Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Biomed Pharmacother ; 143: 112160, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34560546

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease. Bu-Shen-Tong-Du prescription (BSP) has traditionally been used in to treat RA but its underlying mechanisms remain unclear. In this study, we explored the potential mechanisms of BSP in collagen-induced arthritis (CIA) rats, a classic animal model of RA. We employed an integrated pharmacology approach in combination with network pharmacology, 1H-nuclear magnetic resonance (NMR) metabolomics, and biochemical analyses to determine the mechanisms of BSP for treating RA. We found that BSP can regulate immunity and inflammation by decreasing the spleen index; inhibiting hyperplasia of the white pulp; reducing the levels of IL-1ß, IL-6, IL-17A, and IFN-γ; and increasing the levels of IL-10 in the serum. Network pharmacology was utilized to predict related signal transduction pathways of BSP in RA treatment. 1H NMR metabolomics of the serum confirmed that BSP regulated energy metabolism and amino acid metabolism. Finally, we validated the Toll-like receptor 4 (TLR4)/nuclear factor (NF)-κB signaling pathway using immunohistochemical methods, which demonstrated that BSP controlled RA-induced inflammation by inhibiting the TLR4/NF-κB signaling pathway. These results confirm the therapeutic effect of BSP in a CIA rat model, which is exerted via the inhibition of the inflammation and the improvement of the immune function, balancing energy metabolism and amino acid metabolism, and inhibiting the TLR4/NF-κB signaling pathway. This study provides an experimental basis for using BSP as a combinatorial drug to inhibit inflammation and regulate immunity in the treatment of RA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Drugs, Chinese Herbal/pharmacology , Joints/drug effects , Network Pharmacology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Collagen Type II , Cytokines/metabolism , Energy Metabolism/drug effects , Joints/immunology , Joints/metabolism , Joints/pathology , Male , Medicine, Chinese Traditional , NF-kappa B/metabolism , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 4/metabolism
2.
Int Immunopharmacol ; 100: 108107, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34482265

ABSTRACT

Activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome plays a crucial role in the inflammatory responses of monosodium urate (MSU) crystal-induced gouty arthritis. Therefore, the molecular basis of NLRP3 inflammasome is very valuable in developing potential therapeutic drugs for gout. Tetrahydropalmatine (THP), the main active component of the traditional Chinese medicinal herb Corydalis yanhusuo, has shown prominent anti-inflammatory and analgesic activities, but to date, these effects have not been investigated exhaustively on gout. This study indicated that THP attenuated pain and swelling in an MSU-induced acute gout model by decreasing pro-inflammatory cytokine production and inflammatory cell infiltration. THP exerted its actions by suppressing NLRP3 inflammasome activation and subsequent formation of caspase-1. Furthermore, results showed that THP alleviated MSU-induced reactive oxygen species (ROS) generation, upstream of NLRP3 inflammasome activation, by an increase in the activities of antioxidant enzymes both in vitro and in vivo. In conclusion, our study suggests that THP suppressed ROS-mediated NLRP3 inflammasome activation in MSU-induced inflammatory responses, which highlights its therapeutic potential in gouty arthritis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Arthritis, Gouty/prevention & control , Berberine Alkaloids/pharmacology , Inflammasomes/metabolism , Joints/drug effects , Macrophages/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Analgesics/pharmacology , Animals , Arthritis, Gouty/chemically induced , Arthritis, Gouty/immunology , Arthritis, Gouty/metabolism , Caspase 1/metabolism , Cytokines/metabolism , Disease Models, Animal , Joints/immunology , Joints/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , RAW 264.7 Cells , Signal Transduction , Uric Acid
3.
Pharmacol Res ; 170: 105696, 2021 08.
Article in English | MEDLINE | ID: mdl-34052360

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease which affects about 0.5-1% of people with symptoms that significantly impact a sufferer's lifestyle. The cells involved in propagating RA tend to display pro-inflammatory and cancer-like characteristics. Medical drug treatment is currently the main avenue of RA therapy. However, drug options are limited due to severe side effects, high costs, insufficient disease retardation in a majority of patients, and therapeutic effects possibly subsiding over time. Thus there is a need for new drug therapies. Endoplasmic reticulum (ER) stress, a condition due to accumulation of misfolded proteins in the ER, and subsequent cellular responses have been found to be involved in cancer and inflammatory pathologies, including RA. ER stress protein markers and their modulation have therefore been suggested as therapeutic targets, such as GRP78 and CHOP, among others. Some current RA therapeutic drugs have been found to have ER stress-modulating properties. Traditional Chinese Medicines (TCMs) frequently use natural products that affect multiple body and cellular targets, and several medicines and/or their isolated compounds have been found to also have ER stress-modulating capabilities, including TCMs used in RA treatment by Chinese Medicine practitioners. This review encourages, in light of the available information, the study of these RA-treating, ER stress-modulating TCMs as potential new pharmaceutical drugs for use in clinical RA therapy, along with providing a list of other ER stress-modulating TCMs utilized in treatment of cancers, inflammatory diseases and other diseases, that have potential use in RA treatment given similar ER stress-modulating capacity.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Drugs, Chinese Herbal/therapeutic use , Endoplasmic Reticulum Stress/drug effects , Joints/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Drugs, Chinese Herbal/adverse effects , Humans , Joints/immunology , Joints/metabolism , Medicine, Chinese Traditional
4.
Biomed Pharmacother ; 134: 111166, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33373915

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease. Strong evidence supports that excessive activation of B cells plays a critical role in the pathogenesis of RA. Fc gamma receptor b (FcγRIIb) is the B cell inhibitory receptor and inhibits BCR (B cell receptor) signalling in part by selectively dephosphorylating CD19 which is considered a co-receptor for BCR and is essential for B cell activation. Our previous study demonstrated that a FcγRIIb I232T polymorphism presented a strong genetic link to RA and may lead to the excessive activation of B cells. Therefore, novel therapeutic strategies and drugs that can effectively inhibit the excessive activation of B cells by regulating the FcγRIIb are necessary for the treatment of RA. Therefore, we used Burkitt's lymphoma ST486 human B cells (lacking endogenous FcγRIIb) transfected with the 232Thr loss-of-function mutant to construct a FcγRIIb mutant cell line (ST486), and we demonstrated that YSTB treatment not only reduced proliferation and promoted apoptosis in ST486 cells but also did so in a dose-dependent manner. Furthermore, the intracellular Ca2+ flux of ST486 cells was decreased after treatment with YSTB, inhibiting the excessive activation of ST486 cells, and these effects correlated with the CD19/FcγRIIb-Lyn-SHP-1 pathways. Our data showed that YSTB treatment inhibited the expression of phosphorylated CD19 and upregulated the protein expression of FcγRIIb, Lyn, and SHP-1. Additionally, the CIA model was established to explore the anti-inflammatory and inhibitory effects of YSTB on bone destruction, and we found that YSTB decreased the paw oedema and arthritis index (AI) in CIA rats. It is worth mentioning that YSTB clearly decreased the AI earlier than methotrexate (MTX) (day 10 vs 16). Moreover, synovial hyperplasia, inflammatory cell infiltration and cartilage surface erosion in CIA rats were noticeably reduced after treatment with YSTB as evidenced by histopathological examination. Finally, we found that YSTB treatment suppressed bone erosion and joint space score (JNS) in CIA rats as evidenced by radiographic assessment. In summary, these data suggest that YSTB has great therapeutic potential for RA treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/prevention & control , B-Lymphocytes/drug effects , Drugs, Chinese Herbal/pharmacology , Joints/drug effects , Lymphocyte Activation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, IgG/metabolism , src-Family Kinases/metabolism , Animals , Apoptosis/drug effects , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen Type II , Female , Humans , Joints/immunology , Joints/metabolism , Joints/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Rats, Wistar , Receptors, IgG/genetics , Signal Transduction , src-Family Kinases/genetics
5.
Biomed Pharmacother ; 133: 111039, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33254019

ABSTRACT

Assessment of the potential therapeutic benefits offered by naturally occurring phytoestrogens necessitate inspection of their potency and sites of action in impeding the chronic, systemic, autoimmune, joint destructing disorder Rheumatoid arthritis (RA). Possessing structural and functional similarity with human estrogen, phytoestrogen promisingly replaces the use of hormone therapy in eradicating RA symptoms with their anti-inflammatory, anti-oxidative, anti-proliferative, anti-angiogenesis, immunomodulatory, joint protection properties abolishing the harmful side effects of synthetic drugs. Scientific evidences revealed that use of phytoestrogens from different chemical categories including flavonoids, alkaloids, stilbenoids derived from different plant species manifest beneficial effects on RA through various cellular mechanisms including suppression of pro-inflammatory cytokines in particular tumor necrosis factor (TNF-α), interleukin(IL-6) and nuclear factor kappa B (NF-κB) and destructive metalloproteinases, inhibition of oxidative stress, suppressing inflammatory signalling pathways, attenuating osteoclastogenesis ameliorating cartilage degradation and bone erosion. This review summarizes the evidences of different phytoestrogen treatment and their pharmacological mechanisms in both in vitro and in vivo studies along with discussing clinical evaluations in RA patients showing phytoestrogen as a promising agent for RA therapy. Further investigations and more clinical trials are mandatory to clarify the utility of these plant derived compounds in RA prevention and in managing oestrogen deficient diseases in patients.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Inflammation Mediators/antagonists & inhibitors , Joints/drug effects , Phytoestrogens/therapeutic use , Animals , Anti-Inflammatory Agents/adverse effects , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Evidence-Based Medicine , Humans , Inflammation Mediators/metabolism , Joints/immunology , Joints/metabolism , Phytoestrogens/adverse effects , Signal Transduction , Treatment Outcome
6.
Mol Biol Rep ; 48(1): 969-974, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33289909

ABSTRACT

Haemophilic arthropathy (HA), caused by intra-articular haemorrhage, is one of the most common complications in patients with haemophilia. Factor replacement therapy provides missing coagulation factors to prevent children with haemophilia from joint bleeding and decreases their risk for HA. However, haemophilia patients in developing countries are still suffering from HA due to insufficient replacement therapy. Symptoms such as pain and activity limitations caused by HA seriously affect the functional abilities and quality of life of patients with HA, causing a high disability rate in the haemophilia cohort. The pathological mechanism of HA is complicated because the whole pathological mainly involves hypertrophic synovitis, osteopenia, cartilage and bone destruction, and these pathological changes occur in parallel and interact with each other. Inflammation plays an important role in the whole complex pathological process, and iron, cytokines, growth factors and other factors are involved. This review summarizes the pathological mechanism of HA to provide background for clinical and basic research.


Subject(s)
Arthritis/pathology , Bone Diseases, Metabolic/pathology , Hemarthrosis/pathology , Hemophilia A/pathology , Osteonecrosis/pathology , Synovitis/pathology , Adult , Arthritis/genetics , Arthritis/immunology , Arthritis/metabolism , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/immunology , Bone Diseases, Metabolic/metabolism , Child , Cytokines/genetics , Cytokines/immunology , Factor VIII/therapeutic use , Gene Expression Regulation , Hemarthrosis/genetics , Hemarthrosis/immunology , Hemarthrosis/metabolism , Hemophilia A/genetics , Hemophilia A/immunology , Hemophilia A/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Iron/immunology , Iron/metabolism , Joints/immunology , Joints/metabolism , Joints/pathology , Osteonecrosis/genetics , Osteonecrosis/immunology , Osteonecrosis/metabolism , Quality of Life , Synovitis/genetics , Synovitis/immunology , Synovitis/metabolism
7.
Immunol Lett ; 228: 112-121, 2020 12.
Article in English | MEDLINE | ID: mdl-33137380

ABSTRACT

Disease-modifying osteoarthritis (OA) therapy is not yet available. Several adjuvant therapies have demonstrated promising results in the treatment of OA. The present study aimed to investigate the therapeutic effects and underlying mechanisms of a combination of Lactobacillus acidophilus, vitamin B, and curcumin in the treatment of OA. Monosodium iodoacetate (MIA)-induced arthritis of the knee joint in rat was used as an animal model of human OA. The combination of L. acidophilus LA-1, vitamin B, and curcumin or a saline solution was given orally. Pain was measured according to the paw withdrawal latency, and paw withdrawal threshold. Cartilage destruction was analyzed using histomorphological techniques and the Mankin scoring system. Protein expression in the joint was examined using immunohistochemistry. The effects of the combination of L. acidophilus LA-1, vitamin B, and curcumin on mRNA levels in chondrocytes stimulated with interleukin (IL)-1ß were analyzed using real-time polymerase chain reaction. The combination of L. acidophilus, vitamin B, and curcumin effectively downregulated Th17 cells and the related cytokine IL-17, thereby maintained the Treg population, and increased the expression of the Treg-related cytokine IL-10 in human peripheral blood mononuclear cells. The OA animal model exhibited reduced pain and preservation of cartilage in response to the combination treatment. The expression levels of pro-inflammatory cytokines and the catabolic, matrix metalloproteinase-13 (MMP-13), were decreased, whereas the expression of the anabolic tissue inhibitors of metalloproteinases (TIMPs) were upregulated in response to the drug combination. The combination of L. acidophilus, vitamin B, and curcumin was beneficial in OA treatment, controlling the inflammatory response via regulation of the Th17/Treg population and reducing the expression of pro-inflammatory cytokines in human peripheral blood mononuclear cells. The combination treatment also preserved cartilage, suppressed osteoclastogenesis, and regulated the anabolic/catabolic imbalance. These findings indicate the therapeutic potential of combination use of L. acidophilus, vitamin B, and curcumin in patients with OA.


Subject(s)
Antirheumatic Agents/pharmacology , Curcumin/pharmacology , Cytokines/metabolism , Inflammation Mediators/metabolism , Joints/drug effects , Lactobacillus acidophilus/physiology , Osteoarthritis/drug therapy , Probiotics/pharmacology , Vitamin B Complex/pharmacology , Adult , Animals , Cells, Cultured , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Joints/immunology , Joints/metabolism , Joints/pathology , Male , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Osteoarthritis/immunology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteogenesis/drug effects , Rats, Wistar , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism
8.
Front Immunol ; 11: 1932, 2020.
Article in English | MEDLINE | ID: mdl-32973796

ABSTRACT

Cationic host defense peptides (CHDP) are immunomodulatory molecules that control infections and contribute to immune homeostasis. CHDP such as cathelicidin and calprotectin expression is altered in the arthritic synovium, and in the lungs of asthma and COPD patients. Recent studies suggest a link between airway inflammation and the immunopathology of arthritis. Therefore, in this study we compared the abundance of mouse cathelicidin (CRAMP), defensins, and calprotectin subunits (S100A8 and S100A9) in murine models of collagen-induced arthritis (CIA) and allergen house dust mite (HDM)-challenged airway inflammation. CRAMP, S100A8, and S100A9 abundance were significantly elevated in the joint tissues of CIA mice, whereas these were decreased in the lung tissues of HDM-challenged mice, compared to naïve. We further compared the effects of administration of two different synthetic immunomodulatory peptides, IG-19 and IDR-1002, on cathelicidin and calprotectin abundance in the two models. Administration of IG-19, which controls disease progression and inflammation in CIA mice, significantly decreased CRAMP, S100A8, and S100A9 levels to baseline in the joints of the CIA mice, which correlated with the decrease in cellular influx in the joints. However, administration of IDR-1002, which suppresses HDM-induced airway inflammation, did not prevent the decrease in the levels of cathelicidin and calprotectin in the lungs of HDM-challenged mice. Cathelicidin and calprotectin levels did not correlate with leukocyte accumulation in the lungs of the HDM-challenged mice. Results of this study suggest that endogenous cathelicidin and calprotectin abundance are disparately altered, and may be differentially regulated, within local tissues in airway inflammation compared to arthritis.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Arthritis, Experimental/metabolism , Asthma/metabolism , Joints/metabolism , Leukocyte L1 Antigen Complex/metabolism , Leukocytes/metabolism , Lung/metabolism , Allergens , Animals , Antigens, Dermatophagoides , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Asthma/chemically induced , Asthma/drug therapy , Asthma/immunology , Calgranulin A/metabolism , Calgranulin B/metabolism , Collagen Type II , Female , Immunologic Factors/pharmacology , Joints/drug effects , Joints/immunology , Leukocytes/drug effects , Leukocytes/immunology , Lipopolysaccharides , Lung/drug effects , Lung/immunology , Male , Mice, Inbred BALB C , Mice, Inbred DBA , Cathelicidins
9.
J Ethnopharmacol ; 261: 113022, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-32569719

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Traditional plant-derived medicines have enabled the mankind in curing the wide spectrum of diseases throughout the ages. Andrographis paniculata (Burm.f.) Nees, is one of the traditional plant used as a folk medicine for the management of inflammation, arthritis, viral-bacterial infections and other ailments in India, China, Malaysia and other South-East Asian countries. Its major bioactive compound; andrographolide, a diterpenoid, also exerts cytoprotective properties and is reported to be effective in neuroprotection, hepatoprotection, etc. AIM: The study is aimed to explore the role of andrographolide in treatment of complete freund's adjuvant (CFA) induced arthritis. MATERIALS AND METHODS: The influx of immune cells, release of pro-inflammatory cytokines and subsequent accumulation of synovial fluid (swelling) and pain manifest into the disease. The present study used CFA induced Balb/c mice model and treated them intraperitoneally with andrographolide and dexamethasone (used as a positive control) on alternate days for six days. After 6 days, blood and peritoneal macrophages were collected to evaluate the expression of various arthritic markers and paw edema was measured on all days. RESULTS: The in vitro and ex vivo experiments showed that andrographolide treated animal group had reduced paw edema, cell cytotoxicity and nitric oxide production than dexamethasone treated animal group. Further, the study revealed the mechanistic role of andrographolide in treatment of arthritis by suppressing battery of molecules like COX-2, NF-κB, p-p38, CD40, TNF-α, IL-1ß and IL-6 involved in arthritis. CONCLUSION: The study showed the potent anti-arthritic effects of andrographolide and warrants further investigations on andrographolide for the development of safe and effective anti-arthritic drug.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antirheumatic Agents/pharmacology , Cytokines/antagonists & inhibitors , Diterpenes/pharmacology , Inflammation Mediators/antagonists & inhibitors , Joints/drug effects , Macrophages, Peritoneal/drug effects , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Cells, Cultured , Cytokines/metabolism , Female , Freund's Adjuvant , Inflammation Mediators/metabolism , Joints/immunology , Joints/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred BALB C , Signal Transduction
10.
Biochem Biophys Res Commun ; 522(3): 743-748, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31791581

ABSTRACT

Rheumatoid arthritis is a chronic and systemic autoimmune disease, which affects approximately 1% of the adult population worldwide. The present study investigated the therapeutic effect of theacrine (TC) on arthritis and its mechanisms in Freund's incomplete adjuvant (FIA)-induced SD rats. Rats were randomly divided into 5 groups: i) healthy control; ii) model; iii) positive control with methotrexate (MTX); iv) treatment with 12.5 mg/kg TC; and v) treatment with 25.0 mg/kg TC. The apparent scores, including changes in body weights, degree of paw swelling and arthritis indicators, were analyzed to evaluate the anti-chronic inflammatory effect of TC. The levels of interleukin (IL)-6 and transforming growth factor-ß (TGF-ß) in serum were measured by enzyme-linked immunosorbent assay. The protein and RNA expression levels of the critical factors in rats were measured to elucidate the mechanisms responsible for chronic inflammation and to verify molecular indexes of chronic inflammatory conditions. TC notably suppressed the severity of FIA-induced rat by attenuating the apparent scores, animal weight and inflammatory indexes in the 25 mg/kg TC group compared with the FIA rat model. Furthermore, TC significantly decreased the levels of IL-6 and increased the levels of TGF-ß. Histopathological examinations indicated that TC rescued the synovial hyperplasia and inflammatory cell infiltration in joint tissues. In addition, TC enhanced TGF-ß-mediated shifts in inflammatory marker expression in joint tissue. Overall, the present study demonstrated that TC exerted a superior anti-arthritic effect via the suppression of IL-6 and the activation of TGF-ß by the TGF-ß/SMAD pathway.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Smad Proteins/immunology , Transforming Growth Factor beta/immunology , Uric Acid/analogs & derivatives , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Chronic Disease , Freund's Adjuvant , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Joints/drug effects , Joints/immunology , Joints/pathology , Lipids , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Smad Proteins/analysis , Transforming Growth Factor beta/analysis , Uric Acid/therapeutic use
11.
Int Immunopharmacol ; 72: 292-300, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31005039

ABSTRACT

Curcumin is a natural herbal product that has been popularly used to treat autoimmune diseases in China; however, its effects on rheumatoid arthritis and its mechanism are not clear. The main purposes of this study are to explore the therapeutic effects of curcumin on collagen-induced arthritis (CIA) rats and the pharmacological mechanism. In the present study, CIA rats were established by injecting bovine type II collagen. Curcumin and methotrexate were then orally administered daily, and the swelling degree of the hind limb joints was scored every two days. Histopathological changes were observed by hematoxylin-eosin staining. The levels of cytokines (TNF-α, IL-1ß, IL-17 and TGF-ß) were detected by radioimmunoassay, while the expression of IκBα and COX-2 was detected by Western blot. In addition, cell viability was detected by CCK-8 assay, and the effect of curcumin on macrophage apoptosis was detected by flow cytometry and TUNEL assay. The results indicated that in vivo curcumin attenuated the degree of joint swelling of rats and the further development of joint histopathology. Moreover, it downregulated the levels of cytokines. In vitro curcumin inhibited the degradation of IκBα and reduced the production of COX-2 in LPS-induced inflammatory RAW264.7 cells. Importantly, curcumin significantly induced macrophage apoptosis. In conclusion, in this study, we have demonstrated that curcumin exerts therapeutic effects on arthritis in CIA rats and has a strong pharmacological activity on reducing the inflammatory response in macrophages. Its mechanism may be related to the inhibition of the NF-κB signaling pathway and the promotion of macrophage apoptosis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Curcumin/therapeutic use , Animals , Apoptosis/drug effects , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cell Survival/drug effects , Cytokines/immunology , Joints/drug effects , Joints/immunology , Joints/pathology , Male , Mice , RAW 264.7 Cells , Rats, Sprague-Dawley
12.
Arch Immunol Ther Exp (Warsz) ; 67(3): 153-160, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30874838

ABSTRACT

Rheumatoid arthritis (RA) is a common autoimmune disease which impacts a large number of patients worldwide, and new drugs are required for lower the disease burden. Theaflavin-3, 3'-digallate (TFDG) is polyphenol exhibiting inhibition on inflammatory factors. This study aimed to explore the attenuation of TFDG on RA. The collagen-induced arthritis (CIA) mouse model was established and administered with TFDG. The arthritis score and incidence was recorded to assess the amelioration of TFDG on arthritis. Histopathological change of the mouse joint tissues was evaluated by haemotoxylin and eosin staining. The expression of pro-inflammatory mediators including interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and IL-6 was quantified by ELISA. The activation of nuclear factor (NF)-κB and mitogen-activated protein kinase (MAPK) signaling pathways in the synovium were determined by Western blotting. In comparison with the control, administration of TFDG significantly reduced arthritis score and incidence in the CIA mouse model. TFDG significantly suppressed the expression of IL-1ß, TNF-α, and IL-6, as well as the levels of MMP-1, MMP-2, and MMP-3 in the synovium. TFDG also showed remarkable inhibition on the activation of NF-κB and the phosphorylation of P38, JNK2, and ERK. This study puts up evidence that TFDG exerts protection on RA via inhibiting the activation of NF-κB- and MAPK-signaling pathways.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Biflavonoids/pharmacology , Catechin/pharmacology , Gallic Acid/analogs & derivatives , MAP Kinase Signaling System/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Biflavonoids/therapeutic use , Catechin/therapeutic use , Collagen/administration & dosage , Collagen/immunology , Drug Evaluation, Preclinical , Gallic Acid/pharmacology , Gallic Acid/therapeutic use , Humans , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Joints/drug effects , Joints/immunology , Joints/pathology , MAP Kinase Signaling System/immunology , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
13.
Immunol Lett ; 197: 63-69, 2018 05.
Article in English | MEDLINE | ID: mdl-29545108

ABSTRACT

Yin Yang 1 (YY1) is a ubiquitously expressed transcription factor that functions in cooperation with various cofactors to regulate gene expression. In the immune system, YY1 enhances cytokine production and T helper (Th) 2 effector cell differentiation, resulting in the activation of inflammation. However, no studies have reported the role of YY1 in Th17 cell regulation, which is implicated in rheumatoid arthritis (RA). We investigated the expression of YY1 in Th17 cells in vitro and revealed increased levels of YY1 mRNA and protein. To elucidate the function of YY1 pathogenesis in RA, we used a collagen-induced arthritis (CIA) mouse model with YY1 deficiency. Deficiency of YY1 reduced the severity of arthritis and joint destruction. Moreover, Th17 cells were dramatically reduced in YY1-deficient mice. The cytokine interleukin (IL)-17 was decreased in YY1-deficient CD4+ T cells ex vivo and in vivo. Interestingly, the level of signal transducer and activator of transcription 3 (STAT3), tumor necrosis factor-α, IL-17, IL-6, and IL-1ß were markedly decreased in YY1-deficient mice with CIA. The cytokine-inducing function of YY1 was more specific to IL-17 than to interferon-γ. YY1 plays a role in Th17 cell differentiation and RA pathogenesis. Our findings suggest that future RA therapies should target the regulatory mechanism involved in Th17 cell differentiation, in which YY1 may cooperate with the STAT3 signaling pathway.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Inflammation/immunology , Joints/immunology , Th17 Cells/immunology , Th2 Cells/immunology , YY1 Transcription Factor/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Humans , Immunomodulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT3 Transcription Factor/metabolism , YY1 Transcription Factor/genetics
14.
J Pharmacol Sci ; 133(3): 130-138, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28258822

ABSTRACT

Abnormal store-operated calcium uptake has been observed in peripheral T lymphocytes of rheumatoid arthritis (RA) patients, and sustained intracellular calcium signalling is known to mediate the functions of many types of immune cells. Thus, it is hypothesized that regulating calcium entry through CRACM1 (the pore-forming subunit of calcium release-activated calcium (CRAC) channels; also known as ORAI1) may be beneficial for the management of RA. Localized CRACM1 knockdown in the joints and draining lymph nodes (DLNs) of mice with collagen-induced arthritis (CIA) was achieved via lentiviral-based delivery of shRNA targeting mouse CRACM1. Consistent with CRACM1 knockdown, calcium influx in synovial cells and the histopathological features of CIA were reduced. These effects were also associated with reduced levels of several notable inflammatory cytokines, such as IL-6, IL-17A, and IFN-γ, in the joints. Additionally, CRACM1-shRNA reduced the number of bone marrow-derived osteoclasts in vitro as well as osteoclasts in CIA joints, which was associated with reduced RANKL levels in the serum and joints. In summary, inhibiting calcium entry by CRACM1 knockdown suppressed arthritis development and may be therapeutically beneficial for RA patients.


Subject(s)
Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , Genetic Therapy , ORAI1 Protein/genetics , Animals , Arthritis, Experimental/blood , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Cytokines/blood , Cytokines/immunology , Gene Silencing , Joints/immunology , Joints/pathology , Lentivirus/genetics , Lymph Nodes , Male , Mice, Inbred DBA , RANK Ligand/blood , RANK Ligand/immunology , RNA, Small Interfering/genetics , Spleen/cytology , Synovial Membrane/cytology
15.
Int Immunopharmacol ; 44: 43-52, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28073043

ABSTRACT

Periplocoside A (PSA) has been extracted from the Chinese herbal medicine Periploca sepium Bge to treat rheumatoid arthritis (RA) via immune regulation. We previously found that PSA exhibits immunosuppressive activity both in vitro and in vivo. Balanced regulation of helper T 17 (Th17)/regulatory T (Treg) cells is the current therapeutic direction for the treatment of RA. The present study investigated the mechanism of PSA in treating collagen-induced arthritis (CIA). The therapeutic effects and potential pharmacological mechanisms of PSA were specifically clarified by examining its effects on CIA in DBA/1 mice. PSA administration significantly relieved the severity of the arthritis, and preventive administration of PSA reduced the incidence of arthritis in the mice with CIA and relieved joint damage in terms of morphology. PSA was also able to reduce the levels of anti-collagen II (CII) antibodies and pro-inflammatory cytokines in the serum. As a result, the proportion of Th17 cells decreased, and the proportion of Treg cells increased. A follow-up study of the ex vivo immunological reactions induced by a specific antigen found that PSA suppressed lymphocyte proliferation, inhibited the differentiation and reactivity of Th17 cells, and promoted the proportion of Treg cells among helper T cells. PSA also exhibited pharmacological effect in regulating the balance between Th17 and Treg cells in CIA through relevant signalling pathways. Thus, PSA played a specific role in CIA treatment. In particular, our results suggest that the therapeutic effects of PSA on RA are partially realized via the regulation of the balance of Th17/Treg cells.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Glycosides/therapeutic use , Immunosuppressive Agents/therapeutic use , Joints/drug effects , Pregnenes/therapeutic use , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Antibody Formation/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Collagen Type II/immunology , Humans , Joints/immunology , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred DBA
16.
Clin Exp Immunol ; 188(2): 293-298, 2017 05.
Article in English | MEDLINE | ID: mdl-28090641

ABSTRACT

Mouse collagen-induced arthritis (CIA) is the most commonly used animal model to investigate underlying pathogenesis of autoimmune arthritis and to demonstrate the therapeutic efficacy of novel drugs in autoimmune arthritis. The conventional read-outs of CIA are clinical score and histopathology, which have several limitations, including (i) subjected to observer bias; and (ii) longitudinal therapeutic efficacy of a new drug cannot be determined. Thus, a robust, non-invasive, in-vivo drug screening tool is currently an unmet need. Here we have assessed the utility of 18 F-fluorodeoxyglucose positron emission tomography (18 F-FDG) as an in-vivo screening tool for anti-inflammatory drugs using the mouse CIA model. The radiotracer 18 F-FDG and a PET scanner were employed to monitor CIA disease activity before and after murine anti-tumour necrosis factor (TNF)-α antibody (CNTO5048) therapy in the mouse CIA model. Radiotracer concentration was derived from PET images for individual limb joints and on a per-limb basis, and Spearman's correlation coefficient (ρ) was determined with clinical score and histology of the affected limbs. CNTO5048 improved arthritis efficiently, as evidenced by clinical score and histopathology. PET showed an increased uptake of 18 F-FDG with the progression of the disease and a significant decrease in the post-treatment group. 18 F-FDG uptake patterns showed a strong correlation with clinical score (ρ = 0·71, P < 0·05) and histopathology (ρ = 0·76, P < 0·05). This study demonstrates the potential of 18 F-FDG PET as a tool for in-vivo drug screening for inflammatory arthritis and to monitor the therapeutic effects in a longitudinal setting.


Subject(s)
Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/diagnostic imaging , Arthritis, Experimental/drug therapy , Animals , Anti-Inflammatory Agents/analysis , Collagen/administration & dosage , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical/methods , Fluorodeoxyglucose F18 , Joints/diagnostic imaging , Joints/immunology , Joints/pathology , Longitudinal Studies , Male , Mice , Mice, Inbred DBA , Positron-Emission Tomography , Radiopharmaceuticals , Tumor Necrosis Factor-alpha/antagonists & inhibitors
17.
Int J Rheum Dis ; 20(2): 203-213, 2017 Feb.
Article in English | MEDLINE | ID: mdl-25964045

ABSTRACT

AIM: Recently, there has been an increasing interest in tea (Camellia sinensis) as a protective agent against inflammatory diseases. Here, we evaluated/compared the anti-inflammatory activity of two different doses (0.5 and 1.0 g/kg body weight) of green tea aqueous extract (GTE, rich in catechins) and black tea aqueous extract (BTE, rich in theaflavins and thearubigins) in rat adjuvant-induced arthritis (AIA). METHODS: Adjuvant-induced arthritis rat model received orally/daily distilled water as vehicle, indomethacin (1.0 mg/kg body weight; a non-steroidal/anti-inflammatory drug), or tea aqueous extracts (for 28 or 14 consecutive days starting from day 0 or 14 of arthritis induction, respectively). RESULTS: The present study showed that only the high dose of GTE (from day 0) significantly alleviated (P < 0.05-0.001) all complications shown in arthritic rats, including synovial joint inflammation, elevation in erythrocyte sedimentation rate, blood leukocytosis (due to lymphocytosis and neutrocytosis), and changes in weight/cellularity of lymphoid organs. The anti-arthritic activity of the high dose of GTE (from day 0) was comparable (P > 0.05) with that of indomethacin (12.9-53.8 vs. 9.5-48.4%, respectively) and mediated by significantly decreasing and down-regulating (P < 0.001) the systemic production of pro-inflammatory cytokines and the expression of chemokine receptor-5 in synovial tissues, respectively. Moreover, the anti-arthritic activity of tea aqueous extracts was in the following order: high dose of GTE > low dose of GTE ≥ high dose of BTE > low dose of BTE. CONCLUSION: The present study proved the anti-inflammatory activity of GTE over BTE and equal to that of indomethacin in AIA rat model.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental/prevention & control , Camellia sinensis , Joints/drug effects , Tea , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/blood , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Blood Sedimentation , Cytokines/blood , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/prevention & control , Freund's Adjuvant , Indomethacin/pharmacology , Inflammation Mediators/blood , Joints/immunology , Joints/metabolism , Male , Phytotherapy , Plants, Medicinal , Rats, Wistar , Receptors, CCR5/metabolism , Synovial Membrane/drug effects , Synovial Membrane/immunology , Synovial Membrane/metabolism , Time Factors
18.
Int J Rheum Dis ; 20(3): 298-308, 2017 Mar.
Article in English | MEDLINE | ID: mdl-25546349

ABSTRACT

AIM: Rheumatoid arthritis is a chronic inflammatory autoimmune disorder with multi-factorial factors influencing disease alleviation in synovial joints. The aim of this study was to investigate the anti-arthritic efficacy of trikatu, a herbal compound, on biochemical and immunological complications in adjuvant-induced arthritic rats. METHODS: Arthritis was induced in rats by a single intradermal injection of complete Freund's adjuvant (0.1 mL) into the foot pad of the right hind paw. Trikatu (1000 mg/kg body weight, oral) and indomethacin (3 mg/kg body weight, intrap intraperitoneal) were administered for 8 days from days 11 to 18 after adjuvant injection. RESULTS: Our present study results evidenced a significant alteration in the activities/levels of lysosomal enzymes, protein bound carbohydrates, bone collagen, urinary constituents like hydroxyproline and total glycosaminoglycans, lipid peroxidation, antioxidant status, lipid profiles, rheumatoid factor and inflammatory mediators in adjuvant-induced arthritic rats. Trikatu treatment (1000 mg/kg/body weight) reverted back all the above biochemical and immunological parameters to near normal levels in arthritic rats as evidenced by the radiological and histopathological assessements . CONCLUSION: These results suggest that trikatu could be a promising alternative drug for the control and management of rheumatoid arthritis.


Subject(s)
Alkenes/pharmacology , Anti-Inflammatory Agents/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Experimental/prevention & control , Freund's Adjuvant , Joints/drug effects , Piperidines/pharmacology , Plant Extracts/pharmacology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Biomarkers/blood , Biomarkers/urine , Female , Joints/diagnostic imaging , Joints/immunology , Joints/metabolism , Male , Rats, Wistar
19.
J Immunol ; 197(6): 2362-8, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27534559

ABSTRACT

Uncontrolled inflammation is a unifying component of many chronic inflammatory diseases, such as arthritis. Resolvins (Rvs) are a new family from the endogenous specialized proresolving mediators (SPMs) that actively stimulate resolution of inflammation. In this study, using lipid mediator metabololipidomics with murine joints we found a temporal regulation of endogenous SPMs during self-resolving inflammatory arthritis. The SPMs present in self-resolving arthritic joints include the D-series Rvs, for example, RvD1, RvD2, RvD3, and RvD4. Of note, RvD3 levels were reduced in inflamed joints from mice with delayed-resolving arthritis when compared with self-resolving inflammatory arthritis. RvD3 was also reduced in serum from rheumatoid arthritis patients compared with healthy controls. RvD3 administration reduced joint leukocytes as well as paw joint eicosanoids, clinical scores, and edema. Taken together, these findings provide evidence for dysregulated endogenous RvD3 levels in inflamed paw joints and its potent actions in reducing murine arthritis.


Subject(s)
Arthritis/immunology , Arthritis/metabolism , Fatty Acids, Unsaturated/metabolism , Gene Expression Regulation , Lipid Metabolism , Animals , Arthritis/physiopathology , Edema/prevention & control , Eicosanoids/metabolism , Fatty Acids, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/blood , Humans , Joints/immunology , Joints/metabolism , Joints/physiopathology , Metabolomics , Mice
20.
Eur J Immunol ; 46(11): 2629-2638, 2016 11.
Article in English | MEDLINE | ID: mdl-27510283

ABSTRACT

Increased expression of endogenous Toll-like receptor 4 (TLR4) ligands (e.g., Tenascin-C, S100A8/A9, citrullinated fibrinogen (cFb) immune complexes) has been observed in patients with rheumatoid arthritis (RA). However, their roles in RA pathogenesis are not well understood. Here, we investigated the expression kinetics and role of endogenous TLR4 ligands in the murine model of collagen-induced arthritis (CIA). Tenascin-C was upregulated in blood early in CIA, and correlated positively with the clinical score at day 56. Levels of S100A8/A9 increased starting from day 28, peaking at day 42, and correlated positively with joint inflammation. Levels of anti-cFb antibodies increased during the late phase of CIA and correlated positively with both joint inflammation and cartilage damage. Blockade of TLR4 activation at the time of the first TLR4 ligand upregulation prevented clinical and histological signs of arthritis. A TLR4-dependent role was also observed for Tenascin-C and cFb immune complexes in osteoclast differentiation in vitro. Taken together, our data suggests that the pathogenic contribution of TLR4 in promoting joint inflammation and bone erosion during CIA occurs via various TLR4 ligands arising at different stages of disease. The data also suggests that Blockade of TLR4 with monoclonal antibodies is a promising strategy in RA treatment.


Subject(s)
Arthritis, Rheumatoid/immunology , Bone and Bones/pathology , Calgranulin A/blood , Tenascin/blood , Toll-Like Receptor 4/metabolism , Animals , Antigen-Antibody Complex/blood , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/physiopathology , Calgranulin A/genetics , Cell Differentiation , Collagen , Disease Models, Animal , Fibrinogen/immunology , Joints/immunology , Ligands , Mice , Mice, Inbred DBA , Osteoclasts/metabolism , Spatio-Temporal Analysis , Tenascin/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
SELECTION OF CITATIONS
SEARCH DETAIL