiya AU, Saluja AK, Kalaskar MG, Surana SJ, Patil PH. Evaluation of analgesic and anti-inflammatory activity of Bridelia retusa (Spreng) bark. J Tradit Complement Med. 2017;7(4):441–51.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Brooks PM, Day RO. Non-steroidal anti-inflammatory drugs—differences and similarities. N Engl J Med. 1991;324(24):1716–25.PubMedCrossRef Brooks PM, Day RO. Non-steroidal anti-inflammatory drugs—differences and similarities. N Engl J Med. 1991;324(24):1716–25.PubMedCrossRef
43.
Zurück zum Zitat Khan S, Shehzad O, Chun J, Kim YS. Mechanism underlying anti-hyperalgesic and anti-allodynic properties of anomalin in both acute and chronic inflammatory pain models in mice through inhibition of NF-κB, MAPKs and CREB signaling cascades. Eur J Pharmacol. 2013;718(1–3):448–58.PubMedCrossRef Khan S, Shehzad O, Chun J, Kim YS. Mechanism underlying anti-hyperalgesic and anti-allodynic properties of anomalin in both acute and chronic inflammatory pain models in mice through inhibition of NF-κB, MAPKs and CREB signaling cascades. Eur J Pharmacol. 2013;718(1–3):448–58.PubMedCrossRef
44.
45.
Zurück zum Zitat Khan SM, Choi RJ, Lee DU, Kim YS. Sesquiterpene derivatives isolated from Cyperus rotundus L. inhibit inflammatory signaling mediated by NF-κB. Nat Prod Sci. 2011;17(3):250–5. Khan SM, Choi RJ, Lee DU, Kim YS. Sesquiterpene derivatives isolated from Cyperus rotundus L. inhibit inflammatory signaling mediated by NF-κB. Nat Prod Sci. 2011;17(3):250–5.
46.
Zurück zum Zitat Ahmad SF, Ansari MA, Nadeem A, Zoheir KM, Bakheet SA, Alsaad AM, et al. STA-21, a STAT-3 inhibitor, attenuates the development and progression of inflammation in collagen antibody-induced arthritis. Immunobiology. 2017;222(2):206–17.PubMedCrossRef Ahmad SF, Ansari MA, Nadeem A, Zoheir KM, Bakheet SA, Alsaad AM, et al. STA-21, a STAT-3 inhibitor, attenuates the development and progression of inflammation in collagen antibody-induced arthritis. Immunobiology. 2017;222(2):206–17.PubMedCrossRef
47.
Zurück zum Zitat Ahmad SF, Ansari MA, Nadeem A, Zoheir KM, Bakheet SA, Al-Shabanah OA, et al. The tyrosine kinase inhibitor tyrphostin AG126 reduces activation of inflammatory cells and increases Foxp3+ regulatory T cells during pathogenesis of rheumatoid arthritis. Mol Immunol. 2016;78:65–78.PubMedCrossRef Ahmad SF, Ansari MA, Nadeem A, Zoheir KM, Bakheet SA, Al-Shabanah OA, et al. The tyrosine kinase inhibitor tyrphostin AG126 reduces activation of inflammatory cells and increases Foxp3+ regulatory T cells during pathogenesis of rheumatoid arthritis. Mol Immunol. 2016;78:65–78.PubMedCrossRef
48.
Zurück zum Zitat Feldmann M, Maini SRN. Role of cytokines in rheumatoid arthritis: an education in pathophysiology and therapeutics. Immunol Rev. 2008;223(1):7–19.PubMedCrossRef Feldmann M, Maini SRN. Role of cytokines in rheumatoid arthritis: an education in pathophysiology and therapeutics. Immunol Rev. 2008;223(1):7–19.PubMedCrossRef
49.
Zurück zum Zitat Khan S, Shin EM, Choi RJ, Jung YH, Kim J, Tosun A, et al. Suppression of LPS-induced inflammatory and NF-κB responses by anomalin in RAW 264.7 macrophages. J Cell Biochem. 2011;112(8):2179–88.PubMedCrossRef Khan S, Shin EM, Choi RJ, Jung YH, Kim J, Tosun A, et al. Suppression of LPS-induced inflammatory and NF-κB responses by anomalin in RAW 264.7 macrophages. J Cell Biochem. 2011;112(8):2179–88.PubMedCrossRef
50.
Zurück zum Zitat Li M, Shi J, Tang JR, Chen D, Ai B, Chen J, et al. Effects of complete Freund's adjuvant on immunohistochemical distribution of IL-1β and IL-1R I in neurons and glia cells of dorsal root ganglion 1. Acta Pharmacol Sin. 2005;26(2):192–8.PubMedCrossRef Li M, Shi J, Tang JR, Chen D, Ai B, Chen J, et al. Effects of complete Freund's adjuvant on immunohistochemical distribution of IL-1β and IL-1R I in neurons and glia cells of dorsal root ganglion 1. Acta Pharmacol Sin. 2005;26(2):192–8.PubMedCrossRef
51.
Zurück zum Zitat Parada C, Tambeli C, Cunha F, Ferreira S. The major role of peripheral release of histamine and 5-hydroxytryptamine in formalin-induced nociception. Neuroscience. 2001;102(4):937–44.PubMedCrossRef Parada C, Tambeli C, Cunha F, Ferreira S. The major role of peripheral release of histamine and 5-hydroxytryptamine in formalin-induced nociception. Neuroscience. 2001;102(4):937–44.PubMedCrossRef
52.
Zurück zum Zitat A Ansari M, Nadeem A, A Bakheet S, M Attia S, Shahid M, S Alyousef F, et al. Chemokine receptor 5 antagonism causes reduction in joint inflammation in a collagen-induced arthritis mouse model. Molecules. 2021;26(7):1839.CrossRef A Ansari M, Nadeem A, A Bakheet S, M Attia S, Shahid M, S Alyousef F, et al. Chemokine receptor 5 antagonism causes reduction in joint inflammation in a collagen-induced arthritis mouse model. Molecules. 2021;26(7):1839.CrossRef
53.
Zurück zum Zitat Vaculin S, Franek M, Vejrazka M. Role of oxidative stress in animal model of visceral pain. Neurosci Lett. 2010;477(2):82–5.PubMedCrossRef Vaculin S, Franek M, Vejrazka M. Role of oxidative stress in animal model of visceral pain. Neurosci Lett. 2010;477(2):82–5.PubMedCrossRef
55.
Zurück zum Zitat Schett G, Zwerina J, Firestein G. The p38 mitogen-activated protein kinase (MAPK) pathway in rheumatoid arthritis. Ann Rheum Dis. 2008;67(7):909–16.PubMedCrossRef Schett G, Zwerina J, Firestein G. The p38 mitogen-activated protein kinase (MAPK) pathway in rheumatoid arthritis. Ann Rheum Dis. 2008;67(7):909–16.PubMedCrossRef
56.
Zurück zum Zitat Kazmi Z, Zeeshan S, Khan A, Malik S, Shehzad A, Seo EK, et al. Anti-epileptic activity of daidzin in PTZ-induced mice model by targeting oxidative stress and BDNF/VEGF signaling. NeuroToxicology. 2020. Kazmi Z, Zeeshan S, Khan A, Malik S, Shehzad A, Seo EK, et al. Anti-epileptic activity of daidzin in PTZ-induced mice model by targeting oxidative stress and BDNF/VEGF signaling. NeuroToxicology. 2020.
57.
Zurück zum Zitat Zwerina J, Redlich K, Schett G, Smolen JS. Pathogenesis of rheumatoid arthritis: targeting cytokines. Ann N Y Acad Sci. 2005;1051(1):716–29.PubMedCrossRef Zwerina J, Redlich K, Schett G, Smolen JS. Pathogenesis of rheumatoid arthritis: targeting cytokines. Ann N Y Acad Sci. 2005;1051(1):716–29.PubMedCrossRef
58.
Zurück zum Zitat Khan A, Shal B, Naveed M, Nasir B, Irshad N, Ali H, et al. Matrine alleviates neurobehavioral alterations via modulation of JNK-mediated caspase-3 and BDNF/VEGF signaling in a mouse model of burn injury. Psychopharmacology. 2020. Khan A, Shal B, Naveed M, Nasir B, Irshad N, Ali H, et al. Matrine alleviates neurobehavioral alterations via modulation of JNK-mediated caspase-3 and BDNF/VEGF signaling in a mouse model of burn injury. Psychopharmacology. 2020.
59.
Zurück zum Zitat Zhong Z, Zhang Q, Tao H, Sang W, Cui L, Qiang W, et al. Anti-inflammatory activities of Sigesbeckia glabrescens Makino: combined in vitro and in silico investigations. Chin Med. 2019;14(1):1–12.CrossRef Zhong Z, Zhang Q, Tao H, Sang W, Cui L, Qiang W, et al. Anti-inflammatory activities of Sigesbeckia glabrescens Makino: combined in vitro and in silico investigations. Chin Med. 2019;14(1):1–12.CrossRef
60.
Zurück zum Zitat Ullah H, Khan A, Bibi T, Ahmad S, Shehzad O, Ali H, et al. Comprehensive in vivo and in silico approaches to explore the hepatoprotective activity of poncirin against paracetamol toxicity. Naunyn-Schmiedeberg’s Arch Pharmacol. 2022:1–21. Ullah H, Khan A, Bibi T, Ahmad S, Shehzad O, Ali H, et al. Comprehensive in vivo and in silico approaches to explore the hepatoprotective activity of poncirin against paracetamol toxicity. Naunyn-Schmiedeberg’s Arch Pharmacol. 2022:1–21.
61.
Zurück zum Zitat Khan AU, Khan A, Khan A, Shal B, Aziz A, Ahmed MN, et al. Inhibition of NF-κB signaling and HSP70/HSP90 proteins by newly synthesized hydrazide derivatives in arthritis model. Naunyn-Schmiedeberg’s Arch Pharmacol. 2021:1–23. Khan AU, Khan A, Khan A, Shal B, Aziz A, Ahmed MN, et al. Inhibition of NF-κB signaling and HSP70/HSP90 proteins by newly synthesized hydrazide derivatives in arthritis model. Naunyn-Schmiedeberg’s Arch Pharmacol. 2021:1–23.
Metadaten
Titel
RETRACTED ARTICLE: Suppression of NF-κB signaling by ECN in an arthritic model of inflammation
verfasst von
Amna Khan
Li Zhang
Chang Hu Li
Ashraf Ullah Khan
Bushra Shal
Adnan Khan
Sajjad Ahmad
Fakhar ud Din
Zia ur rehman
Feng Wang
Salman Khan
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Complementary Medicine and Therapies / Ausgabe 1/2022
Elektronische ISSN: 2662-7671
DOI
https://doi.org/10.1186/s12906-022-03629-7

Weitere Artikel der Ausgabe 1/2022

BMC Complementary Medicine and Therapies 1/2022 Zur Ausgabe