Abstract
Background/Aim: Rheumatoid arthritis (RA) is an inflammatory autoimmune disease, and management of it is still a challenge. The present investigation assessed the potential preventive effect of phlorizin on rats with RA. Materials and Methods: A total of 40 healthy Wistar rats were used for this study. Bovine type II collagen and Freund’s incomplete adjuvant (1:1 and 1 mg/ml) were administered on days 1 and 8 of the protocol to induce RA in rats; treatment with phlorizin at 60 or 120 mg/kg was started after the 4th week of the protocol, and its effect on inflammation, level of inflammatory cytokines, and expression of proteins were estimated in RA rats. Moreover, an in vitro study was performed on fibroblast-like synoviocytes (FLSs), and the effects of phlorizin on proliferation, apoptosis, and expression of the mechanistic target of rapamycin kinase pathway protein after stimulating these cells with tumor necrosis factor α (TNF-α) were estimated. Results: The data obtained from the study indicate that phlorizin has the potential to mitigate inflammation and enhance weight management in rats with RA induced by bovine type II collagen (CII). The level of inflammatory cytokines in the serum and the expression of protein kinase B (AKT), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), and mechanistic target of rapamycin kinase (mTOR) proteins in the joint tissue were reduced in phlorizin-treated rats with RA. In this investigation, phlorizin was shown to reverse the histological abnormalities in the joint tissue of rats with RA. The in-vitro study showed that phlorizin reduced proliferation and had no apoptotic effect on TNF-α-stimulated FLSs. Expression of AKT, PI3K, and mTOR proteins was also down-regulated in phlorizin-treated TNF-α-stimulated FLSs. Conclusion: Phlorizin protects against inflammation and reduces injury to synovial tissues in RA by modulating the AKT/PI3K/mTOR pathway.
Rheumatoid arthritis (RA) is a prevalent autoimmune disorder affecting approximately 1% of the global populace (1). It is a systemic inflammatory disorder that involves lesions in multiple organs. Destruction of bone and articular cartilage, pannus formation, and synovial inflammation leads to loss of function associated with bone deformities, which are the clinical features of RA (2). The progression of RA does not include a unique etiology. However, apoptosis, hyperplasia of the synovium, and fibroblast-like synoviocytes (FLSs) contribute to RA. FLSs are responsible for the production of cathepsin, matrix metallopeptidase (MMP), proinflammatory factors, and chemokines. These substances contribute to the infiltration of immune cells into the joint and the subsequent degradation of cartilage and the extracellular matrix. Additionally, the production of these substances may be triggered by exposure to cytokines and growth factors (3).
A proteomics study on the pathogenesis of synovial hyperplasia involved in the progression of RA revealed that eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and the 70-kDa ribosomal protein S6 kinase (P70S6K) proteins control cellular proliferation as they modulate ribosomes and affect the synthesis of proteins (2). The regulation of protein phosphorylation is mediated by the mechanistic target of rapamycin (mTOR), a serine/threonine kinase. mTOR functions as two different complexes, mTORC1 and mTORC2, phosphorylation of which activates the 4E-BP1 and P70S6K proteins, which in turn activate translation and lead to activation of the cellular proliferation process (4). This process is also triggered by heightened levels of cytokines, promoting the release of growth factors, and subsequently initiating proliferation. Thus, inhibition of synovial cell proliferation is an essential target that could be targeted through multiple pathways for the management of RA (5).
Secondary metabolites sourced from herbal medicine, specifically flavonoids, are used for the management of several chronic diseases (6). Phlorizin (Figure 1) is a flavonoid isolated from Lithocarpus polystachyus Rehd, which is traditionally used as medicine in China (7). Phlorizin is reported to have strong antidiabetic activity by inhibiting sodium-glucose co-transporter 1/a (8), which balances the level of glucose in multiple ways, as renal and intestinal absorption are down-regulated, and it also improves sensitivity to insulin. It has a beneficial effect against diabetic complications as it ameliorates alterations in lipid levels. Moreover, phlorizin has shown anti-inflammatory properties by regulating the level of inflammatory cytokines in lipopolysaccharide-induced animal models (9). Phlorizin has potent antioxidant and antibacterial capabilities, which are implicated in the pathogenesis of several chronic health problems. Based on this evidence, our study determined the phlorizin effect against RA.
Chemical structure of phlorizin.
Materials and Methods
Animals. A total of 30 healthy Wistar rats (male; aged 7-8 weeks; weight: 120-150 g) purchased from Shanghai SLAC Laboratory Animal Co., Ltd., (Shanghai, PR China) were used for this study and housed under standard conditions (55±5% humidity and 25±2°C) with 12-h light and dark cycles. All the animals were provided unrestricted access to food and hydration. The Second People’s Hospital of Nanyang City in Nanyang, China, approved experimental protocols on these animals (reg no. 2023/RA/758493).
Arthritis model. The animals were divided into two distinct groups: A control group (n=15) and a second group (n=15) which received bovine type II collagen (CII) (Sigma-Aldrich, Shanghai, PR China), to induce RA. CII was prepared at 2 mg/ml by dissolving it in 0.05 M acetic acid and mixing it at 4°C. Further, this solution was then mixed at a ratio of 1:1 with Freund’s incomplete adjuvant to prepare a 1 mg/ml solution. Animals were injected subcutaneously in the tail with this solution on the first day (300 μl) and day (200 μl), whereas the control group received injections with saline (200 μl).
Treatment strategy. The CII-treated animals were further split into three groups: the RA control group (n=5) received saline (200 μl); two RA groups (n=5 each), received phlorizin (Shaanxi Lonier Herb Bio-Technology Co., Ltd, PR China) at 60 or 120 mg/kg per os for 3 weeks from the 21st day after the induction of RA (i.e., total period of the protocol was 7 weeks).
Assessment of clinical signs of inflammation. The rats were weighed and monitored once a week for clinical signs of arthritis from immunization up to 50 days (10).
Estimation of arthritis score. The arthritis severity was assessed by independent researchers was scored 0-4 for each hindlimb as follows: No swelling or redness: 0; redness in the foot or swelling of the ankle: 1; redness and inflammation from midfoot to ankle: 2; entire foot swollen: 3; loss of mobility associated with a swollen foot: 4. The arthritic index was calculated as the total score of the four limbs of each animal (maximum total score=16). An arthritis index greater than 6 points is judged to indicate the successful induction of RA in the model (11). As the arthritis score only provides a subjective quantification of inflammation, it was coupled with the measurement of hind paw diameter using a digital caliper (Fischer Darex, Le Chambon-Feugerolles, France). The values were expressed as the mean of the two hind paw diameters. When indicated, values for individual hind paw diameters are presented.
The mean arthritis index of each group of mice was calculated by dividing the total arthritis score for all mice in a group by the number of mice in the group.
Determination of thymus and spleen index. At 50 days following the induction of RA, the rats were sacrificed. The thymus and spleen were resected from each animal and each of these organs was weighed. Thymus and spleen indices were estimated using the formula given in (12):
Determination of serum interleukin-1 β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α), nuclear factor kappa B (NF-κB), MMP-1 and MMP-3. After rats were sacrificed, TNF-α, IL-1β, IL-6, NFκB, MMP-1, and MMP-3 levels were assessed in the serum of the CII-treated rats by enzyme-linked immunosorbent assay as per the manufacturer’s kit instructions (ThermoFisher, Beijing, PR China) (10).
Histopathological study. Histopathological analysis was performed on sections of paw joints. Ankle joints were analyzed by removing the skin and muscle from the right limb and fixing the tissue in a polyoxymethylene solution (4%) for 1 week at 4°C after isolation. The tissue was subjected to decalcification using a 14% ethylenediaminetetraacetic acid (EDTA) solution, followed by fixation at a temperature of 4°C for 35 days. The joint tissue was dehydrated using alcohol and afterward embedded in paraffin. The tissue was then sectioned into slices with a thickness of 5 μm. The tissue sections were stained with hematoxylin and eosin, and a trinocular microscope (BA210 LED Trinocular; Motic, Wetzlar, Germany) was used for the observation of histopathological changes. All the histopathological alterations were noted quantitatively. The slides were analyzed and histopathologically scored as previously described by López-García et al. (13).
In-vitro cell culture. FLSs were obtained from the American Type Culture Collection (Manassas, VA, USA) and cultured in medium containing 10% fetal bovine serum. The cells were then washed three times using phosphate-buffered saline solution, followed by treatment with trypsin (0.25%) for the purpose of digestion. The digestive process was ended addition of 10% fetal bovine serum. Cells were cultured in an incubator with CO2 at 37°C using Dulbecco’s modified Eagle’s medium/F12 medium.
Determination of cell proliferation. The effect of phlorizin on cellular proliferation induced by TNFα in FLSs was assessed in vitro. Cells (5×103) were seeded onto a 24-well plate and exposed to several doses of phlorizin (25, 50, 75, and 100 μM) for 2 h, TNFα (10 ng/ml) was then used to stimulate the cells for 2 days. CCK-8 solution (10 μl) was then added to each well and plates were incubated for 2 h at 37°C. The absorbance at 450 nm was finally measured using a microplate analyzer. Assessment of FLS proliferation was observed with 5-ethynyl-2-deoxyuridine (EdU); cells were incubated for 2 days. The cells were then fixed using paraformaldehyde, and staining was conducted using an Apollo® staining reaction solution. Further, Hoechst 33342 stain was applied to stain the nucleus, and cellular images were taken with a high-resolution imaging system (EVOS XL Cell Imaging System, Invitrogen™ AMEX1000; ThermoFisher Scientific, Shanghai, PR China). This experiment was conducted three times.
Assessment of cellular apoptosis. Cell apoptosis was determined using a commercial fluorescein isothiocyanate (FITC) Annexin V Apoptosis Detection Kit (BD Pharmingen, San Diego, CA, USA) with annexin V-allophycocyanin/propidium iodide double staining, according to the manufacturer’s instructions. A flow cytometer was used to determine the total number of apoptotic cells.
Immunofluorescence staining. The estimation of microtubule-associated protein light chain 3 (LC3B) expression in FLSs was performed using immunofluorescence staining and immunofluorescence microscopy. Cells were stimulated for 2 days with 50 ng/ml TNFα (Sigma-Aldrich, and fixed for 30 min with para-formaldehyde (4%). Cells were incubated at 4°C with antibody to LC3B (2 μl; Thermo Fisher Scientific, Shanghai, China), overnight, and later incubated with a secondary antibody. Images were then captured after staining the cells with 4’,6-diamidino-2-phenylindole Finally, a quantitative assessment was conducted on the cells.
Western blot assay. The synovial tissues were obtained from all the animals, and protein was extracted from the tissue. The protein concentration was then determined using a bicinchoninic acid assay. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (8-15%) was used to separate the proteins, which were transferred to a nitrocellulose filter membrane (0.22 μm). The membrane was incubated at 4°C overnight with primary antibodies targeting protein kinase B (AKT), phospho-AKT (p-AKT), mTOR, p-mTOR, phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), p-PI3K (ThermoFisher); hypoxia-inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor (VEGF), light chain 3 II (LC3 II), light chain 3 I (LC3 I) and β-actin (Sigma-Aldrich). Following this, the samples were treated for an additional hour with secondary antibodies. An Odyssey Infrared Imaging System, (LI-COR Biosciences, Lincoln, NE, USA) was used to observe the membrane, while ImageJ (National Institutes of Health, Bethesda, MA, USA) tool was deployed for quantitative analysis of the blots relative to standard blots.
Statistical analysis. The data underwent statistical assessment using SPSS software (Version 19.0; IBM, Armonk, NY, USA). The data are reported as the mean±standard deviation. The ratios were compared using chi-square analysis. A one-way analysis of variance (ANOVA) was performed to compare the means of the two groups. Subsequently, the Fisher least significant difference test was conducted to determine significant differences between the groups. The statistical threshold was set at p<0.05.
Results
Effect of phlorizin on body weight of rats. In this research, the body weight of rats with CII-induced RA was assessed every week after treatment with phlorizin. The resulting data are illustrated in Figure 2. During the first 4-week phase of RA induction with CII treatment (phlorizin 60 and 120 mg/kg) and RA control groups, animals did not exhibit many variations in body weight, but the weight of the control group significantly increased. After the completion of the weeks 4-7 of therapy with phlorizin 60 and 120 mg/kg, there was a significant increase in body weight. At the same time, the mean weight of the control RA group moderately increased. The results demonstrated that the administration of phlorizin had a significantly beneficial effect on regulating the body weight of the animals.
Assessment of the effect of phlorizin on body weight of rats with bovine type II collagen-induced rheumatoid arthritis (RA). The experiment was conducted thrice. Data are presented as the mean±standard deviation.
Effect of phlorizin on joint inflammation. Inflammation of joints was assessed by determining the arthritic score and hind paw edema volume in phlorizin-treated CII-induced RA rats. During the first 2-week period, no significant changes in hind paw edema volume were observed in the RA treatment groups or the control RA group. However, from weeks 2-4, a significant rise in the volume of hind paw edema was noted in the RA control and treatment groups. From weeks 4-7, the volume of hind paw edema decreased significantly in the phlorizin-treated groups (Figure 3A). This study reveals that the treatment with phlorizin in experimental groups led to a substantial reduction in hind paw edema volume compared to the control RA group. Furthermore, the reduction in edema volume was shown to be dependent on the dosage of phlorizin administered.
Assessment of the effects of phlorizin on hind paw inflammation (A) and arthritis scores (B) of joints of rats with bovine type II collagen-induced rheumatoid arthritis (RA). Data are presented as the mean±standard deviation.
In this study, the arthritis score was assessed in phlorizin-treated groups and compared to the control group (Figure 3B). In the first week, arthritic scores of 0.5±0.2 were noted for the phlorizin-treated groups and the control RA group. However, the arthritis scores significantly increased from the first to the fourth week in the phlorizin-treated groups. From weeks 4 to 7, the treatment groups (60 mg/kg and 120 mg/kg) exhibited significant reductions in arthritis scores, as did the control RA group and control group. The results demonstrated that animals treated with phlorizin had a significant decrease in arthritis scores after the fourth week of the investigation. Following the completion of week 7, there is a notable similarity between the arthritis scores of the tested animals and those of the control group.
Effect of phlorizin on thymus and spleen indices. Thymus and spleen indices were estimated in phlorizin-treated rats with CII-induced RA (Figure 4). In the control group of rats with RA, the thymus index and spleen indices were significantly higher than the control group. The phlorizin-treated groups had significantly higher thymus indices and spleen indices compared to the control group afflicted with RA. The results of this study demonstrated that the administration of phlorizin efficiently reduced the thymus and spleen indices, consequently restoring them to normal levels in the animals.
Assessment of the effects of phlorizin on thymus and spleen indices in rats with bovine type II collagen-induced rheumatoid arthritis (RA). *Significantly different at p<0.05 from the Control group.
Effect of phlorizin on mediators of inflammation. The levels of inflammatory mediators were determined in the serum of phlorizin-treated rats with RA (Figure 5). The serum of rats in the RA groups, specifically those administered doses of 60 and 120 mg/kg, demonstrated a significant rise in the levels of inflammatory mediators. IL-1β IL-6, TNF-α, NF-κB, MMP-1 and MMP-3 compared with the control group values. The findings of this research indicate that treatment with phlorizin significantly reduced levels of inflammatory mediators, hence returning them to baseline levels in the animal subjects.
Assessment of the effects of phlorizin on inflammatory mediators in the serum of rats with bovine type II collagen-induced rheumatoid arthritis (RA). There were five animals in each group (n=5). IL-1: Interleukin; TNF-α: tumor necrosis factor-α; NF-κB: nuclear factor kappa B; MMP: matrix metallopeptidase. *Significantly different at p<0.05 from the Control group.
Effect of phlorizin on proteins involved in regulation of the AKT/PI3K/mTOR pathway. As shown in Figure 6, the evaluation of AKT/PI3K/mTOR protein expression in the experimental rat groups served to determine the effect of phlorizin on the mTOR pathway. Proteins of the AKT/PI3K/mTOR pathway were up-regulated in groups with RA. The up-regulation of these proteins was seen to be significantly lessened in the synovial tissue of the groups that were treated with phlorizin.
Assessment of the effects of phlorizin on the expression of AKT serine/threonine kinase 1 (AKT)/phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/mechanistic target of rapamycin kinase (mTOR) protein in synovial tissue of rats with bovine type II collagen-induced rheumatoid arthritis (RA). GAPDH: Glyceraldehyde 3-phosphate dehydrogenase; HIF-1α: hypoxia-inducible factor 1α; p-: phosphorylated; VEGF: vascular endothelial growth factor. *Significantly different at p<0.05 from the Control group.
Effect of phlorizin on histopathology. The histopathological alterations in the ankle joint of rats with RA generated by CII were evaluated using the hematoxylin and eosin staining technique, as seen in Figure 7. The control RA group had changes in their histopathology, including necrosis and degenerative changes in the articular cartilage, as well as the presence of inflammatory cells and hyperplasia in the synovial joints. A higher level of alterations was noted in the control RA rats than in the groups treated with phlorizin. The findings of this study demonstrate that the administration of phlorizin had a significant impact and ameliorated the histopathological effects of RA in these animals.
Assessment of the effects of phlorizin on histopathology of ankle joints in the bovine type II collagen-induced rat model of rheumatoid arthritis (RA). *Significantly different at p<0.05 from the Control group.
Effect of phlorizin on cell proliferation. Proliferation of FLSs was determined by the CCK-8 assay and EdU staining, as shown in Figure 8A and B. There was a significant increase in the proliferation of FLSs when stimulated by TNF-α and phlorizin significantly reduced by treatment with phlorizin. Moreover, the effect of phlorizin on the apoptosis of FLSs was estimated using flow cytometry. There was no significant alteration in apoptosis of phlorizin-treated FLSs (Figure 8C).
Assessment of the effects of phlorizin on the cellular proliferation of tumor necrosis factor-α stimulated fibroblast-like synoviocytes. A: Cellular proliferation as determined by Cell Counting Kit-8 (CCK-8) assay. B: Cellular proliferation by 5-ethynyl-2-deoxyuridine (EdU) assay. C: Percentage of apoptotic cells by flow cytometry. *Significantly different at p<0.05 from the TNF-α group.
Effect of phlorizin on autophagy-related proteins. LC3 protein, which is known to play a role in autophagy, was quantified by determination of LC3 fluorescence intensity in FLSs that were activated with TNF-α and treated with phlorizin at different concentrations (25, 50, 75, and 100 μM). The immunohistochemistry data demonstrated that fluorescence intensity was phlorizin concentration-dependent (Figure 9A). The expression of p-AKT/AKT, p-PI3K/PI3K, p-mTOR/mTOR, and LC3II/LC3I proteins were significantly reduced by treatment with 100 μM of phlorizin (Figure 9B).
Assessment of the effects of phlorizin on the cellular autophagy of tumor necrosis factor-α (TNF-α)-stimulated fibroblast-like synoviocytes. A: Fluorescence intensity of microtubule-associated protein light chain 3 (LC3) by immunohistochemistry. B: Expression of AKT serine/threonine kinase 1 (AKT), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), mechanistic target of rapamycin (m-TOR), and LC3 II/LC3 I by western blot assay. Data are presented as the mean±standard deviation. *Significantly different at p<0.05 from the TNF-α group.
Discussion
RA is a persistent autoimmune condition, and there is no proper treatment available for its management. Phlorizin is reported to have anti-inflammatory properties as it reduces inflammatory cytokines (13). Therefore, the current research aimed to assess the impact of phlorizin in the treatment of RA. The effects of phlorizin were evaluated in an CII-induced rat model of RA (in vivo) and in TNF-α-stimulated FLSs (in vitro). Its effect was assessed on the level of inflammatory cytokines and the expression of AKT, PI3K, and mTOR proteins in RA models.
RA is an inflammatory disorder responsible for the development of edema in different body joints. These changes lead to the development of joint movement disorders, due to which patients suffering from RA find it difficult to make movements (14). CII promotes injury to joint tissue in this rat model, inducing RA as it leads to inflammation of the joints and increases the arthritic score (15). The data of our study also support this and showed treatment with phlorizin significantly reduced inflammation and arthritic scores in rats with CII-induced RA; moreover, it also improved body weight in these rats.
The spleen and thymus are the primary organs involved in the process of inflammation, as they are related to immune cell function, and during inflammation, are associated with activation of the immune system, and proliferation in these organs is related to an increase in their weight (16, 17). These changes increase the spleen and thymus index, and the present study also supports this in rats with RA. Effective management of RA is achieved by reducing the weight of the thymus and spleen, and the result of our experiment reveals that treatment with phlorizin significantly reduced the weight of these organs.
Inflammatory changes in RA occur due to a rise in the level of inflammatory cytokines; these cytokines are responsible for the further development of inflammation. TNF-α and NF-κB play a significant role in the process of inflammation in RA, as TNF-α and other cytokines activate NF-κB, which promotes the expression of MMP proteins (18). These promote the proliferation, differentiation, and survival of synovial tissue. Our in-vivo investigations demonstrated an elevation in cytokine levels in rats with CII-induced RA. Our in-vitro studies revealed a notable increase in the proliferation of TNF-α-stimulated FLSs. Effective management of RA is achieved by reducing the level of these cytokines and the proliferation of synovial tissues, and treatment with phlorizin indeed significantly reduced the level of cytokines and the proliferation of synovial tissue in rats with RA. Our results reveal that phlorizin had no effect on apoptosis in TNF-α-stimulated FLSs.
Several signaling pathways play a crucial role in the survival and proliferation of cells, one of which is the PI3K/AKT/mTOR pathway (19). This pathway is reported to be involved in the development of pathological conditions like RA, and agents that inhibit these pathways are considered promising for clinical application (20). Autophagy is also regulated by the PI3K/AKT/mTOR pathway, and its inhibition regulates the death and survival of cells. Autophagy appears to occur via the down-regulation of AKT, PI3K, and mTOR proteins. Additionally, autophagy of synovial tissue was shown to be induced under circumstances resembling RA (21). Our study’s data provide support for this assertion since therapy with phlorizin effectively mitigated the aberrant expression of the PI3K/AKT/mTOR pathway.
Conclusion
In summary, the findings of this research demonstrate the favorable impact of phlorizin on RA via modulation of the PI3K/AKT/mTOR pathway. This mechanism effectively mitigated inflammation and suppressed the growth of synovial tissue in rats with RA.
Acknowledgements
The Authors express their gratitude for the resources and support supplied by the institution’s management.
Footnotes
Authors’ Contributions
The animal experiments were performed by Xiangkun Wu and Quanhui Wan. Cell line-based experiments were carried out by Quanhui Wan and Yuqiang Yang. The research investigation was designed and supervised by Liuyu Wang. Statistical analysis was performed by Chaojie Gao.
Conflicts of Interest
The Authors state that there are no conflicts of interest related to this study.
- Received August 19, 2023.
- Revision received October 6, 2023.
- Accepted November 13, 2023.
- Copyright © 2024 The Author(s). Published by the International Institute of Anticancer Research.
This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).