Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Stem Cell Rev Rep ; 16(3): 596-611, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32112264

ABSTRACT

Osteoarthritis (OA) is a general joint disease. Cartilage damage is associated with a decrease in the density of chondrocytes. Mesenchymal stem cells (MSCs) differentiate into adipocytes, osteocytes and chondrocytes, and are an excellent source of cell therapy. Cartilage-derived extracellular matrix (ECM) promotes chondrogenesis of MSCs. However, the role of MSCs stimulated by ECM is not well known in OA. The purpose of this study is to determine the role of specific factors generated by the application of ECM and umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) in managing OA symptoms. Cartilage acellular matrix (CAM), which is a cartilage-derived ECM, was used to promote the chondrogenesis of UCB-MSCs. Induced MSCs were analyzed using chondrogenic markers (aggrecan, collagen type 2, and SOX9) and bone morphogenic protein 6 (BMP6). BMP6 is known to be involved in early chondrogenesis of MSCs. As a result, treatment with CAM significantly increased the expression of chondrogenic markers and BMP6 in UCB-MSCs. Treatment with recombinant human BMP6 also dramatically increased the levels of chondrogenic markers in UCB-MSCs. In addition, UCB-MSCs and CAM were used to evaluate OA symptom improvement in a rabbit articular cruciate ligament transection (ACLT) model. Application of UCB-MSCs and CAM enhanced not only the structure and synthesis of proteoglycan and collagen type 2 but also anti-inflammatory effects in both rabbit joint and synovial fluid. Moreover, the detection of human cells and involvement of BMP6 were confirmed in rabbit cartilage tissues. This study indicates that therapeutic potential of UCB-MSCs with CAM is mediated via BMP6 in OA.


Subject(s)
Anterior Cruciate Ligament Injuries/therapy , Bone Morphogenetic Protein 6/pharmacology , Cartilage, Articular/pathology , Extracellular Matrix/metabolism , Fetal Blood/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Anterior Cruciate Ligament Injuries/diagnostic imaging , Anterior Cruciate Ligament Injuries/pathology , Behavior, Animal , Cell Tracking , Chondrogenesis , Disease Models, Animal , Humans , Osteoarthritis/pathology , Paracrine Communication , Rabbits , Synovial Fluid/metabolism
2.
J Tissue Eng Regen Med ; 13(7): 1134-1142, 2019 07.
Article in English | MEDLINE | ID: mdl-30959558

ABSTRACT

Rheumatoid arthritis (RA) is a common inflammatory chronic disease. It has been reported that mesenchymal stem cells (MSCs) have the effect of immune suppression in collagen-induced arthritis (CIA) mice model. However, the in vivo therapeutic effect from the long-interval repeated intravenous administration of human umbilical cord blood-derived (hUCB)-MSCs had not been investigated in CIA mice model. This study was undertaken to investigate the effects of long-interval repeated intravenous administration of hUCB-MSCs at different doses in CIA mice model. Mice were intravenously injected with three different doses of hUCB-MSCs once every 2 weeks for three times. RA severity was assessed by clinical joint score and histologic analysis including hematoxylin and eosin staining, safranin-O staining, and toluidine blue staining. We used real-time polymerase chain reaction and flow cytometry to quantify differences in inflammatory cytokines and Tregs. Mice treated with hUCB-MSCs showed significant improvement in clinical joint score. Histologic analysis revealed that hUCB-MSCs definitely reduced joint inflammation, cartilage damage, and formation of pannus in multimedium and multihigh groups. These hUCB-MSCs also significantly decreased IL-1 beta protein levels in multimedium and multihigh groups and IL-6 protein levels in all hUCB-MSCs-treated groups. Furthermore, mRNA levels of IL-1 beta and IL-6 were decreased significantly in all hUCB-MSCs-treated groups, whereas the expression of anti-inflammatory cytokine IL-10 was increased in the multihigh group. Tregs known as suppressor T cells were also significantly increased in the multihigh group. Our findings suggest that long-interval repeated intravenous administration of hUCB-MSCs has therapeutic effects by improving symptoms of RA in CIA mice model in a dose-dependent manner.


Subject(s)
Arthritis, Experimental , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Umbilical Cord/metabolism , Administration, Intravenous , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Experimental/therapy , Female , Heterografts , Humans , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred DBA , Time Factors , Umbilical Cord/pathology
3.
Cell Death Dis ; 9(5): 495, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717113

ABSTRACT

Chemokine CCL4 (MIP-1ß) is released from osteoblast cells to restore the homeostasis of hematopoietic stem cells during the activation of bone marrow. In this study, we investigated the function of CCL4 and its receptor CCR5 during osteoclastogenesis. CCL4 promoted the migration and viability of preosteoclast cells. However, CCL4 had no direct effect on the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation in mouse preosteoclast cells. In addition, CCR5 expression was rapidly reduced by RANKL treatment, which was recovered by IFN-γ during osteoclastogenesis. CCR5 downregulation by RANKL was mediated by MEK and JNK in preosteoclast cells and promoted osteoclastogenesis. These results suggest that CCL4 can enhance the recruitment of preosteoclasts to bone in the early stage, and the reduction of CCR5 promotes osteoclastogenesis when RANKL is prevalent.


Subject(s)
Cell Differentiation/drug effects , Chemokine CCL4/pharmacology , Monocytes/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/pharmacology , Receptors, CCR5/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice, Inbred BALB C , Monocytes/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Receptors, CCR5/genetics , Signal Transduction
4.
Bone ; 103: 287-294, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28739418

ABSTRACT

MicroRNAs (miRNA) significantly contribute to bone formation by post-transcriptional regulation of gene expression. Mature miRNAs are generated following sequential cleavage by DROSHA/DGCR8 and DICER. However, recent studies have identified that some miRNAs require only one of these enzymes. Most studies seeking to clarify the role of miRNA during bone formation have been performed using DICER deletion strategies, but little is known regarding the role of DGCR8. To study the function of DGCR8 in osteogenesis, we generated mice in which Dgcr8 is conditionally deleted in osteoprogenitor cells by Col1a1-Cre. Dgcr8-cKO mice showed increased bone volume (BV/TV), trabecular number (Tb/N), and trabecular thickness (Tb.Th), but decreased trabecular separation (Tb.Sp) in the femur. Von Kossa, tartrate-resistant acid phosphatase staining, and calcein double labeling identified that osteoblast activity is increased in Dgcr8-cKO mice. In an effort to elucidate a detailed cellular mechanism, we found that miR-22 was downregulated in Dgcr8-cKO mice, leading to upregulation of the osteocalcin transcript, a key marker of osteoblasts. Interestingly, the mRNA expression level of Dgcr8 was decreased during osteoblast differentiation. Taken together, these results strongly indicate that DGCR8-dependent generation of miR-22 is essential for bone formation and that miR-22 could be a therapeutic target for individuals with bone disease.


Subject(s)
Gene Expression Regulation/genetics , MicroRNAs/biosynthesis , Osteogenesis/genetics , RNA-Binding Proteins/metabolism , Animals , Down-Regulation , Mice , Mice, Knockout , Osteocalcin/biosynthesis , Osteocalcin/genetics
5.
Cell Commun Signal ; 15(1): 25, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28662672

ABSTRACT

BACKGROUND: Hematopoietic stem cell (HSC) maintenance requires a specific microenvironment. HSC niches can be activated by tissue damaging chemotherapeutic drugs and various cell signaling molecules such as SDF-1 and FGF, which might also result in bone marrow stress. Recent research has insufficiently shown that endosteal osteolineage cells and other niche constituents recover after marrow injury. METHODS: We investigated the role of FGF2 in the osteoblastic niche cells during hematopoietic homeostasis recovery after bone marrow injury. Mice were treated with 5-fluorouracil (5FU) to eliminate actively cycling cells in the bone marrow. Primary osteoblasts were isolated and subjected to cell culture. Real-time PCR, western blot and immunohistochemical staining were performed to study niche-related genes, osteoblast markers, and FGF2 signaling. Proliferation rate were analyzed by marker gene Ki67 and colony formation assay. Also, osterix-positive osteoprogenitor cells were isolated by FACS from Osx-GFP-Cre mice after 5FU treatment, and subjected to RNA-sequencing and analyzed for Fgf receptors and niche markers. RESULTS: The endosteal osteolineage cells isolated from 5FU-treated mice showed increased expression of the niche-related genes Sdf-1, Jagged-1, Scf, N-cad, Angpt1 and Vcam-1 and the osteoblast marker genes Osx, Opn, Runx2, and Alp, indicating that BM stress upon 5FU treatment activated the osteoblastic niche. Endosteal osteoblast expanded from a single layer to several layers 3 and 6 days after 5FU treatment. During the early recovery phase in 5FU-activated osteoblastic niches increased FGF2 expression and activated its downstream pERK. FGF2 treatment resulted in increased proliferation rate and the expression of niche marker genes in 5FU-activated osteoblastic niche cells. RNA-seq analysis in Osterix-positive osteoprogenitor cells isolated from 5FU-treated Osx-GFP mice showed significantly increased expression of Fgf receptors Fgfr1, 2 and 3. Although osteoblastic niche cells were damaged by 5FU treatment in the beginning, the increased number of OB layers in the recovery phase may be derived from resident osteoprogenitor cells by FGF2 activation under stress. CONCLUSIONS: Taken together, FGF2 signaling can regulate osteoblastic niche cells to support HSC homeostasis in response to bone marrow damage.


Subject(s)
Bone Marrow/immunology , Fibroblast Growth Factor 2/metabolism , Hematopoietic Stem Cells/cytology , Homeostasis , Osteoblasts/cytology , Stem Cell Niche , Animals , Bone Marrow/drug effects , Bone Marrow/microbiology , Cell Proliferation/drug effects , Fluorouracil/pharmacology , Hematopoietic Stem Cells/drug effects , Homeostasis/drug effects , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Receptors, Fibroblast Growth Factor/genetics , Stem Cell Niche/drug effects
7.
Exp Cell Res ; 334(2): 301-9, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25814363

ABSTRACT

Lipocalin-2 (LCN2) is a member of the lipocalin superfamily and plays a critical role in the regulation of various physiological processes, such as inflammation and obesity. In this study, we report that LCN2 negatively modulates the proliferation and differentiation of osteoclast precursors, resulting in impaired osteoclast formation. The overexpression of LCN2 in bone marrow-derived macrophages or the addition of recombinant LCN2 protein inhibits the formation of multinuclear osteoclasts. LCN2 suppresses macrophage colony-stimulating factor (M-CSF)-induced proliferation of osteoclast precursor cells without affecting their apoptotic cell death. Interestingly, LCN2 decreases the expression of the M-CSF receptor, c-Fms, and subsequently blocks its downstream signaling cascades. In addition, LCN2 inhibits RANKL-induced osteoclast differentiation and attenuates the expression of c-Fos and nuclear factor of activated T cells c1 (NFATc1), which are important modulators in osteoclastogenesis. Mechanistically, LCN2 inhibits NF-κB signaling pathways, as demonstrated by the suppression of IκBα phosphorylation, nuclear translocation of p65, and NF-κB transcriptional activity. Thus, LCN2 is an anti-osteoclastogenic molecule that exerts its effects by retarding the proliferation and differentiation of osteoclast lineage cells.


Subject(s)
Acute-Phase Proteins/metabolism , Cell Differentiation , Cell Lineage , Lipocalins/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Proliferation , Cells, Cultured , Humans , Lipocalin-2 , Mice , Mice, Inbred C57BL
8.
J Leukoc Biol ; 94(1): 99-107, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23657115

ABSTRACT

LXRs are nuclear receptors that function as important regulators of lipid homeostasis and inflammatory responses. LXR activation has been shown to suppress RANKL-induced osteoclast differentiation, but its underlying mechanisms and its influence on inflammatory bone destruction remain unclear. In this study, we report that the LXR agonists T0901317 and GW3965 inhibit osteoclastogenesis from primary BMMs in a dose-dependent manner. LXR activation suppressed RANKL-induced transcriptional activity of NF-κB without affecting IκBα degradation and the phosphorylation of p38. LXR agonists significantly suppressed RANKL-induced expression of c-Fos and NFATc1, which are crucial transcription factors for osteoclastogenesis. The activation of LXRs also inhibited RANKL-mediated AP-1 transcriptional activity. Furthermore, LXR activation attenuated PPARγ ligand-induced c-Fos expression, and LXR suppressed AP-1 promoter activity by PPARγ. The inhibitory effect of LXR activation on osteoclastogenesis was reversed by overexpression of c-Fos, suggesting that c-Fos is a downstream target of the antiosteoclastogenic action of LXRs. In addition to osteoclast differentiation, LXR activation accelerated apoptosis in mature osteoclasts by the induction of caspase-3 and -9 activity and Bim expression. Consistent with the in vitro effects we observed, the administration of a LXR agonist protected from bone loss induced by LPS in vivo. Together, our data provide evidence that LXRs may have potential as therapeutic targets for bone resorption-associated diseases.


Subject(s)
Alveolar Bone Loss/prevention & control , Bone Development/physiology , Cell Differentiation , NF-kappa B/antagonists & inhibitors , Orphan Nuclear Receptors/physiology , Osteoclasts/cytology , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Animals , Apoptosis , Benzoates/pharmacology , Benzylamines/pharmacology , Blotting, Western , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Proliferation , Cells, Cultured , Hydrocarbons, Fluorinated/pharmacology , Immunoenzyme Techniques , Lipopolysaccharides/pharmacology , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Orphan Nuclear Receptors/agonists , Osteoclasts/physiology , PPAR gamma/genetics , PPAR gamma/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Transcription, Genetic
9.
Life Sci ; 91(19-20): 928-34, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23000100

ABSTRACT

AIMS: Osteoclasts, the unique bone-resorbing polykaryons, are responsible for many bone-destructive diseases, such as osteoporosis and rheumatoid arthritis. Hence, the regulation of osteoclast formation is considered a potential therapeutic approach for these diseases. In this study, we investigated the effect of a novel small compound, C(25)H(32)N(4)O(4)S(2) (NecroX-7) on osteoclast formation. MAIN METHODS: We analyzed the effects of NecoX-7 on receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclast differentiation in vitro and LPS-induced bone loss in vivo. KEY FINDINGS: We observed that NecroX-7 suppressed osteoclast formation from primary bone marrow macrophages (BMMs) in a dose-dependent manner. NecroX-7 significantly inhibited the NF-κB signaling pathway without affecting the activation of the mitogen-activated protein kinases (MAPKs) JNK, p38, and ERK in response to RANKL. In addition, NecroX-7 strongly attenuated the induction of c-Fos and nuclear factor of activated T cells c1 (NFATc1), which are crucial transcription factors for osteoclast differentiation. Mirroring the down-regulation of c-Fos and NFATc1, the expression of osteoclastogenic markers, such as tartrate-resistant acid phosphatase (TRAP) and cathepsin K, was also reduced by the addition of NecroX-7. Furthermore, confirming the in vitro anti-osteoclastogenic effect, NecroX-7 inhibited lipopolysaccharide (LPS)-induced bone loss in vivo. SIGNIFICANCE: Our data imply that NecroX-7 is useful as a therapeutic drug for the treatment of bone resorption-associated diseases.


Subject(s)
Cell Differentiation/drug effects , NF-kappa B/metabolism , Organic Chemicals/pharmacology , Osteoclasts/drug effects , Proto-Oncogene Proteins c-fos/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Resorption/drug therapy , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Organic Chemicals/administration & dosage , Osteoclasts/metabolism , RANK Ligand/metabolism , Signal Transduction/drug effects
10.
Bone ; 51(4): 643-50, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22842221

ABSTRACT

Early growth response 2 (Egr2) is a zinc finger transcription factor that acts as an important modulator of various physiological processes. In this study, we show that Egr2 negatively regulates receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation. The overexpression of Egr2 in bone marrow-derived macrophages (BMMs) suppresses the formation of multinuclear osteoclasts and the expression of osteoclastogenic markers, including nuclear factor of activated T cells c1 (NFATc1). On the other hand, Egr2 overexpression does not impact the phagocytic activity of osteoclast precursors or the expression of macrophage-specific markers in the presence of the osteoclastogenic stimuli, RANKL and M-CSF. We further demonstrate that Egr2 induces the expression of the inhibitors of differentiation/DNA binding (Ids) helix-loop-helix (HLH) transcription factors, which are important repressors in RANKL-mediated osteoclastogenesis. Egr2 transactivates the Id2 promoter and increases its recruitment to the Id2 promoter region. In addition, Egr2-dependent induction of Id2 promoter activity, and its binding to the Id2 promoter is abrogated by the overexpression of the Egr2 repressor, NGFI-A binding protein 2 (Nab2). Accordingly, coexpression with Nab2 restores Egr2-mediated suppression of osteoclast differentiation. Furthermore, knockdown of Egr2 using shRNA enhances osteoclastogenesis and decreases Id2 gene expression. Ectopic expression of Id2 reverses the phenotype mediated by Egr2 silencing. Taken together, our results identify Egr2 as an important modulator of RANKL-induced osteoclast differentiation and provide the link between RANKL, Egr2 and Id proteins in osteoclast-lineage cells.


Subject(s)
Cell Differentiation/physiology , Early Growth Response Protein 2/physiology , Helix-Loop-Helix Motifs , Osteoclasts/cytology , Up-Regulation , Animals , Base Sequence , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , DNA Primers , Mice , Mice, Inbred C57BL , Phagocytosis , Real-Time Polymerase Chain Reaction
11.
Stem Cells Dev ; 21(18): 3391-402, 2012 Dec 10.
Article in English | MEDLINE | ID: mdl-22827607

ABSTRACT

Stem cell maintenance requires a specific microenvironment. Hematopoietic stem cells (HSCs) are mainly maintained by the endosteal osteoblast (OB) niche, which provides a quiescent HSC microenvironment, and the vascular niche, which regulates the proliferation, differentiation, and mobilization of HSCs. The systemic administration of FGF2 failed to induce normal hematopoiesis in bone marrow (BM) by reducing SDF-1, an important factor for hematopoiesis. Interestingly, SDF-1 levels were decreased in the OBs, but increased in vascular endothelial C166 cells when FGF2 was administered. We hypothesized that FGF2 induces changes in HSC migration from BM; therefore, we investigated FGF2-induced factors of HSC migration by a microarray chip. We searched the genes that were decreased in primary OBs, but increased in C166 cells upon FGF2 treatment. We confirmed selected genes that function in the extracellular region and identified the CXCR2-related chemokine candidate LIX/Cxcl5. A chemotaxis assay showed that CXCL5 induced the migration of HSCs (CD34(-/low)LSK). Our data suggest that the differential regulation of the chemokine CXCL5 between OBs and endothelial cells upon FGF2 treatment is involved in HSC mobilization from the OB niche or BM to peripheral blood.


Subject(s)
Chemokine CXCL5/metabolism , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Hematopoietic Stem Cells/metabolism , Osteoblasts/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cell Line , Cell Movement , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/metabolism , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation , Hematopoiesis/drug effects , Mice , Stem Cell Niche
12.
J Cell Physiol ; 220(1): 245-56, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19301256

ABSTRACT

Ets-related molecule (Erm) is a member of the Ets transcription factor family. Erm is known to be an important factor for the self-renewal of Spermatogonial stem cells (SSCs) and the maintenance of spermatogenesis. We investigated the molecular mechanism of Erm regulation on SDF-1 in TM4 Sertoli cells. Erm and Sdf-1 levels were up-regulated after FGF2 treatment in TM4 cells, whereas these levels were significantly decreased by FGF2 in ST2 bone marrow stromal cells. Knockdown of Erm by siRNA in the presence of FGF2 decreased the Sdf-1 levels in TM4 cells. The expression levels of Erm were similar and Erm overexpression increased the Sdf-1 in both TM4 and ST2 cells. FGFR subtype analysis revealed that FGFR4 was expressed in TM4 cells but not in ST2 cells. A blocking experiment also confirmed that FGFR4 is partly responsible for the up-regulation of Erm and SDF-1 induced by FGF2 stimulation in TM4 cells. FGF2 and ERM increased the activity of Sdf-1 gene promoter region in a dose-dependent manner. EMSA revealed that ERM strongly binds to the -846 to -851 nucleotide region of the potential Ets binding site (EBS) in the Sdf-1 promoter. In addition, CXCR4, the SDF-1 receptor, was expressed in spermatogonia and Sertoli cells in the seminiferous tubules of the mouse testis. Our results indicate that ERM directly regulates Sdf-1 gene expression by interacting with its cis-acting element in response to FGF2 stimulation in TM4 cells.


Subject(s)
Chemokine CXCL12/metabolism , DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 2/metabolism , Sertoli Cells/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Bone Marrow Cells/metabolism , COS Cells , Cell Line , Chemokine CXCL12/genetics , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Humans , Male , Mice , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Receptors, CXCR4/metabolism , Recombinant Proteins/metabolism , Spermatogonia/metabolism , Stem Cells/metabolism , Stromal Cells/metabolism , Time Factors , Transcription Factors/genetics , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...