Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 84
1.
Inflamm Regen ; 44(1): 27, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38831448

BACKGROUND: Regeneration of injured tissue is dependent on stem/progenitor cells, which can undergo proliferation and maturation processes to replace the lost cells and extracellular matrix (ECM). Bone has a higher regenerative capacity than other tissues, with abundant mesenchymal progenitor cells in the bone marrow, periosteum, and surrounding muscle. However, the treatment of bone fractures is not always successful; a marked number of clinical case reports have described nonunion or delayed healing for various reasons. Supplementation of exogenous stem cells by stem cell therapy is anticipated to improve treatment outcomes; however, there are several drawbacks including the need for special devices for the expansion of stem cells outside the body, low rate of cell viability in the body after transplantation, and oncological complications. The use of endogenous stem/progenitor cells, instead of exogenous cells, would be a possible solution, but it is unclear how these cells migrate towards the injury site. METHODS: The chemoattractant capacity of the elastin microfibril interface located protein 2 (Emilin2), generated by macrophages, was identified by the migration assay and LC-MS/MS. The functions of Emilin2 in bone regeneration were further studied using Emilin2-/- mice. RESULTS: The results show that in response to bone injury, there was an increase in Emilin2, an ECM protein. Produced by macrophages, Emilin2 exhibited chemoattractant properties towards mesenchymal cells. Emilin2-/- mice underwent delayed bone regeneration, with a decrease in mesenchymal cells after injury. Local administration of recombinant Emilin2 protein enhanced bone regeneration. CONCLUSION: Emilin2 plays a crucial role in bone regeneration by increasing mesenchymal cells. Therefore, Emilin2 can be used for the treatment of bone fracture by recruiting endogenous progenitor cells.

2.
Am J Med Genet A ; : e63631, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38647383

Craniofacial microsomia (CFM), also known as the oculo-auriculo-vertebral spectrum, is a congenital disorder characterized by hypoplasia of the mandible and external ear due to tissue malformations originating from the first and second branchial arches. However, distinguishing it from other syndromes of branchial arch abnormalities is difficult, and causal variants remain unidentified in many cases. In this report, we performed an exome sequencing analysis of a Brazilian family with CFM. The proband was a 12-month-old boy with clinical findings consistent with the diagnostic criteria for CFM, including unilateral mandibular hypoplasia, microtia, and external auditory canal abnormalities. A heterozygous de novo nonsense variant (c.713C>G, p.S238*) in PUF60 was identified, which was predicted to be pathogenic in silico. PUF60 has been reported as a causal gene in Verheij syndrome, but not in CFM. Although the boy showed craniofacial abnormalities and developmental delay that overlapped with Verheij syndrome, the facial asymmetry with unilateral hypoplasia of the mandible observed in this case did not match the previously reported phenotypes of PUF60 variants. Our findings expand the phenotypic range of PUF60 variants that cover CFM and Verheij syndrome.

3.
Nat Commun ; 15(1): 2707, 2024 Mar 28.
Article En | MEDLINE | ID: mdl-38548743

Periodontitis, which is induced by repeated bacterial invasion and the ensuing immune reactions that follow, is the leading cause of tooth loss. Periodontal tissue is comprised of four different components, each with potential role in pathogenesis, however, most studies on immune responses focus on gingival tissue. Here, we present a modified ligature-induced periodontitis model in male mice to analyze the pathogenesis, which captures the complexity of periodontal tissue. We find that the inflammatory response in the peri-root tissues and the expression of IL-6 and RANKL by Thy-1.2- fibroblasts/stromal cells are prominent throughout the bone destruction phase, and present already at an early stage. The initiation phase is characterized by high levels of ST2 (encoded by Il1rl1) expression in the peri-root tissue, suggesting that the IL-33/ST2 axis is involved in the pathogenesis. Both Il1rl1- and Il33-deficient mice exhibit exacerbated bone loss in the acute phase of periodontitis, along with macrophage polarization towards a classically activated phenotype and increased neutrophil infiltration, indicating a protective role of the IL-33/ST2 axis in acute inflammation. Thus, our findings highlight the hidden role of the peri-root tissue and simultaneously advance our understanding of the etiology of periodontitis via implicating the IL-33/ST2 axis.


Alveolar Bone Loss , Periodontitis , Animals , Male , Mice , Inflammation/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/genetics
4.
Biochem Biophys Res Commun ; 684: 149068, 2023 12 03.
Article En | MEDLINE | ID: mdl-37866240

Orthodontic tooth movement (OTM) is accomplished by controlling the mechanical loading onto the bone around the roots of target teeth. The precise orthodontic force induces osteoclastic bone resorption on the compression side and osteoblastic bone formation on the tension side of the alveolar bone. Orthodontic intervention causes inflammation in the periodontal ligament (PDL), which manifests as acute pain. Because inflammation is deeply connected to bone remodeling, it has been indicated that the inflammation after orthodontic intervention affects both the movement of teeth and generation of pain. However, the precise mechanisms underlying the immune regulation of OTM and the related pain are not well elucidated. Here, we found from the search of a public database that the interleukin (IL)-6 family of cytokines are highly expressed in the PDL by mechanical loading. The IL-6 signal was activated in the PDL after orthodontic intervention. The signal promoted OTM by inducing osteoclastic bone resorption. IL-6 was found to increase the number of osteoclasts by suppressing apoptosis and increasing their responsiveness to macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL). Furthermore, IL-6 signal was shown to elicit orthodontic pain by inducing neuroinflammation in the trigeminal ganglion (TG). Taken together, it was demonstrated that the IL-6 signal regulates tooth movement and pain during orthodontic treatment. It was also indicated that local blockade of the IL-6 signal is a promising therapeutic option in orthodontic treatment, targeting both tooth movement and pain.


Bone Resorption , Interleukin-6 , Humans , Tooth Movement Techniques , Osteoclasts , Bone Remodeling , Periodontal Ligament , Pain , Inflammation
5.
J Tissue Eng ; 14: 20417314231187960, 2023.
Article En | MEDLINE | ID: mdl-37529250

Fibroblast growth factor (FGF) signaling plays essential roles in various biological events. FGF18 is one of the ligands to be associated with osteogenesis, chondrogenesis and bone healing. The mouse critical-sized calvarial defect healing induced by the bone morphogenetic protein 2 (BMP2)-hydrogel is stabilized when FGF18 is added. Here, we aimed to investigate the role of FGF18 in the calvarial bone healing model. We first found that FGF18 + BMP2 hydrogel application to the calvarial bone defect increased the expression of anti-inflammatory markers, including those related to tissue healing M2 macrophage (M2-Mø) prior to mineralized bone formation. The depletion of macrophages with clodronate liposome hindered the FGF18 effect. We then examined how FGF18 induces M2-Mø polarization by using mouse primary bone marrow (BM) cells composed of macrophage precursors and BM stromal cells (BMSCs). In vitro studies demonstrated that FGF18 indirectly induces M2-Mø polarization by affecting BMSCs. Whole transcriptome analysis and neutralizing antibody treatment of BMSC cultured with FGF18 revealed that chemoattractant chemokine (c-c motif) ligand 2 (CCL2) is the major mediator for M2-Mø polarization. Finally, FGF18-augmented activity toward favorable bone healing with BMP2 was diminished in the calvarial defect in Ccr2-deleted mice. Altogether, we suggest a novel role of FGF18 in M2-Mø modulation via stimulation of CCL2 production in calvarial bone healing.

6.
Nat Commun ; 13(1): 7194, 2022 11 23.
Article En | MEDLINE | ID: mdl-36424386

Exercise results in mechanical loading of the bone and stimulates energy expenditure in the adipose tissue. It is therefore likely that the bone secretes factors to communicate with adipose tissue in response to mechanical loading. Interleukin (IL)-11 is known to be expressed in the bone, it is upregulated by mechanical loading, enhances osteogenesis and suppresses adipogenesis. Here, we show that systemic IL-11 deletion (IL-11-/-) results in reduced bone mass, suppressed bone formation response to mechanical loading, enhanced expression of Wnt inhibitors, and suppressed Wnt signaling. At the same time, the enhancement of bone resorption by mechanical unloading was unaffected. Unexpectedly, IL-11-/- mice have increased systemic adiposity and glucose intolerance. Osteoblast/osteocyte-specific IL-11 deletion in osteocalcin-Cre;IL-11fl/fl mice have reduced serum IL-11 levels, blunted bone formation under mechanical loading, and increased systemic adiposity similar to IL-11-/- mice. Adipocyte-specific IL-11 deletion in adiponectin-Cre;IL-11fl/fl did not exhibit any abnormalities. We demonstrate that osteoblast/osteocyte-derived IL-11 controls both osteogenesis and systemic adiposity in response to mechanical loading, an important insight for our understanding of osteoporosis and metabolic syndromes.


Interleukin-11 , Osteocytes , Osteogenesis , Animals , Mice , Adipogenesis , Interleukin-11/genetics , Obesity , Osteoblasts , Mice, Knockout
7.
Bone Res ; 10(1): 52, 2022 Aug 03.
Article En | MEDLINE | ID: mdl-35918335

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.

8.
Endocrinology ; 163(10)2022 10 01.
Article En | MEDLINE | ID: mdl-35931046

Semaphorin 3A (Sema3A) coordinates bone resorption and formation under the control of estrogen signaling. However, the contribution of osteoblast lineage cell-derived Sema3A to vertebral homeostasis has remained unclear. Moreover, it is unknown whether androgen signaling is involved in Sema3A expression in osteoblast lineage cells. In this study, we show that osteoblast lineage cell-derived Sema3A plays a key role in bone homeostasis independent of androgen signaling. Sema3a deletion with Sp7-Cre did not alter the trabecular bone mass in lumbar vertebrae, along with there being no significant difference in Sema3a mRNA expression. In contrast, osteoblast lineage cell-specific deletion of Sema3A with BGLAP-Cre led to decreased bone volume in both long bones and lumbar vertebrae. In addition, osteoblast lineage cell-derived Sema3A was not involved in orchidectomy-induced bone loss because androgen deficiency did not affect Sema3A protein expression. Thus, these results indicate that Sema3A derived from osteoblast lineage cells acts as an osteoprotective factor, even in vertebrae, and its expression is controlled in an androgen-independent manner.


Androgens , Semaphorin-3A , Androgens/pharmacology , Bone and Bones/metabolism , Homeostasis , Osteoblasts/metabolism , Semaphorin-3A/genetics , Semaphorin-3A/metabolism
9.
Nat Commun ; 13(1): 4166, 2022 07 18.
Article En | MEDLINE | ID: mdl-35851381

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.


Chondrocytes , Growth Plate , Animals , Hedgehog Proteins/genetics , Mice , Osteogenesis/genetics , Stem Cells
10.
J Oral Biosci ; 64(1): 8-17, 2022 Mar.
Article En | MEDLINE | ID: mdl-35149210

BACKGROUND: The cranial bones provide a structural framework to the head and face, protecting the organs inside. The cranium is endowed with mobility through the temporomandibular joints, with which many oral functions are performed. To exert these functions, the cranial bones undergo a continuous maintenance process of bone formation and resorption, bone remodeling. The remodeling of cranial bones is influenced by the physiological or pathological conditions they are subjected, including periodontitis and tumor. Therefore, oral bone biology has been investigated from multiple viewpoints. HIGHLIGHTS: We overview the physiology and pathology of the bone, centering on oral biology. Recently, many valuable insights have been obtained using novel techniques, including X-ray diffractometry, high-resolution microscopy, DNA/RNA sequencing, and mouse genetics. These insights include hydroxyapatite crystal alignment, cell lineage tracing, bone cell activity, biomechanics, the mechanisms underlying oral diseases and potential therapies for these diseases. These findings have advanced the oral bone biology research field and provided a better awareness of that which is currently known, and that which needs to be investigated. CONCLUSION: Based on the current state of accumulated knowledge, further progress is needed for obtaining a comprehensive picture of oral biology to greatly improve the level of clinical practices.


Bone Remodeling , Bone and Bones , Animals , Biology , Biomechanical Phenomena , Bone Remodeling/physiology , Mice , Osteogenesis
11.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article En | MEDLINE | ID: mdl-34426497

Skeletal muscle atrophy is caused by various conditions, including aging, disuse related to a sedentary lifestyle and lack of physical activity, and cachexia. Our insufficient understanding of the molecular mechanism underlying muscle atrophy limits the targets for the development of effective pharmacologic treatments and preventions. Here, we identified Krüppel-like factor 5 (KLF5), a zinc-finger transcription factor, as a key mediator of the early muscle atrophy program. KLF5 was up-regulated in atrophying myotubes as an early response to dexamethasone or simulated microgravity in vitro. Skeletal muscle-selective deletion of Klf5 significantly attenuated muscle atrophy induced by mechanical unloading in mice. Transcriptome- and genome-wide chromatin accessibility analyses revealed that KLF5 regulates atrophy-related programs, including metabolic changes and E3-ubiquitin ligase-mediated proteolysis, in coordination with Foxo1. The synthetic retinoic acid receptor agonist Am80, a KLF5 inhibitor, suppressed both dexamethasone- and microgravity-induced muscle atrophy in vitro and oral Am80 ameliorated disuse- and dexamethasone-induced atrophy in mice. Moreover, in three independent sets of transcriptomic data from human skeletal muscle, KLF5 expression significantly increased with age and the presence of sarcopenia and correlated positively with the expression of the atrophy-related ubiquitin ligase genes FBXO32 and TRIM63 These findings demonstrate that KLF5 is a key transcriptional regulator mediating muscle atrophy and that pharmacological intervention with Am80 is a potentially preventive treatment.


Benzoates/pharmacology , Drug Development , Gene Expression Regulation/drug effects , Kruppel-Like Transcription Factors/physiology , Muscle, Skeletal/drug effects , Muscular Atrophy/drug therapy , Tetrahydronaphthalenes/pharmacology , Animals , Dexamethasone/toxicity , Glucocorticoids/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Nat Commun ; 12(1): 2046, 2021 04 06.
Article En | MEDLINE | ID: mdl-33824347

Bone formation represents a heritable trait regulated by many signals and complex mechanisms. Its abnormalities manifest themselves in various diseases, including sclerosing bone disorder (SBD). Exploration of genes that cause SBD has significantly improved our understanding of the mechanisms that regulate bone formation. Here, we discover a previously unknown type of SBD in four independent families caused by bi-allelic loss-of-function pathogenic variants in TMEM53, which encodes a nuclear envelope transmembrane protein. Tmem53-/- mice recapitulate the human skeletal phenotypes. Analyses of the molecular pathophysiology using the primary cells from the Tmem53-/- mice and the TMEM53 knock-out cell lines indicates that TMEM53 inhibits BMP signaling in osteoblast lineage cells by blocking cytoplasm-nucleus translocation of BMP2-activated Smad proteins. Pathogenic variants in the patients impair the TMEM53-mediated blocking effect, thus leading to overactivated BMP signaling that promotes bone formation and contributes to the SBD phenotype. Our results establish a previously unreported SBD entity (craniotubular dysplasia, Ikegawa type) and contribute to a better understanding of the regulation of BMP signaling and bone formation.


Bone Morphogenetic Proteins/metabolism , Bone and Bones/pathology , Membrane Proteins/metabolism , Sclerosis/pathology , Signal Transduction , Smad Proteins/metabolism , Animals , Base Sequence , Cell Differentiation , Cell Nucleus/metabolism , Child , Child, Preschool , Female , Humans , Male , Membrane Proteins/genetics , Mice, Mutant Strains , Mutation/genetics , Osteoblasts/pathology , Pedigree , Phosphorylation , Skull/pathology , Young Adult
14.
J Clin Invest ; 131(6)2021 03 15.
Article En | MEDLINE | ID: mdl-33720039

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.


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
16.
J Bone Miner Metab ; 39(1): 34-44, 2021 Jan.
Article En | MEDLINE | ID: mdl-32889574

Receptor activator of nuclear factor-κB ligand (RANKL) is a key mediator of osteoclast differentiation and bone resorption. Osteoblast-lineage cells including osteoblasts and osteocytes express RANKL, which is regulated by several different factors, including hormones, cytokines, and mechanical forces. In vivo and in vitro analyses have demonstrated that various types of mechanosensing proteins on the cell membrane (i.e. mechanosensors) and intracellular mechanosignaling proteins play essential roles in the differentiation and functions of osteoblasts, osteoclasts, and osteocytes via soluble factors, such as sclerostin, Wnt ligands, and RANKL. This section provides an overview of the in vivo and in vitro evidence for the regulation of RANKL expression by mechanosensing and mechanotransduction.


RANK Ligand/metabolism , Animals , Biomechanical Phenomena , Cellular Microenvironment , Humans , Mechanotransduction, Cellular , Models, Biological , Signal Transduction
17.
Proc Natl Acad Sci U S A ; 117(49): 31070-31077, 2020 12 08.
Article En | MEDLINE | ID: mdl-33229551

Osteoporosis is caused by a disequilibrium between bone resorption and bone formation. Therapeutics for osteoporosis can be divided into antiresorptives that suppress bone resorption and anabolics which increase bone formation. Currently, the only anabolic treatment options are parathyroid hormone mimetics or an anti-sclerostin monoclonal antibody. With the current global increases in demographics at risk for osteoporosis, development of therapeutics that elicit anabolic activity through alternative mechanisms is imperative. Blockade of the PlexinB1 and Semaphorin4D interaction on osteoblasts has been shown to be a promising mechanism to increase bone formation. Here we report the discovery of cyclic peptides by a novel RaPID (Random nonstandard Peptides Integrated Discovery) system-based affinity maturation methodology that generated the peptide PB1m6A9 which binds with high affinity to both human and mouse PlexinB1. The chemically dimerized peptide, PB1d6A9, showed potent inhibition of PlexinB1 signaling in mouse primary osteoblast cultures, resulting in significant enhancement of bone formation even compared to non-Semaphorin4D-treated controls. This high anabolic activity was also observed in vivo when the lipidated PB1d6A9 (PB1d6A9-Pal) was intravenously administered once weekly to ovariectomized mice, leading to complete rescue of bone loss. The potent osteogenic properties of this peptide shows great promise as an addition to the current anabolic treatment options for bone diseases such as osteoporosis.


Osteogenesis/drug effects , Peptides, Cyclic/pharmacology , Amino Acid Sequence , Animals , Animals, Newborn , Femur/diagnostic imaging , Humans , Mice, Inbred C57BL , Ovariectomy , Peptide Library , Peptides, Cyclic/chemistry , Protein Multimerization , X-Ray Microtomography
18.
J Biol Chem ; 295(34): 12224-12232, 2020 08 21.
Article En | MEDLINE | ID: mdl-32647011

The supplementation of royal jelly (RJ) is known to provide a variety of health benefits, including anti-inflammatory and anti-obesity effects. RJ treatment also reportedly protects against bone loss, but no single factor in RJ has yet been identified as an anti-osteoporosis agent. Here we fractionated RJ and identified 10-hydroxy-2-decenoic acid (10H2DA) as a key component involved in inhibiting osteoclastogenesis based on mass spectrometric analysis. We further demonstrated free fatty acid receptor 4 (FFAR4) as directly interacting with 10H2DA; binding of 10H2DA to FFAR4 on osteoclasts inhibited receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced activation of NF-κB signaling, thereby attenuating the induction of nuclear factor of activated T cells (NFAT) c1, a key transcription factor for osteoclastogenesis. Oral administration of 10H2DA attenuated bone resorption in ovariectomized mice. These results suggest a potential therapeutic approach of targeting osteoclast differentiation by the supplementation of RJ, and specifically 10H2DA, in cases of pathological bone loss such as occur in postmenopausal osteoporosis.


Fatty Acids, Monounsaturated/pharmacology , Fatty Acids/chemistry , NF-kappa B/metabolism , Osteoclasts/metabolism , Osteoporosis/drug therapy , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Animals , Cell Differentiation/drug effects , Disease Models, Animal , Fatty Acids, Monounsaturated/chemistry , Female , Mice , NFATC Transcription Factors/metabolism , Osteoclasts/pathology , Osteoporosis/metabolism , Osteoporosis/pathology , RANK Ligand/metabolism
19.
Inflamm Regen ; 40: 2, 2020.
Article En | MEDLINE | ID: mdl-32047573

Receptor activator of NF-κB (RANK) ligand (RANKL) induces the differentiation of monocyte/macrophage-lineage cells into the bone-resorbing cells called osteoclasts. Because abnormalities in RANKL, its signaling receptor RANK, or decoy receptor osteoprotegerin (OPG) lead to bone diseases such as osteopetrosis, the RANKL/RANK/OPG system is essential for bone resorption. RANKL was first discovered as a T cell-derived activator of dendritic cells (DCs) and has many functions in the immune system, including organogenesis, cellular development. The essentiality of RANKL in the bone and the immune systems lies at the root of the field of "osteoimmunology." Furthermore, this cytokine functions beyond the domains of bone metabolism and the immune system, e.g., mammary gland and hair follicle formation, body temperature regulation, muscle metabolism, and tumor development. In this review, we will summarize the current understanding of the functions of the RANKL/RANK/OPG system in biological processes.

20.
Mod Rheumatol ; 30(1): 85-92, 2020 Jan.
Article En | MEDLINE | ID: mdl-30486712

Objectives: To investigate the role of non-receptor tyrosine kinases (NRTKs) in inflammation-induced osteoclastogenesis.Methods: Microarray analyses of global mRNA expression during receptor activator of NF-κB ligand (RANKL) and RANKL plus tumor necrosis factor (TNF)-α-induced osteoclast differentiation were performed. The inhibitory effect on TNF-α-induced osteoclast differentiation of A-419259, a potent inhibitor of hematopoietic cell kinase (Hck), was examined. The in vivo therapeutic effect of A-419259 treatment on lipopolysaccharide (LPS)-induced inflammatory bone destruction was evaluated.Results: We confirmed that Hck expression was selectively increased among the NRTKs during the osteoclast differentiation induced by RANKL and TNF-α, but not by RANKL alone. RANKL and TNF-α-induced osteoclast differentiation and they were dose-dependently inhibited by A-419259 treatment through inhibition of the expression of key regulators of osteoclastogenesis, including Prdm1 and Nfatc1. Notably, LPS-induced inflammatory bone loss in murine calvarial bones was ameliorated by the administration of A-419259.Conclusions: Our results demonstrate that the administration of A-419259 is effective for the inhibition of osteoclast differentiation induced by TNF-α in the presence of RANKL. Therefore, an inhibitor of Hck may be useful as a potent anti-osteoclastogenic agent for the treatment of inflammatory bone destruction.


Bone Resorption/genetics , Gene Expression Regulation , Inflammation/genetics , Osteoclasts/metabolism , Osteogenesis/drug effects , Proto-Oncogene Proteins c-hck/genetics , Pyrimidines/pharmacology , Pyrroles/pharmacology , Animals , Blotting, Western , Bone Resorption/drug therapy , Bone Resorption/metabolism , Cell Differentiation , Cells, Cultured , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred BALB C , Osteoclasts/drug effects , Osteoclasts/pathology , Proto-Oncogene Proteins c-hck/biosynthesis , RNA/genetics , src-Family Kinases
...