Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cells ; 41(1): 93-104, 2023 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-36368017

RESUMO

While supplemental angiopoietin-1 (Ang1) improves hematopoiesis, excessive Ang1 induces bone marrow (BM) impairment, hematopoietic stem cell (HSC) senescence, and erythropoietic defect. Here, we examined how excessive Ang1 disturbs hematopoiesis and explored whether hematopoietic defects were related to its level using K14-Cre;c-Ang1 and Col2.3-Cre;c-Ang1 transgenic mice that systemically and locally overexpress cartilage oligomeric matrix protein-Ang1, respectively. We also investigated the impacts of Tie2 inhibitor and AMD3100 on hematopoietic development. Transgenic mice exhibited excessive angiogenic phenotypes, but K14-Cre;c-Ang1 mice showed more severe defects in growth and life span with higher presence of Ang1 compared with Col2.3-Cre;c-Ang1 mice. Dissimilar to K14-Cre;c-Ang1 mice, Col2.3-Cre;c-Ang1 mice did not show impaired BM retention or senescence of HSCs, erythropoietic defect, or disruption of the stromal cell-derived factor 1 (SDF-1)/CXCR4 axis. However, these mice exhibited a defect in platelet production depending on the expression of Tie2 and globin transcription factor 1 (GATA-1), but not GATA-2, in megakaryocyte progenitor (MP) cells. Treatment with Tie2 inhibitor recovered GATA-1 expression in MP cells and platelet production without changes in circulating RBC in transgenic mice. Consecutive AMD3100 administration not only induced irrecoverable senescence of HSCs but also suppressed formation of RBC, but not platelets, via correlated decreases in number of erythroblasts and their GATA-1 expression in B6 mice. Our results indicate that genetic overexpression of Ang1 impairs hematopoietic development depending on its level, in which megakaryopoiesis is preferentially impaired via activation of Ang1/Tie2 signaling, whereas erythropoietic defect is orchestrated by HSC senescence, inflammation, and disruption of the SDF-1/CXCR4 axis.


Assuntos
Anemia , Trombocitopenia , Camundongos , Animais , Proteína de Matriz Oligomérica de Cartilagem/genética , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Camundongos Transgênicos , Anemia/genética , Receptor TIE-2/genética , Receptor TIE-2/metabolismo
2.
Stem Cells ; 39(1): 103-114, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33038284

RESUMO

Although functional association between Wnt signaling and bone homeostasis has been well described through genetic ablation of Wntless (Wls), the mechanisms of how osteoblastic Wls regulates the fate of bone marrow stromal cells (BMSCs) and hematopoietic stem cells (HSCs) in relation to age are not yet understood. Here, we generated Col2.3-Cre;Wlsfl/fl mice that were free from premature lethality and investigated age-related impacts of osteoblastic Wls deficiency on hematopoiesis, BM microenvironment, and maintenance of BMSCs (also known as BM-derived mesenchymal stem/stromal cells) and HSCs. Ablation of osteoblastic Wls deteriorated BM microenvironment and bone mass accrual along with age-independent effects on functions of BMSCs. Osteoblastic Wls deletion impaired HSC repopulation and progeny with skewing toward myeloid lineage cells only at old stage. As proven by hallmarks of stem cell senescence, osteoblastic Wls ablation differentially induced senescence of BMSCs and HSCs in relation to age without alteration in their BM frequency. Our findings support that deletion of Wls in Col2.3-expressing cells induces senescence of BMSCs and impairs BM microenvironment in age-independent manner. Overall, long-term deterioration in BM microenvironment contributes to age-related HSC senescence with impaired progeny and hematopoiesis, which also suggests possible roles of osteoblastic Wls on the maintenance of BM HSCs.


Assuntos
Envelhecimento/metabolismo , Células da Medula Óssea/metabolismo , Deleção de Genes , Osteoblastos/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Células-Tronco/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/metabolismo
3.
Int J Mol Sci ; 23(12)2022 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-35743193

RESUMO

The most prevalent chronic liver disorder in the world is fatty liver disease caused by a high-fat diet. We examined the effects of Lactiplantibacillus plantarum-KCC48 on high-fat diet-induced (HFD) fatty liver disease in mice. We used the transcriptome tool to perform a systematic evaluation of hepatic mRNA transcripts changes in high-fat diet (HFD)-fed animals and high-fat diet with L. plantarum (HFLPD)-fed animals. HFD causes fatty liver diseases in animals, as evidenced by an increase in TG content in liver tissues compared to control animals. Based on transcriptome data, 145 differentially expressed genes (DEGs) were identified in the liver of HFD-fed mice compared to control mice. Moreover, 61 genes were differentially expressed in the liver of mice fed the HFLPD compared to mice fed the HFD. Additionally, 43 common DEGs were identified between HFD and HFLPD. These genes were enriched in metabolic processes, retinol metabolism, the PPAR signaling pathway, fatty acid degradation, arachidonic metabolism, and steroid hormone synthesis. Taking these data into consideration, it can be concluded that L. plantarum-KCC48 treatment significantly regulates the expression of genes involved in hepatosteatosis caused by HFD, which may prevent fatty liver disease.


Assuntos
Dieta Hiperlipídica , Hepatopatia Gordurosa não Alcoólica , Animais , Dieta Hiperlipídica/efeitos adversos , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/metabolismo , Transcriptoma
4.
FASEB J ; 34(9): 12289-12307, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32701200

RESUMO

Given the rising evidence that gut malfunction including changes in the gut microbiota composition, plays a major role in the development of obesity and associated metabolic diseases, the exploring of novel probiotic bacteria with potential health benefits has attracted great attention. Recently Lactobacillus spp., exert potent anti-obesity effects by regulating key transcriptional and translational factors in adipose tissues. However, the molecular mechanism behind the anti-obesity effect of probiotics is not yet fully understood. Therefore, we investigated the effect of Lactobacillus plantarum A29 on the expression of adipogenic and lipogenic genes in 3T3-L1 adipocytes and high-fat diet (HFD)-fed mice. We observed that the treatment of 3T3-L1 adipocytes with the cell-free metabolites of L plantarum inhibited their differentiation and fat depositions via downregulating the key adipogenic transcriptional factors (PPAR-γ, C/EBP-α, and C/EBP-ß) and their downstream targets (FAS, aP2, ACC, and SREBP-1). Interestingly, supplementation with L plantarum reduced the fat mass and serum lipid profile concurrently with downregulation of lipogenic gene expression in the adipocytes, resulting in reductions in the bodyweight of HFD-fed obese mice. L plantarum treatment attenuated the development of obesity in HFD-fed mice via the activation of p38MAPK, p44/42, and AMPK-α by increasing their phosphorylation. Further analysis revealed that A29 modulated gut-associated microbiota composition. Thus, A 29 potential probiotic strain may alleviate the obesity development and its associated metabolic disorders via inhibiting PPARγ through activating the p38MAPK and p44/42 signaling pathways.


Assuntos
Disbiose/terapia , Microbioma Gastrointestinal , Lactobacillus plantarum/fisiologia , Obesidade/terapia , Probióticos/farmacologia , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Diferenciação Celular , Dieta Hiperlipídica , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Obesidade/metabolismo , Obesidade/microbiologia , PPAR gama/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
5.
J Bone Miner Metab ; 37(5): 900-912, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30843129

RESUMO

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.


Assuntos
Suplementos Nutricionais , Fator 2 de Crescimento de Fibroblastos/farmacologia , Oryza/genética , Osteogênese/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Crânio/patologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Biomarcadores/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Escherichia coli/metabolismo , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/isolamento & purificação , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Plantas Geneticamente Modificadas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Crânio/efeitos dos fármacos
6.
J Periodontal Res ; 54(6): 690-701, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31328274

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/complicações , Suplementos Nutricionais , Estresse Oxidativo , Doenças Periodontais/tratamento farmacológico , Propionatos/farmacologia , Administração Oral , Animais , Antioxidantes/metabolismo , Células Cultivadas , Ácidos Cumáricos , Fibroblastos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Periodontais/etiologia , Ligamento Periodontal/citologia , Estreptozocina
7.
Indian J Clin Biochem ; 34(1): 68-75, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30728675

RESUMO

The aims of this study were to explore whether a phenolic acid and flavonoid fraction (named PAFF) isolated from Lolium multiflorum Lam. protects against d-galactosamine (GalN)-induced liver damages in mice and to investigate the associated mechanisms. ICR mice received oral administration with various concentrations (50, 100, and 200 mg/kg body weight) of PAFF once per 2 days for seven times before intraperitoneal injection with 800 mg/kg GalN. After a day of GalN challenge, blood and tissue samples were analyzed by biochemical, histopathological, real time RT-PCR, and Western blot methods. GalN challenge induced severe damage to hepatocytes with hepatocellular vacuolization and necrosis. GalN treatment increased serum ALT, ALP, AST, and LDH levels and hepatic MDA levels and stimulated mRNA and protein expressions of Nrf2 and HO-1 in the liver. GalN treatment also diminished the levels of GSH and the activities of CAT, SOD, and GPx in the liver. However, combined treatment with PAFF inhibited GalN-mediated increases in the histological damages and the levels of serum enzymes and hepatic MDA, restored the activities of hepatic antioxidant enzymes up to those in the control values, and augmented the GalN-stimulated expression of Nrf2 and HO-1 in the liver. Furthermore, PAFF treatment alone increased the cellular SOD activity and the expression of Nrf2 and HO-1 in the liver. Our results suggest that PAFF may protect against GalN-induced liver damage by decreasing oxidative stress and increasing cellular antioxidant activities through an activation of Nrf2/HO-1-dependent pathway.

8.
Biochem Biophys Res Commun ; 499(3): 669-674, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29604278

RESUMO

Supplemental Angiopoietin 1 (Ang1) exerts its therapeutic potential on microvascular regression-associated diseases, and this potential is linked with the function of hematopoietic stem cells (HSCs). However, the underlying mechanisms of the effect of enhanced angiogenesis on the modulation of HSCs are not yet defined. Here, we generated transgenic mice expressing Cartilage Oligomeric Matrix Protein (COMP)-Ang1 in keratin 14-expressing cells. The mutant animals expressed excessive angiogenic characteristics in the skin and bone marrow (BM) along with redder skin with more numerous and branched vessels compared with their wild-type (WT) littermates. The mutants displayed reduced long bone formation and osteoclast activity than did WT littermates and had fewer CD150+CD48-Lineage-Sca-1+c-Kit+ (LSK) cells in the BM. The mutants also exhibited greater senescence-associated (SA) ß-gal activity, p16INK4a protein expression, and superoxide anion levels in CD150+CD48-LSK cells in the BM. Furthermore, transplantation assay revealed that the mutant-derived LSK cells were inferior to the cells derived from WT littermate in inducing competitive repopulating capacity in the recipients. Collectively, our results demonstrate that persistent and prolonged administration of COMP-Ang1 by inducible transgenic expression mediates excessive angiogenesis in the body and impairs BM microenvironment, eventually leading to senescence of BM HSCs.


Assuntos
Angiopoietina-1/genética , Medula Óssea/metabolismo , Proteína de Matriz Oligomérica de Cartilagem/genética , Microambiente Celular , Senescência Celular , Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Proteínas Recombinantes de Fusão/genética , Animais , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos Transgênicos , Mutação/genética , Neovascularização Fisiológica , Osteoclastos , Proteínas Recombinantes de Fusão/metabolismo
9.
Stem Cells ; 34(10): 2601-2607, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27300755

RESUMO

Osteoclasts form a bone marrow (BM) cavity serving as a hematopoietic niche for the maintenance of hematopoietic stem cells (HSCs). However, the role of osteoclasts in the BM has been controversially reported and remains to be further understood. In the present study, we investigated how osteoclasts affect the modulation of hematopoietic stem/progenitor cells in the BM by administering bisphosphate alendronate (ALN) to B6 mice for 21 consecutive days to inhibit osteoclast activity. ALN treatment caused a reduction in the number of tartrate-resistant acid phosphate (TRAP)-positive osteoclast cells and an increase in bone mineral density, particularly in the trabecular zone, but not in the cortical zone of the BM. Osteoclast inhibition caused by ALN treatment decreased mitochondrial reactive oxygen species (ROS) generation and SA-ß-gal activity of CD150+ CD48- Lineage-Sca-1+ c-Kit+ (LSK) cells, eventually leading to an improvement in the engraftment potential and self-renewal activity of HSCs. Moreover, ALN-treated mice exhibited an enhanced resistance of HSCs in response to the genotoxic stress of 5-fluorouracil, as determined by mitochondrial ROS generation, SA-ß-gal activity, and p16INK4a expression in subsets of LSK and CD150+ CD48- LSK cells as well as competitive assay. Collectively, our findings indicate that inhibition of osteoclast activity improves the long-term engraftment potential and stress resistance of HSCs. Stem Cells 2016;34:2601-2607.


Assuntos
Alendronato/administração & dosagem , Alendronato/farmacologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Osteoclastos/metabolismo , Estresse Fisiológico , Animais , Antineoplásicos/efeitos adversos , Densidade Óssea/efeitos dos fármacos , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Osso Esponjoso/efeitos dos fármacos , Osso Esponjoso/fisiologia , Autorrenovação Celular/efeitos dos fármacos , Feminino , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Camundongos Endogâmicos C57BL , Osteoclastos/efeitos dos fármacos , Células-Tronco de Sangue Periférico/citologia , Baço/citologia , Estresse Fisiológico/efeitos dos fármacos , Fatores de Tempo
10.
J Bone Miner Metab ; 35(5): 485-496, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27766421

RESUMO

Fibroblast growth factor 7 (FGF7) plays an important role in regulating the proliferation, migration, and differentiation of cells. However, the role of FGF7 in bone formation is not yet fully understood. We examined the effect of FGF7 on bone formation using a rat model of mandible defects. Rats underwent mandible defect surgery and then either scaffold treatment alone (control group) or FGF7-impregnated scaffold treatment (FGF7 group). Micro-CT and histological analyses revealed that the FGF7 group exhibited greater bone formation than did the control group 10 weeks after surgery. With the exception of total porosity (%), all bone parameters had higher values in the FGF7 group than in the control group at each follow-up after surgery. The FGF7 group showed greater expression of osteogenic markers, such as runt-related transcription factor 2, osterix, osteocalcin, bone morphogenetic protein 2, osteopontin, and type I collagen in newly formed bone than did the control group. The delivery of FGF7 also increased the messenger RNA expression of stromal-cell-derived factor 1 (SDF-1) and CXCR4 in newly formed bone in the FGF7 group compared with the control group. Further, addition of exogenous FGF7 induced migration of rat bone marrow stromal cells and increased the expression of SDF-1 and CXCR4 in the cells. Furthermore, the addition of FGF7 augmented mineralization in the cells with increased expression of osteogenic markers, and this augmentation was significantly suppressed by an inhibitor specific for c-Jun N-terminal kinase (SP600125) or extracellular-signal-regulated kinase (PD98059). Collectively, these results suggest that local delivery of FGF7 increases bone formation in a mandible defect with enhanced osteogenesis and chemoattraction.

11.
Pharm Biol ; 55(1): 611-619, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27937124

RESUMO

CONTEXT: Interest has recently renewed in using Lolium multiflorum Lam. (Poaceae) (called Italian ryegrass; IRG) silage as an antioxidant and anti-inflammatory diet. OBJECTIVE: This study investigated the antioxidant, anti-inflammatory and anti-septic potential of IRG silage and identified the primary components in IRG active fractions. MATERIALS AND METHODS: Total 16 fractions were separated from the chloroform-soluble extract of IRG aerial part using Sephadex LH-20 column before HPLC analysis. Antioxidant and anti-inflammatory activities of the fractions at doses of 0-100 µg/mL were investigated using various cell-free and cell-mediated assay systems. To explore anti-septic effect of IRG fractions, female ICR and BALB/c mice orally received 40 mg/kg of phenolic acid and flavonoid-rich active fractions F7 and F8 every other day for 10 days, respectively, followed by LPS challenge. RESULTS: The active fractions showed greater antioxidant and anti-inflammatory potential compared with other fractions. IC50 values of F7 and F8 to reduce LPS-stimulated NO and TNF-α production were around 15 and 30 µg/mL, respectively. Comparison of retention times with authentic compounds through HPLC analysis revealed the presence of caffeic acid, ferulic acid, myricetin and kaempferol in the fractions as primary components. These fractions inhibited LPS-stimulated MAPK and NF-κB activation. Supplementation with F7 or F8 improved the survival rates of mice to 70 and 60%, respectively, in LPS-injected mice and reduced near completely serum TNF-α and IL-6 levels. DISCUSSION AND CONCLUSION: This study highlights antioxidant, anti-inflammatory and anti-septic activities of IRG active fractions, eventually suggesting their usefulness in preventing oxidative damage and inflammatory disorders.


Assuntos
Anti-Infecciosos Locais/farmacologia , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Flavonoides/farmacologia , Hidroxibenzoatos/farmacologia , Inflamação/prevenção & controle , Lolium/química , Macrófagos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Sepse/prevenção & controle , Animais , Anti-Infecciosos Locais/isolamento & purificação , Anti-Inflamatórios/isolamento & purificação , Antioxidantes/isolamento & purificação , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Feminino , Flavonoides/isolamento & purificação , Hidroxibenzoatos/isolamento & purificação , Inflamação/induzido quimicamente , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fitoterapia , Extratos Vegetais/isolamento & purificação , Plantas Medicinais , Células RAW 264.7 , Sepse/induzido quimicamente , Sepse/metabolismo , Silagem , Solventes/química , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
12.
Mol Cell Biochem ; 416(1-2): 157-68, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27107990

RESUMO

Recombinant COMP-Ang1, a chimera of angiopoietin-1 (Ang1), and a short coiled-coil domain of cartilage oligomeric matrix protein (COMP) can stimulate multiple cellular processes. Proliferative capacity of periodontal ligament (PDL) fibroblasts (PLFs) is important for maintaining PDL integrity and homeostasis. In this study, we explored whether exogenous COMP-Ang1 addition enhances proliferation of human PLFs and the cellular mechanisms therein. We initially isolated and characterized PLFs, where the cells showed highly positive staining for surface markers, CD90, CD105, and CD146. COMP-Ang1 treatment increased proliferation of PLFs by stimulating migration of cells into S and G2/M phases. This increase was coupled with decreased p21(Cip) and p27(Kip) levels and enhanced cyclin D1, cyclin-dependent kinase (CDK) 2, and CDK4 induction. Transfection with si-Tie2 near completely blocked COMP-Ang1-stimulated cell cycle progression in PLFs. Tie2 knockdown also inhibited COMP-Ang1-induced phosphorylation of mitogen-activated protein kinases (MAPKs). In addition, COMP-Ang1-mediated activation of Akt and c-Jun was suppressed by treating each of pharmacological inhibitors specific to phosphoinositide 3-kinase (PI3K) (LY294002 and Wortmannin) or MAPKs (PD98059, SB203580, and SP600125). Similarly, COMP-Ang1-mediated increases in DNA synthesis and cyclin D1 induction were prevented by treating inhibitor of MAPKs and PI3K or by c-Jun knockdown. These results suggest that COMP-Ang1 enhances survival and proliferation of human PLFs through the activation of Tie2-mediated signaling, where PI3K/Akt and MAPK-c-Jun signaling pathways act as downstream effectors. Collectively, COMP-Ang1 may be a useful as a stimulator of human PLFs and therefore improves PDL integrity and homeostasis.


Assuntos
Angiopoietina-1/farmacologia , Proteína de Matriz Oligomérica de Cartilagem/farmacologia , Ciclo Celular/efeitos dos fármacos , DNA/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ligamento Periodontal/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor TIE-2/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Adulto , Angiopoietina-1/genética , Proteína de Matriz Oligomérica de Cartilagem/genética , Células Cultivadas , Humanos , Masculino , Proteínas Recombinantes de Fusão/genética
13.
Mol Cell Biochem ; 419(1-2): 157-63, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27431005

RESUMO

Nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase-1 (HO-1) signal is known to play important roles in controlling bone homeostasis. This study examined how oxidative stress affects the mineralization of embryonic stem (ES) cells by exposing them to glucose oxidase (GO), which continuously generates H2O2 at low concentrations. The roles of Nrf2/HO-1 and mitogen-activated protein kinases on osteogenesis in GO-exposed ES cells were also investigated. GO treatment at relatively low concentrations did not change the viability of ES cells, whereas it enhanced osteogenic differentiation and mineralization in the cells. GO treatment (1 mU/ml) augmented the induction of runt-related transcription factor 2 (Runx2), Nrf2, and HO-1 in ES cells. GO-mediated acceleration of Runx2 expression and mineralization was inhibited either by Nrf2 knockdown or by treating with 5 µM PD98059, an inhibitor of phospho-extracellular signal-regulated kinase (p-ERK). The GO-stimulated mineralization was also suppressed by treating the cells with reduced glutathione or catalase, but not by superoxide dismutase or N-acetyl-cysteine. Collectively, our results demonstrate that a mild oxidative stress activates Nrf2/HO-1 signaling and an ERK-mediated pathway, and facilitates the mineralization of ES cells with a corresponding increase in Runx2.


Assuntos
Calcificação Fisiológica/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Glucose Oxidase/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Osteogênese/efeitos dos fármacos , Animais , Linhagem Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Estresse Oxidativo/efeitos dos fármacos
14.
Mol Cell Biochem ; 411(1-2): 83-94, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26369531

RESUMO

Human periodontal ligament fibroblasts (hPLFs) are exposed to oxidative stress during periodontal inflammation and dental treatments. It is hypothesized that hydrogen peroxide (H2O2)-mediated oxidative stress decreases survival and osteogenic differentiation of hPLFs, whereas these decreases are prevented by activation of the Wnt pathway. However, there has been a lack of reports that define the exact roles of canonical Wnt/ß-catenin signaling in H2O2-exposed hPLFs. Treatment with H2O2 reduced viability and proliferation in hPLFs in a dose- and time-dependent manner and led to mitochondria-mediated apoptosis. Pretreatment with lithium chloride (LiCl) or Wnt1 inhibited the oxidative damage that occurred in H2O2-exposed hPLFs. However, knockout of ß-catenin or treatment with DKK1 facilitated the H2O2-induced decreases in viability, mitochondrial membrane potential, and Bcl-2 induction. Osteoblastic differentiation of hPLFs was also inhibited by combined treatment with 100 µM H2O2, as evidenced by the decreases in alkaline phosphatase (ALP) activity and mineralization. H2O2-mediated inhibition of osteoblast differentiation in hPLFs was significantly attenuated in the presence of 500 ng/ml Wnt1 or 20 mM LiCl. In particular, H2O2 stimulated the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) at protein and mRNA levels in hPLFs, whereas the induction was almost completely suppressed in the presence of Wnt1 or LiCl. Furthermore, siRNA-mediated silencing of Nrf2 blocked H2O2-induced decreases in ALP activity and mineralization of hPLFs with the concomitant restoration of runt-related transcription factor 2 and osteocalcin mRNA expression and ALP activity. Collectively, these results suggest that activation of the Wnt/ß-catenin pathway improves proliferation and mineralization in H2O2-exposed hPLFs by downregulating Nrf2.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Ligamento Periodontal/efeitos dos fármacos , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Adulto , Fosfatase Alcalina/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Masculino , Ligamento Periodontal/citologia , Ligamento Periodontal/enzimologia , Adulto Jovem , beta Catenina/genética
15.
Molecules ; 21(8)2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27490527

RESUMO

Coumaric acid (CA) is a phenolic acid of the hydroxycinnamic acid family, and it has many biological functions such as anti-oxidant, anti-inflammatory, antidiabetic, anti-ulcer, anti-platelet, anti-cancer activities, etc. In the present study, we planned to analyse the potential molecular function of CA on skeletal muscle and preadipocytes differentiation using PCR and Western blot techniques. First, we analysed the impact of CA on C2C12 skeletal muscle differentiation. It revealed that CA treatment inhibited horse serum-induced skeletal muscle differentiation as evidenced by the decreased expression of early myogenic differentiation markers such as Myogenin and myoD via the AMP activated protein kinase- alpha AMPK-α mediated pathway. Furthermore, the level of lipid accumulation and changes in genes and protein expressions that are associated with lipogenesis and lipolysis were analyzed in 3T3-L1 cells. The Oil Red O staining evidenced that CA treatment inhibited lipid accumulation at the concentration of 0.1 and 0.2 mM. Furthermore, coumaric acid treatment decreased the expression of main transcriptional factors such as CCAAT/enhancer binding protein-alpha (C/EBP-α) and peroxisome proliferator-activated receptor gamma-2 (PPAR-γ2). Subsequently, CA treatment decreased the expression of sterol regulatory element binding protein-1 (SREBP-1), fatty acid synthase (FAS), acetyl CoA carboxylase (ACC) and adiponectin. Finally, we identified conformational changes induced by CA in PPAR-γ2 using computational biology tools. It revealed that CA might downregulate the PPAR-γ2 expression by directly binding with amino acids of PPAR-γ2 by hydrogen at 3.26 distance and hydrophobic interactions at 3.90 contact distances. These data indicated that CA suppressed skeletal muscle and preadipocytes differentiation through downregulation of the main transcriptional factors and their downstream targets.


Assuntos
Adipócitos/citologia , Adipogenia/efeitos dos fármacos , Ácidos Cumáricos/farmacologia , Músculo Esquelético/citologia , PPAR gama/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Simulação por Computador , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Lipogênese/efeitos dos fármacos , Lipólise/efeitos dos fármacos , Camundongos , Modelos Moleculares , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , PPAR gama/química , Propionatos
16.
Biochem Biophys Res Commun ; 459(4): 706-12, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25770424

RESUMO

The TGF-ß/BMP family plays an important role in multiple stages of tooth development. TGF-ß/BMP signaling is required for odontoblast differentiation and dentin formation; however, the precise molecular mechanisms underlying dentin formation remain unclear. To address the role of TGF-ß/BMP signaling in dentin formation, we analyzed mice in which Smad4, a key intracellular mediator of TGF-ß/BMP signaling, was subjected to tissue-specific ablation under the control of Dspp, OC, or Col1a1 promoters. Three independent Smad4 conditional knockout mice exhibited various dentin defects in the crowns and roots of their molars depending on the transactivator. In all mutant molars, crown dentin thickness was thinner than that of the control. In addition, impaired dentin was found in the cervical region and root furcation area. Although the initial differentiation of odontoblasts was normal, odontoblast polarity abruptly decreased and the expression of Col1a1, OC, and Dspp was reduced in the odontoblasts of mutant molars. In Dspp-Cre-mediated Smad4 disruption mice, primary dentin formation was slightly delayed, while secondary dentin formation was severely affected in the cervical region of the molars. These results indicate that TGF-ß/BMP signaling is required for odontoblast maturation and dentin formation in a stage- and site-dependent manner.


Assuntos
Dentina/crescimento & desenvolvimento , Odontoblastos/metabolismo , Proteína Smad4/fisiologia , Animais , Camundongos
17.
Mol Cell Biochem ; 402(1-2): 213-23, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25618247

RESUMO

Canonical Wnt signaling is thought to enhance osteogenic differentiation of human periodontal ligament fibroblasts (hPLFs). However, the mechanism of this enhancement has not yet been defined. We investigated the effects of Wnt1 on osteoblast differentiation of hPLFs and explored the mechanisms of the effects. Treating hPLFs with Wnt1 induced cytosolic accumulation and nuclear translocation of ß-catenin with concomitant increases in alkaline phosphatase (ALP) activity and calcium content in a time-dependent and dose-dependent manner. Wnt1-stimulated differentiation of hPLFs was accompanied by augmented phosphorylation of glycogen synthase kinase (GSK)-3ß and expression of the bone-specific factors runt-related transcription factor 2 (Runx2), osterix2 (Osx2), ALP, type I collagen, osteopontin, and osteocalcin. Pretreatment with Dickkopf-1 inhibited Wnt1-stimulated differentiation of hPLFs by suppressing GSK-3ß phosphorylation, nuclear translocation of ß-catenin, and expression of the bone-specific factors. Small interfering (si) RNA-mediated knockdown of ß-catenin, or pretreatment with FH535, markedly prevented Wnt1-stimulated differentiation of cells by blocking Runx2 and its downstream factors at the mRNA and protein levels. siRNA-mediated silencing of Runx2 also inhibited Wnt1-stimulated mineralization of cells, accompanied by a reduction in the levels of Osx2 and other early and late bone-formation regulatory factors. However, Wnt1-mediated nuclear translocation of ß-catenin and GSK-3ß phosphorylation were not inhibited by knockdown of Runx2 or FH535. Collectively, our findings suggested that Wnt1 stimulates osteogenic differentiation and mineralization of hPLFs, mainly by activating the canonical Wnt/ß-catenin pathway, in which Runx2 is a key downstream regulator.


Assuntos
Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Fibroblastos/fisiologia , Osteoblastos/fisiologia , Ligamento Periodontal/citologia , Proteína Wnt1/fisiologia , Adulto , Fosfatase Alcalina/metabolismo , Calcificação Fisiológica , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Via de Sinalização Wnt , Adulto Jovem , beta Catenina/genética , beta Catenina/metabolismo
18.
Mol Cell Biochem ; 410(1-2): 255-66, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26346162

RESUMO

Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) regulates the induction of antioxidant gene expression and protects cells against oxidative injury. However, there are controversial findings regarding the roles of Nrf2 on bone metabolism under oxidative stress. The role of Nrf2 on the differentiation of radiation-exposed osteoblasts is also unclear. We investigated whether Nrf2 negatively or positively affects osteoblast differentiation in response to irradiation. Irradiation inhibited osteoblast differentiation of MC3T3-E1 cells in a dose-dependent manner. This inhibition was evidenced by the irradiation-mediated decreases in bone-like nodule formation, alkaline phosphatase (ALP) activity, calcium accumulation, and expression of osteoblast markers, such as ALP, osteocalcin, osteopontin, bone sialoprotein, osterix, and Runx2. These reductions were accompanied by increased induction of Nrf2 and heme oxygenase-1 (HO-1), accumulation of cellular oxidants, and depletion of antioxidant defense enzymes. siRNA-mediated silencing of Nrf2 markedly reversed the negative effect of irradiation on osteoblast differentiation of the cells, leading to a decrease in HO-1 and an increase in Runx2 levels. Irradiation-mediated decreases in the levels of Runx2 and osteocalcin mRNA, but not of Nrf2 protein, were also significantly inhibited by HO-1 inhibitor, zinc protoporphyrin IX. Furthermore, N-acetyl cysteine restored all of the changes induced by irradiation to near-normal levels in the cells. These results demonstrate that irradiation inhibits osteoblast differentiation and mineralization of MC3T3-E1 cells through the oxidative stress-mediated activation of Nrf2/HO-1 pathway.


Assuntos
Calcificação Fisiológica/efeitos da radiação , Diferenciação Celular/efeitos da radiação , Heme Oxigenase-1/biossíntese , Proteínas de Membrana/biossíntese , Fator 2 Relacionado a NF-E2/metabolismo , Osteoblastos/efeitos da radiação , Osteogênese/efeitos da radiação , Células 3T3 , Animais , Antioxidantes/farmacologia , Biomarcadores/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Relação Dose-Resposta à Radiação , Indução Enzimática , Inibidores Enzimáticos/farmacologia , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/genética , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Fator 2 Relacionado a NF-E2/genética , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , Osteoblastos/patologia , Osteogênese/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos da radiação , Transfecção
19.
J Cell Biochem ; 115(11): 1877-87, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24905050

RESUMO

Because irradiation may cause osteoradionecrosis, antioxidant supplementation is often used to suppress irradiation-mediated injury. This study examined whether a synthetic phenethyl urea compound, (E)-1-(3,4-dihydroxyphenethyl)-3-(3,4-dihydroxystyryl)urea (DPDS-U), prevents irradiation-mediated cellular damage in MC3T3-E1 osteoblastic cells. A relatively high dose of irradiation (>4 Gy) decreased cell viability and proliferation and induced DNA damage and cell cycle arrest at the G(2)/M phase with the attendant increase of cyclin B1. Irradiation with 8 Gy induced intracellular reactive oxygen species (ROS) production and lipid peroxidation, and reduced glutathione content and superoxide dismutase activity in the cells. These events were significantly suppressed by treatment with 200 µM DPDS-U or 5 mM N-acetyl cysteine (NAC). DPDS-U or irradiation alone significantly increased heme oxygenase-1 (HO-1) expression and nuclear factor E2 p45-related factor-2 (Nrf2) nuclear translocation. Interestingly, pretreatment with DPDS-U facilitated irradiation-induced activation of the Nrf2/HO-1 pathway. The potential of DPDS-U to mediate HO-1 induction and protect against irradiation-mediated cellular damage was almost completely attenuated by transient transfection with Nrf2-specific siRNA or treatment with a pharmacological HO-1 inhibitor, zinc protoporphyrin IX. Additional experiments revealed that DPDS-U induced a radioprotective mechanism that differs from that induced by NAC through activation of Nrf2/HO-1 signaling. Collectively, our data suggest that DPDS-U-induced radioprotection is due to its dual function as an antioxidant to remove directly excessive intracellular ROS and as a prooxidant to stimulate intracellular redox-sensitive survival signal.


Assuntos
Acetilcisteína/farmacologia , Antioxidantes/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/efeitos da radiação , Protetores contra Radiação/farmacologia , Ureia/análogos & derivados , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos da radiação , Camundongos , Osteoblastos/citologia , Espécies Reativas de Oxigênio/metabolismo , Estirenos/farmacologia , Ureia/farmacologia
20.
Biochem Biophys Res Commun ; 455(3-4): 371-7, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25446117

RESUMO

Recombinant COMP-Ang1, a chimera of angiopoietin-1 (Ang1) and a short coiled-coil domain of cartilage oligomeric matrix protein (COMP), is under consideration as a therapeutic agent capable of inducing the homing of cells with increased angiogenesis. However, the potentials of COMP-Ang1 to stimulate migration of mesenchymal stem cells (MSCs) and the associated mechanisms are not completely understood. We examined the potential of COMP-Ang1 on bone marrow (BM)-MSCs, human periodontal ligament stem cells (PDLSCs), and calvarial osteoblasts. COMP-Ang1 augmented Tie-2 induction at protein and mRNA levels and increased proliferation and expression of runt-related transcription factor 2 (Runx2), osterix, and CXCR4 in BMMSCs, but not in osteoblasts. The COMP-Ang1-mediated increases were inhibited by Tie-2 knockdown and by treating inhibitors of phosphoinositide 3-kinase (PI3K), LY294002, or p38 mitogen-activated protein kinase (MAPK), SB203580. Phosphorylation of p38 MAPK and Akt was prevented by siRNA-mediated silencing of Tie-2. COMP-Ang1 also induced in vitro migration of BMMSCs and PDLSCs. The induced migration was suppressed by Tie-2 knockdown and by CXCR4-specific peptide antagonist or LY294002, but not by SB203580. Furthermore, COMP-Ang1 stimulated the migration of PDLSCs into calvarial defect site of rats. Collectively, our results demonstrate that COMP-Ang1-stimulated proliferation, differentiation, and migration of progenitor cells may involve the Tie-2-mediated activation of p38 MAPK and PI3K/Akt pathways.


Assuntos
Angiopoietina-1/metabolismo , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Receptor TIE-2/metabolismo , Transdução de Sinais , Adolescente , Adulto , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Inibidores Enzimáticos/química , Fêmur/patologia , Inativação Gênica , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Tíbia/patologia , Adulto Jovem , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA