Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Tissue Eng Regen Med ; 19(6): 1359-1376, 2022 12.
Article in English | MEDLINE | ID: mdl-36207661

ABSTRACT

BACKGROUND: Bone remodeling is tightly regulated through bone resorption and bone formation; imbalances in bone remodeling can cause various pathological conditions such as osteoporosis. Antiresorptive agents commonly used for treating osteoporosis do not substantially reverse osteoporotic bone loss. METHODS: We evaluated the effects of the RVYFFKGKQYWE motif (residues 270-281; VnP-16) of human vitronectin on the osteogenic differentiation of human mesenchymal stem cells (hMSCs) and osteoclastogenesis of bone marrow-derived macrophages. The effects of VnP-16 were also assessed in a mouse model of estrogen deficiency-induced osteoporosis (ovariectomized female C57BL/6 mice). To assay whether VnP-16 can reverse ovariectomy-induced bone loss, synthetic peptides or vehicle were subcutaneously injected into ovariectomized mice once a week for 4 weeks (n = 10/group). To evaluate the bone restorative effects of VnP-16, in-vivo micro-computed tomography analysis and histological staining were performed. RESULTS: VnP-16 induced osteogenic differentiation of hMSCs and inhibited the RANKL-RANK-TRAF6 axis in the osteoclastogenesis signaling pathway. Furthermore, systemic administration of VnP-16 reversed ovariectomy-induced bone loss in the femoral neck, distal femur and lumbar spine by increasing osteoblast differentiation and promoting bone formation, and concomitantly decreasing osteoclastogenesis and inhibiting bone resorption. The bone restorative effect of VnP-16 was observed one week after subcutaneous administration, and although the timing of the effect differed according to bone location, it persisted for at least 3 weeks. CONCLUSION: Our findings suggest that VnP-16 is a potential therapeutic agent for treating osteoporosis that mediates its effects through dual regulation of bone remodeling.


Subject(s)
Bone Resorption , Osteoporosis , Female , Mice , Humans , Animals , Vitronectin/metabolism , Vitronectin/pharmacology , Osteogenesis , Osteoclasts , X-Ray Microtomography , Mice, Inbred C57BL , Ovariectomy/adverse effects , Bone Remodeling , Bone Resorption/drug therapy , Bone Resorption/complications , Bone Resorption/metabolism , Osteoporosis/drug therapy , Peptides/pharmacology , Peptides/metabolism
2.
Exp Cell Res ; 418(1): 113252, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35697077

ABSTRACT

Vitronectin is an abundant multifunctional glycoprotein found in serum, the extracellular matrix, and bone, and is involved in diverse physiological processes. Here, we developed a new bioactive dimeric peptide (VnP-8-DN1 dimer) from a human vitronectin-derived motif (IDAAFTRINCQG; residues 206-217; VnP-8) via removal of an isoleucine residue at the N-terminus of VnP-8 and spontaneous air oxidation. The VnP-8-DN1 dimer potently enhanced cell attachment activity, and this activity was mediated by binding to cellular heparan sulfate proteoglycan receptors. Moreover, the VnP-8-DN1 dimer suppressed osteoclast differentiation by blocking the early stage of osteoclastogenesis induced by macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL). Furthermore, the VnP-8-DN1 dimer decreased the bone-resorbing activity of osteoclasts and increased the survival of osteoclast precursor cells by decreasing the cellular level of c-Fms and reducing RANK expression. Taken together, these results demonstrate that the VnP-8-DN1 dimer inhibits the early stages of M-CSF- and RANK-induced osteoclast differentiation by binding to c-Fms and inhibiting M-CSF signaling.


Subject(s)
Bone Resorption , Macrophage Colony-Stimulating Factor , Bone Resorption/metabolism , Cell Differentiation , Humans , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Membrane Glycoproteins/metabolism , Osteoclasts/metabolism , Osteogenesis , RANK Ligand/metabolism , RANK Ligand/pharmacology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Vitronectin/metabolism , Vitronectin/pharmacology
3.
J Clin Periodontol ; 49(8): 799-813, 2022 08.
Article in English | MEDLINE | ID: mdl-35634689

ABSTRACT

AIM: This study investigated whether a vitronectin-derived peptide (VnP-16) prevents and/or reverses alveolar bone resorption induced by ligature-induced periodontitis in rodents and identified the underlying mechanism. MATERIALS AND METHODS: We evaluated the effects of VnP-16 on osteogenic differentiation in human periodontal ligament cells (hPDLCs), lipopolysaccharide-induced inflammatory responses in gingival fibroblasts, and immune response in T lymphocytes. Ligature-induced periodontitis was induced by ligating the bilateral mandibular first molars for 14 days in rats and for 7 days in mice (n = 10/group). VnP-16 (100 µg/10 µl) was applied topically into the gingival sulcus of rats via intra-sulcular injection, whereas the peptide (50 µg/5 µl) was administered directly into the gingiva of mice via intra-gingival injection. To evaluate the preventive and therapeutic effects of VnP-16, micro-computed tomography analysis and histological staining were then performed. RESULTS: VnP-16 promoted osteogenic differentiation of periodontal ligament cells and inhibited the production of lipopolysaccharide-induced inflammatory mediators in gingival fibroblasts. Concomitantly, VnP-16 modulated the host immune response by reducing the number of receptor activator of NF-κB ligand (RANKL)-expressing lipopolysaccharide-stimulated CD4+ and CD8+ T cells, and by suppressing RANKL and interleukin (IL)-17A production. Furthermore, local administration of VnP-16 in rats and mice significantly prevented and reversed alveolar bone loss induced by ligature-induced periodontitis. VnP-16 enhanced osteoblastogenesis and simultaneously inhibited osteoclastogenesis and suppressed RANKL and IL-17A expression in vivo. CONCLUSIONS: Our findings suggest that VnP-16 acts as a potent therapeutic agent for preventing and treating periodontitis by regulating bone re-modelling and immune and inflammatory responses.


Subject(s)
Alveolar Bone Loss , Periodontitis , Alveolar Bone Loss/drug therapy , Alveolar Bone Loss/prevention & control , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Humans , Interleukin-17/therapeutic use , Ligands , Lipopolysaccharides/pharmacology , Mice , NF-kappa B , Osteogenesis , Periodontitis/drug therapy , Periodontitis/metabolism , Periodontitis/prevention & control , RANK Ligand/metabolism , Rats , Vitronectin/therapeutic use , X-Ray Microtomography
4.
Aging Dis ; 12(6): 1409-1422, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34527418

ABSTRACT

Recent research indicated that extracellular vesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) are a promising alternative to MSCs for immunomodulatory therapy. However, the contents of MSC-EVs would change as their parent MSCs change, hence the therapeutic efficacy of MSC-derived EVs (MSC-EVs) would largely depend on donors, tissue sources and culture conditions of MSCs. To overcome limitations of tissue-derived MSCs, we previously used MSCs derived from human induced pluripotent stem cells (iMSCs) to produce EVs and demonstrated their therapeutic potential in a mouse model of secondary Sjo¨gren's Syndrome. Here, we further found that EVs from early-passage iMSCs had better immunomodulatory potency than EVs from late-passage iMSCs in TLR4-stimulated splenocytes and in a mouse model of primary Sjögren's syndrome. Comparative molecular profiling using proteomics and microRNA sequencing revealed distinctive molecular profiles of iMSC-EVs with or without immunomodulation capacity. Amongst them, manipulation of TGF-ß1, miR-21 and miR-125b levels in iMSC-EVs significantly affected their immunosuppressive effects. These findings would help improve our understanding of the molecular mechanism underlying iMSC-EV-mediated immunomodulation and further provide strategies to improve regulatory function of EVs for the treatment of immune-mediated diseases.

5.
J Tissue Eng Regen Med ; 14(8): 1100-1112, 2020 08.
Article in English | MEDLINE | ID: mdl-32592615

ABSTRACT

We previously reported that the PPFEGCIWN motif (Ln2-LG3-P2-DN3), residues 2678-2686 of the human laminin α2 chain, promotes cell attachment of normal human epidermal keratinocytes (NHEKs) and dermal fibroblasts (NHDFs); however, its in vivo effects on cutaneous wound healing have not yet been examined. In this study, we sought to determine whether Ln2-LG3-P2-DN3 could promote full-thickness cutaneous wound healing by accelerating wound reepithelialization and wound closure in vivo. Ln2-LG3-P2-DN3 had significantly higher cell attachment and spreading activities than vehicle or scrambled peptide control in both NHEKs and NHDFs in vitro. The wound area was significantly smaller in rats treated with Ln2-LG3-P2-DN3 than in those treated with vehicle or scrambled peptide in the early phase of wound healing. Furthermore, Ln2-LG3-P2-DN3 significantly accelerated wound reepithelialization relative to vehicle or scrambled peptide and promoted FAK-Tyr397 phosphorylation and Rac1 activation. Collectively, our findings suggest that the PPFEGCIWN motif has potential as a therapeutic agent for cutaneous regeneration via the acceleration of wound reepithelization and wound closure.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Guanosine Triphosphate/metabolism , Laminin/chemistry , Peptides , Wound Healing/drug effects , Wounds and Injuries , rac1 GTP-Binding Protein/metabolism , Amino Acid Motifs , Animals , Male , Peptides/chemistry , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Wounds and Injuries/metabolism , Wounds and Injuries/therapy
6.
Mol Ther ; 28(7): 1628-1644, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32380062

ABSTRACT

Accumulating evidence indicates that mesenchymal stem/stromal cell-derived extracellular vesicles (MSC-EVs) exhibit immunomodulatory effects by delivering therapeutic RNAs and proteins; however, the molecular mechanism underlying the EV-mediated immunomodulation is not fully understood. In this study, we found that EVs from early-passage MSCs had better immunomodulatory potency than did EVs from late-passage MSCs in T cell receptor (TCR)- or Toll-like receptor 4 (TLR4)-stimulated splenocytes and in mice with ocular Sjögren's syndrome. Moreover, MSC-EVs were more effective when produced from 3D culture of the cells than from the conventional 2D culture. Comparative molecular profiling using proteomics and microRNA sequencing revealed the enriched factors in MSC-EVs that were functionally effective in immunomodulation. Among them, manipulation of transforming growth factor ß1 (TGF-ß1), pentraxin 3 (PTX3), let-7b-5p, or miR-21-5p levels in MSCs significantly affected the immunosuppressive effects of their EVs. Furthermore, there was a strong correlation between the expression levels of TGF-ß1, PTX3, let-7b-5p, or miR-21-5p in MSC-EVs and their suppressive function. Therefore, our comparative strategy identified TGF-ß1, PTX3, let-7b-5p, or miR-21-5p as key molecules mediating the therapeutic effects of MSC-EVs in autoimmune disease. These findings would help understand the molecular mechanism underlying EV-mediated immunomodulation and provide functional biomarkers of EVs for the development of robust EV-based therapies.


Subject(s)
C-Reactive Protein/genetics , Extracellular Vesicles/transplantation , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Serum Amyloid P-Component/genetics , Sjogren-Larsson Syndrome/therapy , Transforming Growth Factor beta1/genetics , Animals , C-Reactive Protein/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Gene Expression Profiling , Humans , Mesenchymal Stem Cells/metabolism , Mice , Proteomics , Serial Passage , Serum Amyloid P-Component/metabolism , Sjogren-Larsson Syndrome/genetics , Sjogren-Larsson Syndrome/metabolism , Transforming Growth Factor beta1/metabolism
7.
J Biomed Mater Res A ; 108(5): 1214-1222, 2020 05.
Article in English | MEDLINE | ID: mdl-32034938

ABSTRACT

Early implant loading is very important for reducing the duration of missing teeth in human patients. The laminin-derived peptide, DLTIDDSYWYRI motif (Ln2-P3), accelerates bone healing. Therefore, to investigate the hypothesis that Ln2-P3 increases the bone response to sandblasted, large-grit, acid-etched (SLA) titanium implants, the effect of the Ln2-P3 peptide on the osseointegration of SLA titanium implants was evaluated in vitro and in vivo. Human osteoblast-like cells were cultured on untreated, scrambled peptide (SP)-treated, and Ln2-P3-treated SLA titanium discs, and the cellular responses of these cells were evaluated. The Ln2-P3 treatment augmented osteoblast attachment and spreading, alkaline phosphatase activity, and the expression of osteogenic marker genes. Furthermore, the untreated and Ln2-P3-treated SLA titanium implants were inserted into the tibiae of rabbits for 9 and 11 days. Compared with the untreated implants, the Ln2-P3-treated implants showed a significantly higher bone-to-implant contact ratio at Day 9 after implantation and an increased bone area. The Ln2-P3 treatment of the SLA titanium implant surface augmented osteoblastic activity and accelerated peri-implant bone formation at the bone-implant interface. Overall, these results indicated that compared with the SLA titanium surface alone, the Ln2-P3 peptide-treated SLA titanium surface enhances initial osseointegration, thereby facilitating earlier implant loading.


Subject(s)
Bone Substitutes/pharmacology , Laminin/pharmacology , Osseointegration/drug effects , Peptides/pharmacology , Titanium/pharmacology , Animals , Bone-Anchored Prosthesis , Cell Line , Female , Humans , Osteoblasts/cytology , Osteogenesis/drug effects , Rabbits , Tibia/drug effects , Tibia/injuries , Tibia/physiology , Tibia/surgery
8.
Materials (Basel) ; 12(20)2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31627447

ABSTRACT

In this study, we evaluated early bone responses to a vitronectin-derived, minimal core bioactive peptide, RVYFFKGKQYWE motif (VnP-16), both in vitro and in vivo, when the peptide was treated on sandblasted, large-grit, acid-etched (SLA) titanium surfaces. Four surface types of titanium discs and of titanium screw-shaped implants were prepared: control, SLA, scrambled peptide-treated, and VnP-16-treated surfaces. Cellular responses, such as attachment, spreading, migration, and viability of human osteoblast-like HOS and MG63 cells were evaluated in vitro on the titanium discs. Using the rabbit tibia model with the split plot design, the implants were inserted into the tibiae of four New Zealand white rabbits. After two weeks of implant insertion, the rabbits were sacrificed, the undecalcified specimens were prepared for light microscopy, and the histomorphometric data were measured. Analysis of variance tests were used for the quantitative evaluations in this study. VnP-16 was non-cytotoxic and promoted attachment and spreading of the human osteoblast-like cells. The VnP-16-treated SLA implants showed no antigenic activities at the interfaces between the bones and the implants and indicated excellent bone-to-implant contact ratios, the means of which were significantly higher than those in the SP-treated implants. VnP-16 reinforces the osteogenic potential of the SLA titanium dental implant.

9.
Int J Oral Maxillofac Implants ; 34(4): 836­844, 2019.
Article in English | MEDLINE | ID: mdl-30521654

ABSTRACT

PURPOSE: This study aimed to investigate the in vitro and in vivo bone-forming potential of a sandblasted, large-grit, acid-etched (SLA) titanium (Ti) surface treated with a laminin-derived functional peptide, PPFEGCIWN (DN3). MATERIALS AND METHODS: Human osteoblast-like MG63 cells were cultured with SLA Ti discs untreated or treated with DN3 or a control scrambled peptide (SP). Cell adhesion, spreading, and viability on the discs were tested. Alkaline phosphatase gene expression and enzyme activity were also evaluated. Four DN3-coated SLA Ti implants and four untreated implants were placed into the tibiae of two rabbits (two implants/tibia). Ten days later, the bone-implant interfaces were subjected to histomorphometry to measure the bone response. The surface properties of the discs and implants were determined using scanning electron, widefield confocal, and confocal laser microscopy and x-ray photoelectron spectroscopy. RESULTS: The peptide-treated and untreated discs and implants were similar in terms of physical surface properties, but the peptide-treated surfaces had significantly higher nitrogen levels (P < .05). The DN3 peptide promoted cell adhesion, spreading, and alkaline phosphatase expression and enzyme activity (P < .05). Histomorphometry of the harvested implants showed rapid bone formation and affinity of the motif. CONCLUSION: This study suggests that treatment with the cell adhesion peptide DN3 promotes bone healing at the SLA Ti surface.


Subject(s)
Dental Implants , Titanium , Animals , Cell Line , Humans , Laminin , Microscopy, Electron, Scanning , Osteoblasts , Osteogenesis , Peptides , Rabbits , Surface Properties
10.
Cell Death Differ ; 25(2): 268-281, 2018 02.
Article in English | MEDLINE | ID: mdl-28937683

ABSTRACT

Osteoporosis affects millions of people worldwide by promoting bone resorption and impairing bone formation. Bisphosphonates, commonly used agents to treat osteoporosis, cannot reverse the substantial bone loss that has already occurred by the time of diagnosis. Moreover, their undesirable side-effects, including osteonecrosis of the jaw, have been reported. Here, we demonstrated that a new bioactive core vitronectin-derived peptide (VnP-16) promoted bone formation by accelerating osteoblast differentiation and activity through direct interaction with ß1 integrin followed by FAK activation. Concomitantly, VnP-16 inhibited bone resorption by restraining JNK-c-Fos-NFATc1-induced osteoclast differentiation and αvß3 integrin-c-Src-PYK2-mediated resorptive function. Moreover, VnP-16 decreased the bone resorbing activity of pre-existing mature osteoclasts without changing their survival rate. Furthermore, VnP-16 had a strong anabolic effect on bone regeneration by stimulating osteoblast differentiation and increasing osteoblast number, and significantly alleviated proinflammatory cytokine-induced bone resorption by restraining osteoclast differentiation and function in murine models. Moreover, VnP-16 could reverse ovariectomy-induced bone loss by both inhibiting bone resorption and promoting bone formation. Given its dual role in promoting bone formation and inhibiting bone resorption, our results suggest that VnP-16 could be an attractive therapeutic agent for treating osteoporosis.


Subject(s)
Bone Resorption/drug therapy , Cell Differentiation/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Ovariectomy , Peptides/pharmacology , Vitronectin/chemistry , Animals , Bone Regeneration/drug effects , Bone Resorption/metabolism , Cell Line , Humans , Male , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Peptides/chemistry
11.
Int J Oncol ; 51(5): 1541-1552, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29048658

ABSTRACT

Numerous studies implicate miR-146a as pleiotropic regulator of carcinogenesis; however, its roles in carcinogenesis are not fully understood. A clue from expression analyses of miR-146a-5p in all 13 oral squamous cell carcinoma (OSCC) cell lines examined and in OSCC tissues, whole blood and whole saliva of OSCC patients in vivo revealed that miR­146a-5p expression was highly upregulated. Particularly, we widened the view of its upregulation in saliva, implicating that high miR-146a-5p expression is not only correlated closely to the development of human oral cancer, but also to a possible candidate as a diagnostic marker of OSCC. Indeed, further examination showed that exogenous miR-146a-5p expression showed pleiotropic effects on cell proliferation and apoptosis which were partially based on the contextual responses of activation of JNK, downstream of TRAF6 that was targeted by miR-146a-5p in normal human keratinocytes and OSCC cell lines. TRAF6 suppression by a TRAF6-specific siRNA resulted in contradictory consequences on cellular processes in normal and OSCC cells. Notably, TRAF6 downregulation by both miR-146a-5p and TRAF6-specific siRNA deactivated JNK in SCC-9, but not in normal human keratinocytes. In support of the proliferation-promoting effect of miR-146a-5p, silencing of endogenous miR-146a-5p significantly reduced proliferation of SCC-9. Together, these results suggest that miR-146a-5p affects proliferation and apoptosis in a cellular context-dependent manner and selectively disarms the TRAF6-mediated branch of the TGF-ß signaling in OSCC cell lines by sparing Smad4 involvement.


Subject(s)
Carcinoma, Squamous Cell/genetics , MicroRNAs/genetics , Mouth Neoplasms/genetics , TNF Receptor-Associated Factor 6/genetics , Apoptosis/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Genetic Variation , Humans , Intracellular Signaling Peptides and Proteins , Mouth Neoplasms/pathology , RNA, Small Interfering , Smad4 Protein/genetics , Transforming Growth Factor beta/genetics
13.
Biomaterials ; 73: 96-109, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26406450

ABSTRACT

Finding bioactive short peptides derived from proteins is a critical step to the advancement of tissue engineering and regenerative medicine, because the former maintains the functions of the latter without immunogenicity in biological systems. Here, we discovered a bioactive core nonapeptide sequence, PPFEGCIWN (residues 2678-2686; Ln2-LG3-P2-DN3), from the human laminin α2 chain, and investigated the role of this peptide in binding to transmembrane proteins to promote intracellular events leading to cell functions. This minimum bioactive sequence had neither secondary nor tertiary structures in a computational structure prediction. Nonetheless, Ln2-LG3-P2-DN3 bound to various cell types as actively as laminin in cell adhesion assays. The in vivo healing tests using rats revealed that Ln2-LG3-P2-DN3 promoted bone formation without any recognizable antigenic activity. Ln2-LG3-P2-DN3-treated titanium (Ti) discs and Ti implant surfaces caused the enhancement of bone cell functions in vitro and induced faster osseointegration in vivo, respectively. These findings established a minimum bioactive sequence within human laminin, and its potential application value for regenerative medicine, especially for bone tissue engineering.


Subject(s)
Laminin/chemistry , Tissue Engineering/methods , Alkaline Phosphatase/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Bone and Bones/pathology , Cell Adhesion , Cell Differentiation , Cell Movement , Cell Survival , Cells, Cultured , Humans , Male , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Molecular Sequence Data , Nanoparticles/chemistry , Osseointegration , Osteoblasts/metabolism , Osteogenesis , PC12 Cells , Peptides/chemistry , Rabbits , Rats , Rats, Sprague-Dawley , Regeneration , Titanium/chemistry , X-Ray Microtomography
14.
Biomed Res Int ; 2013: 638348, 2013.
Article in English | MEDLINE | ID: mdl-23586052

ABSTRACT

Laminin-derived peptide coatings can enhance epithelial cell adhesion to implants, and the positive effect of these peptides on bone cell adhesion has been anticipated. The purpose of this study was to evaluate the improvement in bone cell attachment to and activity on titanium (Ti) scaffolds coated with a laminin-derived functional peptide, Ln2-P3 (the DLTIDDSYWYRI motif). Four Ti disc surfaces were prepared, and a human osteosarcoma (HOS) cell attachment test was performed to select two candidate surfaces for peptide coating. These two candidates were then coated with Ln2-P3 peptide, a scrambled peptide, or left uncoated to measure cell attachment to each surface, following which one surface was chosen to assess alkaline phosphatase (ALP) activity and osteogenic marker gene expression with quantitative real-time PCR. On the commercially pure Ti surface, the Ln2-P3 coating significantly increased cellular ALP activity and the expression levels of ALP and bone sialoprotein mRNA as compared with the scrambled peptide-coated and uncoated surfaces. In conclusion, although further in vivo studies are needed, the findings of this in vitro study indicate that the Ln2-P3-coated implant surface promotes bone cell adhesion, which has clinical implications for reducing the overall treatment time of dental implant therapy.


Subject(s)
Cell Adhesion/drug effects , Dental Implants , Osteogenesis , Cell Differentiation , Cell Line, Tumor , Coated Materials, Biocompatible/administration & dosage , Coated Materials, Biocompatible/chemistry , Humans , Laminin/chemistry , Osteosarcoma/metabolism , Peptides/administration & dosage , Peptides/chemistry , Surface Properties , Titanium/administration & dosage , Titanium/chemistry
15.
Biomaterials ; 34(16): 4027-4037, 2013 May.
Article in English | MEDLINE | ID: mdl-23465831

ABSTRACT

Considerable effort has been directed towards replacing lost teeth using tissue-engineering methods such as titanium implants. A number of studies have tried to modify bioinert titanium surfaces by coating them with functionally bioactive molecules for faster and stronger osseointegration than pure titanium surfaces. Recently, peptides have been recognized as valuable scientific tools in the field of tissue-engineering. The DLTIDDSYWYRI motif of the human laminin-2 α2 chain has been previously reported to promote the attachment of various cell types; however, the in vivo effects of the DLTIDDSYWYRI motif on new bone formation have not yet been studied. To examine whether a laminin-2-derived peptide can promote osseointegration by accelerating new bone formation in vivo, we applied titanium implants coated with the DLTIDDSYWYRI motif in a rabbit tibia model. The application of the DLTIDDSYWYRI motif-treated implant to tibia wounds enhanced collagen deposition and alkaline phosphatase expression. It significantly promoted implant osseointegration compared with treatment with scrambled peptide-treated implants by increasing the bone-to-implant contact ratio and bone area. These findings support the hypothesis that the DLTIDDSYWYRI motif acts as an effective osseointegration accelerator by enhancing new bone formation.


Subject(s)
Implants, Experimental , Laminin/chemistry , Osseointegration/drug effects , Peptides/pharmacology , Alkaline Phosphatase/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Collagen/metabolism , Gene Expression Regulation/drug effects , Humans , Male , Molecular Sequence Data , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoblasts/ultrastructure , Osteogenesis/drug effects , Osteogenesis/genetics , PC12 Cells , Peptides/chemistry , Rabbits , Rats , Surface Properties/drug effects , Titanium/pharmacology
16.
Exp Cell Res ; 319(3): 153-60, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23063429

ABSTRACT

Gö6976 is a nonglycosidic indolocarbazole compound widely used as a specific inhibitor of PKCα/ß. In experiments probing for a role of PKCα in human laminin-2-integrin-mediated cell adhesion and spreading of PC12 cells, we observed unexpected enhancements of adhesion, spreading and stress fiber formation to 1 µM Gö6976 with concomitant increase in membrane translocation of PKCδ and autophosphorylation of focal adhesion kinase (FAK). Importantly, enhanced cellular behavior and membrane translocation of PKCδ induced by Gö6976 was retained in siRNA-transfected PC12 cells to knockdown PKCα expression. Gö6976 also induced laminin-dependent cell adhesion in NIH/3T3 and CV-1 fibroblasts, suggesting of a mechanism that may be common to multiple cell-types. A specific inhibitor of PKCδ, rottlerin, completely abrogated Gö6976-dependent increase in PC12 cell adhesion to laminin as well as the activation of small GTPases, Rac1 and Cdc42, that are downstream of PKCδ in adhesion receptor signaling. siRNA knockdown of Rac1 and Cdc42 expression inhibited cell spreading and lamellipodia formation in PC12 cells. Overall, these results suggest that Gö6976 may stimulate membrane recruitment of PKCδ through a mechanism that is independent of PKCα/ß signaling. In addition, the activation of Rac1 and Cdc42 by human laminin-2-integrin-dependent activation of PKCδ/FAK signaling mediates cell spreading and lamellipodia formation in PC12 cells.


Subject(s)
Carbazoles/pharmacology , Cell Membrane/drug effects , Cell Proliferation/drug effects , Protein Kinase C-delta/physiology , Animals , Cell Adhesion/drug effects , Cell Membrane/metabolism , Cell Membrane/physiology , Cells, Cultured , Chlorocebus aethiops , Enzyme Activation/drug effects , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , PC12 Cells , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-delta/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Up-Regulation/drug effects
17.
Biomaterials ; 33(15): 3967-79, 2012 May.
Article in English | MEDLINE | ID: mdl-22364697

ABSTRACT

Laminin α2 chain plays an important role in basement membrane assembly and peripheral myelinogenesis; however, the integrin binding motif within human laminin α2 chain and the signaling pathways downstream of this ligand-receptor interaction are poorly understood. We identified a motif, RNIPPFEGCIWN (Ln2-LG3-P2), within LG3 domain of human laminin α2 chain as a major site for both α3ß1 integrin and cellular activities such as cell adhesion, spreading, and migration. Binding of α3ß1 integrin with Ln2-LG3-P2 induced the membrane recruitment of protein kinase Cδ (PKCδ) and stimulated its tyrosine phosphorylation. The cellular activities induced by Ln2-LG3-P2 and the phosphorylation of focal adhesion kinase (FAK) were inhibited by rottlerin, a PKCδ inhibitor, but not by Gö6976, a PKCα/ß inhibitor. These results indicate that RNIPPFEGCIWN motif within human laminin α2 chain is a major ligand for α3ß1 integrin, and that binding of α3ß1 integrin mediates cellular activities through membrane recruitment and tyrosine phosphorylation of PKCδ and FAK phosphorylation.


Subject(s)
Biomimetic Materials/pharmacology , Cell Membrane/enzymology , Cell Movement/drug effects , Laminin/pharmacology , Peptides/pharmacology , Protein Kinase C-delta/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Arginine/metabolism , Biomimetic Materials/chemistry , Cell Adhesion/drug effects , Cell Membrane/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Humans , Integrin alpha3beta1/metabolism , Laminin/chemistry , Laminin/isolation & purification , Molecular Sequence Data , PC12 Cells , Peptides/chemistry , Peptides/isolation & purification , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Rats , Stress Fibers/drug effects , Stress Fibers/metabolism
18.
Int J Mol Med ; 28(6): 1001-11, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21879252

ABSTRACT

Although previous studies indicate that skin-derived precursors (SKPs) are multipotent dermal precursors that share similarities with neural crest stem cells (NCSCs), a shared ability for multilineage differentiation toward neural crest lineages between SKPs and NCSCs has not been fully demonstrated. Here, we report the derivation of SKPs from adult mouse skin and their directed multilineage differentiation toward neural crest lineages. Under controlled in vitro conditions, mouse SKPs were propagated and directed toward peripheral nervous system lineages such as peripheral neurons and Schwann cells, and mesenchymal lineages, such as osteogenic, chondrogenic, adipogenic, and smooth muscle cells. To ask if SKPs could generate these same lineages in vivo, a mixture of SKP-derived mesenchymal stem cells and hydroxyapatite/tricalcium phosphate was transplanted into the rat calvarial defects. Over the ensuing 4 weeks, we observed formation of osteogenic structure in the calvarial defect without any evidence of teratomas. These findings demonstrate the multipotency of adult mouse SKPs to differentiate into neural crest lineages. In addition, SKP-derived mesenchymal stem cells represent an accessible, potentially autologous source of precursor cells for tissue-engineered bone repair.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Skin/cytology , Skull/cytology , Stem Cell Transplantation/methods , Tissue Engineering/methods , Adipocytes/cytology , Adipocytes/physiology , Animals , Cell Lineage , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/physiology , Female , Male , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Neural Crest/cytology , Neurons/cytology , Neurons/physiology , Osteoblasts/cytology , Osteoblasts/physiology , Rats , Rats, Sprague-Dawley , Schwann Cells/cytology , Schwann Cells/physiology , Skull/injuries
19.
Colloids Surf B Biointerfaces ; 78(2): 334-42, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20399628

ABSTRACT

A heat-treated PVA nanofibrous matrix containing silver (Ag) was prepared by electrospinning an aqueous 10 wt% PVA solution and followed by heat treatment at 150 degrees C for 10 min. The average diameter of the as-spun and heat-treated PVA nanofibers was 330 nm. The heat-treated PVA nanofibrous matrix containing Ag was irradiated with UV light to transform the Ag ions in the nanofibrous matrix into Ag nanoparticles. The in vitro cytotoxicity of the Ag ions and/or nanoparticles on normal human epidermal keratinocytes (NHEK) and fibroblasts (NHEF) cultures was examined. The PVA nanofibrous matrix containing Ag showed slightly higher level of attachment and spreading in the early stage culture (1 h) than the PVA nanofibers without Ag (control). However, compared with the PVA nanofibers without Ag, the heat-treated and UV-irradiated PVA nanofibers, containing mainly Ag ions and nanoparticles, respectively, showed reduced cell attachment and spreading. This shows that both Ag ions and Ag nanoparticles are cytotoxic to NHEK and NHEF. There was no significant difference in cytotoxicity to NHEK and NHEF between Ag ions and Ag nanoparticles. NHEF appeared to be more sensitive to Ag ions or particles than NHEK. In addition, the residual nitrate ions (NO3(-)) in the PVA nanofibers had an adverse effect on the culture of both cells.


Subject(s)
Fibroblasts/cytology , Keratinocytes/cytology , Metal Nanoparticles/chemistry , Silver/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Child, Preschool , Epidermal Cells , Fibroblasts/ultrastructure , Hot Temperature , Humans , Infant , Keratinocytes/ultrastructure , Male , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nanofibers/chemistry , Nanofibers/radiation effects , Polyvinyl Alcohol/chemistry , Ultraviolet Rays
20.
Int J Oncol ; 36(6): 1491-501, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428774

ABSTRACT

Although >60% of oral cancer cases are not related to human papillomavirus (HPV) infection, most studies of oral carcinogenesis in human cells in vitro are carried out with human oral keratinocytes immortalized by HPV DNA. To explore whether human oral keratinocytes can spontaneously transform without HPV infection, we attempted to establish spontaneously immortalized and tumorigenic-transformed human oral keratinocytes by serial subculture to the post-mitotic stage. Here we report two spontaneously transformed human oral keratinocyte lines from adult human gingival samples. These lines were obviously immortal (>140 passages) and transformed phenotypes in vitro. One of the lines, Spi-HOK1, remained non-tumorigenic in nude mice, whereas the other line, Spt-HOK80, showed tumorigenicity. These lines showed epithelial origi-nality, but did not contain high-risk types of HPV DNAs. On karyotyping, Spi-HOK1 was aneuploid with a unique stable marker chromosome. Both cell lines revealed a mutation in the p53 gene, loss of p21WAF1/Cip1 and overexpression of p-Rb-Ser807/811. These cell lines were resistant to cisplatin-induced apoptosis by suppressing induction of apoptotic proteins. These results clearly demonstrate that spontaneous immortalization and spontaneous tumorigenic transformation of primary human oral keratinocytes can occur in vitro without HPV infection and are associated with chromosomal alterations, p53 mutation and impaired apoptosis. To our knowledge, this is the first report demonstrating that the Spi-HOK1 and Spt-HOK80 lines are novel cell lines that are spontaneously transformed from primary human oral keratinocytes.


Subject(s)
Apoptosis/genetics , Cell Transformation, Neoplastic/genetics , Gingiva/pathology , Keratinocytes/pathology , Mouth Neoplasms/genetics , Adult , Animals , Blotting, Western , Cell Line , Cell Separation , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Nude , Mouth Mucosa/pathology , Mouth Neoplasms/pathology , Papillomaviridae , Retinoblastoma Protein/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...