• 제목/요약/키워드: Receptor activator of nuclear factor kB ligand

검색결과 106건 처리시간 0.032초

Tusc2/Fus1 regulates osteoclast differentiation through NF-κB and NFATc1

  • Kim, Inyoung;Kim, Jung Ha;Kim, Kabsun;Seong, Semun;Kim, Nacksung
    • BMB Reports
    • /
    • 제50권9호
    • /
    • pp.454-459
    • /
    • 2017
  • Tumor suppressor candidate 2 (Tusc2, also known as Fus1) regulates calcium signaling, and $Ca^{2+}$-dependent nuclear factor of activated T-cells (NFAT) and nuclear factor kappa B ($NF-{\kappa}B$) pathways, which play roles in osteoclast differentiation. However, the role of Tusc2 in osteoclasts remains unknown. Here, we report that Tusc2 positively regulates the differentiation of osteoclasts. Overexpression of Tusc2 in osteoclast precursor cells enhanced receptor activator of nuclear factor ${\kappa}B$ ligand (RANKL)-induced osteoclast differentiation. In contrast, small interfering RNA-mediated knockdown of Tusc2 strongly inhibited osteoclast differentiation. In addition, Tusc2 induced the activation of RANKL-mediated $NF-{\kappa}B$ and calcium/calmodulin-dependent kinase IV (CaMKIV)/cAMP-response element (CRE)-binding protein CREB signaling cascades. Taken together, these results suggest that Tusc2 acts as a positive regulator of RANKL-mediated osteoclast differentiation.

L-형 칼슘 이온통로에 의한 파골세포 분화의 조절 (The Regulation of Osteoclastogenesis by L-Type Channel Agonist)

  • 노아롱새미;임미정
    • 약학회지
    • /
    • 제54권6호
    • /
    • pp.461-465
    • /
    • 2010
  • We investigated the role of L-type $Ca^{2+}$ channel in receptor activator of nuclear factor-kappaB ligand (RANKL)-induced osteoclast formation. BayK 8644, a L-type $Ca^{2+}$ channel agonist, was shown to increase the RANKLinduced osteoclastogenesis and actin ring formation in mouse bone marrow-dereived macrophage (BMM) culture system. BayK 8644 stimulated RANKL-induced extracellular signal-regulated kinase (ERK) and p38 MAP kinase (MAPK) activation, which leads to increased nuclear factor of activated T cells (NFAT)c1 expression. Taken together, these data indicate that L-type $Ca^{2+}$ channel regulates osteoclast formation possibly through ERK- and p38-mediated NFATc1 expression.

Design of a RANK-Mimetic Peptide Inhibitor of Osteoclastogenesis with Enhanced RANKL-Binding Affinity

  • Hur, Jeonghwan;Ghosh, Ambarnil;Kim, Kabsun;Ta, Hai Minh;Kim, Hyunju;Kim, Nacksung;Hwang, Hye-Yeon;Kim, Kyeong Kyu
    • Molecules and Cells
    • /
    • 제39권4호
    • /
    • pp.316-321
    • /
    • 2016
  • The receptor activator of nuclear factor ${\kappa}B$ (RANK) and its ligand RANKL are key regulators of osteoclastogenesis and well-recognized targets in developing treatments for bone disorders associated with excessive bone resorption, such as osteoporosis. Our previous work on the structure of the RANK-RANKL complex revealed that Loop3 of RANK, specifically the non-canonical disulfide bond at the tip, performs a crucial role in specific recognition of RANKL. It also demonstrated that peptide mimics of Loop3 were capable of interfering with the function of RANKL in osteoclastogenesis. Here, we reported the structure-based design of a smaller peptide with enhanced inhibitory efficiency. The kinetic analysis and osteoclast differentiation assay showed that in addition to the sharp turn induced by the disulfide bond, two consecutive arginine residues were also important for binding to RANKL and inhibiting osteoclastogenesis. Docking and molecular dynamics simulations proposed the binding mode of the peptide to the RANKL trimer, showing that the arginine residues provide electrostatic interactions with RANKL and contribute to stabilizing the complex. These findings provided useful information for the rational design of therapeutics for bone diseases associated with RANK/RANKL function.

Actin-binding LIM protein 1 regulates receptor activator of NF-κB ligand-mediated osteoclast differentiation and motility

  • Jin, Su Hyun;Kim, Hyunsoo;Gu, Dong Ryun;Park, Keun Ha;Lee, Young Rae;Choi, Yongwon;Lee, Seoung Hoon
    • BMB Reports
    • /
    • 제51권7호
    • /
    • pp.356-361
    • /
    • 2018
  • Actin-binding LIM protein 1 (ABLIM1), a member of the LIM-domain protein family, mediates interactions between actin filaments and cytoplasmic targets. However, the role of ABLIM1 in osteoclast and bone metabolism has not been reported. In the present study, we investigated the role of ABLIM1 in the receptor activator of $NF-{\kappa}B$ ligand (RANKL)-mediated osteoclastogenesis. ABLIM1 expression was induced by RANKL treatment and knockdown of ABLIM1 by retrovirus infection containing Ablim1-specific short hairpin RNA (shAblim1) decreased mature osteoclast formation and bone resorption activity in a RANKL-dose dependent manner. Coincident with the downregulated expression of osteoclast differentiation marker genes, the expression levels of c-Fos and the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), critical transcription factors of osteoclastogenesis, were also decreased in shAblim1-infected osteoclasts during RANKL-mediated osteoclast differentiation. In addition, the motility of preosteoclast was reduced by ABLIM1 knockdown via modulation of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt/Rac1 signaling pathway, suggesting another regulatory mechanism of ABLIM1 in osteoclast formation. These data demonstrated that ABLIM1 is a positive regulator of RANKL-mediated osteoclast formation via the modulation of the differentiation and PI3K/Akt/Rac1-dependent motility.

법랑모세포종에서 Receptor Activatorof Nuclear Factor-${\kappa}B$ Ligand(RANKL)와 Osteoprotegerin(OPG) 발현에 관한 연구 (IDENTIFICATION OF RECEPTOR ACTIVATOR OF NUCLEAR FACTOR-${\kappa}B$ LIGAND(RANKL) AND OSTEOPROTEGERIN(OPG) IN AMELOBLASTOMA)

  • 하우헌;황대석;김용덕;신상훈;김욱규;김종렬;정인교
    • Journal of the Korean Association of Oral and Maxillofacial Surgeons
    • /
    • 제33권2호
    • /
    • pp.94-102
    • /
    • 2007
  • The ameloblastoma is a common odontogenic tumor of the jaw bone and represents approximately 1% of tumor in the jaw. However, it might be able to infiltrate into the adjacent tissue, causing bony destruction and high recurrent rate. In this study, expression of RANKL and OPG in ameloblastoma in relation to age and gender of patient and recurrence, location of the lesion were examined through immunohistochemisry study. The RANKL and OPG antibody staining were used. The obtained result were as follow. 1. Positive immunoreactivity to RANKL/OPG in all specimens was found. 2. 1n recurrenc, location of ameloblastoma and age, gender of patients using immunohistochemical expression of RANKL. There was not significant difference. 3. 1n recurrence, location of ameloblastoma and age, gender of patients using immunohistochemical expression of OPG. There was not significant difference. In summary, it is suggested that activation of osteoclasts by RANKL is an important mechanism by which ameloblastomas cause bone destruction.

Afatinib ameliorates osteoclast differentiation and function through downregulation of RANK signaling pathways

  • Ihn, Hye Jung;Kim, Ju Ang;Bae, Yong Chul;Shin, Hong-In;Baek, Moon-Chang;Park, Eui Kyun
    • BMB Reports
    • /
    • 제50권3호
    • /
    • pp.150-155
    • /
    • 2017
  • Non-small-cell lung cancer (NSCLC) is the third most common cancer that spreads to the bone, resulting in osteolytic lesions caused by hyperactivation of osteoclasts. Activating mutations in epidermal growth factor receptor-tyrosine kinase (EGF-TK) are frequently associated with NSCLC, and afatinib is a first-line therapeutic drug, irreversibly targeting EGF-TK. However, the effects of afatinib on osteoclast differentiation and activation as well as the underlying mechanism remain unclear. In this study, afatinib significantly suppressed receptor activator of nuclear factor ${\kappa}B$ (RANK) ligand (RANKL)-induced osteoclast formation in bone marrow macrophages (BMMs). Consistently, afatinib inhibited the expression of osteoclast marker genes, whereas, it upregulated the expression of negative modulator genes. The bone resorbing activity of osteoclasts was also abrogated by afatinib. In addition, afatinib significantly inhibited RANKL-mediated Akt/protein kinase B and c-Jun N-terminal kinase phosphorylation. These results suggest that afatinib substantially suppresses osteoclastogenesis by downregulating RANK signaling pathways, and thus may reduce osteolysis after bone metastasis.

Osteoclast Activity and Osteoporosis

  • Kim, Hong-Hee
    • 한국응용약물학회:학술대회논문집
    • /
    • 한국응용약물학회 2001년도 춘계학술대회
    • /
    • pp.103-112
    • /
    • 2001
  • Bone homeostasis is maintained by a balance between activities of osteoblasts(bone forming cells) and osteoclasts (bone resorbing cells). The activities of these cells are closely regulated by multiple factors including hormones and cytokines. The cessation of estrogen at menopause disrupts the balanced regulation and is the main cause of osteoporosis in postmenopausal women. Recent molecular biological studies led to a discovery of tumor necrosis factor(TNF) and TNF receptor families genes that play critical roles in the regulation of osteoclast formation and function. RANKL (receptor activator of nuclear factor kappa B ligand; also called ODF, TRANCE, and OPGL) expressed on cells supporting osteoclast is essential for osteoclast differentiation, activation, and survival. RANK, the counter-receptor for RANKL, is expressed on progenitor and mature osteoclasts. The interaction between RANKL and RANK is requlated by a soluble decoy receptor OPG (osteoprotegerin). Gene knock out studies of these molecules showed profound effects on bone. These results prompted development of new strategies for treatment of bone diseases. Inhibition of osteoclast activity by blocking the RANKL-RANK interaction using OPG is being attempted. Research on the signaling pathways of RANK is also actively carried out. Screening natural products that inhibit the RANKL-RANK interaction or the activity of obteoclasts would be another effective means to a new drug target for bone resorbing diseases.

  • PDF

Bone Healing in Ovariectomized-rabbit Calvarial Defect with Tricalcium Phosphate Coated with Recombinant Human Bone Morphogenetic Protein-2 Genetically Engineered in Escherichia coli

  • Kim, Jung-Han;Kim, Chang-Joo;Shin, Sang-Hun
    • Maxillofacial Plastic and Reconstructive Surgery
    • /
    • 제36권2호
    • /
    • pp.37-49
    • /
    • 2014
  • Purpose: This study compares the bone formation ability of tricalcium phosphate (TCP) with and without recombinant human bone morphogenetic protein-2 (rhBMP-2) and assesses TCP as a carrier of rhBMP-2. Methods: Bilateral round defects (diameter: 8.0 mm) were formed in the cranium of eight New Zealand white rabbits. The defects were grafted with TCP only (control group) or with rhBMP-2-coated TCP (experimental group). The animals were sacrificed at 1st week, 2nd week, 4th week, and 8th week postoperatively; two rabbits sacrificed each time. The skulls were harvested and subjected to radiographic and histological examination. Results: Radiologic evaluation showed faster bone remodeling in the experimental group than in the control group. Histologic evaluation (H&E, Masson's trichrome stain) showed rapid bone formation, remodeling and calcification in the 1st and 2nd week in the experimental group. Immunohistochemical evaluation showed higher expression rate of osteoprotegerin, receptor activator of nuclear factor ${\kappa}B$ ligand, and receptor activator of nuclear factor ${\kappa}B$ in the experimental group at the 1st and 2nd week than in the control group. Conclusion: rhBMP-2 coated TCP resulted in rapid bone formation, remodeling, and calcification due to rhBMP-2's osteogenic effect. TCP performed properly as a carrier for rhBMP-2. Thus, the use of an rhBMP-2 coating on TCP had a synergic effect on bone healing and, especially, bone remodeling and maturation.

조골세포내 PDE4에 의한 PTH 신호의 음성적 조절 (The Negative Role of PDE4 on PTH-induced Signaling in Osteoblasts)

  • 박효정;노아롱새미;이정민;임미정
    • 약학회지
    • /
    • 제54권5호
    • /
    • pp.410-415
    • /
    • 2010
  • We explored the role of phosphodiesterase 4 (PDE4) on parathyroid (PTH)-induced signaling in osteoblasts. PTH was shown to increase the activity of PDE, mainly PDE4, in osteoblasts, which is partly attributable to elevated PDE4B and PDE4D mRNA expression. The use of PDE4 inhibitor strengthened the PTH-induced extracellular signal-regulated kinase (ERK) and p38 MAP kinase (MAPK) activation. Furthermore, the PDE4 inhibitor stimulated PTH-induced receptor activator of nuclear factor-${\kappa}B$ ligand (RANKL) expression in osteoblasts, which in turn increased osteoclast formation. Taken together, these data suggest the negative role of PDE4 on PTH-induced signaling in osteoblasts.

Ecklonia cava Extract Containing Dieckol Suppresses RANKL-Induced Osteoclastogenesis via MAP Kinase/NF-κB Pathway Inhibition and Heme Oxygenase-1 Induction

  • Kim, Seonyoung;Kang, Seok-Seong;Choi, Soo-Im;Kim, Gun-Hee;Imm, Jee-Young
    • Journal of Microbiology and Biotechnology
    • /
    • 제29권1호
    • /
    • pp.11-20
    • /
    • 2019
  • Ecklonia cava, an edible marine brown alga (Laminariaceae), is a rich source of bioactive compounds such as fucoidan and phlorotannins. Ecklonia cava extract (ECE) was prepared using 70% ethanol extraction and ECE contained 67% and 10.6% of total phlorotannins and dieckol, respectively. ECE treatment significantly inhibited receptor activator of nuclear $factor-{\kappa}B$ ligand (RANKL)-induced osteoclast differentiation of RAW 264.7 cells and pit formation in bone resorption assay (p <0.05). Moreover, it suppressed RANKL-induced $NF-{\kappa}B$ and mitogen-activated protein kinase signaling in a dose dependent manner. Downregulated osteoclast-specific gene (tartrate-resistant acid phosphatase, cathepsin K, and matrix metalloproteinase-9) expression and osteoclast proliferative transcriptional factors (nuclear factor of activated T cells-1 and c-fos) confirmed ECE-mediated suppression of osteoclastogenesis. ECE treatment ($100{\mu}g/ml$) increased heme oxygenase-1 expression by 2.5-fold and decreased intercellular reactive oxygen species production during osteoclastogenesis. The effective inhibition of RANKL-stimulated osteoclast differentiation and oxidative stress by ECE suggest that ECE has therapeutic potential in alleviating osteoclast-associated disorders.