Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
Appl Microbiol Biotechnol ; 108(1): 397, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922350

ABSTRACT

Functional M cells are differentiated by receptor activator of NF-κB ligand (RANKL) and capture of luminal antigens to initiate immune responses. We aimed to use postbiotic-based recombinant chicken RANKL (cRANKL) to promote M cell differentiation and test the efficacy of oral vaccines. Chicks were divided into three groups that were administered phosphate-buffered saline (PBS), cell extracts of wild-type Lactococcus lactis subsp. lactis IL1403 (WT_CE), or cell extracts of recombinant L. lactis expressing cRANKL (cRANKL_CE). The expression of the M cell marker was measured, and the gut microbiome was profiled. The efficiency of the infectious bursal disease (IBD) vaccine was tested after 12 consecutive days of administering cRANKL_CE. The chickens that were administered cRANKL_CE (p = 0.038) had significantly higher Annexin A5 (ANXA5) mRNA expression levels than those in the PBS group (PBS vs. WT_CE, p = 0.657). In the gut microbiome analysis, no significant changes were observed. However, the relative abundance of Escherichia-Shigella was negatively correlated (r = - 0.43, p = 0.019) with ANXA5 mRNA expression in Peyer's patches. cRANKL_CE/IBD (p = 0.018) had significantly higher IBD-specific faecal IgA levels than PBS/IBD (PBS/IBD vs. WT_CE/IBD, p = 0.217). Postbiotic-based recombinant cRANKL effectively improved the expression of M cell markers and the efficiency of oral vaccines. No significant changes were observed in the gut microbiome after administration of postbiotic-based recombinant cRANKL. This strategy can be used for the development of feed additives and adjuvants. KEY POINTS: • Postbiotic-based recombinant cRANKL enhanced the expression of ANXA5 in chicken. • The relative abundance of Escherichia-Shigella was negatively correlated with ANXA5 expression. • Postbiotic-based recombinant cRANKL effectively improved the efficiency of oral vaccine.


Subject(s)
Chickens , Gastrointestinal Microbiome , Lactococcus lactis , RANK Ligand , Recombinant Proteins , Animals , Chickens/immunology , Administration, Oral , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Lactococcus lactis/immunology , RANK Ligand/immunology , RANK Ligand/genetics , RANK Ligand/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/administration & dosage , Birnaviridae Infections/prevention & control , Birnaviridae Infections/immunology , Birnaviridae Infections/veterinary , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Poultry Diseases/microbiology , Infectious bursal disease virus/immunology , Infectious bursal disease virus/genetics , Cell Differentiation , Peyer's Patches/immunology
2.
Expert Opin Biol Ther ; 23(8): 705-715, 2023.
Article in English | MEDLINE | ID: mdl-36892190

ABSTRACT

BACKGROUND: MW031 is a biosimilar candidate of denosumab (Prolia®). This study aimed to compare the pharmacokinetics, pharmacodynamics, safety and immunogenicity of MW031 to denosumab in healthy Chinese participants. RESEARCH DESIGN AND METHODS: In this single-center, randomized, double-blind, parallel-controlled, single-dose trial, participants were given 60 mg MW031 (N = 58) or denosumab (N = 61) by subcutaneous injection and observed for 140 days. The primary endpoint was the bioequivalence of PK parameters (Cmax, AUC0-∞), and secondary endpoints including PD parameter, safety, and immunogenicity. RESULTS: A comparison of main PK parameters showed that the geometric mean ratios (GMR) (90% confidence intervals [CIs]) of AUC0-∞ and Cmax for MW031 over denosumab were 105.48% (98.96%, 112.43%) and 98.58% (92.78%, 104.75%), respectively. The inter-CV values of AUC0-∞ and Cmax for MW031 ranged from 19.9% to 23.1%. PD parameter (sCTX) in the MW031 and denosumab groups were similar, and the positivity rates of immunogenicity were 0% in both groups. This study also showed similar safety profiles in both groups, and there were no drug-related, high-incidence and previously unreported adverse reactions. CONCLUSION: This trial confirmed similar pharmacokinetic profiles of MW031 and denosumab in healthy male participants, and pharmacodynamic profile, immunogenicity and safety were comparable for both drugs. TRIAL REGISTRATION: NCT04798313; CTR20201149.


Subject(s)
Antibodies, Monoclonal, Humanized , Biosimilar Pharmaceuticals , Denosumab , Humans , Male , Area Under Curve , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/metabolism , Biosimilar Pharmaceuticals/pharmacokinetics , Denosumab/adverse effects , Denosumab/immunology , Denosumab/pharmacokinetics , Double-Blind Method , East Asian People , Healthy Volunteers , Therapeutic Equivalency , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , RANK Ligand/antagonists & inhibitors , RANK Ligand/immunology , Injections, Subcutaneous
3.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830316

ABSTRACT

Dental calculus (DC) is a common deposit in periodontitis patients. We have previously shown that DC contains both microbial components and calcium phosphate crystals that induce an osteoclastogenic cytokine IL-1ß via the NLRP3 inflammasome in macrophages. In this study, we examined the effects of cytokines produced by mouse macrophages stimulated with DC on osteoclastogenesis. The culture supernatants from wild-type (WT) mouse macrophages stimulated with DC accelerated osteoclastogenesis in RANKL-primed mouse bone marrow macrophages (BMMs), but inhibited osteoclastogenesis in RANKL-primed RAW-D cells. WT, but not NLRP3-deficient, mouse macrophages stimulated with DC produced IL-1ß and IL-18 in a dose-dependent manner, indicating the NLRP3 inflammasome-dependent production of IL-1ß and IL-18. Both WT and NLRP3-deficient mouse macrophages stimulated with DC produced IL-10, indicating the NLRP3 inflammasome-independent production of IL-10. Recombinant IL-1ß accelerated osteoclastogenesis in both RANKL-primed BMMs and RAW-D cells, whereas recombinant IL-18 and IL-10 inhibited osteoclastogenesis. These results indicate that DC induces osteoclastogenic IL-1ß in an NLRP3 inflammasome-dependent manner and anti-osteogenic IL-18 and IL-10 dependently and independently of the NLRP3 inflammasome, respectively. DC may promote alveolar bone resorption via IL-1ß induction in periodontitis patients, but suppress resorption via IL-18 and IL-10 induction in some circumstances.


Subject(s)
Dental Calculus/genetics , Interleukin-10/genetics , Interleukin-18/genetics , Interleukin-1beta/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Osteogenesis/genetics , Alveolar Bone Loss/genetics , Alveolar Bone Loss/immunology , Alveolar Bone Loss/pathology , Animals , Cell Line , Culture Media, Conditioned/pharmacology , Dental Calculus/immunology , Dental Calculus/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-10/immunology , Interleukin-10/pharmacology , Interleukin-18/immunology , Interleukin-18/pharmacology , Interleukin-1beta/immunology , Interleukin-1beta/pharmacology , Macrophage Activation , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Osteoclasts/immunology , Osteoclasts/pathology , Osteogenesis/immunology , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/pathology , Primary Cell Culture , RANK Ligand/genetics , RANK Ligand/immunology , Signal Transduction
4.
Int Immunol ; 33(11): 573-585, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34498703

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are tissue-resident cells that play different roles in different organs by sensing surrounding environmental factors. Initially, it was thought that ILC2s in bone marrow (BM) are progenitors for systemic ILC2s, which migrate to other organs and acquire effector functions. However, accumulating evidence that ILC2s differentiate in peripheral tissues suggests that BM ILC2s may play a specific role in the BM as a unique effector per se. Here, we demonstrate that BM ILC2s highly express the receptor activator of nuclear factor κB ligand (RANKL), a robust cytokine for osteoclast differentiation and activation, and RANKL expression on ILC2s is up-regulated by interleukin (IL)-2, IL-7 and all-trans retinoic acid (ATRA). BM ILC2s co-cultured with BM-derived monocyte/macrophage lineage cells (BMMs) in the presence of IL-7 induce the differentiation of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts in a RANKL-dependent manner. In contrast, BM ILC2s stimulated with IL-33 down-regulate RANKL expression and convert BMMs differentiation into M2 macrophage-like cells rather than osteoclasts by granulocyte macrophage colony-stimulating factor (GM-CSF) and IL-13 production. Intravital imaging using two-photon microscopy revealed that a depletion of ILC2s prominently impaired in vivo osteoclast activity in an IL-7 plus ATRA-induced bone loss mouse model. These results suggest that ILC2s regulate osteoclast activation and contribute to bone homeostasis in both steady state and IL-33-induced inflammation.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunity, Innate/immunology , Interleukin-13/immunology , Lymphocytes/immunology , Osteoclasts/immunology , RANK Ligand/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques , Inflammation/immunology , Interleukin-13/biosynthesis , Lymphocytes/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Osteogenesis/immunology
5.
J Clin Invest ; 131(18)2021 09 15.
Article in English | MEDLINE | ID: mdl-34343136

ABSTRACT

IL-1ß is a proinflammatory mediator with roles in innate and adaptive immunity. Here we show that IL-1ß contributes to autoimmune arthritis by inducing osteoclastogenic capacity in Tregs. Using mice with joint inflammation arising through deficiency of the IL-1 receptor antagonist (Il1rn-/-), we observed that IL-1ß blockade attenuated disease more effectively in early arthritis than in established arthritis, especially with respect to bone erosion. Protection was accompanied by a reduction in synovial CD4+Foxp3+ Tregs that displayed preserved suppressive capacity and aerobic metabolism but aberrant expression of RANKL and a striking capacity to drive RANKL-dependent osteoclast differentiation. Both Il1rn-/- Tregs and wild-type Tregs differentiated with IL-1ß accelerated bone erosion upon adoptive transfer. Human Tregs exhibited analogous differentiation, and corresponding RANKLhiFoxp3+ T cells could be identified in rheumatoid arthritis synovial tissue. Together, these findings identify IL-1ß-induced osteoclastogenic Tregs as a contributor to bone erosion in arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Interleukin-1beta/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/pathology , Cell Differentiation/immunology , Female , Humans , Interleukin 1 Receptor Antagonist Protein/deficiency , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Osteoclasts/immunology , Osteoclasts/pathology , Osteogenesis/immunology , RANK Ligand/immunology , T-Lymphocytes, Regulatory/metabolism
6.
Sci Rep ; 11(1): 12186, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108600

ABSTRACT

Prostate cancer (PCa) morbidity in the majority of patients is due to metastatic events, which are a clinical obstacle. Therefore, a better understanding of the mechanism underlying metastasis is imperative if we are to develop novel therapeutic strategies. Receptor activator of nuclear factor kappa-B (NF-κB) ligand (RANKL) regulates bone remodelling. Thus, agents that suppress RANKL signalling may be useful pharmacological treatments. Here, we used preclinical experimental models to investigate whether an inactive form of RANKL affects bone metastasis in RANKL-induced PCa. RANKL was associated with epithelial-mesenchymal transition (EMT) and expression of metastasis-related genes in PC3 cells. Therefore, we proposed a strategy to induce anti-cytokine antibodies using mutant RANKL as an immunogen. RANKL promoted migration and invasion of PC3 cells through EMT, and induced a significant increase in binding of ß-catenin to TCF-4, an EMT-induced transcription factor in PCa cells, via mitogen-activated protein kinase and ß-catenin/TCF-4 signalling. Thus, RANKL increased EMT and the metastatic properties of PC3 cells, suggesting a role as a therapeutic target to prevent PCa metastasis. Treatment with mutant RANKL reduced EMT and metastasis of PC3 PCa cells in an experimental metastasis model. Thus, mutant RANKL could serve as a potential vaccine to prevent and treat metastatic PCa.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Neoplasms/prevention & control , Epithelial-Mesenchymal Transition , Prostatic Neoplasms/drug therapy , RANK Ligand/antagonists & inhibitors , Animals , Apoptosis , Bone Neoplasms/immunology , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Movement , Cell Proliferation , Humans , Immunization , Male , Mice , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RANK Ligand/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33720039

ABSTRACT

In rheumatoid arthritis (RA), osteoclastic bone resorption causes structural joint damage as well as periarticular and systemic bone loss. Periarticular bone loss is one of the earliest indices of RA, often preceding the onset of clinical symptoms via largely unknown mechanisms. Excessive osteoclastogenesis induced by receptor activator of NF-κB ligand (RANKL) expressed by synovial fibroblasts causes joint erosion, whereas the role of RANKL expressed by lymphocytes in various types of bone damage has yet to be elucidated. In the bone marrow of arthritic mice, we found an increase in the number of RANKL-expressing plasma cells, which displayed an ability to induce osteoclastogenesis in vitro. Genetic ablation of RANKL in B-lineage cells resulted in amelioration of periarticular bone loss, but not of articular erosion or systemic bone loss, in autoimmune arthritis. We also show conclusive evidence for the critical contribution of synovial fibroblast RANKL to joint erosion in collagen-induced arthritis on the arthritogenic DBA/1J background. This study highlights the importance of plasma-cell RANKL in periarticular bone loss in arthritis and provides mechanistic insight into the early manifestation of bone lesion induced by autoimmunity.


Subject(s)
Arthritis, Experimental/immunology , Osteogenesis/immunology , Plasma Cells/immunology , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Resorption/immunology , Bone Resorption/pathology , Female , Humans , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Plasma Cells/pathology , RANK Ligand/deficiency , RANK Ligand/genetics , RANK Ligand/immunology , Synovial Membrane/immunology , Synovial Membrane/pathology
8.
Biosci Trends ; 14(5): 342-348, 2020 Nov 04.
Article in English | MEDLINE | ID: mdl-32908076

ABSTRACT

The human immune system has evolved to recognize and eradicate pathogens, a process that is known as "host defense". If, however, the immune system does not work properly, it can mistakenly attack the body's own tissues and induce autoimmune diseases. Rheumatoid arthritis (RA) is such an autoimmune disease in which the synovial joints are predominately attacked by the immune system. Moreover, RA is associated with bone destruction and joint deformity. Although biologic agents have propelled RA treatment forward dramatically over the past 30 years, a considerable number of patients with RA still experience progressive bone damage and joint disability. That is to be expected since current RA therapies are all intended to halt inflammation but not to alleviate bone destruction. A better understanding of bone erosions is crucial to developing a novel strategy to treat RA-associated erosions. This review provides insights into RA-associated bone destruction and perspectives for future clinical interventions.


Subject(s)
Arthritis, Rheumatoid/complications , Biological Factors/pharmacology , Bone Density Conservation Agents/pharmacology , Osteoporosis/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Biological Factors/therapeutic use , Bone Density Conservation Agents/therapeutic use , Cadherins/pharmacology , Cadherins/therapeutic use , Humans , Joint Capsule/drug effects , Joint Capsule/immunology , Joint Capsule/pathology , Osteoblasts/drug effects , Osteoblasts/immunology , Osteoclasts/drug effects , Osteoclasts/immunology , Osteogenesis/drug effects , Osteogenesis/immunology , Osteoporosis/drug therapy , Osteoporosis/pathology , Proteins/antagonists & inhibitors , Proteins/metabolism , RANK Ligand/antagonists & inhibitors , RANK Ligand/immunology , RANK Ligand/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Signal Transduction/immunology , Synovial Fluid/drug effects , Synovial Fluid/immunology
9.
Sci Rep ; 10(1): 13751, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792620

ABSTRACT

Bone tissues have trabecular bone with a high bone turnover and cortical bone with a low turnover. The mechanisms by which the turnover rate of these bone tissues is determined remain unclear. Osteocytes secrete sclerostin, a Wnt/ß-catenin signaling antagonist, and inhibit bone formation. We found that sclerostin expression in cortical bone is more marked than in trabecular bone in Sost reporter mice. Leukemia inhibitory factor (LIF) secreted from osteoclasts reportedly suppressed sclerostin expression and promoted bone formation. Here, we report that osteoclasts downregulate sclerostin expression in trabecular bone and promote bone turnover. Treatment of C57BL/6 mice with an anti-RANKL antibody eliminated the number of osteoclasts and LIF-positive cells in trabecular bone. The number of sclerostin-positive cells was increased in trabecular bone, while the number of ß-catenin-positive cells and bone formation were decreased in trabecular bone. Besides, Tnfsf11 heterozygous (Rankl+/-) mice exhibited a decreased number of LIF-positive cells and increased number of sclerostin-positive cells in trabecular bone. Rankl+/- mice exhibited a decreased number of ß-catenin-positive cells and reduced bone formation in trabecular bone. Furthermore, in cultured osteoclasts, RANKL stimulation increased Lif mRNA expression, suggesting that RANKL signal increased LIF expression. In conclusion, osteoclasts downregulate sclerostin expression and promote trabecular bone turnover.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Remodeling/physiology , Cancellous Bone/metabolism , Osteoclasts/metabolism , RANK Ligand/genetics , Animals , Antibodies/immunology , Biomarkers, Tumor/metabolism , Bone Density , Cortical Bone/metabolism , Leukemia Inhibitory Factor/metabolism , Male , Mice , Mice, Inbred C57BL , RANK Ligand/immunology , Up-Regulation/genetics , Wnt Signaling Pathway/physiology
10.
Front Immunol ; 11: 886, 2020.
Article in English | MEDLINE | ID: mdl-32477357

ABSTRACT

Macrophages host Leishmania major infection, which causes cutaneous Leishmaniasis in humans. In the murine model, resistance to infection depends on the host immunity mediated by CD4 T-cell cytokines and macrophages. In association to other stimuli, the Th1 cytokine IFN-γ induces NO-mediated microbial killing by M1/classically-activated macrophages. By contrast, the Th2 cytokine IL-4 promotes M2/alternatively activated macrophages, which express arginase-1 and shelter infection. Other cytokines, such as RANKL, might also participate in the crosstalk between T cells and macrophages to restrict parasite infection. RANKL and its receptor RANK are known to play an essential role in bone remodeling, by inducing osteoclatogenesis. It has also been shown that RANKL stimulates antigen-presenting cells, such as DCs and macrophages, to enhance T cell responses. Here we investigated how RANKL directly modulates the effector macrophage phenotypes and immunity to L. major parasites. We found that inflammatory peritoneal macrophages from B6 mice express RANK and M2 features, such as CD301 (MGL) and CD206 (mannose receptor). Nonetheless, treatment with RANKL or IFN-γ induced macrophage differentiation into more mature F40/80hi macrophages able to produce IL-12 and TNF-α. In parallel, macrophages treated with RANKL, IFN-γ, or RANKL along with IFN-γ progressively downregulated the expression of the M2 hallmarks MGL, arginase-1, and CCL17. Moreover, a synergism between IFN-γ and RANKL enhanced inducible NO synthase (iNOS) expression and NO production by macrophages. These results are consistent with the idea that RANKL helps IFN-γ to induce a M2-like to M1 phenotype shift. Accordingly, concomitant treatment with RANKL and IFN-γ promoted macrophage-mediated immunity to L. major, by inducing NO and ROS-dependent parasite killing. Furthermore, by cooperating with IFN-γ, endogenous RANKL engages CD4 T-cell help toward L. major-infected macrophages to upregulate M1 and Th1 cytokine responses. Therefore, RANKL, in combination with IFN-γ, is a potential local therapeutic tool to improve immune responses in Leishmaniasis, by skewing M2-like into effector M1 macrophages.


Subject(s)
Cell Differentiation/immunology , Macrophage Activation , Macrophages/immunology , Macrophages/parasitology , RANK Ligand/immunology , Animals , Leishmania major , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction
11.
J Agric Food Chem ; 68(39): 10664-10677, 2020 Sep 30.
Article in English | MEDLINE | ID: mdl-32530618

ABSTRACT

Glycine tabacina (Labill.) Benth is an edible medicinal herb for rheumatoid arthritis (RA) treatment in folk medicine. Current phytochemical research on this dried herb led to the isolation of eight new coumestans, named glytabastan A-H (1-8), and twenty-three known compounds 9-31. Their structures were elucidated using spectroscopic methods. The antiarthritic activities of all isolates were evaluated, and the results showed that coumestans 1-6 and 8-10 could inhibit arthritic inflammation in vitro, while coumestans 1, 2, 9, and 10 significantly blocked the osteoclastogenesis induced by receptor activator of nuclear factor (NF) κB ligand (RANKL). Moreover, network pharmacological analysis revealed that the anti-RA effect of G. tabacina involved multitargets, multipathways such as PI3K/Akt and MAPK signaling pathways, and various biological processes such as inflammatory response and cytokine-mediated signaling pathways. These results suggested that this species and its novel coumestans could serve as potential antiarthritic agents for functional food or medicinal use.


Subject(s)
Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Fabaceae/chemistry , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/physiopathology , Drugs, Chinese Herbal/isolation & purification , Humans , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/immunology , Mice , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/immunology , Osteogenesis/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , RANK Ligand/genetics , RANK Ligand/immunology , RAW 264.7 Cells , Synovial Membrane/drug effects , Synovial Membrane/immunology
13.
Proc Jpn Acad Ser B Phys Biol Sci ; 96(4): 159-169, 2020.
Article in English | MEDLINE | ID: mdl-32281551

ABSTRACT

Bone is a critically important part of the skeletal system that is essential for body support and locomotion. The immune system protects against pathogens and is active in host defense. These two seemingly distinct systems in fact interact with each other, share molecules and create a collaborative regulatory system called the "osteoimmune system". The most representative osteoimmune molecule is receptor activator of NF-κB ligand (RANKL), which plays multiple roles in the osteoimmune system under both physiological and pathological conditions such as rheumatoid arthritis and cancer metastasis to bone. Based on accumulating evidence for such mutual dependence, it is concluded that the relationship between bone and the immune system did not develop by accident but as a necessary consequence of evolution. Here I describe the history of and recent advances in osteoimmunology, providing a perspective in the contexts of both science and medicine.


Subject(s)
Allergy and Immunology/history , Arthritis, Rheumatoid/immunology , Bone Neoplasms/immunology , Bone and Bones/immunology , Neoplasm Proteins/immunology , RANK Ligand/immunology , Animals , Arthritis, Rheumatoid/pathology , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Bone and Bones/pathology , History, 20th Century , History, 21st Century , Humans , Neoplasm Metastasis
14.
Article in English | MEDLINE | ID: mdl-32338225

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune inflammatory disease comparing the inflammation of synovium. Macrophage-like synoviocytes and fibroblast-like synoviocytes (synoviocytes) are crucial ingredients of synovium. Therein, a lot of research has focused on synoviocytes. Researches demonstrated that TLR1, TLR2, TLR3, TLR4, TLR5, TLR6 TLR7 and TLR9 are expressed in synoviocyte. Additionally, the expression of TLR2, TLR3, TLR4 and TLR5 is increased in RA synoviocyte. In this paper, we review the exact role of TLR2, TLR3, TLR4 and TLR5 participate in regulating the production of inflammatory factors in RA synoviocyte. Furthermore, we discuss the role of vasoactive intestinal peptide (VIP), MicroRNA, Monome of Chinese herb and other cells (Monocyte and T cell) influence the function of synoviocyte by regulating TLRs. The activation of toll-like receptors (TLRs) in synoviocyte leads to the aggravation of arthritis, comparing with angiogenesis and bone destruction. Above all, TLRs are promising targets for managing RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Synoviocytes/physiology , Toll-Like Receptors/physiology , Animals , Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Humans , MicroRNAs/immunology , MicroRNAs/metabolism , RANK Ligand/immunology , RANK Ligand/metabolism , Synovial Membrane/drug effects , Synovial Membrane/physiology , Synoviocytes/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vasoactive Intestinal Peptide/immunology , Vasoactive Intestinal Peptide/metabolism
15.
Eur J Pharmacol ; 879: 173129, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32339516

ABSTRACT

The main aims of this study were: (1) to investigate whether a blockade of the interaction between the receptor activator of nuclear factor-κB (NF-ĸB) ligand (RANKL) and its receptor RANK may have potential as a novel therapeutic strategy for allergic asthma; (2) to compare the efficacies of the blockade of RANKL/RANK interaction as well as the blockade of NF-κB inhibitor kinase (IKK) and of NF-κB translocation to the nucleus, also in comparison with glucocorticosteroid treatment, in terms of the development of a mouse model of allergic airway inflammation (AAI) and accompanying immune response. The blockade of each of the targets fully prevented the development of AAI. All the tested therapeutic strategies seemed to have a certain advantage over glucocorticosteroids with regard to counteracting the development of AAI. Prevention of the activation and clonal expansion of CD4+ effector T (Teff) cells in the mediastinal lymph nodes (MLNs) constitutes a fundamental event underlying the anti-asthmatic effect induced by the blockade of IKK, NF-κB translocation or of RANKL/RANK interaction. The results indicate that attenuation of the CD11b+CD103-CD11chigh dendritic cell response in the MLNs is an initial but not the main mechanism responsible for this effect. In turn, the direct anti-proliferative action on CD4+ Teff cells seems to constitute the chief mechanism responsible for the anti-asthmatic effect of all the tested therapeutic strategies. A clinical implication is that local inhibition of RANKL/RANK interaction achieved via inhalatory administration of a RANKL antagonist can be considered as a novel therapeutic strategy in treatment of allergic asthma.


Subject(s)
Asthma/drug therapy , NF-kappa B/antagonists & inhibitors , RANK Ligand/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/antagonists & inhibitors , Animals , Asthma/immunology , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Lung/drug effects , Lung/immunology , Lung/pathology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice, Inbred BALB C , NF-kappa B/immunology , Ovalbumin , RANK Ligand/immunology , Receptor Activator of Nuclear Factor-kappa B/immunology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
16.
Vet Microbiol ; 242: 108604, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32122610

ABSTRACT

Here, we examined the efficacy of are combinant subunit antigen-based oral vaccine for preventing porcine epidemic diarrhea virus (PEDV). First, we generated a soluble recombinant partial spike S1 protein (aP2) from PEDV in E. coli and then evaluated the utility of aP2 subunit vaccine-loaded hydroxypropyl methylcellulose phthalate microspheres (HPMCP) and RANKL-secreting L. lactis (LLRANKL) as a candidate oral vaccine in pregnant sows. Pregnant sows were vaccinated twice (with a 2 week interval between doses) at 4 weeks before farrowing. Titers of virus-specific IgA antibodies in colostrum, and neutralizing antibodies in serum, of sows vaccinated with HPMCP (aP2) plus LL RANKL increased significantly at 4 weeks post-first vaccination. Furthermore, the survival rate of newborn suckling piglets delivered by sows vaccinated with HPMCP (aP2) plus LL RANKL was similar to that of piglets delivered by sows vaccinated with a commercial killed porcine epidemic diarrhea virus (PED) vaccine. The South Korean government promotes a PED vaccine program (live-killed-killed) to increase the titers of IgA and IgG antibodies in pregnant sows and prevent PEDV. The oral vaccine strategy described herein, which is based on a safe and efficient recombinant subunit antigen, is an alternative PED vaccination strategy that could replace the traditional strategy, which relies on attenuated live oral vaccines or artificial infection with virulent PEDV.


Subject(s)
Coronavirus Infections/veterinary , Lactobacillus/immunology , Methylcellulose/analogs & derivatives , RANK Ligand/immunology , Swine Diseases/prevention & control , Viral Vaccines/immunology , Administration, Oral , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Colostrum/immunology , Coronavirus Infections/prevention & control , Female , Methylcellulose/administration & dosage , Microspheres , Porcine epidemic diarrhea virus , Pregnancy , RANK Ligand/administration & dosage , Swine , Swine Diseases/virology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/administration & dosage
17.
Front Immunol ; 11: 588522, 2020.
Article in English | MEDLINE | ID: mdl-33613513

ABSTRACT

IL-17 is critical in lung lymphoid neogenesis in COPD, but the cellular and molecular mechanisms remain to be elucidated. Receptor activator of nuclear factor-κB ligand (RANKL) functions in lymphoid follicle formation in other organs, whether it is involved in IL-17A-dependent lymphoid neogenesis in COPD is unknown. To elucidate the expression and functional role of IL-17A/RANKL pathway in COPD. We first quantified and localized RANKL, its receptor RANK and IL-17A in lungs of patients with COPD, smokers and non-smokers. Next, IL-17A-/- and wild-type (WT) mice were exposed to air or cigarette smoke (CS) for 24 weeks, and lung lymphoid follicles and RANKL-RANK expression were measured. Lastly, we studied the in vitro biological function of RANKL pertaining to lymphoid neogenesis. We found that the expressions of RANKL-RANK and IL-17A, together with lymphoid follicles, were increased in lung tissues from patients with COPD. In WT mice exposed to CS, RANKL-RANK expressions were prominent in lung lymphoid follicles, which were absent in IL-17A-/- mice exposed to CS. In the lymphoid follicles, RANKL+ cells were identified mostly as B cells and RANK was localized in dendritic cells (DCs). In vitro IL-17A increased the expressions of RANKL in B cells and RANK in DCs, which in turn responded to RANKL stimulation by upregulation of CXCL13. Altogether, these results suggest that B lymphocyte RANKL pathway is involved in IL-17A-dependent lymphoid neogenesis in COPD.


Subject(s)
Interleukin-17/immunology , Lymphoid Tissue/immunology , Nicotiana , Pulmonary Disease, Chronic Obstructive/immunology , RANK Ligand/immunology , Smoke/adverse effects , Animals , B-Lymphocytes/immunology , Chemokine CXCL13/immunology , Dendritic Cells/immunology , Female , Humans , Interleukin-17/genetics , Lung/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Signal Transduction , Smoking/immunology
18.
Front Immunol ; 11: 623265, 2020.
Article in English | MEDLINE | ID: mdl-33552088

ABSTRACT

Thymic epithelial cells (TECs) provide essential clues for the proliferation, survival, migration, and differentiation of thymocytes. Recent advances in mouse and human have revealed that TECs constitute a highly heterogeneous cell population with distinct functional properties. Importantly, TECs are sensitive to thymic damages engendered by myeloablative conditioning regimen used for bone marrow transplantation. These detrimental effects on TECs delay de novo T-cell production, which can increase the risk of morbidity and mortality in many patients. Alike that TECs guide the development of thymocytes, reciprocally thymocytes control the differentiation and organization of TECs. These bidirectional interactions are referred to as thymic crosstalk. The tumor necrosis factor receptor superfamily (TNFRSF) member, receptor activator of nuclear factor kappa-B (RANK) and its cognate ligand RANKL have emerged as key players of the crosstalk between TECs and thymocytes. RANKL, mainly provided by positively selected CD4+ thymocytes and a subset of group 3 innate lymphoid cells, controls mTEC proliferation/differentiation and TEC regeneration. In this review, I discuss recent advances that have unraveled the high heterogeneity of TECs and the implication of the RANK-RANKL signaling axis in TEC differentiation and regeneration. Targeting this cell-signaling pathway opens novel therapeutic perspectives to recover TEC function and T-cell production.


Subject(s)
Epithelial Cells/physiology , Receptor Activator of Nuclear Factor-kappa B/immunology , Regeneration/immunology , Signal Transduction , Thymus Gland/physiology , Humans , RANK Ligand/immunology , Thymocytes/immunology
19.
Cell Death Dis ; 12(1): 45, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33414409

ABSTRACT

Glucocorticoids (GC) are widely used clinically, despite the presence of significant side effects, including glucocorticoid-induced osteoporosis (GIOP). While GC are believed to act directly on osteoblasts and osteoclasts to promote osteoporosis, the detailed underlying molecular mechanism of GC-induced osteoporosis is still not fully elucidated. Here, we show that lymphocytes play a pivotal role in regulating GC-induced osteoporosis. We show that GIOP could not be induced in SCID mice that lack T cells, but it could be re-established by adoptive transfer of splenic T cells from wild-type mice. As expected, T cells in the periphery are greatly reduced by GC; instead, they accumulate in the bone marrow where they are protected from GC-induced apoptosis. These bone marrow T cells in GC-treated mice express high steady-state levels of NF-κB receptor activator ligand (RANKL), which promotes the formation and maturation of osteoclasts and induces osteoporosis. Taken together, these findings reveal a critical role for T cells in GIOP.


Subject(s)
Glucocorticoids/adverse effects , Osteoporosis/chemically induced , Osteoporosis/immunology , T-Lymphocytes/immunology , Animals , Apoptosis/immunology , Chemokines/immunology , Dexamethasone/adverse effects , Dexamethasone/pharmacology , Female , Femur/drug effects , Femur/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Osteoporosis/metabolism , Osteoporosis/pathology , RANK Ligand/immunology , T-Lymphocytes/drug effects
20.
Behav Brain Res ; 379: 112397, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31790783

ABSTRACT

Multiple lines of evidence suggest a link between depression and osteoporosis in elderly people. Receptor activator of nuclear factor-κB ligand (RANKL) plays a role in the pathology of osteoporosis, and anti-RANKL antibody has been used in the treatment of osteoporosis. In this study, we investigated whether anti-mouse RANKL antibody could attenuate depression-like phenotypes, inflammatory bone markers and bone mineral density (BMD) in male susceptible mice after chronic social defeat stress (CSDS). We measured plasma levels of inflammatory bone markers, including osteoprotegerin (OPG), RANKL, and osteopontin. A single intravenous injection of anti-RANKL (2 mg/kg) elicited rapid antidepressant effects in CSDS susceptible mice. Furthermore, anti-RANKL significantly improved the increased plasma levels of RANKL and decreased OPG/RANKL ratio in CSDS susceptible mice. Moreover, anti-RANKL significantly attenuated the decreased BMD in CSDS susceptible mice. Interestingly, there is a positive correlation between anhedonia-like behavior and OPG/RANKL ratio in mice. These findings demonstrate that anti-RANKL may have beneficial effects in depression-like phenotype and abnormalities in bone functions of CSDS susceptible mice. It is, therefore, likely that anti-human RANKL antibody (i.e., Denosumab) would be a potential therapeutic drug for depression and osteoporosis.


Subject(s)
Autoantibodies/pharmacology , Bone Density/drug effects , Depression/drug therapy , Osteitis/drug therapy , Osteopontin/drug effects , Osteoporosis/drug therapy , Osteoprotegerin/drug effects , RANK Ligand/immunology , Social Defeat , Stress, Psychological/immunology , Animals , Autoantibodies/administration & dosage , Behavior, Animal/drug effects , Behavior, Animal/physiology , Depression/physiopathology , Disease Models, Animal , Disease Susceptibility , Male , Mice , Mice, Inbred C57BL , Osteitis/blood , Osteopontin/blood , Osteoporosis/blood , Osteoprotegerin/blood , Phenotype , RANK Ligand/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...