Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Antioxidants (Basel) ; 11(12)2022 Nov 23.
Article in English | MEDLINE | ID: mdl-36552528

ABSTRACT

Hyperglycemia has various adverse health effects, some of which are due to chronic oxidative and inflammatory impairment of bone marrow (BM), hematopoietic stem cells (HSCs), and mesenchymal stem cells (MSCs). Astaxanthin (ASTX) has been shown to ameliorate hyperglycemia-associated systemic complications and acute mortality, and this effect is partially associated with restoration of normal hematopoiesis. Here, the effects of ASTX on diabetes-induced complications in BM and BM stem cells were investigated, and the underlying molecular mechanisms were elucidated. Ten-week-old C57BL/6 mice received a single intraperitoneal injection of streptozotocin (STZ; 150 mg/kg) in combination with oral gavage of ASTX (12.5 mg/kg) for 30 or 60 consecutive days. Supplemental ASTX ameliorated acute mortality and restored the STZ-impaired bone mass accrual and BM microenvironment in STZ-injected mice. Oral gavage of ASTX suppressed osteoclast formation in the BM of STZ-injected mice. Specifically, supplementation with ASTX inhibited oxidative stress and senescence induction of BM HSCs and MSCs and ameliorated hematopoietic disorders in STZ-injected mice. These effects of ASTX were associated with BM restoration of angiopoietin 1, stromal cell-derived factor 1, ß-catenin, and Nrf2. Long-term ASTX gavage also recovered the STZ-induced dysfunction in migration, colony formation, and mineralization of BM-derived stromal cells. Further, a direct addition of ASTX exhibited direct and dose-dependent inhibition of osteoclastic activation without cytotoxic effects. Collectively, these results indicate that ASTX protects against diabetes-induced damage in the BM microenvironment in BM, HSCs, and MSCs and restores normal hematopoiesis and bone accrual in STZ-injected mice.

2.
Mater Sci Eng C Mater Biol Appl ; 135: 112673, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35581065

ABSTRACT

Scaffolds combined with bioactive agents can enhance bone regeneration at therapeutic sites. We explore whether combined supplementation with coumaric acid and recombinant human-cartilage oligomeric matrix protein-angiopoietin 1 (rhCOMP-Ang1) is an ideal approach for bone tissue engineering. We developed coumaric acid-conjugated absorbable collagen scaffold (CA-ACS) and investigated whether implanting CA-ACS in combination with rhCOMP-Ang1 facilitates ACS- or CA-ACS-mediated bone formation using a rat model of critically sized mandible defects. We examined the mechanisms by which coumaric acid and rhCOMP-Ang1 regulate behaviors of human periodontal ligament fibroblasts (hPLFs). The CA-ACS exhibits greater anti-degradation and mechanical strength properties than does ACS alone. Implanting CA-ACS loaded with rhCOMP-Ang1 greatly enhances bone regeneration at the defect via the activation of angiogenic, osteogenic, and anti-osteoclastic responses compared with other rat groups implanted with an ACS alone or CA-ACS. Treatment with both rhCOMP-Ang1 and coumaric acid increases proliferation, mineralization, and migration of cultured hPLFs via activation of the Ang1/Tie2 signaling axis at a greater rate than treatment with either of them alone. Collectively, this study demonstrates that CA-ACS impregnated with rhCOMP-Ang1 enhances bone regeneration at therapeutic sites, and this enhancement is associated with a synergistic interaction between rhCOMP-Ang1-mediated angiogenesis and coumaric acid-related antioxidant responses.


Subject(s)
Angiopoietin-1 , Antioxidants , Angiopoietin-1/metabolism , Angiopoietin-1/pharmacology , Animals , Antioxidants/pharmacology , Cartilage Oligomeric Matrix Protein , Collagen/pharmacology , Coumaric Acids , Mandible , Rats
3.
Nutrients ; 13(10)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34684576

ABSTRACT

Numerous studies highlight that astaxanthin (ASTX) ameliorates hyperglycemic condition and hyperglycemia-associated chronic complications. While periodontitis and periodontic tissue degradation are also triggered under chronic hyperglycemia, the roles of ASTX on diabetes-associated periodontal destruction and the related mechanisms therein are not yet fully understood. Here, we explored the impacts of supplemental ASTX on periodontal destruction and systemic complications in type I diabetic mice. To induce diabetes, C57BL/6 mice received a single intraperitoneal injection of streptozotocin (STZ; 150 mg/kg), and the hyperglycemic mice were orally administered with ASTX (12.5 mg/kg) (STZ+ASTX group) or vehicle only (STZ group) daily for 60 days. Supplemental ASTX did not improve hyperglycemic condition, but ameliorated excessive water and feed consumptions and lethality in STZ-induced diabetic mice. Compared with the non-diabetic and STZ+ASTX groups, the STZ group exhibited severe periodontal destruction. Oral gavage with ASTX inhibited osteoclastic formation and the expression of receptor activator of nuclear factor (NF)-κB ligand, 8-OHdG, γ-H2AX, cyclooxygenase 2, and interleukin-1ß in the periodontium of STZ-injected mice. Supplemental ASTX not only increased the levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and osteogenic transcription factors in the periodontium, but also recovered circulating lymphocytes and endogenous antioxidant enzyme activity in the blood of STZ-injected mice. Furthermore, the addition of ASTX blocked advanced glycation end products-induced oxidative stress and growth inhibition in human-derived periodontal ligament cells by upregulating the Nrf2 pathway. Together, our results suggest that ASTX does not directly improve hyperglycemia, but ameliorates hyperglycemia-triggered periodontal destruction and oxidative systemic complications in type I diabetes.


Subject(s)
Antioxidants/metabolism , Diabetes Mellitus, Experimental/complications , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Periodontitis/drug therapy , Periodontitis/etiology , Streptozocin/administration & dosage , Adolescent , Alveolar Process/pathology , Animals , Blood Glucose/metabolism , Catalase/blood , Cell Proliferation , Cytokines/metabolism , DNA Damage , Diabetes Mellitus, Experimental/blood , Dietary Supplements , Feeding Behavior , Glycation End Products, Advanced/metabolism , Humans , Hyperglycemia/complications , Inflammation Mediators/metabolism , Injections , Lymphocytes/immunology , Male , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoclasts/pathology , Periodontal Ligament/pathology , Periodontitis/blood , Reactive Oxygen Species/metabolism , Superoxide Dismutase/blood , Up-Regulation , Xanthophylls/pharmacology , Xanthophylls/therapeutic use , Young Adult
4.
J Periodontal Res ; 54(6): 690-701, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31328274

ABSTRACT

OBJECTIVE: Dietary bioactive materials having anti-inflammatory and antioxidant potentials are able to inhibit diabetes-associated periodontal complications. Although numerous studies indicate that administration of p-coumaric acid (p-CA) ameliorates diabetes and diabetes-related complications, the roles of p-CA on periodontal tissue destruction in diabetic mice and the possible mechanisms therein are not completely understood. In this study, we evaluated whether supplementation with p-CA protects mice against diabetes-associated spontaneous periodontal destruction and also explored the associated mechanism therein using in vivo and in vitro experimental systems. MATERIALS AND METHODS: C57BL/6 male mice were divided into sham, streptozotocin (STZ), and STZ+CA groups (n = 5/group). Sham group was intraperitoneally injected with sodium buffer, whereas other two groups were injected with the buffer containing 160 mg/kg of STZ. STZ-induced diabetic mice received oral gavage with p-CA (50 mg/kg) (STZ+CA group) or with buffer only (STZ group) daily for 6 weeks. The effect of p-CA on diabetes-associated spontaneous periodontal destruction was evaluated using µCT analysis, hematoxylin and eosin staining, tartrate-resistant acid phosphatase staining, and immunohistochemical staining methods. The efficacies of p-CA on cell proliferation, osteoblast differentiation, reactive oxygen species (ROS) accumulation, and antioxidant-related marker expression were examined using human periodontal ligament fibroblasts (hPLFs) cultured under high glucose condition. RESULTS: Streptozotocin group exhibited periodontal tissue destruction along with increased inflammation, oxidative stress, and osteoclast formation, as well as with decreased osteogenesis. However, oral administration with p-CA protected mice against STZ-induced periodontal destruction by inhibiting inflammation and osteoclastic activation. STZ+CA group also showed higher expression of antioxidant and osteogenic markers in periodontal tissue than did STZ group. Treatment with high glucose concentration (30 mmol/L) impaired proliferation and osteoblast differentiation of hPLFs along with cellular ROS accumulation, whereas these impairments were almost completely disappeared by supplementation with p-CA. CONCLUSION: These findings demonstrate that supplementation with p-CA inhibits diabetes-associated spontaneous destruction of periodontal tissue by enhancing anti-inflammatory, anti-osteoclastogenic, and antioxidant defense systems in STZ-treated mice.


Subject(s)
Diabetes Mellitus, Experimental/complications , Dietary Supplements , Oxidative Stress , Periodontal Diseases/drug therapy , Propionates/pharmacology , Administration, Oral , Animals , Antioxidants/metabolism , Cells, Cultured , Coumaric Acids , Fibroblasts , Humans , Male , Mice , Mice, Inbred C57BL , Periodontal Diseases/etiology , Periodontal Ligament/cytology , Streptozocin
5.
J Bone Miner Metab ; 37(5): 900-912, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30843129

ABSTRACT

Numerous studies have demonstrated the advantages of plant cell suspension culture systems in producing bioactive recombinant human growth factors. This study investigated the biological activity of recombinant basic human fibroblast growth factor (rhFGF2) protein produced by a plant culture system to enhance new bone formation in a bone defect mouse model. The human FGF2 cDNA gene was cloned into a plant expression vector driven by the rice α-amylase 3D promoter. The vector was introduced into rice calli (Oryza sativa L. cv. Dongjin), and the clone with the highest expression of rhFGF2 was selected. Maximum accumulation of rhFGF2 protein (approximately 28 mg/l) was reached at 13 day post-incubation. Male C57BL/6 mice underwent calvarial defect surgery and the defects were loaded with absorbable collagen sponge (ACS) only (ACS group) or ACS impregnated with 5 µg of plant-derived rhFGF2 (p-FGF2) protein or E. coli-derived rhFGF2 (e-FGF2) protein. Similar to the effects of e-FGF2, local delivery with p-FGF2 enhanced bone healing in the damaged region to higher levels than the ACS group. Exogenous addition of p-FGF2 or e-FGF2 exhibited similar effects on proliferation, mineralization, and osteogenic marker expression in MC3T3-E1 cells. Together, the current findings support the usefulness of this plant-based expression system for the production of biologically active rhFGF2.


Subject(s)
Dietary Supplements , Fibroblast Growth Factor 2/pharmacology , Oryza/genetics , Osteogenesis/drug effects , Recombinant Proteins/pharmacology , Skull/pathology , Amino Acid Sequence , Animals , Base Sequence , Biomarkers/metabolism , Calcification, Physiologic/drug effects , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Escherichia coli/metabolism , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/isolation & purification , Gene Expression Regulation/drug effects , Humans , Male , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Plants, Genetically Modified , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Skull/drug effects
6.
J Periodontol ; 85(4): 645-54, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23805819

ABSTRACT

BACKGROUND: Periodontal ligament fibroblasts (PLFs) maintain homeostasis of periodontal ligaments by producing paracrine factors that affect various functions of stem-like cells. It is hypothesized that PLFs induce proliferation and differentiation of stem cells more effectively than gingival fibroblasts (GFs) and skin fibroblasts (SFs). METHODS: PLFs and GFs were isolated from extracted teeth and cultured in the presence and absence of osteogenesis-inducing factors. Mouse embryonic stem (mES) cells and SFs were purchased commercially. mES cells were incubated with culture supernatants of these fibroblasts or cocultured directly with the cells. Proliferation and mineralization in mES cells were determined at various times of incubation. Immunostaining and polymerase chain reaction were performed. The activity of mitogen-activated protein kinase and alkaline phosphatase (ALP) was also measured. RESULTS: In cocultures, PLFs stimulated proliferation of mES cells more effectively than GFs or SFs. Similarly, the addition of culture supernatant of PLFs induced the most prominent proliferation of mES cells, and this was significantly inhibited by treatment with antibody against fibroblast growth factor (FGF)4 or the c-Jun N-terminal kinase inhibitor SP600125 (anthra[1,9-cd]pyrazol-6(2H)-one). Supplementation with culture supernatant from the fibroblasts induced osteogenic differentiation of mES cells in the order PLFs > GFs > SFs. These activities of PLFs were related to their potential to produce osteogenic markers, such as ALP and runt-related transcription factor-2 (Runx2), and to secrete FGF7. Pretreatment of mES cells with the extracellular signal-regulated kinase inhibitor PD98059 [2-(2-amino-3-methyoxyphenyl)-4H-1-benzopyran-4-one] or SP600125 clearly attenuated mineralization induced by culture supernatant of PLF with attendant decreases in mRNA levels of Runx2, bone sialoprotein, osteocalcin, and osteopontin. CONCLUSION: PLFs regulate the proliferation and osteogenic differentiation of mES cells more strongly than GFs and SFs via the secretion of FGF through a mechanism that involves mitogen-activated protein kinase-mediated signaling.


Subject(s)
Embryonic Stem Cells/physiology , Fibroblast Growth Factors/physiology , Fibroblasts/physiology , Osteogenesis/physiology , Periodontal Ligament/cytology , Alkaline Phosphatase/analysis , Animals , Anthracenes/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Culture Techniques , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Coculture Techniques , Core Binding Factor Alpha 1 Subunit/analysis , Culture Media, Conditioned , Fibroblast Growth Factor 4/antagonists & inhibitors , Fibroblast Growth Factor 7/analysis , Fibroblast Growth Factors/analysis , Flavonoids/pharmacology , Gingiva/cytology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Signaling System/physiology , Mice , Mitogen-Activated Protein Kinases/analysis , Osteocalcin/analysis , Osteopontin/analysis , Skin/cytology
7.
Phytother Res ; 25(8): 1166-73, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21290442

ABSTRACT

Quercetin is known to inhibit tyrosinase activity and melanin production in melanocytes. However, several reports suggest that quercetin has different and opposite effects on melanogenesis. This study examined the precise effects of quercetin on melanogenesis using cell-free assay systems and melanocytes. Quercetin inhibited the monophenolase and diphenolase activities of tyrosinase, and melanin synthesis in cell-free assay systems. Quercetin induced mild stimulation of the tyrosinase activity and dihydroxyphenylalaminechrome tautomerase (TRP-2) expression but only at low concentrations (<20 µm) in B16F10 melanoma cells. In contrast, the addition of 50 µm quercetin to the cells led to a significant decrease in the activity and synthesis of tyrosinase, as well as a decrease in the expression of tyrosinase-related protein-1 and TRP-2 proteins, regardless of the presence or absence of α-melanocyte stimulating hormone (α-MSH). Quercetin also reduced the intracellular cAMP and the phosphorylated protein kinase A levels in α-MSH-stimulated B16F10 cells. Moreover, quercetin (20 µm) diminished the expression and activity of tyrosinase, and melanin content in cultured normal human epidermal melanocytes. These effects were not related to its cytotoxic action. Although the in vivo effects of quercetin are still unclear, these results suggest that quercetin could play important roles in controlling melanogenesis.


Subject(s)
Melanins/biosynthesis , Melanoma, Experimental/metabolism , Quercetin/pharmacology , alpha-MSH/antagonists & inhibitors , Animals , Cell Differentiation/drug effects , Cell Growth Processes/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Intramolecular Oxidoreductases/biosynthesis , Melanins/antagonists & inhibitors , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma, Experimental/pathology , Mice , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Oxidoreductases/biosynthesis , Phosphorylation , Pigmentation/drug effects , Tumor Cells, Cultured , alpha-MSH/pharmacology
8.
Apoptosis ; 12(7): 1289-98, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17356895

ABSTRACT

Flavonoids have antioxidant and antitumor promoting effects. Rhus verniciflua Stokes (RVS) is a flavonoid-rich herbal medicine that has long been used in Korea as both a food additive and antitumor agent. It was previous reported that a purified flavonoid fraction prepared from RVS, herein named RCMF (the RVS chloroform-methanol fraction), inhibited the proliferation and induced apoptosis in human osteosarcoma (HOS) cells. This study examined the mechanisms involved in the RCMF-mediated apoptosis in HOS cells. RCMF was shown to be capable of inducing apoptosis in HOS cells by inducing p53 in the cells resulting in the decrease in Bcl-2 level, activation of Bax, and cytoplasmic release of cytochrome c, which led to the translocation of apoptosis-inducing factor (AIF) and endonuclease G (EndoG) into the nucleus. However, the RCMF-induced apoptosis was suppressed by transfecting the cells with antisense p53 oligonucleotides but not by treating them with a MAPK or caspase inhibitor. This suppression occurred through the regulation of Bcl-2 members as well as by preventing the nuclear translocation of the mitochondrial apoptogenic factors. Overall, it appears that p53-mediated mitochondrial stress and the nuclear translocation of AIF and EndoG are mainly required for the apoptosis induced by RCMF.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/physiology , Cell Line, Tumor/metabolism , Endodeoxyribonucleases/metabolism , Mitochondria/metabolism , Tumor Suppressor Protein p53/metabolism , Caspases/metabolism , Cell Line, Tumor/cytology , Flavonoids , Humans , Korea , Membrane Potential, Mitochondrial , Mitogen-Activated Protein Kinases/metabolism , Osteosarcoma , Rhus
9.
Biochim Biophys Acta ; 1726(3): 309-16, 2005 Nov 30.
Article in English | MEDLINE | ID: mdl-16213662

ABSTRACT

Many studies have suggested that dietary flavonoids are anticancer agents that induce the apoptosis of cancer cells. However, the effects of flavonoids on the induction of apoptosis in osteosarcoma cells are unclear. Previously, a flavonoid fraction, consisting mainly of protocatechuic acid, fustin, fisetin, sulfuretin, and butein, herein named RCMF (the RVS chloroform-methanol fraction), was prepared from a crude acetone extract of Rhus verniciflua Stokes (RVS). This study evaluated the effects of RCMF on the proliferation and apoptosis using human osteosarcoma (HOS) cells. The mechanism of growth inhibition of the HOS cells by the flavonoid fraction, RCMF, was also assessed. The results demonstrated that RCMF exhibited sensitive growth inhibition and induced apoptosis in HOS cells. PARP cleavage was closely associated with the RCMF-induced apoptosis of the HOS cells. Furthermore, the activation of caspase 8 and Bax, the inhibition of Bcl-2 expression, and the release of cytochrome c are believed to be involved in the RCMF-mediated apoptosis. Collectively, these findings suggest that RCMF is an agent which may be capable of inducing sensitive growth inhibition and apoptosis in HOS cells.


Subject(s)
Anticarcinogenic Agents/toxicity , Apoptosis , Flavonoids/toxicity , Anticarcinogenic Agents/isolation & purification , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Flavonoids/isolation & purification , Humans , Mitochondria/metabolism , Osteosarcoma/pathology , Plant Extracts/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Rhus/chemistry
10.
Toxicol Lett ; 155(1): 115-25, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15585366

ABSTRACT

Considerable attention is being concentrated on dietary flavonoids in developing novel cancer-preventive approaches due to their potential ability to induce selective apoptosis of cancer cells. In this study, we prepared a flavonoid-containing fraction from a crude acetone extract of Rhus verniciflua Stokes (RVS), traditionally used as a food additive and as an herbal medicine, and named RVS chloroform-methanol fraction (RCMF). We evaluated the effects of RCMF on proliferation and apoptosis using mouse embryonic primary hepatic cells (MPHC), embryonic normal hepatic cell line (BNL CL.2), and its SV40-mediated transformed cell line (BNL SV A.8). We also investigated the effects of RCMF on the antioxidant defense system in those cells. This study demonstrated that RCMF exhibited a selective growth inhibition and apoptosis induction on transformed cells. BNL SV A.8 cells were more sensitive to RCMF-mediated cytotoxicity than were MPHC or BNL CL.2. RCMF-mediated reduction of MnSOD activity and glutathione (GSH) content in BNL SV A.8 cells is thought to be associated with RCMF-induced apoptosis. Our findings suggest that RCMF is an agent which may be capable of inducing growth inhibition and apoptosis of hepatic tumor cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavonoids/pharmacology , Liver/cytology , Rhus/chemistry , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Cells, Cultured , DNA/biosynthesis , DNA Fragmentation/drug effects , Flavonoids/chemistry , Flavonoids/isolation & purification , Hepatocytes/drug effects , In Situ Nick-End Labeling , Liver/drug effects , Mice , Mice, Inbred BALB C , Structure-Activity Relationship
11.
Mol Cells ; 18(2): 163-70, 2004 Oct 31.
Article in English | MEDLINE | ID: mdl-15528991

ABSTRACT

Excessive breakdown of extracellular matrix by metalloproteinases (MMPs) occurs in many pathological conditions. Consequently, methods for inhibiting MMP activity have therapeutic potential. In this study, we investigated the effect of G-120, a 120 kDa glycoprotein purified from the Oriental herbal plant, Ulmus davidiana Nakai (UDN), on the activity and production of several MMPs by evaluating its growth inhibitory effect on NIH 3T3 cells. Tritium uptake assays showed that proliferation of NIH 3T3 cells was strongly suppressed, and G-120-mediated inhibition of DNA synthesis proved to involve a cytostatic, rather than a cytotoxic, effect, as shown by cytotoxicity and apoptosis assays. More importantly, G-120 strongly reduced the gelatinolytic and collagenase activities of MMP proteins, as well as expression of MMP-2 and MMP-9. Electrophoretic mobility shift assays revealed that it suppressed the DNA binding activity of NF-kappaB. Collectively, our observations show that G-120 strongly inhibits the activation of MMPs and NF-kappaB.


Subject(s)
Cell Proliferation/drug effects , Matrix Metalloproteinase Inhibitors , Plant Proteins/pharmacology , Ulmus/chemistry , Animals , DNA/biosynthesis , Dose-Response Relationship, Drug , Glycoproteins/pharmacology , Matrix Metalloproteinases/metabolism , Mice , NF-kappa B/antagonists & inhibitors , NIH 3T3 Cells , Plants, Medicinal/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL