Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
1.
J Pharm Pharmacol ; 76(3): 236-244, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38183672

ABSTRACT

OBJECTIVES: The mechanisms underlying the therapeutic effects of Si-Zhi Wan (SZW), a traditional Chinese medicine used to treat osteoporosis (OP), remain unknown. This study investigated the therapeutic effects of SZW on mice that underwent ovariectomy (OVX) and underlying mechanisms thereof. METHODS: We established an in vivo model of OP by performing OVX in mice. Microcomputed tomography (Micro-CT) was used to assess changes in bone characteristics of mice following SZW administration for 4 weeks. H&E staining revealed alterations in bone tissues of mice. Osteoclastogenesis in mouse bone tissue was observed using tartrate-resistant acid phosphatase staining and western blotting. Furthermore, we examined the impact of SZW on osteoclastogenesis in vitro using receptor activator of nuclear factor kappa-B ligand (RANKL). Finally, we explored the regulatory effects of SZW on osteoclast autophagy and the AMPK pathway. KEY FINDINGS: The results demonstrated that high-dose SZW reversed changes in bone density parameters caused by OVX, including bone volume (BV), BV/total volume, trabecular number, and trabecular spacing (P = 0.0007, 0.0035, 0.0114, and 0.0182, respectively), and stimulated the formation of bone trabeculae in mice (P < 0.0001). Furthermore, SZW suppressed osteoclast formation in mice with OVX and inhibited osteoclast formation induced by RANKL. Mechanistically, SZW inhibited osteoclast precursor cell autophagy through the AMPK pathway. CONCLUSIONS: SZW effectively inhibited the autophagy of osteoclast precursors by regulating the AMPK pathway, thereby exerting anti-osteoclastogenic effects and serving as an alternative therapy for OP.


Subject(s)
Osteoclasts , Osteoporosis , Female , Mice , Animals , Humans , Osteoclasts/metabolism , Osteogenesis , AMP-Activated Protein Kinases/metabolism , X-Ray Microtomography , Osteoporosis/drug therapy , Osteoporosis/metabolism , Signal Transduction , Autophagy , RANK Ligand/metabolism , RANK Ligand/pharmacology , RANK Ligand/therapeutic use , Ovariectomy , Cell Differentiation
2.
J Cancer Res Ther ; 19(3): 768-772, 2023.
Article in English | MEDLINE | ID: mdl-37470608

ABSTRACT

Background: Giant cell tumor (GCT) of the bone is a locally aggressive primary bone tumor, that can rarely metastasize. Arising mostly in epiphysis of the long bones in young adults, the tumor is composed of mononuclear cells that are admixed with osteoclastic giant cells(OLGCs), which express RANK ligand and RANK respectively. Denosumab a monoclonal antibody against RANK ligand has been shown to reduce the tumor by causing bone lysis by inhibiting RANKL. Histological changes in 11 patients of GCT who were treated with denosumab are presented here. Materials and Methods: Clinical records and slides of 11 patients of GCT who had been administered neoadjuvant denosumab were included in the study. Evaluation of pre and post therapy GCT specimens was performed by two pathologists (RK and VM). There were 4 males and 7 females. Their mean age was 30 years. All the patients received 120 mg denosumab subcutaneously every week with additional 120 mg on days 8 and 15 of therapy. The histological slides were reviewed and following points noted: 1) degree of ossification,2) fibrosis,3) loss of osteoclastic giant cells,4) proliferation of mononuclear cells,5) atypia,6) Permeation of osteoid by malignant cells. Results: Out of 11 cases, 2 cases did not show any significant histological improvement. 7 cases showed reduction in giant cells, increased fibrosis, enhanced mononuclear cell proliferation and ossification consistent with a pathological response. Atypia and osteoid permeation were noted in 2 cases which showed transformation to osteosarcoma. Conclusion: Denosumab treated giant cell tumor show dramatic histological changes. The post therapy lesions may bear no resemblance to pretherapy lesion. There may be complete resolution or may be confused with benign or malignant lesions Rarely they may show sarcomatous transformation. It is imperative that the pathologist is aware of these changes to prevent diagnostic pitfalls as it poses therapeutic and prognostic implications.


Subject(s)
Bone Density Conservation Agents , Bone Neoplasms , Giant Cell Tumor of Bone , Male , Female , Young Adult , Humans , Adult , Denosumab/pharmacology , Denosumab/therapeutic use , RANK Ligand/therapeutic use , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Fibrosis , Bone Density Conservation Agents/therapeutic use
3.
Nat Rev Rheumatol ; 19(5): 307-317, 2023 05.
Article in English | MEDLINE | ID: mdl-37024711

ABSTRACT

Denosumab, a human monoclonal antibody against receptor activator of nuclear factor-κB ligand (RANKL), is a potent inhibitor of osteoclast differentiation and activity. As the first biologic drug used to treat osteoporosis, denosumab has shown potent anti-resorptive properties and anti-fracture efficacy. The effects of this drug are also unique compared with the effects of bisphosphonates: namely, long-term treatment with this drug results in a continuous gain of bone mineral density, whereas withdrawal of the drug results in a transient overshoot in bone turnover and rapid bone loss. Although the mechanisms for these specific effects remain incompletely understood, emerging experimental and clinical data have started to highlight potential biological and pharmacological mechanisms by which denosumab might affect osteoclasts, as well as osteoblasts, and cause both sustained bone gain and bone loss upon treatment cessation. This Perspective discusses those potential mechanisms and the future studies and clinical implications that might ensue from these findings.


Subject(s)
Bone Density Conservation Agents , Osteoporosis , Humans , Denosumab/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , RANK Ligand/pharmacology , RANK Ligand/therapeutic use , Antibodies, Monoclonal/therapeutic use , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Bone and Bones , Bone Density , Osteoclasts , Bone Density Conservation Agents/adverse effects
4.
Gan To Kagaku Ryoho ; 50(3): 299-303, 2023 Mar.
Article in Japanese | MEDLINE | ID: mdl-36927895

ABSTRACT

Bisphosphonates were introduced in the late 1990s to reduce the frequency of skeletal-related events(SREs)in patients with metastatic bone disease. In the 2010s, anti-RANKL antibodies were launched as another type of bone modifying agents(BMA). Both induce osteoclast apoptosis and have been used in Japan for solid tumors with bone metastases and multiple myeloma. In addition to osteoclasts, bone microenvironment includes a wide variety of cells such as osteoblasts, vascular endothelial cells, and immune cells, which spatiotemporally interact with cancer cells, resulting in the colonization of dissociated tumor cells(DTCs). Understanding the pathways and microenvironmental factors through which cancer evolves to promote bone colony formation will lead to better planning of the therapeutic strategies in individual cases and to further elucidation of molecular drivers.


Subject(s)
Bone Neoplasms , Endothelial Cells , Humans , Endothelial Cells/metabolism , Endothelial Cells/pathology , Bone Neoplasms/drug therapy , Bone and Bones/metabolism , Bone and Bones/pathology , Diphosphonates/therapeutic use , Osteoclasts/pathology , RANK Ligand/metabolism , RANK Ligand/therapeutic use , Tumor Microenvironment
5.
Postgrad Med J ; 99(1175): 976-984, 2023 Aug 22.
Article in English | MEDLINE | ID: mdl-36841226

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by systemic inflammation and synovitis which evolve into joint destruction and deformity. Bone abnormalities are represented by marginal bone erosions and iuxta-articular and generalized osteoporosis. Overactivation of osteoclasts along with dysregulation of osteoblasts are the key events. Bone resorption is mediated by the receptor activator of nuclear factor (NF)-κB (RANK) ligand (RANK-L), responsible for the differentiation, proliferation, and activation of osteoclasts. RANK-L binds its receptor RANK, localized on the surface of preosteoclasts and mature osteoclasts promoting osteoclastogenesis. High levels of RANK-L were demonstrated in active RA patients. Denosumab, a fully human monoclonal antibody, binds RANK-L and suppresses the RANK-RANK-L signaling pathway leading to the inhibition of osteoclastogenesis. A retrospective analysis of published studies such as clinical trials evidenced the efficacy of denosumab in preventing bone erosion progression in RA patients. Key messages Key questions to answer in future include the following: Could denosumab be associated with other biologic therapies in RA patients? Could denosumab block the progression of bone damage in RA? Could denosumab be used for the prevention of bone erosion in RA?


Subject(s)
Arthritis, Rheumatoid , Bone Density Conservation Agents , Humans , Denosumab/therapeutic use , Retrospective Studies , Bone Density Conservation Agents/therapeutic use , RANK Ligand/metabolism , RANK Ligand/therapeutic use , Arthritis, Rheumatoid/drug therapy
6.
Lab Invest ; 103(5): 100067, 2023 05.
Article in English | MEDLINE | ID: mdl-36801641

ABSTRACT

Increased bone resorption caused by excessive number or activity of osteoclasts is the main cause of osteoporosis. Osteoclasts are multinucleated cells that are formed by the fusion of precursor cells. Although osteoclasts are primarily characterized by bone resorption, our understanding of the mechanisms that regulate the formation and function of osteoclasts is poor. Here we showed that the expression level of Rab interacting lysosomal protein (RILP) was strongly induced by receptor activator of NF-κB ligand in mouse bone marrow macrophages. Inhibition of RILP expression induced a drastic decrease in the number, size, F-actin ring formation of osteoclasts, and the expression level of osteoclast-related genes. Functionally, inhibition of RILP reduced the migration of preosteoclasts through PI3K-Akt signaling and suppressed bone resorption by inhibiting the secretion of lysosome cathepsin K. Treatments with siRNA-RILP attenuated pathologic bone loss in disease models induced by lipopolysaccharide. Thus, this work indicates that RILP plays an important role in the formation and bone resorption function of osteoclasts and may have a therapeutic potential to treat bone diseases caused by excessive or hyperactive osteoclasts.


Subject(s)
Bone Resorption , Osteogenesis , Animals , Mice , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation , Osteoclasts , Phosphatidylinositol 3-Kinases/metabolism , RANK Ligand/metabolism , RANK Ligand/pharmacology , RANK Ligand/therapeutic use , Signal Transduction
7.
Ann Intern Med ; 176(2): 224-238, 2023 02.
Article in English | MEDLINE | ID: mdl-36592456

ABSTRACT

DESCRIPTION: This guideline updates the 2017 American College of Physicians (ACP) recommendations on pharmacologic treatment of primary osteoporosis or low bone mass to prevent fractures in adults. METHODS: The ACP Clinical Guidelines Committee based these recommendations on an updated systematic review of evidence and graded them using the GRADE (Grading of Recommendations Assessment, Development and Evaluation) system. AUDIENCE AND PATIENT POPULATION: The audience for this guideline includes all clinicians. The patient population includes adults with primary osteoporosis or low bone mass. RECOMMENDATION 1A: ACP recommends that clinicians use bisphosphonates for initial pharmacologic treatment to reduce the risk of fractures in postmenopausal females diagnosed with primary osteoporosis (strong recommendation; high-certainty evidence). RECOMMENDATION 1B: ACP suggests that clinicians use bisphosphonates for initial pharmacologic treatment to reduce the risk of fractures in males diagnosed with primary osteoporosis (conditional recommendation; low-certainty evidence). RECOMMENDATION 2A: ACP suggests that clinicians use the RANK ligand inhibitor (denosumab) as a second-line pharmacologic treatment to reduce the risk of fractures in postmenopausal females diagnosed with primary osteoporosis who have contraindications to or experience adverse effects of bisphosphonates (conditional recommendation; moderate-certainty evidence). RECOMMENDATION 2B: ACP suggests that clinicians use the RANK ligand inhibitor (denosumab) as a second-line pharmacologic treatment to reduce the risk of fractures in males diagnosed with primary osteoporosis who have contraindications to or experience adverse effects of bisphosphonates (conditional recommendation; low-certainty evidence). RECOMMENDATION 3: ACP suggests that clinicians use the sclerostin inhibitor (romosozumab, moderate-certainty evidence) or recombinant PTH (teriparatide, low-certainty evidence), followed by a bisphosphonate, to reduce the risk of fractures only in females with primary osteoporosis with very high risk of fracture (conditional recommendation). RECOMMENDATION 4: ACP suggests that clinicians take an individualized approach regarding whether to start pharmacologic treatment with a bisphosphonate in females over the age of 65 with low bone mass (osteopenia) to reduce the risk of fractures (conditional recommendation; low-certainty evidence).


Subject(s)
Bone Density Conservation Agents , Fractures, Bone , Osteoporosis , Physicians , Adult , Female , Humans , Male , Bone Density Conservation Agents/adverse effects , Denosumab/therapeutic use , Diphosphonates/adverse effects , Fractures, Bone/prevention & control , Osteoporosis/complications , Osteoporosis/drug therapy , RANK Ligand/therapeutic use
8.
ACS Biomater Sci Eng ; 8(9): 3810-3818, 2022 Sep 12.
Article in English | MEDLINE | ID: mdl-36005299

ABSTRACT

The imbalance in the bone remodeling process with more bone resorption by osteoclasts compared to bone formation by osteoblasts results in a metabolic bone disorder known as osteoporosis. This condition reduces the bone mineral density and increases the risk of fractures due to low bone mass and disrupted bone microarchitecture. Osteoclastogenesis increases when the receptor activator NFκB ligand (RANKL) on the osteoblast surface binds to the receptor activator NFκB (RANK) on the osteoclast surface and the function of the decoy receptor of RANKL, osteoprotegrin, is compromised due to external stimuli such as heparin and lipopolysaccharides. The RANK/RANKL axis promotes the nuclear factor kappa B (NFκB) expression, which in turn increases the histone methyltransferase activity of EzH2 and EzH1 for the epigenetic regulation of osteoclastogenesis-related genes. Genistein counteracts NFκB-induced osteoclastogenesis and downstream signaling through the direct regulation of histone methyltransferase, EzH2 and EzH1, transcription. However, genistein possesses limitations like low bioavailability, low water solubility, high estrogen activity, and thyroid side effects, which obstruct its therapeutic usage. Here, the nanoemulsified formulation of genistein with vitamin D was utilized to circumvent the limitations of genistein so that it can be utilized for therapeutic purposes in osteoporosis management. The nanoemulsification of genistein and vitamin D was performed through the spontaneous emulsification using Tween 80 and medium chain triglyceride oil as an organic phase. The physiologically stable and biocompatible combination of the genistein and vitamin D nanoemulsion (GVNE) exhibited the controlled release pattern of genistein with Korsmeyer-Peppas and Higuchi models under different pH conditions (7.4, 6.5, and 1.2). The GVNE potentially enhanced the therapeutic efficacy under in vitro osteoporosis models and helped restore disease parameters like alkaline phosphatase activity, tartrate-resistant acid phosphatase activity, and the formation of multinuclear giant cells. Molecularly, the GVNE overturned the LPS-induced osteoclastogenesis by downregulation of NFκB expression along with its binding on EzH2 and EzH1 promoters. GVNE effects on the osteoporosis model established it as an efficient antiosteoporotic therapy. This nanonutraceutical-based formulation provides an epigenetic regulation of osteoporosis management and opens new avenues for alternate epigenetic therapies for osteoporosis.


Subject(s)
Genistein , Osteoporosis , Epigenesis, Genetic , Genistein/therapeutic use , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B/therapeutic use , Osteoporosis/drug therapy , Osteoporosis/genetics , Osteoporosis/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RANK Ligand/therapeutic use , Vitamin D/therapeutic use
9.
Spine (Phila Pa 1976) ; 47(16): 1165-1171, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35797625

ABSTRACT

STUDY DESIGN: Rat spine fusion model. OBJECTIVE: The present study aimed to determine whether administration of osteoprotegerin (OPG) in a rat model of spinal fusion increases bone volume, bone density, and decreases osteoclasts in the fusion mass. SUMMARY OF BACKGROUND DATA: OPG is a soluble RANK-ligand inhibitor that blocks osteoclast differentiation and activation. This makes it a potential agent to control the remodeling process and enhance bone mass during spinal fusion. MATERIALS AND METHODS: Forty-eight male Sprague-Dawley rats received a one-level spinal fusion of L4-L5 with bone allograft. Rats were then divided into four groups according to initiation of treatment: (1) saline on day 0 (saline), (2) OPG on day 0 (OPG D0), (3) OPG on day 10 (OPG D10), and (4) OPG on day 21 (OPG D21) postsurgery. After their initial injection, rats received weekly subcutaneous injections of OPG (10 mg/kg) and were euthanized six weeks postsurgery. MicroCT analysis of the fusion site and histological analysis of bone surface for quantification of osteoclast lining was performed. RESULTS: Increased bone volume in the fusion site and around the spinous process was seen in OPG D0 and OPG D10 when compared with saline. Mean trabecular thickness was greater in all groups receiving OPG compared with saline, with OPG D0 and OPG D10 having significantly greater mean trabecular thickness than OPG D21. All OPG groups had less bone surface lined with osteoclasts when compared with Saline, with OPG D0 and OPG D10 having fewer than OPG D21. CONCLUSIONS: This study indicates that OPG inhibited osteoclast bone resorption, which led to greater bone at the fusion site. Future studies investigating OPG on its own or in combination with an osteogenic factor to improve spinal fusion outcomes are warranted to further elucidate its potential therapeutic effect.


Subject(s)
Bone Resorption , Spinal Fusion , Animals , Bone Resorption/drug therapy , Bone Resorption/pathology , Male , Osteoclasts , Osteogenesis , Osteoprotegerin , RANK Ligand/pharmacology , RANK Ligand/therapeutic use , Rats , Rats, Sprague-Dawley
10.
Comput Math Methods Med ; 2022: 2011625, 2022.
Article in English | MEDLINE | ID: mdl-35669373

ABSTRACT

Epimedium is a traditional Chinese medicine that is most commonly prescribed by practitioners of Chinese medicine for the clinical treatment of malignant tumor bone metastasis. The main component of Epimedium is icariin (ICA). Studies have shown that ICA inhibits bone resorption of osteoclasts through the OPG/RANKL/RANK signaling pathway. Osteoclasts are the only cells in the body that have a bone-destroying capability. The OPG/RANKL/RANK system consists of cytokines that play major roles in osteoclast formation. Therefore, our study selected the OPG/RANKL/RANK system as the research target to investigate the effect of ICA on nude mice with lung cancer bone metastasis. We established the model of bone metastasis in nude mice, intervened the model with icariin and zoledronic acid, and detected the levels of OPG and RANKL by ELISA and western blot. The results showed that ICA had a significant inhibitory effect on bone metastases in nude mice. ICA achieved its antibone metastasis effect in nude mice with lung cancer via inhibiting RANKL expression and simultaneously increasing OPG expression. ICA not only alleviated osteolytic bone destruction caused by bone metastases, but it also reduced weight loss in tumor-bearing nude mice at the late stage of the experiment. The role of ICA in preventing bone metastasis of lung cancer merits further investigation.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Flavonoids/pharmacology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Nude , Osteoprotegerin/metabolism , Osteoprotegerin/therapeutic use , RANK Ligand/metabolism , RANK Ligand/therapeutic use
11.
Andes Pediatr ; 93(2): 247-252, 2022 Apr.
Article in Spanish | MEDLINE | ID: mdl-35735304

ABSTRACT

INTRODUCTION: Central Giant Cell Granuloma is an infrequent bone lesion located mainly in the maxillary bone. The main treatment is surgery with wide margins, so it sometimes causes great morbidity and esthetic al terations. Denosumab, a RANK-ligand inhibitor monoclonal antibody, has been presented as a valid therapeutic alternative in the treatment of these lesions. OBJECTIVE: to describe the clinical and radio logical response after treatment with Denosumab in a patient with unresected giant cell granuloma. CLINICAL CASE: 12-year-old boy who consulted due to a 24-hour maxillary swelling, without other associated symptoms. Examination revealed a tumor in the upper left maxilla with bulging of the ip- silateral gingiva. A CT scan was performed which showed a large expansive intraosseous lesion in the maxillary alveolar ridge. The biopsy of the lesion was compatible with Central Giant Cell Granuloma. Due to the size and location of the lesion, initial treatment with Denosumab, a human monoclonal antibody with action on RANK-ligand, was indicated. After 10 months of treatment, the patient showed a favorable clinical and radiological response, with a size decrease of the lesion and metabolic activity. As an adverse effect, the boy presented mild hypocalcemia, resolved after supplementation with calcium. CONCLUSION: the use of Denosumab as the first line of treatment in Giant Cell Granu loma may be an adequate therapeutic option in adolescents with lesions that are difficult to resect.


Subject(s)
Granuloma, Giant Cell , Adolescent , Child , Denosumab/therapeutic use , Granuloma, Giant Cell/diagnostic imaging , Granuloma, Giant Cell/drug therapy , Granuloma, Giant Cell/pathology , Humans , Ligands , Male , RANK Ligand/therapeutic use , Tomography, X-Ray Computed
12.
Cell Death Dis ; 10(5): 353, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040267

ABSTRACT

A variety of osteolytic factors have been identified from breast cancer cells leading to osteolysis, but less is known about which factor plays an essential role in the initiation process prior to the overt vicious osteolytic cycle. Here, we present in vitro and in vivo evidences to clarify the role of interleukin-11 (IL-11) as an essential contributor to breast cancer bone metastasis mediated osteolysis. Animal studies showed that bone specific metastatic BoM-1833 cells induce earlier onset of osteolysis and faster tumor growth compared with MCF7 and parental MDA-MB-231 cells in BALB/c-nu/nu nude mice. IL-11 was further screened and identified as the indispensable factor secreted by BoM-1833 cells inducing osteoclastogenesis independently of receptor activator of nuclear factor κB ligand (RANKL). Mechanistic investigation revealed that the JAK1/STAT3 signaling pathway as a downstream effector of IL-11, STAT3 activation further induces the expression of c-Myc, a necessary factor required for osteoclastogenesis. By inhibiting STAT3 phosphorylation, AG-490 was shown effective in reducing osteolysis and tumor growth in the metastatic niche. Overall, our results revealed the essential role and the underlying molecular mechanism of IL-11 in breast cancer bone metastasis mediated osteolysis. STAT3 targeting through AG-490 is a potential therapeutic strategy for mitigating osteolysis and tumor growth of bone metastatic breast cancer.


Subject(s)
Bone Neoplasms/pathology , Breast Neoplasms/pathology , Interleukin-11/pharmacology , Osteogenesis/drug effects , Osteolysis/prevention & control , RANK Ligand/pharmacology , Animals , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Cell Line, Tumor , Female , Humans , Interleukin-11/metabolism , Interleukin-11/therapeutic use , Janus Kinase 1/metabolism , Kaplan-Meier Estimate , Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Colony-Stimulating Factor/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Osteolysis/pathology , RANK Ligand/therapeutic use , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
13.
Clin Calcium ; 29(3): 337-341, 2019.
Article in Japanese | MEDLINE | ID: mdl-30814379

ABSTRACT

Bone erosions develop early in the course of rheumatoid arthritis(RA)and are predictive of a worse prognosis. They deteriorate gradually and cause joint damage, resulting in impaired functional capacity and disability. Lately, a considerable number of studies have increased our understanding of the pathogenic mechanisms participating in the development of bone erosions in RA. Osteoclasts are responsible cells and multiple factors have been identified to stimulate their differentiation and function. RANKL(receptor activator of NF-κB ligand)and other cytokines have been known for a long time to enhance osteoclastogenesis, but the role of other pathways has also been revealed recently. Besides to excessive ostaoclastogenesis, impair osteoblast differentiation and function also plays part in bone erosion formation in RA. Inflamed synovial membrane products increased levels of cytokines and antagonists of the canonical Wnt signaling pathway, which inhibit osteoblast differentiation and function. It seems that downregulation of this pathway leads to impaired osteoblast differentiation and activity and consequently, to reduced capacity of bone erosion to repair. Preclinical studies show that these findings could have implications in RA treatment, although more studies are required in this direction.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Osteoclasts/metabolism , RANK Ligand/therapeutic use , Wnt Signaling Pathway , Drug Evaluation, Preclinical , Humans , RANK Ligand/administration & dosage
14.
Sci Rep ; 8(1): 17770, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30538259

ABSTRACT

Although the regulation of post-ischemic inflammation is an important strategy to treat ischemic stroke, all clinical trials have failed to show its efficacy. To solve the problem, we previously developed a novel partial peptide of RANKL, microglial healing peptide 1 (MHP1), which could reduce ischemic injury by inhibiting Toll-like receptor (TLR) induced inflammation. However, optimization of the peptide was necessary to increase the stability and efficacies for clinical use. According to information gathered through HPLC/MS in serum, we have newly designed a series of modified MHP1 peptides and have found that N-terminal acetylation and C-terminal amidation in MHP1 (MHP1-AcN), can strengthen its anti-inflammatory effects and increase its stability with anti-osteoclastogenic effects. Anti-TLR activity was reported to be reduced in MHP1 when incubated at 37 °C for 24 hrs, but MHP1-AcN could keep the activity under the same condition. The therapeutic effect of MHP1-AcN was observed in transient ischemic stroke model at lower dose than MHP1. Importantly, MHP1-AcN did not affect thrombolytic effects of tissue plasminogen activator (tPA) and inhibited tPA-induced hemorrhagic transformation. These findings indicated that MHP1-AcN was stable and effective anti-TLR signal peptide and could be a promising agent for treating stroke patients receiving tPA and endovascular therapy.


Subject(s)
Brain Ischemia/drug therapy , RANK Ligand/therapeutic use , Stroke/therapy , Amino Acid Sequence , Animals , Fibrinolytic Agents/therapeutic use , Male , Mice , Mice, Inbred ICR , Microglia/physiology , Peptides/therapeutic use , Tissue Plasminogen Activator/administration & dosage
15.
Diabetes Obes Metab ; 20(8): 1817-1828, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29687585

ABSTRACT

Type 2 diabetes mellitus is now a worldwide health problem with increasing prevalence. Mounting efforts have been made to treat, prevent and predict this chronic disease. In recent years, increasing evidence from mice and clinical studies suggests that bone-derived molecules modulate glucose metabolism. This review aims to summarize our current understanding of the interplay between bone and glucose metabolism and to highlight potential new means of therapeutic intervention. The first molecule recognized as a link between bone and glucose metabolism is osteocalcin (OCN), which functions in its active form, that is, undercarboxylated OCN (ucOC). ucOC acts in promoting insulin expression and secretion, facilitating insulin sensitivity, and favouring glucose and fatty acid uptake and utilization. A second bone-derived molecule, lipocalin2, functions in suppressing appetite in mice through its action on the hypothalamus. Osteocytes, the most abundant cells in bone matrix, are suggested to act on the browning of white adipose tissue and energy expenditure through secretion of bone morphogenetic protein 7 and sclerostin. The involvement of bone resorption in glucose homeostasis has also been examined. However, there is evidence indicating the implication of the receptor activator of nuclear factor κ-B ligand, neuropeptide Y, and other known and unidentified bone-derived factors that function in glucose homeostasis. We summarize recent advances and the rationale for treating, preventing and predicting diabetes by skeleton intervention.


Subject(s)
Bone and Bones/drug effects , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Models, Biological , Prediabetic State/drug therapy , Animals , Appetite Depressants/metabolism , Appetite Depressants/pharmacology , Appetite Depressants/therapeutic use , Appetite Regulation/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/prevention & control , Energy Metabolism/drug effects , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin Secretion/drug effects , Lipocalin-2/genetics , Lipocalin-2/metabolism , Lipocalin-2/pharmacology , Lipocalin-2/therapeutic use , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Neuropeptide Y/therapeutic use , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteocalcin/pharmacology , Osteocalcin/therapeutic use , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Prediabetic State/metabolism , Prediabetic State/pathology , Prediabetic State/prevention & control , RANK Ligand/genetics , RANK Ligand/metabolism , RANK Ligand/pharmacology , RANK Ligand/therapeutic use , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Secretagogues/metabolism , Secretagogues/pharmacology , Secretagogues/therapeutic use
16.
Phytother Res ; 32(3): 452-458, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29226489

ABSTRACT

Safe and efficient therapeutic agents for bone diseases are required in natural sources. We previously found that edible seaweed-derived polysaccharide porphyran exhibited anti-inflammatory effects through the down regulation of nuclear factor-κB. The aim of this study was to investigate the availability of porphyran as a therapeutic agent for bone diseases. The effects of porphyran on receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclastogenesis in RAW264.7 cells were examined. Porphyran suppressed RANKL-induced osteoclast formation in a concentration-dependent manner (6.25-50 µg/ml) without any cytotoxic effects. Furthermore, real-time polymerase chain reaction analyses indicated that porphyran at 50 µg/ml significantly attenuated the RANKL-induced increase in the mRNA levels of osteoclastogenesis-related marker genes such as nuclear factor of activated T cells, tartrate-resistant acid phosphatase, cathepsin K, and matrix metalloproteinase-9 in RAW264.7 cells. To our knowledge, this is the first report showing that edible-seaweed-derived polysaccharide porphyran can suppress RANKL-induced osteoclastogenesis. Our results suggest that porphyran can be used as a safe therapeutic agent to improve osteoclast-related pathological conditions.


Subject(s)
Osteoclasts/metabolism , RANK Ligand/therapeutic use , RAW 264.7 Cells/metabolism , Sepharose/analogs & derivatives , Animals , Cell Differentiation , Mice , RANK Ligand/pharmacology , Sepharose/pharmacology , Sepharose/therapeutic use
17.
Tissue Eng Part C Methods ; 23(11): 781-794, 2017 11.
Article in English | MEDLINE | ID: mdl-28741427

ABSTRACT

The canine mandible is useful for studying bone regeneration after dental implant placement. However, it is limited in investigations of peri-implant osteogenesis under osteoporotic conditions due to the insignificant osteoporotic effect of ovariectomy. This study aimed at establishing a local osteoporotic model without ovariectomy by using receptor activator of nuclear factor kappa-B ligand (RANKL) in a canine mandible model. This new model was used to evaluate the effects of injectable ß-tricalcium phosphate (TCP) microsphere bone grafts on peri-implant bone regeneration under osteoporotic conditions with combinations of recombinant human bone morphogenetic protein-2 (rhBMP-2). A local osteoporotic canine mandible model was designed by creating a hole in the mandibular alveolar bone, then implanting a collagen sponge soaked with 20, 40, or 60 µg RANKL into the hole, and leaving it for 2 weeks. After the establishment of the dose for maximum osteoporotic bone loss at 40 µg of RANKL, the main surgery was performed. RANKL-soaked collagen sponges were removed, and dental implants were placed with bone grafts in five groups: implant only, TCP, and TCP + rhBMP-2 at 5, 15, and 45 µg. Peri-implant bone generation was determined by radiologic and histologic evaluations at 6 weeks after dental implant placement. On performing micro-computed tomography analysis, the group with TCP + 5 µg rhBMP-2 showed the highest bone volume than the other groups and a 22% increase (p < 0.05) compared with the implant-only group. In the histologic analysis, the TCP-only and TCP + 5 µg BMP-2 groups showed higher bone areas (14% and 16% increase, respectively) and bone-implant contact (12% and 7% increase, respectively) compared with the implant-only group, but there was no significant difference among the groups. In this study, the RANKL-induced local osteoporotic canine mandible model was useful for peri-implant bone regeneration under osteoporotic conditions such as those found in geriatric patients. The injectable ß-TCP bone grafts used in this study were effective in peri-implant bone generation under osteoporotic conditions, and their efficiency was enhanced at 5 µg BMP-2 compared with higher concentrations of BMP-2.


Subject(s)
Bone Regeneration , Dental Implants , Mandible/pathology , Osteoporosis/drug therapy , RANK Ligand/therapeutic use , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/therapeutic use , Bone Regeneration/drug effects , Disease Models, Animal , Dogs , Humans , Imaging, Three-Dimensional , Mandible/diagnostic imaging , Mandible/drug effects , Osteoporosis/pathology , RANK Ligand/pharmacology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/therapeutic use , X-Ray Microtomography
18.
J Negat Results Biomed ; 13: 18, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25495344

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is an inflammatory disease that leads to destruction of both articular cartilage and bone tissues. In rheumatic joints, synoviocytes and T-lymphocytes as well as bone cells produce the receptor activator of nuclear factor κ-B (RANK) ligand (RANKL), which binds to RANK on the surface of osteoclasts and their precursor cells to induce differentiation and activation of osteoclasts. Hence, inhibition of RANKL may be a promising approach to suppress osteolysis in RA. On the other hand, RANKL production by lymphocytes indicates the possibility that its inhibition would be effective to suppress inflammation in RA. In addition, it has been reported that cathepsin K, a predominant cysteine protease in osteoclasts, is involved in cartilage destruction in RA model mice. Here, we evaluated the effects of an anti-RANKL antibody on inflammation in footpads and degradation of articular cartilage in RA model mice. RESULTS: We induced arthritis in mice by injection of anti-type II collagen antibodies and lipopolysaccharide (LPS). Inhibition of RANKL by an anti-RANKL antibody (OYC1, Oriental Yeast, Tokyo, Japan) was confirmed by increased bone volume in the metaphysis of tibias. Swelling in either limb until day 14 was seen in 5 of 6 mice injected with anti-collagen antibodies and LPS without treatment with OYC1, while that was seen in 4 of 5 mice treated with OYC1. The average arthritis scores on day 14 in those groups were 2.17 and 3.00, respectively, indicating that OYC1 did not ameliorate inflammation in the limbs. Histological analyses indicated that OYC1 does not protect articular cartilage from destruction in mice with arthritis. CONCLUSIONS: Our present study failed to show the effectiveness of an anti-RANKL antibody to ameliorate inflammation in the limbs or protect articular cartilage from degradation in a collagen antibody-induced arthritis mouse model.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Cartilage/pathology , RANK Ligand/therapeutic use , Animals , Arthritis, Experimental/immunology , Cartilage/drug effects , Cartilage/immunology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred DBA , RANK Ligand/pharmacology , Random Allocation , Treatment Outcome
19.
Biomaterials ; 33(32): 8177-85, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22884682

ABSTRACT

Targeting tumor necrosis factor-α (TNF-α) and activator of NF-κB ligand (RANKL) has been proved highly successful in rheumatoid arthritis (RA) models and patients. This raises a possibility whether a single agent simultaneously targeting TNF-α and RANKL provides a potential therapeutic opportunity. This study aimed to design a dual functional vaccine and evaluate its therapeutic effects in RA mice model. Standard molecular biological techniques were used to generate human RANKL-TNF-like core fusion protein (RTFP-2) vaccine. High titers of antibodies against human TNF-α and RANKL were elicited and the RTFP-2 antiserum decreased TNF-α mediated apoptosis of L929 cells to 41% compared with 90% in positive controls. In addition, the antiserum completely abrogated osteoclastogenesis in vitro. Immunization with RTFP-2 also reduced the mortality of TNF-α induced cachexia from 56% to 28%. The RANKL-mediated hypercalcemic effects were significantly attenuated in RTFP-2 vaccinated mice. Furthermore, RTFP-2 vaccine significantly mitigated the incidence and severity of CIA via inhibition of inflammation and bone resorption. Our results showed the RTFP-2 vaccine of dual targets ameliorated the symptoms of CIA mice, suggesting the potential possibility to treat inflammatory bone diseases such as RA.


Subject(s)
Arthritis, Experimental/prevention & control , RANK Ligand/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use , Vaccines/therapeutic use , Amino Acid Sequence , Animals , Antibody Formation , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Bone and Bones/immunology , Bone and Bones/pathology , Cell Line , Female , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RANK Ligand/chemistry , RANK Ligand/genetics , RANK Ligand/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccines/chemistry , Vaccines/genetics , Vaccines/immunology
20.
J Bone Miner Res ; 27(12): 2501-10, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22836362

ABSTRACT

In the last decades the molecular basis of monogenic diseases has been largely unraveled, although their treatment has often remained unsatisfactory. Autosomal recessive osteopetrosis (ARO) belongs to the small group of genetic diseases that are usually treated with hematopoietic stem cell transplantation (HSCT). However, this approach is not effective in the recently identified form carrying mutations in the receptor activator of NF-κB ligand (RANKL) gene. In this subset, therapy replacement approach based on RANKL delivery has a strong rationale. Here we demonstrate that the systematic administration of RANKL for 1 month to Rankl(-/-) mice, which closely resemble the human disease, significantly improves the bone phenotype and has beneficial effects on bone marrow, spleen and thymus; major adverse effects arise only when mice are clearly overtreated. Overall, we provide evidence that the pharmacological administration of RANKL represents the appropriate treatment option for RANKL-deficient ARO patients, to be validated in a pilot clinical trial.


Subject(s)
Osteopetrosis/drug therapy , Osteopetrosis/genetics , RANK Ligand/therapeutic use , Animals , Bone Marrow Cells/drug effects , Bone Resorption/chemically induced , Bone and Bones/drug effects , Disease Models, Animal , Female , Humans , Male , Mice , Osteopetrosis/pathology , Phenotype , RANK Ligand/administration & dosage , RANK Ligand/adverse effects , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/deficiency , Receptor Activator of Nuclear Factor-kappa B/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...