Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Arthritis Rheumatol ; 74(7): 1139-1146, 2022 07.
Article in English | MEDLINE | ID: mdl-35166055

ABSTRACT

OBJECTIVE: Genome-wide association studies have connected PADI4, encoding peptidylarginine deiminase 4 (PAD4), with rheumatoid arthritis (RA). PAD4 promotes neutrophil extracellular trap (NET) formation. This study was undertaken to investigate the origin of PAD4 and the importance of NET formation in a C57BL/6 mouse model of arthritis. METHODS: To permit the effective use of C57BL/6 mice in the collagen-induced arthritis (CIA) model, we introduced the administration of granulocyte colony-stimulating factor (G-CSF) for 4 consecutive days in conjunction with the booster immunization on day 21. Mice with global Padi4 deficiency (Padi4-/- ) and mice with hematopoietic lineage-specific Padi4 deficiency (Padi4Vav1Cre/+ ) were evaluated in the model. RESULTS: G-CSF significantly increased the incidence and severity of CIA. G-CSF-treated mice showed elevated citrullinated histone H3 (Cit-H3) levels in plasma, while vehicle-treated mice did not. Immunofluorescence microscopy revealed deposition of Cit-H3 in synovial tissue in G-CSF-treated mice. Padi4-/- mice developed less severe arthritis and had lower levels of serum interleukin-6 and plasma Cit-H3, lower levels of Cit-H4 in synovial tissue, and less bone erosion on micro-computed tomography than Padi4+/+ mice in the G-CSF-modified CIA model. Similarly, Padi4Vav1Cre/+ mice developed less severe arthritis, compared with Padi4fl/fl mice, and presented the same phenotype as Padi4-/- mice. CONCLUSION: We succeeded in developing an arthritis model suitable for use in C57BL/6 mice that is fully compliant with high animal welfare standards. We observed a >90% incidence of arthritis in male mice and detectable NET markers. This model, with some features consistent with human RA, demonstrates that hematopoietic PAD4 is an important contributor to arthritis development and may prove useful in future RA research.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Protein-Arginine Deiminase Type 4 , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/enzymology , Collagen , Genome-Wide Association Study , Granulocyte Colony-Stimulating Factor , Male , Mice , Mice, Inbred C57BL , Protein-Arginine Deiminase Type 4/metabolism , Protein-Arginine Deiminases , X-Ray Microtomography
2.
J Ethnopharmacol ; 267: 113514, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33223115

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Rhamnella gilgitica Mansf. et Melch. (སེང་ལྡེང་།, RG) is a traditional Tibetan medicinal plant that is currently grown throughout Tibet. According to the theory of Tibetan medicine, RG is efficient for removing rheumatism, reducing swelling, and relieving pain. Hence, it has been used for the treatment of rheumatoid arthritis (RA) in Tibet for many years. However, there are no previous reports on the anti-RA activities of ethyl acetate extract of RG (RGEA). AIM OF THE STUDY: This study aimed to explore the anti-RA effect and mechanism of RGEA on collagen-induced arthritis (CIA) in rats. MATERIALS AND METHODS: The CIA model was established in male Wister rats by intradermal injection of bovine type II collagen and Complete Freund's Adjuvant at the base of the tail and left sole, respectively. The rats were orally administered with RGEA (9.71, 19.43, or 38.85 mg/kg) for 23 days. The body weight, swelling volume, arthritis index score, thymus and spleen indices, and pathological changes were observed to evaluate the effect of RGEA on RA. Furthermore, the inflammatory cytokines in serum, such as interleukin1 beta (IL-1ß), tumor necrosis factor alpha (TNF-α), interleukin6 (IL-6), interleukin17 (IL-17), interferon-γ (INF-γ), interleukin4 (IL-4), and interleukin10 (IL-10) were measured by enzyme linked immunosorbent assay (ELISA) to explore the anti-inflammatory effects of RGEA. The terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) staining was used to examine apoptosis. Finally, the protein and gene expression of B-cell lymphoma-2-associated X (Bax), B-cell lymphoma 2 (Bcl-2), Caspase3, janus-activated kinase 2 (JAK2), signal transducer and activator of transcription 3 (STAT3), suppressor of cytokine signaling1 (SOCS1), and 3 (SOCS3) in synovial tissue were detected using immunohistochemistry and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS: After the treatment with RGEA, the body weight of rats was restored, both the arthritis index and paw swelling were suppressed, and spleen and thymus indices were decreased. RGEA reduced the inflammatory cells and synovial hyperplasia in the synovial tissue of the knee joint, and suppressed bone erosion. Meanwhile, RGEA decreased the levels of IL-1ß, IL-6, IL-17, TNF-α, and INF-γ, while increased the levels of IL-4 and IL-10. TUNEL fluorescence apoptosis results confirmed that RGEA obviously promoted the apoptosis of synovial cells. Further studies showed that RGEA inhibited the proteins and mRNAs expression of JAK2 and STAT3 as well as increased the proteins and mRNAs expression of SOCS1 and SOCS3. In addition, RGEA upregulated the expression of Bax and Caspase3, and downregulated the expression of Bcl-2. CONCLUSION: The anti-RA effectof RGEA might be related to the promotion of apoptosis and inhibition of inflammation, which regulated the JAK-STAT pathway.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental/prevention & control , Janus Kinase 2/metabolism , Joints/drug effects , Plant Extracts/pharmacology , Rhamnaceae , STAT3 Transcription Factor/metabolism , Acetates/chemistry , Animals , Antirheumatic Agents/isolation & purification , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Collagen Type II , Cytokines/metabolism , Inflammation Mediators/metabolism , Janus Kinase 2/genetics , Joints/enzymology , Joints/pathology , Male , Medicine, Tibetan Traditional , Plant Extracts/isolation & purification , Rats, Wistar , Rhamnaceae/chemistry , STAT3 Transcription Factor/genetics , Signal Transduction , Solvents/chemistry , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism
3.
Cell Rep ; 32(11): 108158, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32937124

ABSTRACT

The traditional Chinese medicinal herb Notopterygium incisum Ting ex H.T. Chang has anti-rheumatism activity, and a mass spectrometry assay of patients' serum after administration of the herb revealed that notopterol is the most abundant component enriched. However, the functions of notopterol and its molecular target in rheumatoid arthritis (RA) treatment remain unknown. Here, we show in different RA mouse strains that both oral and intraperitoneal administration of notopterol result in significant therapeutic effects. Mechanistically, notopterol directly binds Janus kinase (JAK)2 and JAK3 kinase domains to inhibit JAK/signal transducers and activators of transcription (JAK-STAT) activation, leading to reduced production of inflammatory cytokines and chemokines. Critically, combination therapy using both notopterol and tumor necrosis factor (TNF) blocker results in enhanced therapeutic effects compared to using TNF blocker alone. We demonstrate that notopterol ameliorates RA pathology by targeting JAK-STAT signaling, raising the possibility that notopterol could be effective in treating other diseases characterized by aberrant JAK-STAT signaling pathway.


Subject(s)
Arthritis, Rheumatoid/pathology , Coumarins/pharmacology , Inflammation/pathology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , Biological Products/administration & dosage , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/therapeutic use , Chemokines/metabolism , Coumarins/administration & dosage , Coumarins/chemistry , Coumarins/therapeutic use , Etanercept/pharmacology , Inflammation/drug therapy , Inflammation/enzymology , Inflammation Mediators/metabolism , Interferon-gamma/pharmacology , Janus Kinase 2/chemistry , Janus Kinase 3/metabolism , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Inbred DBA , Protein Domains , STAT Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology
4.
Biomed Pharmacother ; 129: 110471, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32768958

ABSTRACT

Huoxuezhitong capsule (HXZT, activating blood circulation and relieving pain capsule), has been applied for osteoarthritis since 1974. It consists of Angelica sinensis (Oliv.) Diels, Panax notoginseng (Burkill) F. H. Chen ex C. H., Boswellia sacra, Borneol, Eupolyphaga sinensis Walker, Pyritum. However, the direct effects of HXZT on osteoarthritis and the underlying mechanisms were poorly understood. In this study, we aimed to explore the analgesia effect of HXZT on MIA-induced osteoarthritis rat and the underlying mechanisms. The analgesia and anti-inflammatory effect of HXZT on osteoarthritis in vivo were tested by the arthritis model rats induced by monosodium iodoacetate (MIA).. Mechanistic studies confirmed that HXZT could inhibit the activation of NF-κB and down-regulate the mRNA expression of related inflammatory factors in LPS-induced RAW264.7 and ATDC5 cells. Furtherly, in LPS-induced RAW264.7 cells, HXZT could suppress NF-κB via inhibiting PI3K/Akt pathway. Taken together, HXZT capsule could ameliorate MIA-induced osteoarthritis of rats through suppressing PI3K/ Akt/ NF-κB pathway.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental/prevention & control , Drugs, Chinese Herbal/pharmacology , Knee Joint/drug effects , NF-kappa B/metabolism , Osteoarthritis, Knee/prevention & control , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Capsules , Cytokines/metabolism , Inflammation Mediators/metabolism , Iodoacetic Acid , Knee Joint/enzymology , Knee Joint/pathology , Male , Mice , Osteoarthritis, Knee/chemically induced , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/pathology , Phosphorylation , RAW 264.7 Cells , Rats, Sprague-Dawley , Signal Transduction
5.
Pharmacol Res Perspect ; 8(4): e00565, 2020 08.
Article in English | MEDLINE | ID: mdl-32790160

ABSTRACT

Bruton's tyrosine kinase (BTK) plays a central and pivotal role in controlling the pathways involved in the pathobiology of cancer, rheumatoid arthritis (RA), and other autoimmune disorders. ZYBT1 is a potent, irreversible, specific BTK inhibitor that inhibits the ibrutinib-resistant C481S BTK with nanomolar potency. ZYBT1 is found to be a promising molecule to treat both cancer and RA. In the present report we profiled the molecule for in-vitro, in-vivo activity, and pharmacokinetic properties. ZYBT1 inhibits BTK and C481S BTK with an IC50 of 1 nmol/L and 14 nmol/L, respectively, inhibits the growth of various leukemic cell lines with IC50 of 1 nmol/L to 15 µmol/L, blocks the phosphorylation of BTK and PLCγ2, and inhibits secretion of TNF-α, IL-8 and IL-6. It has favorable pharmacokinetic properties suitable for using as an oral anti-cancer and anti-arthritic drug. In accordance with the in-vitro properties, it demonstrated robust efficacy in murine models of collagen-induced arthritis (CIA) and streptococcal cell wall (SCW) induced arthritis. In both models, ZYBT1 alone could suppress the progression of the diseases. It also reduced the growth of TMD8 xenograft tumor. The results suggested that ZYBT1 has high potential for treating RA, and cancer.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/enzymology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , Humans , Inhibitory Concentration 50 , Mice , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics
6.
J Ethnopharmacol ; 261: 113074, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-32534115

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Kyung-Bang Gumiganghwal-tang tablet (GMGHT) is a standardized Korean Medicine that could treat a cold, headache, arthralgia and fever. Although GMGHT has been used for arthritis-related diseases including a sprain, arthralgia, unspecified arthritis and knee arthritis, there is no pre-clinical evidence to treat osteoarthritis (OA). This study determined the drug dosage and the mechanisms of GMGHT for OA. METHODS: OA was induced by intra-articular monoiodoacetic acid (MIA) injection in Sprague-Dawley rats. As calculated from the human equivalent dose formula, GMGHT was orally administered at the doses of 9.86, 98.6 and 986 mg/kg for 4 weeks. The arthritis score was performed by a blind test, and histological changes in articular cartilage were indicated by hematoxylin and eosin, Safranin O and toluidine blue staining. SW1353 chondrocytes were stimulated by interleukin (IL)-1ß recombinant to analyze the expressions of Type II collagen, matrix metalloproteinases (MMPs) and nuclear factor (NF)-κB. RESULTS: Rough and punctate surfaces of the femoral condyle induced by MIA, were recovered by the GMGHT treatment. The arthritis score was significantly improved in the 968 mg/kg of GMGHT-treated cartilage. Loss of chondrocytes and proteoglycan were ameliorated at the deep zone of the subchondral bone plate by the GMGHT administration in OA rats. The expression of Type II collagen was increased, while MMP-1, -3 and -13 levels were decreased in the GMGHT-treated SW1353 chondrocytes. In addition, the GMGHT treatment regulated NF-κB activation along with IL-6, transforming growth factor-ß and IL-12 production. CONCLUSIONS: GMGHT promoted the recovery of articular cartilage damage by inhibiting MMPs, accompanied with its anti-inflammatory effects in OA. GMGHT might be an alternative therapeutic treatment for OA.


Subject(s)
Arthritis, Experimental/prevention & control , Cartilage, Articular/drug effects , Joints/drug effects , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinases, Secreted/antagonists & inhibitors , Osteoarthritis/prevention & control , Plant Extracts/pharmacology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Cartilage, Articular/enzymology , Cartilage, Articular/pathology , Cell Line, Tumor , Chondrocytes/drug effects , Chondrocytes/enzymology , Chondrocytes/pathology , Collagen Type II/metabolism , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Iodoacetic Acid , Joints/enzymology , Joints/pathology , Male , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/metabolism , Osteoarthritis/chemically induced , Osteoarthritis/enzymology , Osteoarthritis/pathology , Rats, Sprague-Dawley
7.
Ann Rheum Dis ; 78(3): 421-428, 2019 03.
Article in English | MEDLINE | ID: mdl-30610061

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) appears to be associated with various metabolic disorders, but the potential contribution of amino acid metabolism to OA pathogenesis has not been clearly elucidated. Here, we explored whether alterations in the amino acid metabolism of chondrocytes could regulate OA pathogenesis. METHODS: Expression profiles of amino acid metabolism-regulating genes in primary-culture passage 0 mouse chondrocytes were examined by microarray analysis, and selected genes were further characterised in mouse OA chondrocytes and OA cartilage of human and mouse models. Experimental OA in mice was induced by destabilisation of the medial meniscus (DMM) or intra-articular (IA) injection of adenoviruses expressing catabolic regulators. The functional consequences of arginase II (Arg-II) were examined in Arg2-/- mice and those subjected to IA injection of an adenovirus encoding Arg-II (Ad-Arg-II). RESULTS: The gene encoding Arg-II, an arginine-metabolising enzyme, was specifically upregulated in chondrocytes under various pathological conditions and in OA cartilage from human patients with OA and various mouse models. Adenovirus-mediated overexpression of Arg-II in mouse joint tissues caused OA pathogenesis, whereas genetic ablation of Arg2 in mice (Arg2-/-) abolished all manifestations of DMM-induced OA. Mechanistically, Arg-II appears to cause OA cartilage destruction at least partly by upregulating the expression of matrix-degrading enzymes (matrix metalloproteinase 3 [MMP3] and MMP13) in chondrocytes via the nuclear factor (NF)-κB pathway. CONCLUSIONS: Our results indicate that Arg-II is a crucial regulator of OA pathogenesis in mice. Although chondrocytes of human and mouse do not identically, but similarly, respond to Arg-II, our results suggest that Arg-II could be a therapeutic target of OA pathogenesis.


Subject(s)
Arginase/physiology , Arthritis, Experimental/enzymology , Cartilage, Articular/enzymology , Chondrocytes/enzymology , Osteoarthritis/enzymology , Animals , Arthritis, Experimental/chemically induced , Disease Models, Animal , Humans , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Mice , Osteoarthritis/chemically induced , Up-Regulation
8.
Biomed Pharmacother ; 106: 1616-1623, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119237

ABSTRACT

This study investigated the effect of eugenol on arginase, nucleotidase and adenosine deaminase activities in platelets of carrageenan-induced arthritic rat model to explain a possible anti-arthritic mechanism of eugenol. Fifty adult female rats (140-250 g) were divided into ten (10) groups (n = 5). Group I received oral administration of corn oil, group II received 2.50 mg/kg of eugenol, group III and IV rats received oral administration of 5.0 and 10.0 mg/kg of eugenol respectively, group V received 0.20 mg/kg of dexamethasone orally, group VI rats was injected with 1% carrageenan (arthritic rats) and received saline solution orally (arthritic control rat group), group VII, VIII and IX: arthritic rats received 2.50, 5.0 or 10 mg/kg of eugenol orally respectively, group X: arthritic rats was administered with 0.20 mg/kg of dexamethasone orally. The animals were treated for 21 days, thereafter, tibiofemoral histological examination, thiobabituric acid reactive substances level, arginase, nucleoside triphosphate diphosphohydrolase, 5´-nucleotidase and adenosine deaminase activities were assessed. Tibiofemoral histological examination result showed that infiltration of inflammatory cells was significantly decreased with an increase in eugenol dose. Activities of arginase, adenosine triphosphate and adenosine monophosphate hydrolyses were significantly decreased while adenosine diphosphate hydrolysis and adenosine deaminase activities were significantly increased in arthritic rat groups administered with different doses of eugenol. Therefore, eugenol might be a natural complement and alternative promising anti-arthritic agent. These possible anti-arthritic mechanisms may be partly through the modulation of arginase and adenosine nucleotides hydrolyzing enzyme activities as well as the antioxidative action of eugenol.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Adenosine Deaminase/metabolism , Anti-Inflammatory Agents/pharmacology , Arginase/antagonists & inhibitors , Arthritis, Experimental/prevention & control , Blood Platelets/drug effects , Carrageenan , Enzyme Inhibitors/pharmacology , Eugenol/pharmacology , Joints/drug effects , 5'-Nucleotidase/metabolism , Adenosine Diphosphate/blood , Adenosine Monophosphate/blood , Adenosine Triphosphate/blood , Animals , Antioxidants/pharmacology , Arginase/metabolism , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Blood Platelets/enzymology , Dexamethasone/pharmacology , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Hydrolysis , Joints/metabolism , Joints/pathology , Oxidative Stress/drug effects , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
9.
Ann Rheum Dis ; 77(6): 935-943, 2018 06.
Article in English | MEDLINE | ID: mdl-29555825

ABSTRACT

OBJECTIVES: To investigate the role of tyrosine kinase Fyn in the development of osteoarthritis (OA) and the underlying mechanisms, and to define whether targeting Fyn could prevent OA in mice. METHODS: Cartilage samples from normal and aged mice were analysed with proteome-wide screening. Fyn expression was examined with immunofluorescence in human and age-dependent or experimental mouse OA cartilage samples. Experimental OA in Fyn-knockout mice was induced by destabilisation of the medial meniscus. Primary cultured mouse chondrocytes were treated with proinflammatory cytokine interleukin-1ß. The inhibitor of Src kinase family, AZD0530 (saracatinib), and inhibitor of Fyn, PP1, were used to treat experimental OA in mice. RESULTS: Fyn expression was markedly upregulated in human OA cartilage and in cartilage from aged mice and those with post-traumatic OA. Fyn accumulates in articular chondrocytes and interacts directly with and phosphorylates ß-catenin at Tyr142, which stabilises ß-catenin and promotes its nuclear translocation. The deletion of Fyn effectively delayed the development of post-traumatic and age-dependent OA in mice. Fyn inhibitors AZD0530 and PP1 significantly attenuated OA progression by blocking the ß-catenin pathway and reducing the levels of extracellular matrix catabolic enzymes in the articular cartilage. CONCLUSIONS: Fyn accumulates and activates ß-catenin signalling in chondrocytes, accelerating the degradation of the articular cartilage and OA development. Targeting Fyn is a novel and potentially therapeutic approach to the treatment of OA.


Subject(s)
Arthritis, Experimental/enzymology , Osteoarthritis/enzymology , Proto-Oncogene Proteins c-fyn/physiology , beta Catenin/metabolism , Aging/metabolism , Animals , Arthritis, Experimental/prevention & control , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Cartilage, Articular/enzymology , Cells, Cultured , Chondrocytes/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Knockout Techniques , Humans , Mice, Knockout , Molecular Targeted Therapy/methods , Osteoarthritis/prevention & control , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/genetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology
10.
FASEB J ; 32(6): 3398-3410, 2018 06.
Article in English | MEDLINE | ID: mdl-29401630

ABSTRACT

Tetrandrine, a bisbenzylisoquinoline alkaloid, was previously demonstrated to attenuate inflammation and cartilage destruction in the ankles of mice with collagen-induced arthritis (CIA). Here, we explored the underlying mechanism by which tetrandrine prevented arthritis-induced bone erosion by focusing on the differentiation and function of osteoclasts. We found that daily administration of tetrandrine (30 mg/kg) markedly reduced the bone damage and decreased the number of osteoclasts in CIA rats. In vitro, tetrandrine inhibited receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis at the early stage and reduced the expressions of osteoclast-related marker genes. In bone marrow-derived macrophages and RAW264.7 cells, tetrandrine inhibited RANKL-induced translocation of NF-κB-p65 and nuclear factor of activated T cell 1 (NFATc1) through suppressing spleen tyrosine kinase (Syk)-Bruton's tyrosine kinase-PLCγ2-Ca2+ signaling. Of interest, tetrandrine did not affect the phosphorylation of immunoreceptor tyrosine-based activation motifs, the conventional upstream of Syk, but it inhibited the activity of Syk by enhancing its ubiquitination and degradation. The anti-osteoclastogenesis effect of tetrandrine nearly disappeared when it was used in combination with the Syk inhibitor piceatannol or in constitutively activated Syk-overexpressing cells. Taken together, tetrandrine attenuated CIA-induced bone destruction by inhibiting osteoclastogenesis through hindering the translocation of NF-κB-p65 and NFATc1 via reducing the activation of Syk.-Jia, Y., Miao, Y., Yue, M., Shu, M., Wei, Z., Dai, Y. Tetrandrine attenuates the bone erosion in collagen-induced arthritis rats by inhibiting osteoclastogenesis via spleen tyrosine kinase.


Subject(s)
Arthritis, Experimental/enzymology , Benzylisoquinolines/pharmacology , Bone Resorption/enzymology , Calcium Signaling/drug effects , Osteoclasts/enzymology , Syk Kinase/metabolism , Animals , Arthritis, Experimental/pathology , Bone Resorption/drug therapy , Bone Resorption/pathology , Female , Osteoclasts/pathology , Proteolysis/drug effects , Rats , Rats, Wistar , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Ubiquitination/drug effects
11.
Atherosclerosis ; 266: 136-144, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29024866

ABSTRACT

BACKGROUND AND AIMS: We aimed at investigating the effect of celecoxib (COX-2 selective inhibitor) and diclofenac (non-selective COX inhibitor) on endothelial function, and at identifying the underlying mechanisms in adjuvant-induced arthritis (AIA). METHODS: At the first signs of AIA, diclofenac (5 mg/kg twice a day, i.p), celecoxib (3 mg/kg/day, i.p) or saline (Vehicle) was administered for 3 weeks. Endothelial function was studied in aortic rings relaxed with acetylcholine (Ach) with or without inhibitors of NOS, arginase, EDHF and superoxide anions (O2-°) production. Aortic expression of eNOS, Ser1177-phospho-eNOS, COX-2, arginase-2, p22phox and p47phox was evaluated by Western blotting analysis. Arthritis scores, blood pressure, glycaemia and serum ADMA levels were measured. RESULTS: Diclofenac and celecoxib significantly reduced arthritis score to the same extent (p<0.05). As compared to vehicle-treated AIA, celecoxib did not change whereas diclofenac improved endothelial function (p<0.05) through increased EDHF production, decreased arginase activity and expression, decreased superoxide anions production and expression of p22phox and p47phox. Diclofenac but not celecoxib significantly enhanced blood pressure and serum ADMA levels. Glycaemia was unchanged by both treatments. CONCLUSIONS: Our study reveals that the effect of NSAIDs on endothelial function cannot be extrapolated from their impact on arthritis severity and suggest that changes in blood pressure and plasma ADMA levels may not be useful to predict CV risk of NSAIDs in RA.


Subject(s)
Aorta, Thoracic/drug effects , Arthritis, Experimental/drug therapy , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Diclofenac/pharmacology , Endothelium, Vascular/drug effects , Vasodilation/drug effects , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Arginase/metabolism , Arginine/analogs & derivatives , Arginine/blood , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Experimental/physiopathology , Biological Factors/metabolism , Blood Pressure/drug effects , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Freund's Adjuvant , In Vitro Techniques , Male , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Rats, Inbred Lew , Vasodilator Agents/pharmacology
12.
Biomed Pharmacother ; 96: 466-470, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29031206

ABSTRACT

BACKGROUD: Dipeptidyl peptidase I (DPPI), a lysosomal cysteine protease is derived from granule immune cells including mast cell, neutrophils, and toxicity T cells. DPPI can activate serine proteases by removal of dipeptides from N-termini of the pro-proteases, resulting in granule immune cells activation which involved in physiological or pathological responses. Triperygium Wilfordii Polyglucoside (TWP) is one of the traditional Chinese medicines, and commonly used in rheumatoid arthritis (RA) treatment. The present study intended to evaluate the effects of TWP on DPPI activity. METHODS: In vivo and in vitro studies were carried out to investigate the functions of TWP or triptolide (TP) on DPPI activities in serum, tissues of CIA rats. Rats were divided into five groups randomly: normal group, untreated CIA rat group, TWP treatment CIA groups (the low dose 2.5mg/100g body-weight and high dose 5mg/100g body-weight), and TP treatment CIA group (4µg/100g body-weight). Arthritis development was monitored visually, and joint pathology was examined radiologically. Total protein concentrations in synovial fluids (SFs) were determined by BCA method. Serums and tissue homogenates from CIA rats were collected and DPPI activities were detected using fluorescence substrate GF-AFC. The in vitro interactions between DPPI in serums or in tissue homogenates and TWP or TP were assessed. RESULTS: TWP-treated CIA rats showed a significant improvement in bone erosion. TWP significantly suppressed paw swelling and total protein concentration in the SFs of CIA rats compared with untreated CIA rats. The elevated activities of DPPI in serums or tissues of CIA rats were significantly inhibited by TWP, but not by TP in vivo. The inhibitory effects of TWP on DPPI activities were also confirm by in vitro study. CONCLUSION: One of the therapeutic functions of TWP in RA treatment could be inhibiting DPPI activity in serums and synovial tissue produced during RA development, and then reducing inflammatory serine proteases activities and further recovering CIA rats from RA symptoms.


Subject(s)
Arthritis, Experimental/drug therapy , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucosides/pharmacology , Plant Extracts/pharmacology , Tripterygium , Animals , Arthritis, Experimental/diagnostic imaging , Arthritis, Experimental/enzymology , Dipeptidyl-Peptidase IV Inhibitors/isolation & purification , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dose-Response Relationship, Drug , Glucosides/isolation & purification , Glucosides/therapeutic use , Male , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Rats , Rats, Wistar , Synovial Fluid/diagnostic imaging , Synovial Fluid/drug effects , Synovial Fluid/enzymology
13.
Osteoarthritis Cartilage ; 25(10): 1719-1728, 2017 10.
Article in English | MEDLINE | ID: mdl-28694081

ABSTRACT

OBJECTIVE: 12/15-Lipoxygenase (12/15-LOX) catalyzes the generation of various anti-inflammatory lipid mediators, and has been implicated in several inflammatory and degenerative diseases. However, there is currently no evidence that 12/15-LOX has a role in osteoarthritis (OA). The aim of this study was to investigate the role of 12/15-LOX in the pathogenesis of OA. METHODS: The development of aging-associated and destabilization of the medial meniscus (DMM)-induced OA were compared in 12/15-LOX-deficient (12/15-LOX-/-) and wild-type (WT) mice. The extent of cartilage damage was evaluated by histology. The expression of OA markers was evaluated by immunohistochemistry and RT-PCR. Cartilage explants were stimulated with IL-1α in the absence or presence of the 12/15-LOX metabolites, 15-hydroxyeicosatetraenoic acids (15-HETE), 13-hydroxyoctadecadienoic acid (13-HODE) or lipoxin A4 (LXA4), and the levels of matrix metalloproteinases-13 (MMP-13), Nitric oxide (NO) and prostaglandin E2 (PGE2) were determined. The effect of LXA4 on the progression of OA was evaluated in wild type (WT) mice. RESULTS: The expression of 12/15-LOX in cartilage increased during the progression of DMM-induced OA and with aging in WT mice. Cartilage degeneration was more severe in 12/15-LOX-/- mice compared to WT mice in both models of OA, and this was associated with increased expression of MMP-13, a disintegrin and metalloproteinase with thrombospondin motifs, aggrecanases (ADAMTS5), inducible NO synthases (iNOS), and mPGES-1. Treatment of cartilage explants with 12/15-LOX metabolites, suppressed IL-1α-induced production of MMP-13, NO and PGE2, with LXA4 being the most potent. Intra-peritoneal injection of LXA4 reduced the severity of DMM-induced cartilage degradation. CONCLUSIONS: These data suggest an important role of 12/15-LOX in the pathogenesis of OA. They also suggest that activation of this pathway may provide a novel strategy for prevention and treatment of OA.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Arthritis, Experimental/enzymology , Osteoarthritis/enzymology , Aging/metabolism , Aging/pathology , Animals , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/genetics , Arthritis, Experimental/etiology , Arthritis, Experimental/prevention & control , Cartilage, Articular/metabolism , Disease Progression , Inflammation Mediators/metabolism , Joint Instability/complications , Lipoxins/therapeutic use , Male , Mice, Knockout , Osteoarthritis/etiology , Osteoarthritis/prevention & control , Tibial Meniscus Injuries/complications , Tissue Culture Techniques , Up-Regulation
14.
Cell Biol Int ; 41(10): 1093-1102, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28665049

ABSTRACT

Dipeptidyl peptidase I (DPPI), a lysosomal cysteine protease, required for activation of serine proteases of granulocytes including mast cells (MCs), neutrophils (NPs) and others, which were found in synovial tissue of patients with rheumatoid arthritis (RA). But, the role of DPPI associated with those cells in RA development is unclear. In this study, the collagen-induced-arthritis (CIA) rat-model was employed to investigate the expression and activity levels of DPPI and its association with RA progress. Primary granulocytes were freshly extracted from bone-marrows of normal or CIA rats, human mast cell line LAD-2 and primary neutrophils, human-recombinant-DPPI, DPPI-inhibitor Gly-Phe-CHN2 , LTB4, anti-IgE antibody, calcium ionophore were used to study the regulatory role of DPPI in cell activations. The increased DPPI activities in synovial fluids, serum, and bone-marrow homogenates of CIA rats associated with RA severities progress were observed after injections. MMP2/9 expressions in SFs and bone-marrow were in different patterns. Regular-Blood-Tests have shown the high leveled DPPI activities associated with granulocytes differentiations in-vivo in blood of CIA rats. In-vitro cell models, DPPI up-regulated the proliferation of primary bone-marrow granulocytes of normal rats, but inhibited that of CIA rats. DPPI up-regulated and Gly-Phe-CHN2 down-regulated MCs intracellular DPPI and chymase activities. Gly-Phe-CHN2 also inhibited the LTB4 -activated-NPs and NP-elastase activities. Following stimulation of calcium ionophore, the net-releases of DPPI and ß-hexosaminidase from MCs were increased over a time-course, while Gly-Phe-CHN2 down-regulated MCs and NPs activation. Our findings demonstrate the role of DPPI in regulating MCs and NPs activation, and modulating proteolysis in the process of RA.


Subject(s)
Cathepsin C/metabolism , Granulocytes/enzymology , Animals , Antibodies, Anti-Idiotypic , Arthritis, Experimental/enzymology , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Cathepsin C/blood , Disease Models, Animal , Disease Progression , Granulocytes/immunology , Granulocytes/metabolism , Male , Mast Cells/metabolism , Neutrophils/metabolism , Rats , Rats, Wistar , Synovial Fluid/enzymology , Synovial Fluid/immunology , Synovial Fluid/metabolism
15.
Lasers Med Sci ; 32(5): 1071-1079, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28429194

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by chronic and systemic inflammation, which leads to the destruction of the cartilage and bone and affects tissues in multiple joints. Oxidative stress has been implicated with regards to involvement in various disease conditions, such as diabetes mellitus and neurodegenerative, respiratory, cardiovascular, and RA diseases. In vivo experimental studies using photobiomodulation therapy (PBMT) have shown positive effects in reducing lipid peroxidation and in increasing antioxidant activity. The regular practice of physical exercise has also been reported to be a beneficial treatment capable of reducing oxidative damage. Thus, the aim of this study was to analyze the effects of photobiomodulation therapy at 2- and 4-J doses associated with physical exercise on oxidative stress in an experimental model of RA in protein expression involving superoxide dismutase (SOD), glutathione peroxidase (GPX), and/or catalase (CAT) on thiobarbituric acid reactive substances (TBARS). In this study, 24 male Wistar rats divided into four groups were submitted to an RA model (i.e., collagen-induced arthritis, CIA), with the first immunization performed at the base of the tail on days 0 and 7 were included. After 28 days, a third intraarticular dose was administered in both knees of the animals. After the last induction, PBMT was started immediately, transcutaneously at two points (i.e., the medial and lateral), with a total of 15 applications. Treadmill exercise was also started the day after the last induction, and lasted for 5 weeks. With respect to results, we obtained the decreases in the lipid peroxidation and the increases of the antioxidant activities of SOD, GPX and CAT, with physical exercise associated to PBMT in doses of 2 and 4 J. In conclusion, physical exercise associated with PBMT decreases lipid peroxidation and increases antioxidant activity.


Subject(s)
Arthritis, Experimental/pathology , Arthritis, Experimental/radiotherapy , Low-Level Light Therapy , Oxidative Stress/radiation effects , Physical Conditioning, Animal , Animals , Antioxidants/metabolism , Arthritis, Experimental/enzymology , Catalase/metabolism , Disease Models, Animal , Glutathione Peroxidase/metabolism , Lipid Peroxidation/radiation effects , Male , Malondialdehyde/metabolism , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
16.
Arthritis Res Ther ; 19(1): 76, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28399896

ABSTRACT

BACKGROUND: The recent findings of cancer-specific metabolic changes, including increased glucose and glutamine consumption, have provided new therapeutic targets for consideration. Fibroblast-like synoviocytes (FLS) from rheumatoid arthritis (RA) patients exhibit several tumor cell-like characteristics; however, the role of glucose and glutamine metabolism in the aberrant proliferation of these cells is unclear. Here, we evaluated the role of these metabolic pathways in RA-FLS proliferation and in autoimmune arthritis in SKG mice. METHODS: The expression of glycolysis- or glutaminolysis-related enzymes was evaluated by real-time polymerase chain reaction (PCR) and Western blotting, and the intracellular metabolites were evaluated by metabolomic analyses. The effects of glucose or glutamine on RA-FLS cell growth were investigated using glucose- or glutamine-free medium. Glutaminase (GLS)1 small interfering RNA (siRNA) and the GLS1 inhibitor compound 968 were used to inhibit GLS1 in RA-FLS, and compound 968 was used to study the effect of GLS1 inhibition in zymosan A-injected SKG mice. RESULTS: GLS1 expression was increased in RA-FLS, and metabolomic analyses revealed that glutamine metabolism was increased in RA-FLS. RA-FLS proliferation was reduced under glutamine-deprived, but not glucose-deprived, conditions. Cell growth of RA-FLS was inhibited by GLS1 siRNA transfection or GLS1 inhibitor treatment. Treating RA-FLS with either interleukin-17 or platelet-derived growth factor resulted in increased GLS1 levels. Compound 968 ameliorated the autoimmune arthritis and decreased the number of Ki-67-positive synovial cells in SKG mice. CONCLUSIONS: Our results suggested that glutamine metabolism is involved in the pathogenesis of RA and that GLS1 plays an important role in regulating RA-FLS proliferation, and may be a novel therapeutic target for RA.


Subject(s)
Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Fibroblasts/pathology , Glutaminase/metabolism , Synoviocytes/pathology , Animals , Arthritis, Experimental/enzymology , Arthritis, Rheumatoid/enzymology , Blotting, Western , Cell Proliferation/physiology , Female , Fibroblasts/enzymology , Gene Knockdown Techniques , Glutamine/metabolism , Immunohistochemistry , Mice , Real-Time Polymerase Chain Reaction , Synoviocytes/enzymology
17.
J Transl Med ; 15(1): 31, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28193225

ABSTRACT

BACKGROUND: Leflunomide is a low-molecular-weight compound that is widely used in the treatment of rheumatoid arthritis. Although leflunomide is thought to act through the inhibition of the de novo pyrimidine synthesis, the molecular mechanism of the drug remains largely unknown. We investigated the antiarthritis effects and mechanisms of action of the active metabolite of leflunomide, A77 1726, in interleukin-1 receptor antagonist-knockout (IL-1Ra-KO) mice. METHODS: 14- to 15-week-old male IL-1Ra-KO mice were treated with 10 or 30 mg/kg A77 1726 via intraperitoneal injection three times per week for 6 weeks. The effects of A77 1726 on arthritis severities were assessed by clinical scoring and histological analysis. The serum concentrations of IL-1ß, tumor necrosis factor-α (TNF-α), and malondialdehyde were measured by enzyme-linked immunosorbent assay. Histologic analysis of the joints was performed using Safranin O, and immunohistochemical staining. The frequencies of interleukin-17-producing CD4+ T (Th17) cells were analyzed by flow cytometry. Heme oxygenase-1 (HO-1) expression in splenic CD4+ T cells isolated from A77 1726-treated arthritis mice were assessed by western blotting. RESULTS: A77 1726 treatment induced heme oxygenase-1 (HO-1) in Jurkat cells and primary mouse T cells. Interestingly, A77 1726 inhibited Th17 cell differentiation. In vivo, A77 1726 reduced the clinical arthritis severity of histological inflammation and cartilage destruction. The joints isolated from A77 1726-treated mice showed decreased expression of inducible nitric oxide synthase, nitrotyrosine, TNF-α, and IL-1ß. The serum levels of TNF-α, IL-1ß, and malondialdehyde were also decreased in A77 1726-treated mice. Whereas the number of Th17 cells in spleens was decreased in A77 1726-treated arthritis mice, a significant increase in the number of Treg cells in spleens was observed. Interestingly, HO-1 expression was significantly higher in splenic CD4+ T cells isolated from A77 1726-treated mice compared with those from vehicle-treated mice, whereas HO-1 expression of splenic non-CD4+ T cells did not differ between groups. CONCLUSION: The inhibitory effects of A77 1726 on joint inflammation and oxidative stress in autoimmune arthritis may be associated with HO-1 induction in CD4+ T cells.


Subject(s)
Aniline Compounds/therapeutic use , Arthritis, Experimental/complications , Arthritis, Experimental/drug therapy , Heme Oxygenase-1/metabolism , Hydroxybutyrates/therapeutic use , Inflammation/complications , Inflammation/drug therapy , Isoxazoles/metabolism , Aniline Compounds/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/enzymology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Cell Differentiation/drug effects , Crotonates , Forkhead Transcription Factors/metabolism , Humans , Hydroxybutyrates/pharmacology , Inflammation/enzymology , Jurkat Cells , Leflunomide , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitriles , Oxidative Stress/drug effects , Signal Transduction/drug effects , Spleen/pathology , Th17 Cells/cytology , Toluidines , Tyrosine/analogs & derivatives , Tyrosine/metabolism
18.
Bioorg Med Chem Lett ; 26(22): 5562-5567, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27789138

ABSTRACT

A series of furano[3,2-d]pyrimidine Syk inhibitors were synthesized and optimized for their enzyme potency and selectivity versus other kinases. In addition, ADME properties were assessed and compounds were prepared with optimized profiles for in vivo experiments. Compound 23 was identified as having acceptable pharmacokinetic properties and demonstrated efficacy in a rat collagen induced arthritis model.


Subject(s)
Arthritis, Experimental/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Syk Kinase/antagonists & inhibitors , Animals , Arthritis, Experimental/enzymology , Dogs , Furans/chemical synthesis , Furans/chemistry , Furans/pharmacology , Furans/therapeutic use , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Syk Kinase/metabolism
19.
Indian J Pharmacol ; 48(4): 441-444, 2016.
Article in English | MEDLINE | ID: mdl-27756958

ABSTRACT

OBJECTIVES: The aim of the present work was to study the anti-inflammatory and anti-arthritic activities of petroleum ether extract of fenugreek seeds. MATERIALS AND METHODS: Fenugreek seed powder was extracted in petroleum ether by cold maceration. This fenugreek seed petroleum ether extract (FSPEE) was analyzed by gas-liquid chromatography (GLC) and tested on rats against carrageenan and formaldehyde-induced paw edema, complete Freund's adjuvant (CFA)-induced arthritis and cotton pellet-induced granuloma. Changes in serum glutamic oxaloacetic tansaminase (SGOT), serum glutamate-pyruvate transaminase (SGPT), and alkaline phosphatase (ALP) activities in liver and serum were also studied in cotton pellet-induced arthritic rats. Data were analyzed by Student's t-test. P <0.05 was considered statistically significant. RESULTS: GLC of FSPEE showed oleic (33.61%), linoleic (40.37%), and linolenic (12.51%) acids. With 0.5 mL/kg FSPEE treatment, there was 37% (P < 0.05) and 85% (P < 0.05) reduction in inflammation of the paw in carrageenan and formaldehyde-induced paw edema. In CFA-induced arthritis, a biphasic increase in paw volume followed by decrease was seen. There was 42.5% (P < 0.01) reduction in the weight of cotton pellets and significant (P < 0.01) reductions in the elevated SGPT and ALP activities in serum and liver of FSPEE (0.5 mL/kg) treated rats. CONCLUSION: Thus, petroleum ether extract of fenugreek seeds has significant anti-inflammatory and anti-arthritic activities which are due to the presence of linolenic and linoleic acids.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Plant Extracts/therapeutic use , Seeds/chemistry , Trigonella/chemistry , Alkanes/chemistry , Animals , Anti-Inflammatory Agents/isolation & purification , Arthritis, Experimental/blood , Arthritis, Experimental/drug therapy , Arthritis, Experimental/enzymology , Chromatography, Gas , Edema/blood , Edema/drug therapy , Edema/enzymology , Female , Granuloma, Foreign-Body/blood , Granuloma, Foreign-Body/drug therapy , Granuloma, Foreign-Body/enzymology , Liver/drug effects , Liver/enzymology , Male , Plant Extracts/isolation & purification , Rats, Wistar
20.
Biochem Pharmacol ; 106: 56-69, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26883061

ABSTRACT

Class IA phosphoinositide 3-kinases (PI3Ks) are essential to function of normal and tumor cells, and to modulate immune responses. T lymphocytes express high levels of p110α and p110δ class IA PI3K. Whereas the functioning of PI3K p110δ in immune and autoimmune reactions is well established, the role of p110α is less well understood. Here, a novel dual p110α/δ inhibitor (ETP-46321) and highly specific p110α (A66) or p110δ (IC87114) inhibitors have been compared concerning T cell activation in vitro, as well as the effect on responses to protein antigen and collagen-induced arthritis in vivo. In vitro activation of naive CD4(+) T lymphocytes by anti-CD3 and anti-CD28 was inhibited more effectively by the p110δ inhibitor than by the p110α inhibitor as measured by cytokine secretion (IL-2, IL-10, and IFN-γ), T-bet expression and NFAT activation. In activated CD4(+) T cells re-stimulated through CD3 and ICOS, IC87114 inhibited Akt and Erk activation, and the secretion of IL-2, IL-4, IL-17A, and IFN-γ better than A66. The p110α/δ inhibitor ETP-46321, or p110α plus p110δ inhibitors also inhibited IL-21 secretion by differentiated CD4(+) T follicular (Tfh) or IL-17-producing (Th17) helper cells. In vivo, therapeutic administration of ETP-46321 significantly inhibited responses to protein antigen as well as collagen-induced arthritis, as measured by antigen-specific antibody responses, secretion of IL-10, IL-17A or IFN-γ, or clinical symptoms. Hence, p110α as well as p110δ Class IA PI3Ks are important to immune regulation; inhibition of both subunits may be an effective therapeutic approach in inflammatory autoimmune diseases like rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , CD4-Positive T-Lymphocytes/drug effects , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Subunits/antagonists & inhibitors , Pyrazines/pharmacology , Animals , Antibodies/pharmacology , Arthritis, Experimental/enzymology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/genetics , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/immunology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Gene Expression , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Lymph Nodes/drug effects , Lymph Nodes/enzymology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Protein Subunits/genetics , Protein Subunits/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL