Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
J Immunol ; 206(10): 2386-2392, 2021 05 15.
Article En | MEDLINE | ID: mdl-33952619

Periodontal disease (PD) is a chronic destructive inflammatory disease of the tooth-supporting structures that leads to tooth loss at its advanced stages. Although the disease is initiated by a complex organization of oral microorganisms in the form of a plaque biofilm, it is the uncontrolled immune response to periodontal pathogens that fuels periodontal tissue destruction. IL-17A has been identified as a key cytokine in the pathogenesis of PD. Despite its well documented role in host defense against invading pathogens at oral barrier sites, IL-17A-mediated signaling can also lead to a detrimental inflammatory response, causing periodontal bone destruction. In this study, we developed a local sustained delivery system that restrains IL-17A hyperactivity in periodontal tissues by incorporating neutralizing anti-IL-17A Abs in poly(lactic-coglycolic) acid microparticles (MP). This formulation allowed for controlled release of anti-IL-17A in the periodontium of mice with ligature-induced PD. Local delivery of anti-IL-17A MP after murine PD induction inhibited alveolar bone loss and osteoclastic activity. The anti-IL-17A MP formulation also decreased expression of IL-6, an IL-17A target gene known to induce bone resorption in periodontal tissues. This study demonstrates proof of concept that local and sustained release of IL-17A Abs constitutes a promising therapeutic strategy for PD and may be applicable to other osteolytic bone diseases mediated by IL-17A-driven inflammation.


Alveolar Bone Loss/drug therapy , Alveolar Bone Loss/immunology , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Drug Delivery Systems/methods , Interleukin-17/immunology , Periodontitis/drug therapy , Periodontitis/immunology , Animals , Capsules , Disease Models, Animal , Drug Compounding/methods , Drug Liberation , Male , Mice , Mice, Inbred BALB C , Osteolysis/drug therapy , Osteolysis/immunology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Treatment Outcome
2.
Acta Biomater ; 73: 559-566, 2018 06.
Article En | MEDLINE | ID: mdl-29684620

Magnesium (Mg) medical devices are currently being marketed for orthopedic applications and have a complex degradation process which includes the evolution of hydrogen gas (H2). The effect of H2 exposure on relevant cell types has not been studied; and the concentration surrounding degrading Mg devices has not been quantified to enable such mechanistic studies. A simple and effective method to measure the concentration of H2 in varying microenvironments surrounding Mg implants is the first step to understanding the biological impact of H2 on these cells. Here, the in vivo measurement of H2 surrounding fracture fixation devices implanted in vivo is demonstrated. An electrochemical H2 microsensor detected increased levels of H2 at three anatomical sites with a response time of about 30 s. The sensor showed the H2 concentration in the bone marrow at 1 week post-implantation (1460 ±â€¯320 µM) to be much higher than measured in the subcutaneous tissue (550 ±â€¯210 µM) and at the skin surface (120 ±â€¯50 µM). Additionally, the H2 concentrations measured in the bone marrow exceeded the concentration in a H2 saturated water solution (∼800 µM). These results suggest that H2 emanating from Mg implants in bone during degradation pass through the bone marrow and become at least partially trapped because of slow permeation through the bone. This study is the first to identify H2 concentrations in the bone marrow environment and will enable in vitro experiments to be executed at clinically relevant H2 concentrations to explore possible biological effects of H2 exposure. STATEMENT OF SIGNIFICANCE: An electrochemical H2 sensor was used to monitor the degradation of a Mg fracture fixation system in a lapine ulna fracture model. Interestingly, the H2 concentration in the bone marrow is 82% higher than H2 saturated water solution. This suggests H2 generated in situ is trapped in the bone marrow and bone is less permeable than the surrounding tissues. The detectable H2 at the rabbit skin also demonstrates a H2 sensor's ability to monitor the degradation process under thin layers of tissue. H2 sensing shows promise as a tool for monitoring the degradation of Mg alloy in vivo and creating in vitro test beds to more mechanistically evaluate the effects of varying H2 concentrations on cell types relevant to osteogenesis.


Bone Marrow/metabolism , Bone Plates , Electrochemistry/methods , Fracture Fixation/instrumentation , Hydrogen/chemistry , Magnesium/chemistry , Ulna Fractures/surgery , Absorbable Implants , Alloys , Animals , Bone Screws , Fracture Fixation/methods , Fracture Healing/drug effects , Gases , Osteogenesis/drug effects , Prostheses and Implants , Rabbits , X-Ray Microtomography
3.
Acta Biomater ; 28: 234-239, 2015 Dec.
Article En | MEDLINE | ID: mdl-26318803

Magnesium (Mg) alloys have many unique qualities which make them ideal candidates for bone fixation devices, including biocompatibility and degradation in vivo. Despite a rise in Mg alloy production and research, there remains no standardized system to assess their degradation or biological effect on human stem cells in vivo. In this study, we developed a novel in vivo model to assess Mg alloys for craniofacial and orthopedic applications. Our model consists of a collagen sponge seeded with human bone marrow stromal cells (hBMSCs) around a central Mg alloy rod. These scaffolds were implanted subcutaneously in mice and analyzed after eight weeks. Alloy degradation and biological effect were determined by microcomputed tomography (microCT), histological staining, and immunohistochemistry (IHC). MicroCT showed greater volume loss for pure Mg compared to AZ31 after eight weeks in vivo. Histological analysis showed that hBMSCs were retained around the Mg implants after 8 weeks. Furthermore, immunohistochemistry showed the expression of dentin matrix protein 1 and osteopontin around both pure Mg and AZ31 with implanted hBMSCs. In addition, histological sections showed a thin mineral layer around all degrading alloys at the alloy-tissue interface. In conclusion, our data show that degrading pure Mg and AZ31 implants are cytocompatible and do not inhibit the osteogenic property of hBMSCs in vivo. These results demonstrate that this model can be used to efficiently assess the biological effect of corroding Mg alloys in vivo. Importantly, this model may be modified to accommodate additional cell types and clinical applications. STATEMENT OF SIGNIFICANCE: Magnesium (Mg) alloys have been investigated as ideal candidates for bone fixation devices due to high biocompatibility and degradation in vivo, and there is a growing need of establishing an efficient in vivo material screening system. In this study, we assessed degradation rate and biological effect of Mg alloys by transplanting Mg alloy rod with human bone marrow stromal cells seeded on collagen sponge subcutaneously in mice. After 8 weeks, samples were analyzed by microcomputed tomography and histological staining. Our data show that degrading Mg alloys are cytocompatible and do not inhibit the osteogenic property of hBMSCs in vivo. These results demonstrate that this model can be used to efficiently assess the biological effect of corroding Mg alloys in vivo.


Alloys , Magnesium/pharmacology , Mesenchymal Stem Cells/drug effects , Models, Biological , Cells, Cultured , Humans
4.
Stem Cell Res ; 14(3): 297-306, 2015 May.
Article En | MEDLINE | ID: mdl-25814434

Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) are fibroblastic reticular cells, a subset of which is composed of multipotent skeletal stem cells (SSCs). SSCs/BMSCs are able to recreate a bone/marrow organ in vivo. To determine differences between clonogenic multipotent SSCs and similarly clonogenic but non-multipotent BMSCs, we established single colony-derived strains (SCDSs, initiated by individual Colony Forming Unit-Fibroblasts) and determined their differentiation capacity by vivo transplantation. In this series of human SCDSs (N=24), 20.8% formed fibrous tissue (F), 66.7% formed bone (B), and 12.5% formed a bone/marrow organ, and thus were multipotent (M). RNA isolated from 12 SCDSs just prior to transplantation was analyzed by microarray. Although highly similar, there was variability from one SCDS to another, and SCDSs did not strictly segregate into the three functional groups (F, B or M) by unsupervised hierarchical clustering. We then compared 3 F-SCDSs to 3 M-SCDSs that did segregate. Genes associated with skeletogenesis, osteoblastogeneis, hematopoiesis, and extracellular matrix were over-represented in M-SCDSs compared with F-SCDSs. These results highlight the heterogeneity of SSCs/BMSCs, even between functionally similar SCDSs, but also indicate that differences can be detected that may shed light on the character of the SSC.


Mesenchymal Stem Cells/metabolism , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Clone Cells/cytology , Clone Cells/metabolism , Gene Expression Profiling , Humans , Mesenchymal Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Principal Component Analysis
5.
Acta Biomater ; 18: 262-9, 2015 May.
Article En | MEDLINE | ID: mdl-25712384

Each year, millions of Americans suffer bone fractures, often requiring internal fixation. Current devices, like plates and screws, are made with permanent metals or resorbable polymers. Permanent metals provide strength and biocompatibility, but cause long-term complications and may require removal. Resorbable polymers reduce long-term complications, but are unsuitable for many load-bearing applications. To mitigate complications, degradable magnesium (Mg) alloys are being developed for craniofacial and orthopedic applications. Their combination of strength and degradation make them ideal for bone fixation. Previously, we conducted a pilot study comparing Mg and titanium devices with a rabbit ulna fracture model. We observed Mg device degradation, with uninhibited healing. Interestingly, we observed bone formation around degrading Mg, but not titanium, devices. These results highlighted the potential for these fixation devices. To better assess their efficacy, we conducted a more thorough study assessing 99.9% Mg devices in a similar rabbit ulna fracture model. Device degradation, fracture healing, and bone formation were evaluated using microcomputed tomography, histology and biomechanical tests. We observed device degradation throughout, and calculated a corrosion rate of 0.40±0.04mm/year after 8 weeks. In addition, we observed fracture healing by 8 weeks, and maturation after 16 weeks. In accordance with our pilot study, we observed bone formation surrounding Mg devices, with complete overgrowth by 16 weeks. Bend tests revealed no difference in flexural load of healed ulnae with Mg devices compared to intact ulnae. These data suggest that Mg devices provide stabilization to facilitate healing, while degrading and stimulating new bone formation.


Bone Plates , Bone Screws , Fracture Healing/drug effects , Magnesium/pharmacology , Ulna Fractures/pathology , Animals , Bone Development/drug effects , Materials Testing , Rabbits , Ulna/diagnostic imaging , Ulna/drug effects , Ulna/pathology , Ulna Fractures/diagnostic imaging , X-Ray Microtomography
6.
J Oral Maxillofac Surg ; 73(2): 295-305, 2015 Feb.
Article En | MEDLINE | ID: mdl-25579013

PURPOSE: Internal bone fixation devices made with permanent metals are associated with numerous long-term complications and may require removal. We hypothesized that fixation devices made with degradable magnesium alloys could provide an ideal combination of strength and degradation, facilitating fracture fixation and healing while eliminating the need for implant removal surgery. MATERIALS AND METHODS: Fixation plates and screws were machined from 99.9% pure magnesium and compared with titanium devices in a rabbit ulnar fracture model. Magnesium device degradation and the effect on fracture healing and bone formation were assessed after 4 weeks. Fracture healing with magnesium device fixation was compared with that of titanium devices using qualitative histologic analysis and quantitative histomorphometry. RESULTS: Micro-computed tomography showed device degradation after 4 weeks in vivo. In addition, 2-dimensional micro-computed tomography slices and histologic staining showed that magnesium degradation did not inhibit fracture healing or bone formation. Histomorphology showed no difference in bone-bridging fractures fixed with magnesium and titanium devices. Interestingly, abundant new bone was formed around magnesium devices, suggesting a connection between magnesium degradation and bone formation. CONCLUSION: Our results show potential for magnesium fixation devices in a loaded fracture environment. Furthermore, these results suggest that magnesium fixation devices may enhance fracture healing by encouraging localized new bone formation.


Bone Plates , Bone Screws , Fracture Healing , Internal Fixators , Animals , Osteogenesis , Rabbits , X-Ray Microtomography
7.
J Bone Miner Res ; 30(3): 412-22, 2015 Mar.
Article En | MEDLINE | ID: mdl-25264308

Inflammatory bone resorption is a hallmark of periodontitis, and Tregs and Th2 cells are independently associated with disease progression attenuation. In this study, we employed an infection-triggered inflammatory osteolysis model to investigate the mechanisms underlying Treg and Th2 cell migration and the impact on disease outcome. Aggregatibacter actinomycetemcomitans-infected C57Bl/6 (wild-type [WT]) mice develop an intense inflammatory reaction and alveolar bone resorption, and Treg and Th2 cell migration is temporally associated with disease progression attenuation. Tregs extracted from the lesions preferentially express CCR4 and CCR8, whereas Th2 cells express CCR3, CCR4, and CCR8. The absence of CCR5 and CCR8 did not significantly impact the migration of Tregs and Th2 cells or affect the disease outcome. CCR4KO mice presented a minor reduction in Th2 cells in parallel with major impairment of Treg migration, which was associated with increased inflammatory bone loss and higher proinflammatory and osteoclastogenic cytokine levels. The blockade of the CCR4 ligand CCL22 in WT mice resulted in an increased inflammatory bone loss phenotype similar to that in the CCR4KO strain. Adoptive transfer of CCR4(+) Tregs to the CCR4KO strain revert the increased disease phenotype to WT mice-like levels; also, the in situ production of CCL22 in the lesions is mandatory for Tregs migration and the consequent bone loss arrest. The local release of exogenous CCL22 provided by poly(lactic-co-glycolic acid) (PLGA) microparticles promotes migration of Tregs and disease arrest in the absence of endogenous CCL22 in the IL-4KO strain, characterized by the lack of endogenous CCL22 production, defective migration of Tregs, and exacerbated bone loss. In summary, our results show that the IL-4/CCL22/CCR4 axis is involved in the migration of Tregs to osteolytic lesion sites, and attenuates development of lesions by inhibiting inflammatory migration and the production of proinflammatory and osteoclastogenic mediators.


Chemokine CCL22/metabolism , Interleukin-4/metabolism , Osteitis/pathology , Osteoporosis/pathology , Periodontitis/pathology , T-Lymphocytes, Regulatory/pathology , Animals , Male , Mice , Mice, Inbred C57BL , Osteitis/metabolism , Periodontitis/metabolism
8.
Blood ; 125(5): 793-802, 2015 Jan 29.
Article En | MEDLINE | ID: mdl-25499762

Dyskeratosis congenita (DC) is an inherited multisystem disorder, characterized by oral leukoplakia, nail dystrophy, and abnormal skin pigmentation, as well as high rates of bone marrow (BM) failure, solid tumors, and other medical problems such as osteopenia. DC and telomere biology disorders (collectively referred to as TBD here) are caused by germline mutations in telomere biology genes leading to very short telomeres and limited proliferative potential of hematopoietic stem cells. We found that skeletal stem cells (SSCs) within the BM stromal cell population (BMSCs, also known as BM-derived mesenchymal stem cells), may contribute to the hematologic phenotype. TBD-BMSCs exhibited reduced clonogenicity, spontaneous differentiation into adipocytes and fibrotic cells, and increased senescence in vitro. Upon in vivo transplantation into mice, TBD-BMSCs failed to form bone or support hematopoiesis, unlike normal BMSCs. TERC reduction (a TBD-associated gene) in normal BMSCs by small interfering TERC-RNA (siTERC-RNA) recapitulated the TBD-BMSC phenotype by reducing proliferation and secondary colony-forming efficiency, and by accelerating senescence in vitro. Microarray profiles of control and siTERC-BMSCs showed decreased hematopoietic factors at the messenger RNA level and decreased secretion of factors at the protein level. These findings are consistent with defects in SSCs/BMSCs contributing to BM failure in TBD.


Bone Marrow Cells/metabolism , Dyskeratosis Congenita/genetics , Mesenchymal Stem Cells/metabolism , RNA/genetics , Telomerase/genetics , Telomere/metabolism , Adolescent , Adult , Animals , Base Sequence , Bone Marrow Cells/pathology , Cell Differentiation , Cell Proliferation , Cellular Senescence , Child , Child, Preschool , Colony-Forming Units Assay , DNA Helicases/genetics , DNA Helicases/metabolism , Dyskeratosis Congenita/pathology , Female , Hematopoiesis/genetics , Humans , Male , Mesenchymal Stem Cells/pathology , Mice , Middle Aged , Molecular Sequence Data , Mutation , RNA/antagonists & inhibitors , RNA/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Telomere/chemistry , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
9.
Ann Biomed Eng ; 43(3): 593-602, 2015 Mar.
Article En | MEDLINE | ID: mdl-25245220

Disease and injury perturb the balance of processes associated with inflammation and tissue remodeling, resulting in positive feedback loops, exacerbation of disease and compromised tissue repair. Conversely, under homeostatic healthy conditions, these processes are tightly regulated through the expansion and/or recruitment of specific cell populations, promoting a balanced steady-state. Better understanding of these regulatory processes and recent advances in biomaterials and biotechnology have prompted strategies to utilize cells for the treatment and prevention of disease through regulation of inflammation and promotion of tissue repair. Herein, we describe how cells that regulate these processes can be increased in prevalence at a site of disease or injury. We review several relevant cell therapy approaches as well as new strategies for directing endogenous regulatory cells capable of promoting environmental homeostasis and even the establishment of a pro-regenerative micro-environment. Collectively, these examples may provide a blueprint for next-generation "medicine" that spurs the body's own cells to action and replaces conventional drugs.


Cell- and Tissue-Based Therapy , Animals , Homeostasis , Humans , Mesenchymal Stem Cells/physiology , Regeneration , T-Lymphocytes, Regulatory/physiology
10.
Connect Tissue Res ; 55 Suppl 1: 155-9, 2014 Aug.
Article En | MEDLINE | ID: mdl-25158202

Biodegradable magnesium (Mg) alloys have been investigated for craniofacial and orthopedic bone fracture fixation due to their initial mechanical strength and high biocompatibility. Although Mg alloys have been reported to enhance bone regeneration in vivo, and enhanced osteogenic marker expression in human bone marrow stromal cells (hBMSCs) cultured in Mg alloy extract was reported, however, the biological mechanism is not fully understood. Thus, it is important to elucidate which signaling pathway in the hBMSCs are activated by Mg(2+) that enhances bone formation. We investigated possible mechanisms underlying effects of Mg(2+) on bone regeneration by culturing differentiated and undifferentiated hBMSCs in the presence of culture medium containing 10 mM MgSO4 both with or without osteogenic factors. mRNA expression of osteogenic genes was analyzed using quantitative PCR arrays. Quantitative PCR array data indicated increased mRNA expression of collagen type X and insulin-like growth factor 2, and decreased expression of integrin alpha 3 in the presence of 10 mM MgSO4. Moreover, Western blotting analysis showed enhanced expression of collagen type X, vascular endothelial growth factor (VEGF), hypoxia-inducible factor (HIF)-2α, and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in the presence of 10 mM MgSO4. In conclusion, 10 mM of MgSO4 enhanced the production of collagen type X and VEGF by hBMSCs. These results also suggest that Mg(2+) released from bone fixation devices may promote bone regeneration by enhancing the production of collagen type X and VEGF of osteogenic cells in bone tissue.


Cell Differentiation/physiology , Magnesium/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis/physiology , Signal Transduction/physiology , Cells, Cultured , Humans , Vascular Endothelial Growth Factor A/metabolism
11.
Acta Biomater ; 10(6): 2834-42, 2014 Jun.
Article En | MEDLINE | ID: mdl-24512978

Magnesium alloys are being investigated for load-bearing bone fixation devices due to their initial mechanical strength, modulus similar to native bone, biocompatibility and ability to degrade in vivo. Previous studies have found Mg alloys to support bone regeneration in vivo, but the mechanisms have not been investigated in detail. In this study, we analyzed the effects of Mg(2+) stimulation on intracellular signaling mechanisms of human bone marrow stromal cells (hBMSCs). hBMSCs were cultured in medium containing 0.8, 5, 10, 20 and 100mM MgSO4, either with or without osteogenic induction factors. After 3weeks, mineralization of extracellular matrix (ECM) was analyzed by Alizarin red staining, and gene expression was analyzed by quantitative polymerase chain reaction array. Mineralization of ECM was enhanced at 5 and 10mM MgSO4, and collagen type X mRNA (COL10A1, an ECM protein deposited during bone healing) expression was increased at 10mM MgSO4 both with and without osteogenic factors. We also confirmed the increased production of collagen type X protein by Western blotting. Next, we investigated the mechanisms of intracellular signaling by analyzing the protein production of hypoxia-inducible factor (HIF)-1α and 2α (transcription factors of COL10A1), vascular endothelial growth factor (VEGF) (activated by HIF-2α) and peroxisome proliferator-activated receptor gamma coactivator (PGC)-1α (transcription coactivator of VEGF). We observed that 10mM MgSO4 stimulation enhanced COL10A1 and VEGF expression, possibly via HIF-2α in undifferentiated hBMSCs and via PGC-1α in osteogenic cells. These data suggest possible ECM proteins and transcription factors affected by Mg(2+) that are responsible for the enhanced bone regeneration observed around degradable Mg orthopedic/craniofacial devices.


Alloys , Magnesium/pharmacology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Animals , Cells, Cultured , Mesenchymal Stem Cells/cytology , Mice , Polymerase Chain Reaction
12.
Proc Natl Acad Sci U S A ; 110(46): 18525-30, 2013 Nov 12.
Article En | MEDLINE | ID: mdl-24167272

The hallmark of periodontal disease is the progressive destruction of gingival soft tissue and alveolar bone, which is initiated by inflammation in response to an invasive and persistent bacterial insult. In recent years, it has become apparent that this tissue destruction is associated with a decrease in local regulatory processes, including a decrease of forkhead box P3-expressing regulatory lymphocytes. Accordingly, we developed a controlled release system capable of generating a steady release of a known chemoattractant for regulatory lymphocytes, C-C motif chemokine ligand 22 (CCL22), composed of a degradable polymer with a proven track record of clinical translation, poly(lactic-co-glycolic) acid. We have previously shown that this sustained presentation of CCL22 from a point source effectively recruits regulatory T cells (Tregs) to the site of injection. Following administration of the Treg-recruiting formulation to the gingivae in murine experimental periodontitis, we observed increases in hallmark Treg-associated anti-inflammatory molecules, a decrease of proinflammatory cytokines, and a marked reduction in alveolar bone resorption. Furthermore, application of the Treg-recruiting formulation (fabricated with human CCL22) in ligature-induced periodontitis in beagle dogs leads to reduced clinical measures of inflammation and less alveolar bone loss under severe inflammatory conditions in the presence of a diverse periodontopathogen milieu.


Alveolar Bone Loss/immunology , Alveolar Bone Loss/prevention & control , Chemokine CCL22/pharmacology , Drug Delivery Systems/methods , Periodontitis/complications , T-Lymphocytes, Regulatory/immunology , Aggregatibacter actinomycetemcomitans/drug effects , Alveolar Bone Loss/etiology , Animals , Chemokine CCL22/administration & dosage , Delayed-Action Preparations/pharmacology , Dogs , Lactic Acid , Mice , Periodontitis/microbiology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Porphyromonas gingivalis/drug effects
14.
Lab Invest ; 90(12): 1747-56, 2010 Dec.
Article En | MEDLINE | ID: mdl-20680009

Fibroblasts act as important immune regulatory cells via their ability to cross-talk with T cells accumulating in lesions. Our previous study showed that fibroblasts produce several cytokines and chemokines by crosslinking HLA class II (HLA-II) molecules with monoclonal antibodies or by making T-cell receptor-peptide-HLA complexes. It is thus conceivable that the interaction of T cells and fibroblasts via HLA-II affects fibroblast responses to stimuli. This study used human gingival fibroblasts (HGF) to investigate possible effects of these fibroblast-derived soluble factors on the differentiation of naïve T cells and on the subsequent fibroblast responses. After mixed lymphocyte reaction culture between naïve T cells and allogeneic dendritic cells in the presence of culture supernatant from HGF stimulated via HLA-DQ molecules (DQ-sup), but not via DR, T cells exhibited a Th2-shifted phenotype, thereby producing quantitatively more IL-13 and IL-5 compared with interferon-γ. Astonishingly, analyses to identify possible factors affecting the Th2 polarization secreted from HLA-II-stimulated HGF, prostaglandin E2, was detected only in DQ-sup. The Th2 polarization of naïve T cells was blocked in the presence of supernatants from indomethacin-treated HGF with HLA-DQ stimulation. In addition, we found that the culture supernatants of Th cells activated following mixed lymphocyte reaction culture in the presence of DQ-sup had the potential to induce gene expression of type I and III collagens in HGF. These results suggested that fibroblasts stimulated via HLA-DQ molecules promote Th2 polarization in Th-cell responses and showed the counter activation of collagen synthesis, implicating orchestrated responses among these cells in the fibrosis of chronic inflammatory lesions.


Cytokines/biosynthesis , Fibroblasts/immunology , Histocompatibility Antigens Class II/immunology , Prostaglandins E/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Adult , Cell Differentiation/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Fibroblasts/drug effects , Gingiva/immunology , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , HLA-DQ Antigens/metabolism , Histocompatibility Antigens Class II/genetics , Humans , In Vitro Techniques , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-5/immunology , Interleukin-5/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Male , Prostaglandins E/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
J Biomed Mater Res B Appl Biomater ; 92(1): 95-101, 2010 Jan.
Article En | MEDLINE | ID: mdl-19924692

In recent years, calcium phosphate cements (CPCs) have frequently been used as bone substitutes in the field of orthopedic surgery. When CPC is used as a bone substitute in vivo, blood contamination is unavoidable. To date, however, no detailed study has been conducted focusing on how the physical properties of CPCs would change under the influence of blood. In this study, the effects of blood contamination on Biopex-R (BPR, PENTAX, Tokyo) are examined in vitro and in vivo. The compressive strength of BPR after setting decreased depending on the amount of contaminating blood. The BPR, which has set in vivo, not only has a fragile surface due to the contamination by blood, but also has a propensity to shorten and be destroyed during the early postoperative stage, especially in the bone exposed to loads. On the other hand, radiographic and histological features in vivo indicated that the absorption and the bone replacement of BPR were stimulated by blood contamination. In the clinical evaluation, the patient's own peripheral venous blood was added to the BPR. One year after the surgery, the absorption was noted around the hardened BPR. To advance CPCs (including BPR) as bioabsorbable bone replaceable materials, it is essential to utilize the patient's own blood in combination with the CPC.


Blood , Bone Cements , Calcium Phosphates , Animals , Rabbits , Surface Properties
16.
Obesity (Silver Spring) ; 15(11): 2549-52, 2007 Nov.
Article En | MEDLINE | ID: mdl-18070744

OBJECTIVE: Recent studies suggested macrophages were integrated in adipose tissues, interacting with adipocytes, thereby exacerbating inflammatory responses. Persistent low-grade infection by gram-negative bacteria appears to promote atherogenesis. We hypothesized a ligand for toll-like receptor 4 (TLR4), bacterial lipopolysaccharide (LPS), would further exaggerate macrophage-adipocyte interaction. RESEARCH METHODS AND PROCEDURES: RAW264.7 macrophage cell line and differentiated 3T3-L1 preadipocytes were co-cultured using transwell system. As a control, each cell was cultured independently. After incubation of the cells with or without Escherichia coli LPS, tumor necrosis factor (TNF)-alpha and interleukin (IL)-6 production was evaluated. RESULTS: Co-culture of macrophages and adipocytes with low concentration of Escherichia coli LPS (1 ng/mL) markedly up-regulated IL-6 production (nearly 100-fold higher than that of adipocyte culture alone, p < 0.01), whereas TNF-alpha production was not significantly influenced. This increase was partially inhibited by anti-TNF-alpha neutralizing antibody. Recombinant TNF-alpha and LPS synergistically up-regulated IL-6 production in adipocytes. However, this increase did not reach the level of production observed in co-cultures stimulated with LPS. DISCUSSION: A ligand for TLR-4 stimulates macrophages to produce TNF-alpha. TNF-alpha, thus produced, cooperatively up-regulates IL-6 production with other soluble factors secreted either from adipocytes or macrophages in these cells. Markedly up-regulated IL-6 would greatly influence the pathophysiology of diabetes and its vascular complications.


Adipocytes/metabolism , Cell Communication/physiology , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/pathology , Animals , Cell Line , Cells, Cultured , Coculture Techniques , Drug Synergism , Macrophages/drug effects , Macrophages/pathology , Mice , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
17.
J Endotoxin Res ; 13(4): 227-34, 2007.
Article En | MEDLINE | ID: mdl-17956941

Diabetic subjects are susceptible to atherosclerosis. It has been postulated that inflammation plays a crucial role in atherogenesis. Since previous studies suggested persistent low-grade infection by Gram-negative bacteria such as Chlamydia spp. and/or periodontal infection is associated with increased atherogenesis among diabetic subjects, we hypothesized that macrophages under hyperglycemia respond to lipopolysaccharide (LPS) challenge in a more exaggerated manner than under normal glucose conditions. Therefore, we examined cytokine productivity and associated signal transduction molecules in LPS-stimulated the monocytic cell line THP-1, under conditions of hyperglycemia. Differentiated THP-1 cells were cultured under normal and high glucose conditions without fetal bovine serum, and were stimulated with Escherichia coli LPS in the presence of LPS binding protein. Following stimulation, activated signal transduction molecules were detected by protein microarray and confirmed thereafter. Results indicated that c-jun N-terminal kinase (JNK) was highly-phosphorylated at high glucose concentrations, and this was confirmed by Western-immunoblotting. Tumor necrosis factor-alpha and monocyte chemo-attractant protein-1 production were significantly enhanced under these conditions. SP600125, a selective inhibitor of JNK, dose-dependently suppressed the production of these cytokine. Therefore, we suggest that this may be one of the mechanisms by which sub-clinical infection by Gram-negative bacteria promotes atherosclerosis in diabetic subjects.


Cytokines/biosynthesis , Cytokines/genetics , Glucose/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL5/genetics , Humans , Inflammation , Interleukin-1beta/genetics , Interleukin-2 , Monocytes , Oligonucleotide Array Sequence Analysis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Tumor Necrosis Factor-alpha/genetics
...