Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Int J Oral Sci ; 16(1): 18, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38413562

ABSTRACT

The immune-stromal cell interactions play a key role in health and diseases. In periodontitis, the most prevalent infectious disease in humans, immune cells accumulate in the oral mucosa and promote bone destruction by inducing receptor activator of nuclear factor-κB ligand (RANKL) expression in osteogenic cells such as osteoblasts and periodontal ligament cells. However, the detailed mechanism underlying immune-bone cell interactions in periodontitis is not fully understood. Here, we performed single-cell RNA-sequencing analysis on mouse periodontal lesions and showed that neutrophil-osteogenic cell crosstalk is involved in periodontitis-induced bone loss. The periodontal lesions displayed marked infiltration of neutrophils, and in silico analyses suggested that the neutrophils interacted with osteogenic cells through cytokine production. Among the cytokines expressed in the periodontal neutrophils, oncostatin M (OSM) potently induced RANKL expression in the primary osteoblasts, and deletion of the OSM receptor in osteogenic cells significantly ameliorated periodontitis-induced bone loss. Epigenomic data analyses identified the OSM-regulated RANKL enhancer region in osteogenic cells, and mice lacking this enhancer showed decreased periodontal bone loss while maintaining physiological bone metabolism. These findings shed light on the role of neutrophils in bone regulation during bacterial infection, highlighting the novel mechanism underlying osteoimmune crosstalk.


Subject(s)
Alveolar Bone Loss , Periodontitis , Humans , Mice , Animals , Neutrophils/metabolism , Neutrophils/pathology , Cytokines , Alveolar Bone Loss/microbiology , Osteogenesis , RANK Ligand
2.
Inflamm Regen ; 43(1): 44, 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37726797

ABSTRACT

BACKGROUND: The types of bone damage in rheumatoid arthritis (RA) include joint erosion, periarticular osteoporosis, and systemic osteoporosis. Janus kinase (JAK) inhibitors ameliorate inflammation and joint erosion in RA, but their effect on the three types of bone loss have not been reportedly explored in depth. We aimed to clarify how JAK inhibitors influence the various types of bone loss in arthritis by modulating osteoclastic bone resorption and/or osteoblastic bone formation. METHODS: Collagen-induced arthritis (CIA) mice were treated with a JAK inhibitor after the onset of arthritis. Micro-computed tomography (µCT) and histological analyses (bone morphometric analyses) on the erosive calcaneocuboid joint, periarticular bone (distal femur or proximal tibia), and vertebrae were performed. The effect of four different JAK inhibitors on osteoclastogenesis under various conditions was examined in vitro. RESULTS: The JAK inhibitor ameliorated joint erosion, periarticular osteopenia and systemic bone loss. It reduced the osteoclast number in all the three types of bone damage. The JAK inhibitor enhanced osteoblastic bone formation in the calcaneus distal to inflammatory synovium in the calcaneocuboid joints, periarticular region of the tibia and vertebrae, but not the inflamed calcaneocuboid joint. All the JAK inhibitors suppressed osteoclastogenesis in vitro to a similar extent in the presence of osteoblastic cells. Most of the JAK inhibitors abrogated the suppressive effect of Th1 cells on osteoclastogenesis by inhibiting IFN-γ signaling in osteoclast precursor cells, while a JAK inhibitor did not affect this effect due to less ability to inhibit IFN-γ signaling. CONCLUSIONS: The JAK inhibitor suppressed joint erosion mainly by inhibiting osteoclastogenesis, while it ameliorated periarticular osteopenia and systemic bone loss by both inhibiting osteoclastogenesis and promoting osteoblastogenesis. These results indicate that the effect of JAK inhibitors on osteoclastogenesis and osteoblastogenesis depends on the bone damage type and the affected bone area. In vitro studies suggest that while JAK inhibitors inhibit osteoclastic bone resorption, their effects on osteoclastogenesis in inflammatory environments vary depending on the cytokine milieu, JAK selectivity and cytokine signaling specificity. The findings reported here should contribute to the strategic use of antirheumatic drugs against structural damages in RA.

3.
Bone Res ; 11(1): 43, 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37563119

ABSTRACT

The bony skeleton is continuously renewed throughout adult life by the bone remodeling process, in which old or damaged bone is removed by osteoclasts via largely unknown mechanisms. Osteocytes regulate bone remodeling by producing the osteoclast differentiation factor RANKL (encoded by the TNFSF11 gene). However, the precise mechanisms underlying RANKL expression in osteocytes are still elusive. Here, we explored the epigenomic landscape of osteocytic cells and identified a hitherto-undescribed osteocytic cell-specific intronic enhancer in the TNFSF11 gene locus. Bioinformatics analyses showed that transcription factors involved in cell death and senescence act on this intronic enhancer region. Single-cell transcriptomic data analysis demonstrated that cell death signaling increased RANKL expression in osteocytic cells. Genetic deletion of the intronic enhancer led to a high-bone-mass phenotype with decreased levels of RANKL in osteocytic cells and osteoclastogenesis in the adult stage, while RANKL expression was not affected in osteoblasts or lymphocytes. These data suggest that osteocytes may utilize a specialized regulatory element to facilitate osteoclast formation at the bone surface to be resorbed by linking signals from cellular senescence/death and RANKL expression.

4.
Bone ; 168: 116675, 2023 03.
Article in English | MEDLINE | ID: mdl-36638904

ABSTRACT

Bone and immune systems mutually influence each other by sharing a variety of regulatory molecules and the tissue microenvironment. The interdisciplinary research field "osteoimmunology" has illuminated the complex and dynamic interactions between the two systems in the maintenance of tissue homeostasis as well as in the development of immune and skeletal disorders. T cells play a central role in the immune response by secreting various immune factors and stimulating other immune cells and structural cells such as fibroblasts and epithelial cells, thereby contributing to pathogen elimination and pathogenesis of immune diseases. The finding on regulation of osteoclastic bone resorption by activated CD4+ T cells in rheumatoid arthritis was one of the driving forces for the development of osteoimmunology. With advances in research on helper T cell subsets and rare lymphoid cells such as γδ T cells in the immunology field, it is becoming clear that various types of T cells exert multiple effects on bone metabolism depending on immune context. Understanding the diverse effects of T cells on bone is essential for deciphering the osteoimmune regulatory network in various biological settings.


Subject(s)
Arthritis, Rheumatoid , Bone Resorption , Humans , Bone and Bones/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , T-Lymphocytes/metabolism
5.
Nat Immunol ; 23(9): 1330-1341, 2022 09.
Article in English | MEDLINE | ID: mdl-35999392

ABSTRACT

Fibroblasts, the most abundant structural cells, exert homeostatic functions but also drive disease pathogenesis. Single-cell technologies have illuminated the shared characteristics of pathogenic fibroblasts in multiple diseases including autoimmune arthritis, cancer and inflammatory colitis. However, the molecular mechanisms underlying the disease-associated fibroblast phenotypes remain largely unclear. Here, we identify ETS1 as the key transcription factor governing the pathological tissue-remodeling programs in fibroblasts. In arthritis, ETS1 drives polarization toward tissue-destructive fibroblasts by orchestrating hitherto undescribed regulatory elements of the osteoclast differentiation factor receptor activator of nuclear factor-κB ligand (RANKL) as well as matrix metalloproteinases. Fibroblast-specific ETS1 deletion resulted in ameliorated bone and cartilage damage under arthritic conditions without affecting the inflammation level. Cross-tissue fibroblast single-cell data analyses and genetic loss-of-function experiments lent support to the notion that ETS1 defines the perturbation-specific fibroblasts shared among various disease settings. These findings provide a mechanistic basis for pathogenic fibroblast polarization and have important therapeutic implications.


Subject(s)
Arthritis, Rheumatoid , Fibroblasts , Proto-Oncogene Protein c-ets-1 , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Matrix Metalloproteinases/metabolism , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , RANK Ligand/genetics , Transcription Factors/metabolism
6.
Bone Res ; 10(1): 52, 2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35918335

ABSTRACT

Impaired locomotion has been extensively studied worldwide because those afflicted with it have a potential risk of becoming bedridden. Physical exercise at times can be an effective remedy for frailty, but exercise therapy cannot be applied in all clinical cases. Medication is safer than exercise, but there are no drugs that reinforce both muscle and bone when administered alone. Multiple medications increase the risk of adverse events; thus, there is a need for individual drugs targeting both tissues. To this end, we established a novel sequential drug screening system and identified an aminoindazole derivative, locamidazole (LAMZ), which promotes both myogenesis and osteoblastogenesis while suppressing osteoclastogenesis. Administration of this drug enhanced locomotor function, with muscle and bone significantly strengthened. Mechanistically, LAMZ induced Mef2c and PGC-1α in a calcium signaling-dependent manner. As this signaling is activated upon physical exercise, LAMZ mimics physical exercise. Thus, LAMZ is a promising therapeutic drug for locomotor diseases, including sarcopenia and osteoporosis.

7.
Nat Commun ; 13(1): 4166, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35851381

ABSTRACT

The ontogeny and fate of stem cells have been extensively investigated by lineage-tracing approaches. At distinct anatomical sites, bone tissue harbors multiple types of skeletal stem cells, which may independently supply osteogenic cells in a site-specific manner. Periosteal stem cells (PSCs) and growth plate resting zone stem cells (RZSCs) critically contribute to intramembranous and endochondral bone formation, respectively. However, it remains unclear whether there is functional crosstalk between these two types of skeletal stem cells. Here we show PSCs are not only required for intramembranous bone formation, but also for the growth plate maintenance and prolonged longitudinal bone growth. Mice deficient in PSCs display progressive defects in intramembranous and endochondral bone formation, the latter of which is caused by a deficiency in PSC-derived Indian hedgehog (Ihh). PSC-specific deletion of Ihh impairs the maintenance of the RZSCs, leading to a severe defect in endochondral bone formation in postnatal life. Thus, crosstalk between periosteal and growth plate stem cells is essential for post-developmental skeletal growth.


Subject(s)
Chondrocytes , Growth Plate , Animals , Hedgehog Proteins/genetics , Mice , Osteogenesis/genetics , Stem Cells
8.
Cytokine ; 143: 155521, 2021 07.
Article in English | MEDLINE | ID: mdl-33863633

ABSTRACT

OBJECTIVES: Our study aimed to evaluate the cytokine levels in pediatric chronic non-bacterial osteomyelitis (CNO) patients and compare these with other immune-mediated diseases and healthy controls. METHODS: In this prospective study, we included 42 children with CNO, 28 patients with non-systemic juvenile idiopathic arthritis (JIA), 17 children with insulin-dependent diabetes mellitus (IDDM), and 30 healthy age-matched controls. In each of the CNO patients and comparison groups, the levels of 14-3-3-η protein, S100A8/A9 protein, interleukin-4 (IL-4), interleukin-17 (IL-17), interleukin-18 (IL-18), interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) were measured by ELISA assay. RESULTS: All studied cytokines in the CNO patients were significantly higher than controls, and IDDM, 14-3-3-η protein, IL-18, IL-4, IL-17, IL-1ß, and TNF-α were less than in JIA patients. In the discriminant analysis, ESR, 14-3-3 protein, S100A8/A9, IL-18, IL-4, and TNF-α can discriminate CNO from JIA, and 14-3-3 protein, S100A8/A9, IL-18, IL-17, IL-4, and TNF-α can distinguish CNO from other diseases and HC. CONCLUSION: The increased level of pro-inflammatory cytokines confirms the role of monocyte-driven inflammation in CNO patients. Cytokines may prove valuable as biomarkers and potential therapeutic targets for CNO.


Subject(s)
Arthritis, Juvenile/blood , Cytokines/blood , Diabetes Mellitus, Type 1/blood , Osteomyelitis/blood , Case-Control Studies , Child , Child, Preschool , Chronic Disease , Female , Humans , Immunocompromised Host , Male , Multivariate Analysis , Sensitivity and Specificity
9.
J Bone Miner Metab ; 39(1): 71-81, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33387063

ABSTRACT

Bone metastasis involves tumor-induced osteoclast activation, resulting in skeletal tumor progression as well as skeletal disorders. Aberrant expression of receptor activator of NF-κB ligand (RANKL), an essential cytokine for osteoclast differentiation, induced by the metastatic tumor cells is responsible for the pathological bone resorption in bone metastasis. A fully human anti-RANKL neutralizing antibody has been developed to block osteoclast activation and is now used for the treatment of patients with bone metastasis and multiple myeloma. On the other hand, numerous studies have revealed that the RANKL/RANK system also contributes to primary tumorigenesis as well as metastasis through osteoclast-independent processes. Furthermore, emerging clinical and preclinical evidence has suggested anti-tumor immune effects of RANKL blockade when added to immune checkpoint inhibitor therapies. Study on the pleiotropic functions of RANKL in tumorigenesis and metastasis is now expanding beyond the bone field and has been established as one of the most important areas of "RANKL biology".


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/pathology , RANK Ligand/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Clinical Trials as Topic , Humans , Neoplasm Metastasis , Osteoblasts/metabolism , Osteoblasts/pathology
10.
Nat Metab ; 2(12): 1382-1390, 2020 12.
Article in English | MEDLINE | ID: mdl-33288951

ABSTRACT

Osteoclasts are the exclusive bone-resorbing cells, playing a central role in bone metabolism, as well as the bone damage that occurs under pathological conditions1,2. In postnatal life, haematopoietic stem-cell-derived precursors give rise to osteoclasts in response to stimulation with macrophage colony-stimulating factor and receptor activator of nuclear factor-κB ligand, both of which are produced by osteoclastogenesis-supporting cells such as osteoblasts and osteocytes1-3. However, the precise mechanisms underlying cell fate specification during osteoclast differentiation remain unclear. Here, we report the transcriptional profiling of 7,228 murine cells undergoing in vitro osteoclastogenesis, describing the stepwise events that take place during the osteoclast fate decision process. Based on our single-cell transcriptomic dataset, we find that osteoclast precursor cells transiently express CD11c, and deletion of receptor activator of nuclear factor-κB specifically in CD11c-expressing cells inhibited osteoclast formation in vivo and in vitro. Furthermore, we identify Cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 (Cited2) as the molecular switch triggering terminal differentiation of osteoclasts, and deletion of Cited2 in osteoclast precursors in vivo resulted in a failure to commit to osteoclast fate. Together, the results of this study provide a detailed molecular road map of the osteoclast differentiation process, refining and expanding our understanding of the molecular mechanisms underlying osteoclastogenesis.


Subject(s)
Osteoclasts/physiology , Osteogenesis/physiology , Signal Transduction/physiology , Animals , Bone Marrow Cells , CD11c Antigen/metabolism , Cell Proliferation , Databases, Factual , Female , Mice , Mice, Inbred C57BL , Osteogenesis/genetics , Pregnancy , Repressor Proteins/metabolism , Signal Transduction/genetics , Trans-Activators/metabolism , p300-CBP Transcription Factors
11.
Cell Rep ; 32(10): 108124, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32905763

ABSTRACT

Osteoprotegerin (OPG) is a circulating decoy receptor for RANKL, a multifunctional cytokine essential for the differentiation of tissue-specific cells in bone and immune systems such as osteoclasts, medullary thymic epithelial cells (mTECs), and intestinal microfold cells (M cells). However, it is unknown whether OPG functions only at the production site or circulates to other tissues acting in an endocrine fashion. Here we explore the cellular source of OPG by generating OPG-floxed mice and show that locally produced OPG, rather than circulating OPG, is crucial for bone and immune homeostasis. Deletion of OPG in osteoblastic cells leads to severe osteopenia without affecting serum OPG. Deletion of locally produced OPG increases mTEC and M cell numbers while retaining the normal serum OPG level. This study shows that OPG limits its functions within the tissue where it was produced, illuminating the importance of local regulation of the RANKL system.


Subject(s)
Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoprotegerin/metabolism , Animals , Mice
12.
Cell Metab ; 29(3): 627-637.e5, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30661929

ABSTRACT

Osteocyte survival is key to bone homeostasis and is perturbed in menopause and aging. However, it remains unknown how osteocyte-mediated maintenance of the skeleton is regulated by the osteoprotective factor semaphorin 3A (Sema3A), a secreted protein that is known to reduce bone resorption and enhance bone formation. Here, we show that estrogen induces osteocyte expression of Sema3A, which acts on its receptor on osteocytes to promote their survival and maintain bone homeostasis. Postnatal global and conditional deletion of Sema3a in osteoblastic cells resulted in a severe osteoporotic phenotype marked by fewer osteocytes. This phenotype was recapitulated by osteocyte-specific deficiency of either Sema3A or its receptor component neuropilin-1 (Nrp1). A stimulator of soluble guanylate cyclase-cGMP signaling mimicked Sema3A action and ameliorated bone loss after ovariectomy. We further show that serum levels of SEMA3A decreased with age or after menopause in humans. Thus, we provide a mechanistic insight into the estrogen action and a promising therapeutic approach to protect against bone-related aging.


Subject(s)
Aging/metabolism , Estrogens/metabolism , Menopause/metabolism , Osteocytes/metabolism , Semaphorin-3A/physiology , Animals , Bone Resorption/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Middle Aged , Neuropilin-1/metabolism , Osteocytes/cytology , Osteogenesis/physiology
13.
Bone Res ; 7: 1, 2019.
Article in English | MEDLINE | ID: mdl-30622830

ABSTRACT

Bone is one of the preferred sites for the metastasis of malignant tumours, such as breast cancer, lung cancer and malignant melanoma. Tumour cells colonizing bone have the capacity to induce the expression of receptor activator of nuclear factor-κB ligand (RANKL), which promotes osteoclast differentiation and activation. Tumour-induced osteoclastic bone resorption leads to a vicious cycle between tumours and bone cells that fuels osteolytic tumour growth, causing bone pain and hypercalcaemia. Furthermore, RANKL contributes to bone metastasis by acting as a chemoattractant to bone for tumour cells that express its receptor, RANK. Thus inhibition of the RANKL-RANK pathway is a promising treatment for bone metastasis, and a human monoclonal anti-RANKL antibody, denosumab, has been used in the clinic. However, orally available drugs targeting RANKL must be developed to increase the therapeutic benefits to patients. Here we report the efficacy of the small-molecule RANKL inhibitor AS2676293 in treating bone metastasis using mouse models. Oral administration of AS2676293 markedly inhibited bone metastasis of human breast cancer cells MDA-MB-231-5a-D-Luc2 as well as tumour-induced osteolysis. AS2676293 suppressed RANKL-mediated tumour migration in the transwell assay and inhibited bone metastasis of the murine cell line B16F10, which is known not to trigger osteoclast activation. Based on the results from this study, RANKL inhibition with a small-molecule compound constitutes a promising therapeutic strategy for treating bone metastasis by inhibiting both osteoclastic bone resorption and tumour migration to bone.

14.
Article in English | MEDLINE | ID: mdl-29610150

ABSTRACT

Bone is a crucial element of the skeletal-locomotor system, but also functions as an immunological organ that harbors hematopoietic stem cells (HSCs) and immune progenitor cells. Additionally, the skeletal and immune systems share a number of regulatory molecules, including cytokines and signaling molecules. Osteoimmunology was created as an interdisciplinary field to explore the shared molecules and interactions between the skeletal and immune systems. In particular, the importance of an inseparable link between the two systems has been highlighted by studies on the pathogenesis of rheumatoid arthritis (RA), in which pathogenic helper T cells induce the progressive destruction of multiple joints through aberrant expression of receptor activator of nuclear factor (NF)-κB ligand (RANKL). The conceptual bridge of osteoimmunology provides not only a novel framework for understanding these biological systems but also a molecular basis for the development of therapeutic approaches for diseases of bone and/or the immune system.


Subject(s)
Bone Diseases/immunology , Bone and Bones/immunology , Immune System/metabolism , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Bone Diseases/metabolism , Bone Remodeling/immunology , Humans , Mice , RANK Ligand/immunology , Receptor Cross-Talk , Signal Transduction , T-Lymphocytes/immunology
15.
Nat Metab ; 1(9): 868-875, 2019 09.
Article in English | MEDLINE | ID: mdl-32694743

ABSTRACT

Receptor activator of NF-κB ligand (RANKL) is a multifunctional cytokine known to affect immune and skeletal systems, as well as oncogenesis and metastasis1-4. RANKL is synthesized as a membrane-bound molecule, and cleaved into its soluble form by proteases5-7. As the soluble form of RANKL does not contribute greatly to bone remodelling or ovariectomy-induced bone loss8, whether soluble RANKL has a role in pathological settings remains unclear. Here we show that soluble RANKL promotes the formation of tumour metastases in bone. Mice that selectively lack soluble RANKL (Tnfsf11ΔS/ΔS)5-7,9 have normal bone homoeostasis and develop a normal immune system but display markedly reduced numbers of bone metastases after intracardiac injection of RANK-expressing melanoma and breast cancer cells. Deletion of soluble RANKL does not affect osteoclast numbers in metastatic lesions or tumour metastasis to non-skeletal tissues. Therefore, soluble RANKL is dispensable for physiological regulation of bone and immune systems, but has a distinct and pivotal role in the promotion of bone metastases.


Subject(s)
Bone Neoplasms/secondary , Neoplasms/pathology , Receptor Activator of Nuclear Factor-kappa B/physiology , Animals , Bone Remodeling/physiology , Cell Differentiation/physiology , Female , Humans , Male , Mice , Mice, Knockout , Neoplasm Metastasis , Osteoclasts/cytology , Receptor Activator of Nuclear Factor-kappa B/genetics
16.
Nat Immunol ; 19(11): 1265-1276, 2018 11.
Article in English | MEDLINE | ID: mdl-30323341

ABSTRACT

The methylation of arginine residues in proteins is a post-translational modification that contributes to a wide range of biological processes. Many cytokines involved in T cell development and activation utilize the common cytokine receptor γ-chain (γc) and the kinase JAK3 for signal transduction, but the regulatory mechanism that underlies the expression of these factors remains unclear. Here we found that the arginine methyltransferase PRMT5 was essential for the maintenance of invariant natural killer T cells (iNKT cells), CD4+ T cells and CD8+ T cells. T cell-specific deletion of Prmt5 led to a marked reduction in signaling via γc-family cytokines and a substantial loss of thymic iNKT cells, as well as a decreased number of peripheral CD4+ T cells and CD8+ T cells. PRMT5 induced the symmetric dimethylation of Sm proteins that promoted the splicing of pre-mRNA encoding γc and JAK3, and this critically contributed to the expression of γc and JAK3. Thus, arginine methylation regulates strength of signaling via γc-family cytokines by facilitating the expression of signal-transducing components.


Subject(s)
Arginine/metabolism , Interleukin Receptor Common gamma Subunit/immunology , Protein-Arginine N-Methyltransferases/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Interleukin Receptor Common gamma Subunit/metabolism , Methylation , Mice , Protein-Arginine N-Methyltransferases/immunology , T-Lymphocytes/metabolism
17.
Materials (Basel) ; 11(4)2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29642614

ABSTRACT

Multicomponent polymer particles with specific morphology are promising materials exhibiting novel functionality which cannot be obtained with single-component polymer particles. Particularly, the preparation of such kinds of polymer particles involving electrically or optically active conjugated polymers with uniform size is a challenging subject due to their intense demands. Here, microspheres of binary polymer blend consisting of poly(4-butyltriphenylamine) (PBTPA)/poly(methyl methacrylate) (PMMA) (1:1 in weight) were produced via a microfluidic emulsification with a Y-shaped microreactor, and a subsequent solvent evaporation method. The flow rate of the dispersed phase (polymer solution) was fixed to 7 µL/min, and 140 or 700 µL/min of the flow rate of the continuous phase (aqueous 0.6 wt % of poly(vinyl alcohol) (PVA) solution) was utilized to produce the dispersion with different diameter. The concentration of dispersed phase was adjusted to 0.1 or 1.0 w/v%. Core-shell, Janus and dumbbell type microspheres were obtained dependent on the flow rate of continuous phase. Incomplete core-shell type microspheres were produced for the blend involving low molecular weight PMMA. Complex Janus and core-shell type microspheres were fabricated by the addition of sodium dodecyl sulfate (SDS) to continuous phase. It is found that final morphologies are strongly dependent on the initial conditions of dispersion including the particle size suggesting that the morphologies are governed by the kinetical factors together with the conventionally accepted thermodynamic ones.

18.
Nat Commun ; 9(1): 701, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29453398

ABSTRACT

The immune system evolved to efficiently eradicate invading bacteria and terminate inflammation through balancing inflammatory and regulatory T-cell responses. In autoimmune arthritis, pathogenic TH17 cells induce bone destruction and autoimmune inflammation. However, whether a beneficial function of T-cell-induced bone damage exists is unclear. Here, we show that bone-damaging T cells have a critical function in the eradication of bacteria in a mouse model of periodontitis, which is the most common infectious disease. Bacterial invasion leads to the generation of specialized TH17 cells that protect against bacteria by evoking mucosal immune responses as well as inducing bone damage, the latter of which also inhibits infection by removing the tooth. Thus, bone-damaging T cells, which may have developed to stop local infection by inducing tooth loss, function as a double-edged sword by protecting against pathogens while also inducing skeletal tissue degradation.


Subject(s)
Alveolar Bone Loss/immunology , Bacteremia/microbiology , Periodontitis/immunology , Th17 Cells/physiology , Tooth Loss/immunology , Animals , Disease Models, Animal , Female , Interleukin-6/metabolism , Mice, Inbred C57BL , Microbiota , Mouth/microbiology , Periodontitis/complications , Periodontitis/metabolism , Periodontitis/microbiology , RANK Ligand/metabolism
19.
Nihon Rinsho Meneki Gakkai Kaishi ; 40(5): 361-366, 2017.
Article in Japanese | MEDLINE | ID: mdl-29238018

ABSTRACT

  Bone is a component of the skeletal-locomotor system but also functions as an immunological organ that harbors hematopoietic stem and progenitor cells. Since the immune and skeletal systems are closely related through a number of shared regulatory molecules including cytokines and receptors, bone can be affected in various immune disorders. Rheumatoid arthritis is a typical disease in which the immune system affects the bone metabolism. The enhanced activity of osteoclasts by the activation of Th17 cells causes the joint destruction in rheumatoid arthritis. Studies on bone destruction associated rheumatoid arthritis have highlighted the importance of the interplay between the immune and bone systems, and promoted the new interdisciplinary field of "osteoimmunology". Furthermore, recent studies have suggested that regulation of bone tissues by IL-17 is more complicated than we had expected. IL-17-prodcuing cells contribute to new bone formation at the enthesis in ankylosing spondylitis, and IL-17-producing γδ T cells promote bone regeneration by acting on the mesenchymal stem cells in bone fracture healing. It would be necessary to comprehensively understand the interplay between the immune and bone systems for elucidation of the molecular mechanisms underlying the pathogenesis of various diseases that involves the two systems.


Subject(s)
Bone Regeneration/genetics , Bone and Bones/immunology , Interleukin-17/physiology , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/physiology , Osteogenesis/genetics , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Bone and Bones/cytology , Bone and Bones/metabolism , Bone and Bones/pathology , Hematopoietic Stem Cells , Humans , Interleukin-17/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/physiology , Mice , Osteoblasts/immunology , Osteoblasts/physiology , Osteoclasts/immunology , Osteoclasts/physiology , Stem Cells , Th17 Cells/immunology
20.
Physiol Rev ; 97(4): 1295-1349, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28814613

ABSTRACT

The immune and skeletal systems share a variety of molecules, including cytokines, chemokines, hormones, receptors, and transcription factors. Bone cells interact with immune cells under physiological and pathological conditions. Osteoimmunology was created as a new interdisciplinary field in large part to highlight the shared molecules and reciprocal interactions between the two systems in both heath and disease. Receptor activator of NF-κB ligand (RANKL) plays an essential role not only in the development of immune organs and bones, but also in autoimmune diseases affecting bone, thus effectively comprising the molecule that links the two systems. Here we review the function, gene regulation, and signal transduction of osteoimmune molecules, including RANKL, in the context of osteoclastogenesis as well as multiple other regulatory functions. Osteoimmunology has become indispensable for understanding the pathogenesis of a number of diseases such as rheumatoid arthritis (RA). We review the various osteoimmune pathologies, including the bone destruction in RA, in which pathogenic helper T cell subsets [such as IL-17-expressing helper T (Th17) cells] induce bone erosion through aberrant RANKL expression. We also focus on cellular interactions and the identification of the communication factors in the bone marrow, discussing the contribution of bone cells to the maintenance and regulation of hematopoietic stem and progenitors cells. Thus the time has come for a basic reappraisal of the framework for understanding both the immune and bone systems. The concept of a unified osteoimmune system will be absolutely indispensable for basic and translational approaches to diseases related to bone and/or the immune system.


Subject(s)
Immunity , Skeleton/immunology , Allergy and Immunology , Animals , Arthritis, Rheumatoid/immunology , Cell Communication , Hematopoietic Stem Cells/physiology , Humans , Osteoclasts/metabolism , Osteology , Osteoprotegerin/metabolism , RANK Ligand/immunology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction , Skeleton/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...