Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
Small ; 18(6): e2106035, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34923744

ABSTRACT

Transition metal dichalcogenide (TMD) nanosheets exfoliated in the liquid phase are of significant interest owing to their potential for scalable and flexible photoelectronic applications. Although various dispersants such as surfactants, oligomers, and polymers are used to obtain highly exfoliated TMD nanosheets, most of them are electrically insulating and need to be removed; otherwise, the photoelectric properties of the TMD nanosheets degrade. Here, inorganic halide perovskite nanocrystals (NCs) of CsPbX3  (X = Cl, Br, or I) are presented as non-destructive dispersants capable of dispersing TMD nanosheets in the liquid phase and enhancing the photodetection properties of the nanosheets, thus eliminating the need to remove the dispersant. MoSe2 nanosheets dispersed in the liquid phase are adsorbed with CsPbCl3  NCs. The CsPbCl3 nanocrystals on MoSe2 efficiently withdraw electrons from the nanosheets, and suppress the dark current of the MoSe2 nanosheets, leading to flexible near-infrared MoSe2  photodetectors with a high ON/OFF photocurrent ratio and detectivity. Moreover, lanthanide ion-doped CsPbCl3  NCs enhance the ON/OFF current ratio to >106 . Meanwhile, the dispersion stability of the MoSe2  nanosheets exfoliated with the perovskite NCs is sufficiently high.

2.
Int J Mol Sci ; 23(1)2022 Jan 01.
Article in English | MEDLINE | ID: mdl-35008917

ABSTRACT

The ubiquitin proteasome system (UPS) is a protein degradation machinery that is crucial for cellular homeostasis in eukaryotes. Therefore, it is not surprising that the UPS coordinates almost all host cellular processes, including host-pathogen interactions. This protein degradation machinery acts predominantly by tagging substrate proteins designated for degradation with a ubiquitin molecule. These ubiquitin tags have been involved at various steps of the innate immune response. Hence, herpesviruses have evolved ways to antagonize the host defense mechanisms by targeting UPS components such as ubiquitin E3 ligases and deubiquitinases (DUBs) that establish a productive infection. This review delineates how herpesviruses usurp the critical roles of ubiquitin E3 ligases and DUBs in innate immune response to escape host-antiviral immune response, with particular focus on retinoic acid-inducible gene I (RIG-I)-like receptors (RLR), cyclic-GMP-AMP (cGAMP) synthase (cGAS), stimulator of interferon (IFN) genes (STING) pathways, and inflammasome signaling.


Subject(s)
Herpesviridae/immunology , Immunity, Innate , Signal Transduction , Ubiquitin/metabolism , Animals , Humans , Immunologic Factors/metabolism , Inflammation/pathology
3.
J Cell Mol Med ; 25(1): 473-483, 2021 01.
Article in English | MEDLINE | ID: mdl-33230972

ABSTRACT

Osteoporosis is a disease in which bone mineral density decreases due to abnormal activity of osteoclasts, and is commonly found in post-menopausal women who have decreased levels of female hormones. Sphingosylphosphorylcholine (SPC) is an important biological lipid that can be converted to sphingosine-1-phosphate (S1P) by autotaxin. S1P is known to be involved in osteoclast activation by stimulating osteoblasts, but bone regulation by SPC is not well understood. In this study, we found that SPC strongly inhibits RANKL-induced osteoclast differentiation. SPC-induced inhibitory effects on osteoclast differentiation were not affected by several antagonists of S1P receptors or pertussis toxin, suggesting cell surface receptor independency. However, SPC inhibited RANKL-induced calcineurin activation and subsequent NFATc1 activity, leading to decrease of the expression of Trap and Ctsk. Moreover, we found that bone loss in an experimental osteoporosis mouse model was recovered by SPC injection. SPC also blocked ovariectomy-induced body weight increase and Nfatc1 gene expression in mice. We also found that SPC inhibits RANKL-induced osteoclast differentiation in human macrophages. Since currently available treatments for osteoporosis, such as administration of female hormones or hormone receptor modulators, show serious side effects, SPC has potential as a new agent for osteoporosis treatment.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Osteoclasts/metabolism , Osteoporosis/metabolism , Ovariectomy/adverse effects , Phosphorylcholine/analogs & derivatives , Sphingosine/analogs & derivatives , Animals , Blotting, Western , Bone Resorption/drug therapy , Bone Resorption/metabolism , Cell Survival/drug effects , Female , Mice , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoporosis/drug therapy , Phosphorylcholine/therapeutic use , Real-Time Polymerase Chain Reaction , Sphingosine/therapeutic use , X-Ray Microtomography
4.
BMC Cancer ; 21(1): 137, 2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33549040

ABSTRACT

BACKGROUND: Prostate cancers frequently metastasize to bone, where the best microenvironment for distant colonization is provided. Since osteotropic metastasis of prostate cancer is a critical determinant of patients' survival, searches for preventive measures are ongoing in the field. Therefore, it is important to dissect the mechanisms of each step of bone metastasis, including the epithelial-mesenchymal transition (EMT) and cross-talk between metastatic niches and cancer cells. METHODS: In this study, we established a highly bone-metastatic subline of human prostate cancer cells by selecting bone-homing population of PC3 cells after cardiac injection of eight-week-old male BALB/c-nude mice. Then we assessed the proliferation, EMT characteristics, and migration properties of the subline (mtPC3) cells in comparison with the parental PC3 cells. To investigate the role of S100A4, we performed gene knock-down by lentiviral transduction, or treated cells with recombinant S100A4 protein or a S100A4-neutralizing antibody. The effect of cancer cells on osteoclastogenesis was evaluated after treatment of pre-osteoclasts with conditioned medium (CM) from cancer cells. RESULTS: The mtPC3 cells secreted a markedly high level of S100A4 protein and showed elevated cell proliferation and mesenchymal properties. The increased proliferation and EMT traits of mtPC3 cells was inhibited by S100A4 knock-down, but was not affected by exogenous S100A4. Furthermore, S100A4 released from mtPC3 cells stimulated osteoclast development via the cell surface receptor RAGE. Down-regulation or neutralization of S100A4 in the CM of mtPC3 cells attenuated cancer-induced osteoclastogenesis. CONCLUSION: Altogether, our results suggest that intracellular S100A4 promotes cell proliferation and EMT characteristics in tumor cells, and that secreted S100A4 activates osteoclastogenesis, contributing to osteolytic bone metastasis. Thus, S100A4 upregulation in cancer cells highly metastatic to bone might be a key element in regulating bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Cell Proliferation , Epithelial-Mesenchymal Transition , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , S100 Calcium-Binding Protein A4/metabolism , Animals , Cell Communication , Cell Differentiation , Cell Movement , Culture Media, Conditioned/pharmacology , Down-Regulation , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Osteoclasts/cytology , Osteoclasts/drug effects , Osteogenesis/drug effects , PC-3 Cells , S100 Calcium-Binding Protein A4/genetics , Up-Regulation
5.
J Immunol ; 202(12): 3359-3369, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31076532

ABSTRACT

Haptoglobin (Hp), a type of acute-phase protein, is known to have a systemic anti-inflammatory function and to modulate inflammation by directly affecting immune cells, such as T cells, dendritic cells, and macrophages. However, the effects of Hp on osteoclast differentiation are not well studied, even though osteoclast precursor cells belong to a macrophage-monocyte lineage. In this study, we found that the bone volume was reduced, and the number of osteoclasts was increased in Hp-deficient mice compared with wild-type mice. Moreover, our in vitro studies showed that Hp inhibits osteoclastogenesis by reducing the protein level of c-Fos at the early phase of osteoclast differentiation. We revealed that Hp-induced suppression of c-Fos was mediated by increased IFN-ß levels. Furthermore, Hp stimulated IFN-ß via a TLR4-dependent mechanism. These results demonstrate that Hp plays a protective role against excessive osteoclastogenesis via the Hp-TLR4-IFN-ß axis.


Subject(s)
Haptoglobins/metabolism , Interferon-beta/metabolism , Osteoclasts/physiology , Acute-Phase Reaction , Animals , Bone Resorption/genetics , Cell Differentiation , Cells, Cultured , Haptoglobins/genetics , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction
6.
Biochem Biophys Res Commun ; 516(1): 202-208, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31204051

ABSTRACT

Mitofusin 2 (Mfn2) is a mitochondrial outer membrane protein that participates in tethering mitochondria to the ER. Mitochondria-ER tethering has been demonstrated to play important roles in many cellular activities by regulating homeostasis of metabolites and calcium. Intracellular calcium signaling is crucial for the differentiation of osteoclasts, the bone-resorbing cells. In this study, we investigated whether Mfn2 plays a role in osteoclastogenesis by receptor activator of nuclear factor kappa B (RANKL) in primary cells. We found that RANKL increased Mfn2 expression during osteoclast formation from mouse bone marrow-derived macrophages (BMMs). When Mfn2 expression was suppressed in BMMs by using a siRNA-mediated gene knock-down system, osteoclast differentiation and activity of mature osteoclasts were reduced. Mfn2 knock-down also decreased the RANKL-mediated induction of NFATc1, the key transcription factor for osteoclast gene expression, without affecting c-Fos level. This effect on NFATc1 was associated with decreased calcium oscillation and calcineurin activity in Mfn2-deficient osteoclasts. Taken together, our results indicate that Mfn2 positively contributes to RANKL-induced osteoclast differentiation by regulating the calcium-calcieurin-NFATc1 axis, raising the importance of a previously under-recognized role of mitochondria in osteoclastogenesis.


Subject(s)
Calcineurin/metabolism , Calcium/metabolism , GTP Phosphohydrolases/metabolism , NFATC Transcription Factors/metabolism , Osteogenesis , Signal Transduction , Animals , Calcium Signaling , Cells, Cultured , Male , Mice , Mice, Inbred ICR , Mitochondria/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism
7.
Nanotechnology ; 30(3): 035207, 2019 Jan 18.
Article in English | MEDLINE | ID: mdl-30452390

ABSTRACT

ZnO nanoparticles (NPs) of 4-5 nm, widely adopted as an electron transport layer (ETL) in quantum dot light emitting diodes (QD-LEDs), were synthesized using the solution-precipitation process. It is notable that synthesized ZnO NPs are highly degenerate intrinsic semiconductors and their donor concentration can be increased up to N D = 6.9 × 1021 cm-3 by annealing at 140 °C in air. An optical bandgap increase of as large as 0.16-0.33 eV by degeneracy is explained well by the Burstein-Moss shift. In order to investigate the influence of intrinsic defects of ZnO NP ETLs on the performance of QD-LED devices without a combined annealing temperature between ZnO NP ETLs and the emissive QD layer, pre-annealed ZnO NPs at 60 °C, 90 °C, 140 °C, and 180 °C were spin-coated on the annealed QD layer without further post-annealing. As the annealing temperature increases from 60 °C to 180 °C, the defect density related to oxygen vacancy (V O) in ZnO NPs is reduced from 34.4% to 17.8%, whereas the defect density of interstitial Zn (Zni) is increased. Increased Zni reduces the width (W) of the depletion region from 0.21 to 0.12 nm and lowers the Schottky barrier (ФB) between ZnO NPs and the Al electrode from 1.19 to 0.98 eV. We reveal for the first time that carrier conduction between ZnO NP ETLs and the Al electrode is largely affected by the concentration of Zni above the conduction band minimum, and effectively described by space charge limited current and trap charge limited current models.

8.
J Cell Mol Med ; 22(2): 849-860, 2018 02.
Article in English | MEDLINE | ID: mdl-29077264

ABSTRACT

The G12 family of G protein alpha subunits has been shown to participate in the regulation of various physiological processes. However, the role of Gα12 in bone physiology has not been well described. Here, by micro-CT analysis, we discovered that Gα12-knockout mice have an osteopetrotic phenotype. Histological examination showed lower osteoclast number in femoral tissue of Gα12-knockout mice compared to wild-type mice. Additionally, in vitro osteoclastic differentiation of precursor cells with receptor activator of nuclear factor-κB ligand (RANKL) showed that Gα12 deficiency decreased the number of osteoclast generated and the bone resorption activity. The induction of nuclear factor of activated T-cell c1 (NFATc1), the key transcription factor of osteoclastogenesis, and the activation of RhoA by RANKL was also significantly suppressed by Gα12 deficiency. We further found that the RANKL induction of NFATc1 was not dependent on RhoA signalling, while osteoclast precursor migration and bone resorption required RhoA in the Gα12-mediated regulation of osteoclasts. Therefore, Gα12 plays a role in differentiation through NFATc1 and in cell migration and resorption activity through RhoA during osteoclastogenesis.


Subject(s)
NFATC Transcription Factors/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Resorption/pathology , Cell Differentiation/genetics , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Deletion , Humans , Macrophages/metabolism , Male , Mice, Knockout , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis , Osteopetrosis/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , rho GTP-Binding Proteins/metabolism
9.
J Bone Miner Metab ; 36(5): 537-546, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29027021

ABSTRACT

Bone homeostasis is achieved through coordinated activities of bone-forming osteoblasts and bone-resorbing osteoclasts. When the balance is skewed in favor of osteoclasts due to hormonal or inflammatory issues, pathologic bone loss occurs leading to conditions such as osteoporosis, rheumatoid arthritis, and periodontitis. Bortezomib is the first in-class of proteasome inhibitors used as an anti-myeloma agent. In the present study, we show that bortezomib directly inhibited the receptor activator of nuclear factor κB ligand (RANKL)-dependent osteoclast differentiation of mouse bone marrow macrophages. Bortezomib significantly reduced the induction of osteoclast marker genes and proteins including nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1). The intraperitoneal injection of bortezomib reduced ovariectomy-induced osteoclastogenesis and protected the mice from bone loss. These data propose novel use of bortezomib as a potential anti-resorptive agent.


Subject(s)
Bortezomib/therapeutic use , Cell Differentiation , Osteoclasts/pathology , Osteoporosis/drug therapy , Osteoporosis/prevention & control , Ovariectomy , Animals , Biomarkers/metabolism , Bone Resorption/complications , Bone Resorption/drug therapy , Bone Resorption/pathology , Bone Resorption/prevention & control , Bortezomib/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Female , Humans , Mice, Inbred ICR , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoporosis/complications , Osteoporosis/pathology
10.
J Immunol ; 196(9): 3887-95, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27016605

ABSTRACT

A balance between bone formation and bone resorption is critical for the maintenance of bone mass. In many pathological conditions, including chronic inflammation, uncontrolled activation of osteoclast differentiation often causes excessive bone resorption that results in osteoporosis. In this study, we identified the osteopenia phenotype of mice lacking Usp18 (also called Ubp43), which is a deISGylating enzyme and is known as a negative regulator of type I IFN signaling. The expression of Usp18 was induced in preosteoclasts upon receptor activator of NF-κB ligand (RANKL) treatment. In an in vitro osteoclast-differentiation assay, bone marrow macrophages from Usp18-deficient mice exhibited an enhanced differentiation to multinucleated cells, elevated activation of NFATc1, and an increased expression of osteoclast marker genes upon RANKL treatment. Furthermore, in vitro quantification of bone resorption revealed a great increase in osteoclastic activities in Usp18-deficient cells. Interestingly, proinflammatory cytokine genes, such as IP-10 (CXCL10), were highly expressed in Usp18-deficient bone marrow macrophages upon RANKL treatment compared with wild-type cells. In addition, serum cytokine levels, especially IP-10, were significantly high in Usp18-knockout mice. In sum, we suggest that, although type I IFN is known to restrict osteoclast differentiation, the exaggerated activation of the type I IFN response in Usp18-knockout mice causes an osteopenia phenotype in mice.


Subject(s)
Macrophages/physiology , Osteoclasts/physiology , Osteogenesis , Osteoporosis/immunology , Ubiquitin Thiolesterase/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chemokine CXCL10/metabolism , Interferon Type I/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , NFATC Transcription Factors/metabolism , Osteogenesis/genetics , Osteogenesis/immunology , RANK Ligand/metabolism , Ubiquitin Thiolesterase/genetics
11.
Nano Lett ; 16(11): 6946-6953, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27733041

ABSTRACT

Although various colloidal quantum dot (QD) coating and patterning techniques have been developed to meet the demands in optoelectronic applications over the past years, each of the previously demonstrated methods has one or more limitations and trade-offs in forming multicolor, high-resolution, or large-area patterns of QDs. In this study, we present an alternative QD patterning technique using conventional photolithography combined with charge-assisted layer-by-layer (LbL) assembly to solve the trade-offs of the traditional patterning processes. From our demonstrations, we show repeatable QD patterning process that allows multicolor QD patterns in both large-area and microscale. Also, we show that the QD patterns are robust against additional photolithography processes and that the thickness of the QD patterns can be controlled at each position. To validate that this process can be applied to actual device applications as an active material, we have fabricated inverted, differently colored, active QD light-emitting device (QD-LED) on a pixelated substrate, which achieved maximum electroluminescence intensity of 23 770 cd/m2, and discussed the results. From our findings, we believe that our process provides a solution to achieving both high-resolution and large-scale QD pattern applicable to not only display, but also to practical photonic device research and development.

12.
J Biol Chem ; 290(10): 6522-30, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25561739

ABSTRACT

Lipid raft microdomains have important roles in various cellular responses. Caveolae are a specialized type of lipid rafts that are stabilized by oligomers of caveolin proteins. Here, we show that caveolin-1 (Cav-1) plays a crucial role in the regulation of osteoclastogenesis. We found that caveolin-1 was dramatically up-regulated by receptor activator of nuclear factor κB ligand (RANKL), the osteoclast differentiation factor. Knockdown of Cav-1 reduced osteoclastogenesis and induction of NFATc1, the master transcription factor for osteoclastogenesis, by RANKL. Consistent with the in vitro results, injection of caveolin-1 siRNA onto mice calvariae showed reduction in RANKL-induced bone resorption and osteoclast formation. Moreover, Cav-1(-/-) female mice had higher bone volume and lower osteoclast number compared with wild type mice. However, Cav-1(-/-) male mice had both osteoclast and osteoblast numbers higher than wild type mice with no difference in bone volume. The sex dependence in the effect of Cav-1 deficiency was partly attributed to decreased receptor activator of nuclear factor κB and increased cFms expression in osteoclast precursors of female and male mice, respectively. Taken together, these data demonstrate that Cav-1 has a complicated but critical role for osteoclastogenesis.


Subject(s)
Bone Development/genetics , Caveolin 1/genetics , Cell Differentiation/genetics , Osteoclasts/metabolism , Animals , Caveolin 1/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Male , Membrane Microdomains/genetics , Membrane Microdomains/metabolism , Mice , NFATC Transcription Factors/biosynthesis , Osteoclasts/chemistry , Osteogenesis/genetics , RANK Ligand/biosynthesis , Sex Characteristics
13.
Int J Cancer ; 138(5): 1232-45, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26413934

ABSTRACT

Elevated expression and aberrant activation of Ras have been implicated in breast cancer aggressiveness. H-Ras, but not N-Ras, induces breast cell invasion. A crucial link between lipid rafts and H-Ras function has been suggested. This study sought to identify the lipid raft protein(s) responsible for H-Ras-induced tumorigenicity and invasiveness of breast cancer. We conducted a comparative proteomic analysis of lipid raft proteins from invasive MCF10A human breast epithelial cells engineered to express active H-Ras and non-invasive cells expressing active N-Ras. Here, we identified a lipid raft protein flotillin-1 as an important regulator of H-Ras activation and breast cell invasion. Flotillin-1 was required for epidermal growth factor-induced activation of H-Ras, but not that of N-Ras, in MDA-MB-231 triple-negative breast cancer (TNBC) cells. Flotillin-1 knockdown inhibited the invasiveness of MDA-MB-231 and Hs578T TNBC cells in vitro and in vivo. In xenograft mouse tumor models of these TNBC cell lines, we showed that flotillin-1 played a critical role in tumor growth. Using human breast cancer samples, we provided clinical evidence for the metastatic potential of flotillin-1. Membrane staining of flotillin-1 was positively correlated with metastatic spread (p = 0.013) and inversely correlated with patient disease-free survival rates (p = 0.005). Expression of flotillin-1 was associated with H-Ras in breast cancer, especially in TNBC (p < 0.001). Our findings provide insight into the molecular basis of Ras isoform-specific interplay with flotillin-1, leading to tumorigenicity and aggressiveness of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Genes, ras , Membrane Proteins/physiology , Adult , Aged , Animals , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Proteomics , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction
14.
Biochem Biophys Res Commun ; 477(4): 1078-1084, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27416754

ABSTRACT

Tetraspanin family proteins regulate morphology, motility, fusion, and signaling in various cell types. We investigated the role of the tetraspanin 7 (Tspan7) isoform in the differentiation and function of osteoclasts. Tspan7 was up-regulated during osteoclastogenesis. When Tspan7 expression was reduced in primary precursor cells by siRNA-mediated gene knock-down, the generation of multinuclear osteoclasts was not affected. However, a striking cytoskeletal abnormality was observed: the formation of the podosome belt structure was inhibited and the microtubular network were disrupted by Tspan7 knock-down. Decreases in acetylated microtubules and levels of phosphorylated Src and Pyk2 in Tspan7 knock-down cells supported the involvement of Tspan7 in cytoskeletal rearrangement signaling in osteoclasts. This cytoskeletal defect interfered with sealing zone formation and subsequently the bone-resorbing activity of mature osteoclasts on dentin surfaces. Our results suggest that Tspan7 plays an important role in cytoskeletal organization required for the bone-resorbing function of osteoclasts by regulating signaling to Src, Pyk2, and microtubules.


Subject(s)
Bone Resorption/metabolism , Bone Resorption/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Podosomes/metabolism , Tetraspanins/metabolism , Animals , Cell Movement , Cell Survival , Cells, Cultured , Female , Mice , Osteogenesis , Podosomes/pathology
15.
Stem Cells ; 32(9): 2467-79, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24801901

ABSTRACT

In bone marrow, bone marrow stromal cells (BMSCs) have the capacity to differentiate into osteoblasts and adipocytes. Age-related osteoporosis is associated with a reciprocal decrease of osteogenesis and an increase of adipogenesis in bone marrow. In this study, we demonstrate that disruption of nuclear factor I-C (NFI-C) impairs osteoblast differentiation and bone formation, and increases bone marrow adipocytes. Interestingly, NFI-C controls postnatal bone formation but does not influence prenatal bone development. We also found decreased NFI-C expression in osteogenic cells from human osteoporotic patients. Notably, transplantation of Nfic-overexpressing BMSCs stimulates osteoblast differentiation and new bone formation, but inhibits adipocyte differentiation by suppressing peroxisome proliferator-activated receptor gamma expression in Nfic(-/-) mice showing an age-related osteoporosis-like phenotype. Finally, NFI-C directly regulates Osterix expression but acts downstream of the bone morphogenetic protein-2-Runx2 pathway. These results suggest that NFI-C acts as a transcriptional switch in cell fate determination between osteoblast and adipocyte differentiation in BMSCs. Therefore, regulation of NFI-C expression in BMSCs could be a novel therapeutic approach for treating age-related osteoporosis.


Subject(s)
NFI Transcription Factors/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Transcription Factors/biosynthesis , Aged , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Gene Expression Profiling , Humans , Male , Mice , Mice, Transgenic , NFI Transcription Factors/genetics , Osteogenesis/physiology , Sp7 Transcription Factor , Transfection
16.
J Cell Physiol ; 229(12): 2166-74, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24891219

ABSTRACT

Cell fusion is a fundamental biological event that is essential for the development of multinucleated cells such as osteoclasts. Fusion failure leads to the accumulation of dense bone such as in osteopetrosis, demonstrating the importance of fusion in osteoclast maturity and bone remodeling. In a recent study, we reported that Pin1 plays a role in the regulation of bone formation and Runx2 regulation. In this study, we explored the role of Pin1 in osteoclast formation and bone resorption. Pin1 null mice have low bone mass and increased TRAP staining in histological sections of long bones, compared to Pin1 wild-type mice. In vitro osteoclast forming assays with bone marrow-derived monocyte/macrophage revealed that Pin1-deficient osteoclasts are larger than wild-type osteoclasts and have higher nuclei numbers, indicating greater extent of fusion. Pin1 deficiency also highly enhanced foreign body giant cell formation both in vitro and in vivo. Among the known fusion proteins, only DC-STAMP was significantly increased in Pin1(-/-) osteoclasts. Immunohistochemistry showed that DC-STAMP expression was also significantly increased in tibial metaphysis of Pin1 KO mice. We found that Pin1 binds and isomerizes DC-STAMP and affects its expression levels and localization at the plasma membrane. Taken together, our data indicate that Pin1 is a determinant of bone mass through the regulation of the osteoclast fusion protein DC-STAMP. The identification of Pin1 as a factor involved in cell fusion contributes to the understanding of osteoclast-associated diseases, including osteoporosis, and opens new avenues for therapeutic targets.


Subject(s)
Cell Fusion , Membrane Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Osteoclasts/metabolism , Peptidylprolyl Isomerase/genetics , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Nerve Tissue Proteins/genetics , Osteoclasts/physiology , Osteogenesis/genetics , Osteogenesis/physiology , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteopetrosis/pathology , Peptidylprolyl Isomerase/metabolism
17.
Exp Cell Res ; 319(4): 436-46, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23246654

ABSTRACT

Apolipoprotein E (ApoE) plays a major role in the transport and metabolism of lipid. Other functions of ApoE include modulation of innate and adaptive immune responses. The expression of ApoE in osteoblasts and its relevance with bone formation have also been reported. However, the effect of ApoE on osteoclasts has not yet been examined. Here, we investigated the role of ApoE in osteoclast differentiation using bone marrow-derived macrophages (BMMs) and RAW264.7 cells. We found a down-regulation of ApoE gene expression during osteoclastic differentiation of those cells. Overexpression of ApoE in BMMs and RAW264.7 cells significantly blocked the induction of c-Fos and nuclear factor of activated T cell c1 (NFATc1), transcription factors critical for expression of osteoclast marker genes, by receptor activator of nuclear factor κB ligand (RANKL), the osteoclast differentiation factor. ApoE inhibited osteoclast differentiation, as measured by decreased number of tartrate-resistant acid phosphatase (TRAP)-positive multinuclear cells (MNCs). In addition, ApoE reduced the expression of dendritic cell-specific transmembrane protein (DC-STAMP) and ATPase, H(+) transporting, lysosomal 38kDa, V0 subunit d2 (ATP6v0d2), genes involved in cell-cell fusion during osteoclastogenesis. Knock-down of ApoE using a specific siRNA promoted the RANKL-mediated induction of osteoclast differentiation. While ApoE did not affect the activation of ERK, JNK, and p38 MAPK signaling pathways by RANKL, the phosphorylation of p65 trans-activation domain on serine 536 and transcription activity of NF-κB were reduced by ApoE overexpression. These findings suggest that ApoE plays an inhibitory role in osteoclast differentiation via the suppression of RANKL-dependent activation of NF-κB and induction of c-Fos and NFATc1.


Subject(s)
Apolipoproteins E/physiology , Cell Differentiation/genetics , Genes, fos , NF-kappa B/genetics , NFATC Transcription Factors/genetics , Osteoclasts/physiology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Apolipoproteins E/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation/drug effects , Genes, fos/drug effects , Genes, fos/genetics , Genes, fos/physiology , Mice , NF-kappa B/metabolism , NF-kappa B/physiology , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/physiology , Osteoclasts/drug effects , Osteoclasts/metabolism , RANK Ligand/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection
18.
Mol Cells ; 47(4): 100059, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38554844

ABSTRACT

Periodontitis (PD) is an inflammatory disease with alveolar bone destruction by osteoclasts (OCs). In PD, both inflammation and OC activation are significantly influenced by periodontal ligament fibroblasts (PDL-Fib). Yet, whether PDL-Fib has heterogeneity and whether distinct PDL-Fib subsets have specific functions have not been investigated. In this study, we discovered the complexity of PDL-Fib in PD, utilizing single-cell RNA sequencing data from human PD patients. We identified distinct subpopulations of PDL-Fib: one expressing interleukin-1 beta (IL-1ß) and another expressing the receptor activator of nuclear factor-kappa B ligand (RANKL), both crucial in OC differentiation and bone resorption. In periodontal tissues of mice with PD, active IL-1ß, cleaved caspase 1, and nucleotide-binding oligomerization domain-like receptor 3 (NLPR3) were significantly elevated, implicating the NLRP3 inflammasome in IL-1ß production. Upon stimulation of PDL-Fib with LPS from Porphyromonas gingivalis (pg), the most well-characterized periodontal bacteria, a more rapid increase in IL-1ß, followed by RANKL induction, was observed. IL-1ß and tumor necrosis factor alpha (TNF-α), another LPS-responsive cytokine, effectively increased RANKL in PDL-Fib, suggesting an indirect effect of pgLPS through IL-1ß and TNF-α on RANKL induction. Immunohistological analyses of mouse periodontal tissues also showed markedly elevated levels of IL-1ß and RANKL upon PD induction and displayed separate locations of IL-1ß-expressing PDL-Fib and RANKL-expressing PDL-Fib in PD. The heterogenic feature of fibroblasts expressing IL-1ß and RANKL was also mirrored in our combined cross-tissue single-cell RNA sequencing datasets analysis. In summary, our study elucidates the heterogeneity of PDL-Fib, highlighting distinct functional groups for producing RANKL and IL-1ß, which collectively promote OC generation and bone destruction in PD.


Subject(s)
Fibroblasts , Interleukin-1beta , Periodontal Ligament , Periodontitis , RANK Ligand , Single-Cell Analysis , Periodontal Ligament/metabolism , Periodontal Ligament/cytology , Periodontal Ligament/pathology , RANK Ligand/metabolism , RANK Ligand/genetics , Fibroblasts/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Periodontitis/metabolism , Periodontitis/genetics , Periodontitis/pathology , Humans , Animals , Mice , Gene Expression Profiling , Osteoclasts/metabolism , Male , Mice, Inbred C57BL , Single-Cell Gene Expression Analysis
19.
J Extracell Vesicles ; 13(1): e12407, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38251423

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is associated with nonalcoholic fatty liver disease (NAFLD) by lipid accumulation in the liver. In this study, we showed that extracellular vesicles (EVs) from the periodontal pathogens Filifactor alocis and Porphyromonas gingivalis induced steatosis by inducing PAI-1 in the liver and serum of mice fed a low-fat diet. PAI-1 induction was not observed in TLR2-/- mice. When tested using HEK-Blue hTLR2 cells, human TLR2 reporter cells, the TLR2-activating ability of serum from NAFLD patients (n = 100) was significantly higher than that of serum from healthy subjects (n = 100). Correlation analysis confirmed that PAI-1 levels were positively correlated with the TLR2-activating ability of serum from NAFLD patients and healthy subjects. Amphiphilic molecules in EVs were involved in PAI-1 induction. Our data demonstrate that the TLR2/PAI-1 axis is important for hepatic steatosis by EVs of periodontal pathogens.


Subject(s)
Extracellular Vesicles , Non-alcoholic Fatty Liver Disease , Plasminogen Activator Inhibitor 1 , Toll-Like Receptor 2 , Animals , Humans , Mice
20.
Redox Biol ; 69: 103023, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38181706

ABSTRACT

The oral colonization of periodontal pathogens onto gingival tissues establishes hypoxic microenvironment, often disrupting periodontal homeostasis in conjunction with oxidative stress. The association between reactive oxygen species (ROS) and osteolytic periodontitis have been suggested by recent studies. PTEN-induced kinase 1 (PINK1), a mitochondrial serine/threonine kinase, is an essential protein for mitochondrial quality control as it protects cells from oxidative stress by promoting degradation of damaged mitochondria through mitophagy. However, the pathophysiological roles of PINK1 in osteoclast-mediated bone loss have not been explored. Here we aimed to determine whether PINK1 plays a role in the regulation of osteoclastogenesis and alveolar bone resorption associated with periodontitis. C57BL/6 wild type (WT) and Pink1 knockout (KO) mice were subjected to ligature-induced periodontitis (LIP), and alveolar bones were evaluated by µCT-analysis and tartrate-resistant acid phosphatase (TRAP) staining. The µCT-analysis showed that bone volume fraction and travecular thickness were lower in Pink1 KO compared to WT mice. The number of TRAP-positive osteoclasts was markedly increased in the periodontal tissues of Pink1 KO mice with LIP. The genetic silencing or deletion of Pink1 promoted excessive osteoclast differentiation and bone resorption in vitro, as respectively indicated by TRAP staining and resorption pits on dentin slices. PINK1 deficiency led to mitochondrial instabilities as indicated by confocal microscopy of mitochondrial ROS, mitochondrial oxygen consumption rate (OCR) analysis, and transmission electron microscopy (TEM). Consequently, a significant increase in Ca2+-nuclear factor of activated T cells 1 (NFATc1) signaling was also found. On the other hand, restoration of mitophagy and autophagy by spermidine (SPD) treatment and the resolution of oxidative stress by N-acetyl-l-cysteine (NAC) treatment protected PINK1 deficiency-induced excessive generation of osteoclasts. Taken together, our findings demonstrate that PINK1 is essential for maintaining mitochondrial homeostasis during osteoclast differentiation. Therefore, targeting PINK1 may provide a novel therapeutic strategy for severe periodontitis with fulminant osteolysis.


Subject(s)
Alveolar Bone Loss , Periodontitis , Animals , Mice , Alveolar Bone Loss/complications , Alveolar Bone Loss/drug therapy , Mice, Inbred C57BL , Mitophagy/genetics , Osteoclasts/metabolism , Periodontitis/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL