Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
J Transl Med ; 20(1): 167, 2022 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-35397552

RESUMO

BACKGROUND: Calcific aortic valve disease (CAVD) is the most commonly valvular disease in the western countries initiated by inflammation and abnormal calcium deposition. Currently, there is no clinical drug for CAVD. Neutrophil elastase (NE) plays a causal role in inflammation and participates actively in cardiovascular diseases. However, the effect of NE on valve calcification remains unclear. So we next explore whether it is involved in valve calcification and the molecular mechanisms involved. METHODS: NE expression and activity in calcific aortic valve stenosis (CAVD) patients (n = 58) and healthy patients (n = 30) were measured by enzyme-linked immunosorbent assay (ELISA), western blot and immunohistochemistry (IHC). Porcine aortic valve interstitial cells (pVICs) were isolated and used in vitro expriments. The effects of NE on pVICs inflammation, apoptosis and calcification were detected by TUNEL assay, MTT assay, reverse transcription polymerase chain reaction (RT-PCR) and western blot. The effects of NE knockdown and NE activity inhibitor Alvelestat on pVICs inflammation, apoptosis and calcification under osteogenic medium induction were also detected by RT-PCR, western blot, alkaline phosphatase staining and alizarin red staining. Changes of Intracellular signaling pathways after NE treatment were measured by western blot. Apolipoprotein E-/- (APOE-/-) mice were employed in this study to establish the important role of Alvelestat in valve calcification. HE was used to detected the thickness of valve. IHC was used to detected the NE and α-SMA expression in APOE-/- mice. Echocardiography was employed to assess the heat function of APOE-/- mice. RESULTS: The level and activity of NE were evaluated in patients with CAVD and calcified valve tissues. NE promoted inflammation, apoptosis and phenotype transition in pVICs in the presence or absence of osteogenic medium. Under osteogenic medium induction, NE silencing or NE inhibitor Alvelestat both suppressed the osteogenic differentiation of pVICs. Mechanically, NE played its role in promoting osteogenic differentiation of pVICs by activating the NF-κB and AKT signaling pathway. Alvelestat alleviated valve thickening and decreased the expression of NE and α-SMA in western diet-induced APOE-/- mice. Alvelestat also reduced NE activity and partially improved the heart function of APOE-/-mice. CONCLUSIONS: Collectively, NE is highly involved in the pathogenesis of valve calcification. Targeting NE such as Alvelestat may be a potential treatment for CAVD.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica , Animais , Valva Aórtica/patologia , Estenose da Valva Aórtica/patologia , Apolipoproteínas E/metabolismo , Calcinose , Células Cultivadas , Humanos , Inflamação/patologia , Elastase de Leucócito/metabolismo , Camundongos , Osteogênese , Suínos
2.
J Cell Mol Med ; 25(1): 132-146, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33191645

RESUMO

It has been reported that chemokine CX3 CL1 can regulate various tumours by binding to its unique receptor CX3 CR1. However, the effect of CX3 CL1-CX3 CR1 on the lung adenocarcinoma and lung squamous cell carcinoma is still unclear. Here, we showed that CX3 CL1 can further invasion and migration of lung adenocarcinoma A549 and lung squamous cell carcinoma H520. In addition, Western blot and immunofluorescence test indicated CX3 CL1 up-regulated the phosphorylation level of cortactin, which is a marker of cell pseudopodium. Meanwhile, the phosphorylation levels of c-Src and c-Abl, which are closely related to the regulation of cortactin phosphorylation, are elevated. Nevertheless, the src/abl inhibitor bosutinib and mutations of cortactin phosphorylation site could inhibit the promotion effect of CX3 CL1 on invasion and migration of A549 and H520. Moreover, these results of MTT, Hoechst staining and Western blot suggested that CX3 CL1 had no effect on the proliferation and apoptosis of A549 and H520 in vitro. The effects of CX3 CL1 were also verified by the subcutaneous tumour formation in nude mice, which showed that it could promote proliferation and invasion of A549 in vivo. In summary, our results indicated that CX3 CL1 furthered invasion and migration in lung cancer cells partly via activating cortactin, and CX3 CL1 may be a potential molecule in regulating the migration and invasion of lung cancer.


Assuntos
Quimiocina CXCL1/metabolismo , Cortactina/metabolismo , Neoplasias Pulmonares/metabolismo , Fosfotirosina/metabolismo , Animais , Apoptose , Receptor 1 de Quimiocina CX3C/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos Nus , Invasividade Neoplásica , Fosforilação , Proteínas Proto-Oncogênicas c-abl/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/metabolismo
3.
J Mol Cell Cardiol ; 141: 93-104, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32247641

RESUMO

Inflammation is implicated in the pathogenesis of calcific aortic valve disease (CAVD) which is a major contributor to cardiovascular mortality and lacks non-surgical treatment. The progranulin (PGRN) is an important immunomodulatory factor in a variety of inflammatory diseases, including rheumatoid arthritis, osteoarthritis, inflammatory bowel disease and pneumonia. However, its role in calcification of aortic valve remains unknown. We firstly found that PGRN was increased in calcified human aortic valve (AV) tissues. Interestingly, in addition to full-length PGRN (68KD), a much stronger band of approximately 45 KD was also significantly increased. The band of 45 KD (45-GRN), was present in wild type (WT) mouse MEFs and AV but absent in grn-/-MEFs, indicating that it was a specific degradation product derived from PGRN. 45-GRN was upregulated whereas PGRN was reduced in human valve interstitial cells (hVICs) under calcifying conditions which is induced by osteogenic medium (OM). In primary porcine VICs (pVICs), PGRN downregulated TNF-α and α-SMA which was accompanied by downregulation of RUNX2, OPN, OCN, alkaline phosphatase activity and calcium deposition, effects pointing to reduced inflammation, myofibroblastic and osteoblastic transition under calcifying conditions. We overexpressed a mimic of 45-GRN which contains p-G-F-B-A-C in pVICs. However, 45-GRN overexpression promoted OM-induced calcification through activating the Smad1/5/8, NF-κB and AKT signaling pathways. Inhibition of the three signaling pathways suppressed 45-GRN's effect on VICs phenotype transition. 45-GRN promoted TNF-α and expressed converse pathogenic signatures with PGRN during TNF-α stimulation. Collectively, this study provides new insight into the pathogenesis of CAVD, indicating that PGRN is a stratagem in mitigating valve fibrosis/osteoblastic differentiation, and also presenting 45-GRN as a potential target for the treatment of CAVD.


Assuntos
Estenose da Valva Aórtica/metabolismo , Valva Aórtica/patologia , Calcinose/metabolismo , Progranulinas/metabolismo , Animais , Valva Aórtica/efeitos dos fármacos , Valva Aórtica/metabolismo , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Meios de Cultura/farmacologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , NF-kappa B/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Fenótipo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Smad/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
4.
J Cell Mol Med ; 24(2): 1399-1412, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31809000

RESUMO

Liver plays an essential role in regulating lipid metabolism, and chronically disturbed hepatic metabolism may cause obesity and metabolic syndrome, which may lead to non-alcoholic fatty liver disease (NAFLD). Increasing evidence indicates long non-coding RNAs (lncRNAs) play an important role in energy metabolism. Here, we investigated the role of lncRNA H19 in hepatic lipid metabolism and its potential association with NAFLD. We found that H19 was up-regulated in oleic acid-induced steatosis and during the development of high-fat diet (HFD)-induced NAFLD. Exogenous overexpression of H19 in hepatocytes induced lipid accumulation and up-regulated the expression of numerous genes involved in lipid synthesis, storage and breakdown, while silencing endogenous H19 led to a decreased lipid accumulation in hepatocytes. Mechanistically, H19 was shown to promote hepatic steatosis by up-regulating lipogenic transcription factor MLXIPL. Silencing Mlxipl diminished H19-induced lipid accumulation in hepatocytes. Furthermore, H19-induced lipid accumulation was effectively inhibited by PI3K/mTOR inhibitor PF-04691502. Accordingly, H19 overexpression in hepatocytes up-regulated most components of the mTORC1 signalling axis, which were inhibited by silencing endogenous H19. In vivo hepatocyte implantation studies further confirm that H19 promoted hepatic steatosis by up-regulating both mTORC1 signalling axis and MLXIPL transcriptional network. Collectively, these findings strongly suggest that H19 may play an important role in regulating hepatic lipid metabolism and may serve as a potential therapeutic target for NAFLD.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Hepatopatia Gordurosa não Alcoólica/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Modelos Animais de Doenças , Inativação Gênica , Células HEK293 , Humanos , Metabolismo dos Lipídeos/genética , Masculino , Camundongos Endogâmicos C57BL , Ácido Oleico , RNA Longo não Codificante/genética , Triglicerídeos/metabolismo , Regulação para Cima/genética
5.
Ecotoxicol Environ Saf ; 203: 110930, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32684523

RESUMO

Benzo[a]pyrene(BaP), a polycyclic aromatic hydrocarbons (PAH) of environmental pollutants, is one of the main ingredients in cigarettes and an agonist of the aryl hydrocarbon receptor (AhR). Mesenchymal stem cells (MSCs) including C3H10T1/2 and MEF cells, adult multipotent stem cells, can be differentiated toward osteoblasts during the induction of osteogenic induction factor-bone morphogenetic protein 2(BMP2). Accumulating evidence suggests that BaP decreases bone development in mammals, but the further mechanisms of BaP on BMP2-induced bone formation involved are unknown. Here, we researched the role of BaP on BMP2-induced osteoblast differentiation and bone formation. We showed that BaP significantly suppressed early and late osteogenic differentiation, and downregulated the runt-related transcription factor 2(Runx2), osteocalcin(OCN) and osteopontin (OPN) during the induction of BMP2 in MSCs. Consistent with in vitro results, administration of BaP inhibited BMP2-induced subcutaneous ectopic osteogenesis in vivo. Interestingly, blocking AhR reversed the inhibition of BaP on BMP2-induced osteogenic differentiation, which suggested that AhR played an important role in this process. Moreover, BaP significantly decreased BMP2-induced Smad1/5/8 phosphorylation. Furthermore, BaP significantly reduced bone morphogenetic protein receptor 2(BMPRII) expression and excessively activated Hey1. Thus, our data demonstrate the role of BaP in BMP2-induced bone formation and suggest that impaired BMP/Smad pathways through AhR regulating BMPRII and Hey1 may be an underlying mechanism for BaP inhibiting BMP2-induced osteogenic differentiation.


Assuntos
Benzo(a)pireno/toxicidade , Proteína Morfogenética Óssea 2/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Benzo(a)pireno/metabolismo , Linhagem Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células HCT116 , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Nus , Osteoblastos/metabolismo
6.
Cell Physiol Biochem ; 41(2): 484-500, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28214873

RESUMO

BACKGROUND: BMP9 induces both osteogenic and adipogenic differentiation of mesenchymal stem cells (MSCs). Nell1 is a secretory glycoprotein with osteoinductive and anti-adipogenic activities. We investigated the role of Nell1 in BMP9-induced osteogenesis and adipogenesis in MSCs. METHODS: Previously characterized MSCs iMEFs were used. Overexpression of BMP9 and NELL1 or silencing of mouse Nell1 was mediated by adenoviral vectors. Early and late osteogenic and adipogenic markers were assessed by staining techniques and qPCR analysis. In vivo activity was assessed in an ectopic bone formation model of athymic mice. RESULTS: We demonstrate that Nell1 expression was up-regulated by BMP9. Exogenous Nell1 potentiated BMP9-induced late stage osteogenic differentiation while inhibiting the early osteogenic marker. Forced Nell1 expression enhanced BMP9-induced osteogenic regulators/markers and inhibited BMP9-upregulated expression of adipogenic regulators/markers in MSCs. In vivo ectopic bone formation assay showed that exogenous Nell1 expression enhanced mineralization and maturity of BMP9-induced bone formation, while inhibiting BMP9-induced adipogenesis. Conversely, silencing Nell1 expression in BMP9-stimulated MSCs led to forming immature chondroid-like matrix. CONCLUSION: Our findings indicate that Nell1 can be up-regulated by BMP9, which in turn accelerates and augments BMP9-induced osteogenesis. Exogenous Nell1 may be exploited to enhance BMP9-induced bone formation while overcoming BMP9-induced adipogenesis in regenerative medicine.


Assuntos
Adipogenia , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular , Glicoproteínas/metabolismo , Fator 2 de Diferenciação de Crescimento/metabolismo , Osteogênese , Adipogenia/efeitos dos fármacos , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Fator 2 de Diferenciação de Crescimento/genética , Células HEK293 , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Osteogênese/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transplante Homólogo
7.
Int J Mol Sci ; 18(4)2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28420207

RESUMO

The transforming growth factor-ß (TGFß) family signaling pathways play an important role in regulatory cellular networks and exert specific effects on developmental programs during embryo development. However, the function of TGFß signaling pathways on the early kidney development remains unclear. In this work, we aim to detect the underlying role of TGFß type II receptor (TßRII) in vitro, which has a similar expression pattern as the crucial regulator Six2 during early kidney development. Firstly, the 5-ethynyl-2'-deoxyuridine (EdU) assay showed knock down of TßRII significantly decreased the proliferation ratio of metanephric mesenchyme (MM) cells. Additionally, real-time Polymerase Chain Reaction (PCR) and Western blot together with immunofluorescence determined that the mRNA and protein levels of Six2 declined after TßRII knock down. Also, Six2 was observed to be able to partially rescue the proliferation phenotype caused by the depletion of TßRII. Moreover, bioinformatics analysis and luciferase assay indicated Smad3 could transcriptionally target Six2. Further, the EdU assay showed that Smad3 could also rescue the inhibition of proliferation caused by the knock down of TßRII. Taken together, these findings delineate the important function of the TGFß signaling pathway in the early development of kidney and TßRII was shown to be able to promote the expression of Six2 through Smad3 mediating transcriptional regulation and in turn activate the proliferation of MM cells.


Assuntos
Proteínas de Homeodomínio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proliferação de Células , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Humanos , Rim/embriologia , Rim/metabolismo , Proteínas do Tecido Nervoso/genética , Organogênese/genética , Fenótipo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo
8.
Tumour Biol ; 37(3): 3479-88, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26449831

RESUMO

Osteosarcoma (OS) is the most common primary malignant bone tumor in young patients. However, treatment paradigms and survival rates have not improved in decades. MicroRNAs have been shown to be critical regulators of physiological homeostasis and pathological process, including bone disease. Nearly half of the microRNA (miRNA) genes are located at genomic regions and fragile sites known to be frequently deleted or amplified in various kinds of cancers. In this study, we investigated the role miR-30a in OS. A negative correlation between miR-30a expression and malignant grade was observed in OS cell lines. The overexpression of miR-30a reduced proliferation, migration, and invasion in 143B cells and the inhibitor of miR-30a increased proliferation, migration, and invasion in Saos2 cells. Further studies revealed that runt-related transcription factors 2 (Runx2) was a regulative target gene of miR-30a. Rescue assay significantly reversed the effects of overexpressing or inhibiting miR-30a. miR-30a also suppressed tumor formation and pulmonary metastasis in vivo. All the results suggest a critical role of miR-30a in suppressing proliferation, migration, and invasion of OS by targeting Runx2.


Assuntos
Neoplasias Ósseas/patologia , Movimento Celular , Proliferação de Células , Subunidade alfa 1 de Fator de Ligação ao Core/genética , MicroRNAs/fisiologia , Osteossarcoma/patologia , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Invasividade Neoplásica
9.
Hum Mutat ; 35(5): 575-84, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24677579

RESUMO

Mutation of the tumor suppressor TP53 gene occurs in greater than half of all human cancers. In addition to loss of tumor suppressor function of wild-type TP53, gain-of-function mutations endow cancer cells with more malignant properties. R273 is a mutation hotspot with the p.R273H, p.R273C, and p.R273G variants occurring most commonly in patient samples. To better understand the consequences of these R273 mutations, we constructed cancer cell lines expressing TP53 p.R273H, p.R273C, or p.R273G and explored their characteristics. We found that p.R273H and p.R273C, but not p.R273G, enhanced proliferation, invasion, and drug resistance in vitro. Furthermore, breast cancer susceptibility protein 1 was upregulated by mutant TP53 p.R273H and p.R273C in response to DNA damage and repair. Transcriptional analysis of the TP53-R273 mutants by RNA-seq confirmed that the apoptosis pathway was less active in p.R273H and p.R273C, compared with R273G. Molecular dynamics simulation further revealed that TP53-R273G binds more tightly to DNA than TP53-R273H or TP53-R273C. These findings indicate that mutation of TP53 at a single codon has different effects, and likely clinical implications. p.R273H and p.R273C lead to a more aggressive phenotype than p.R273G. These findings may contribute to future diagnosis and therapy in TP53 mutant cancers.


Assuntos
Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos/genética , Invasividade Neoplásica/genética , Proteína Supressora de Tumor p53/genética , Apoptose/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Cisplatino/administração & dosagem , Feminino , Humanos , Simulação de Dinâmica Molecular , Mutação , Conformação Proteica , Proteína Supressora de Tumor p53/química
10.
Mol Biol Rep ; 41(3): 1373-83, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24413988

RESUMO

Bone morphogenetic proteins (BMPs), which belong to the transforming growth factor-ß superfamily, regulate a wide range of cellular responses including cell proliferation, differentiation, adhesion, migration, and apoptosis. BMP9, the latest BMP to be discovered, is reportedly expressed in a variety of human carcinoma cell lines, but the role of BMP9 in breast cancer has not been fully clarified. In a previous study, BMP9 was found to inhibit the growth, migration, and invasiveness of MDA-MB-231 breast cancer cells. In the current study, the effect of BMP9 on the bone metastasis of breast cancer cells was investigated. After absent or low expression of BMP9 was detected in the MDA-MB-231 breast cancer cells and breast non-tumor adjacent tissues using Western blot and immunohistochemistry, In our previous study, BMP9 could inhibit the proliferation and invasiveness of breast cancer cells MDA-MB-231 in vitro and in vivo. This paper shows that BMP9 inhibit the bone metastasis of breast cancer cells by activating the BMP/Smad signaling pathway and downregulating connective tissue growth factor (CTGF); however, when CTGF expression was maintained, the inhibitory effect of BMP9 on the MDA-MB-231 cells was abolished. Together, these observations indicate that BMP9 is an important mediator of breast cancer bone metastasis and a potential therapeutic target for treating this deadly disease.


Assuntos
Apoptose/genética , Neoplasias da Mama/genética , Fator de Crescimento do Tecido Conjuntivo/biossíntese , Fatores de Diferenciação de Crescimento/metabolismo , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Fator de Crescimento do Tecido Conjuntivo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento/genética , Humanos
11.
Int J Med Sci ; 11(10): 1065-72, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25136261

RESUMO

Periodontal ligament stem cells (PDLSCs) with bone morphogenic ability are used to treat diseases such as periodontitis. Their treatment potential is increased when used in combination with proteins that induce osteogenic differentiation. For example, bone morphogenetic protein-9 (BMP9) has been found to have potent osteogenic activity. In the present study, PDLSCs were isolated from human periodontal membrane and infected with recombinant adenoviruses expressing BMP9 (Ad-BMP9). Levels of osteogenic markers such as runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), osteopontin (OPN), and osteocalcin (OCN) as well as mineralization ability were measured. The results showed that BMP9 promoted bone formation of PDLSCs. In other experiments, SB203580 and PD98059, which are inhibitors of p38 and ERK1/2, respectively, were used to determine if these kinases are involved in the osteogenic differentiation process. The resulting protein expression profiles and osteogenic markers of PDLSCs revealed that the mitogen-activated protein kinase (MAPK) signaling pathway might play an important role in the process of BMP9-induced osteogenic differentiation of PDLSCs.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 2 de Diferenciação de Crescimento/metabolismo , Osteogênese/efeitos dos fármacos , Ligamento Periodontal/citologia , Células-Tronco/citologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adolescente , Fosfatase Alcalina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Criança , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Flavonoides/farmacologia , Humanos , Imidazóis/farmacologia , Imuno-Histoquímica , Osteocalcina/metabolismo , Osteopontina/metabolismo , Piridinas/farmacologia , Células-Tronco/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
12.
Int J Med Sci ; 10(9): 1181-92, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935395

RESUMO

Mesenchymal progenitor stem cells (MPCs) are a group of bone marrow stromal progenitor cells processing osteogenic, chondrogenic, adipogenic and myogenic lineages differentiations. Previous studies have demonstrated that bone morphogeneic protein 9(BMP9) is one of the most osteogenic BMPs both in vitro and in vivo, however, the underlying molecular mechanism of osteogenesis induced by BMP9 is needed to be deep explored. Here, we used the recombinant adenoviruses assay to introduce BMP9 into C3H10T1/2 mesenchymal stem cells to elucidate the role of CXCL12/CXCR4 signal axis during BMP9-incuced osteogenic differentiation. The results showed that CXCL12 and CXCR4 expressions were down-regulated at the stage of BMP9-induced osteogenic differentiation, in a dose- and time-dependent. Pretreatment of C3H10T1/2 cells with CXCL12/CXCR4 could significantly affect the early and mid osteogenic markers alkaline phosphatase (ALP), osteocalcin (OCN), the transcription factors of Runx2, Osx, Plzf and Dlx5 expression, through activating the Smad, MAPK signaling pathway. Addition of exogenous CXCL12 did not affect the changes of the late osteogenic marker calcium deposition. Thus, our findings suggest a co-requirement of the CXCL12/CXCR4 signal axis in BMP9-induced the early- and mid-process of osteogenic differentiation of MSCs.


Assuntos
Quimiocina CXCL12/metabolismo , Fator 2 de Diferenciação de Crescimento/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese/fisiologia , Receptores CXCR4/metabolismo , Fosfatase Alcalina/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/farmacologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Fator 2 de Diferenciação de Crescimento/genética , Células HCT116 , Proteínas de Homeodomínio/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Osteocalcina/metabolismo , Osteogênese/genética , Proteína com Dedos de Zinco da Leucemia Promielocítica , Receptores CXCR4/genética , Fator de Transcrição Sp7 , Fatores de Transcrição/metabolismo
13.
Artigo em Chinês | MEDLINE | ID: mdl-24053920

RESUMO

OBJECTIVE: To investigate the protective effect of suppressive oligodeoxynucleotides (Sup ODN) on interferon-γ (IFN-γ) and signal transducers and activators of transcription (pSTAT4) expression of Silica-induced pulmonary inflammation in Mice. METHODS: Sixty Balb/c mice were randomly divided into 4 groups, normal control group, silicious group, suppressive oligodeoxynucleotides (Sup ODN) group, control oligodeoxynucleotides (Con ODN) group. Except the normal control group injected normal saline, the rest groups were induced by the intratracheal instillation of 0.1 ml (5 g/L) of sterilized silica suspension. Sup ODN group and Con ODN group were treated by i.p. injection of 0.3 ml (1mg/mL) of suppressive or control ODN 3 h before silica administration. After 7 days, the animals were killed and levels of IFN-γ were detected by ELISA. The pathologic changes in lung tissues of mice were observed with HE staining. Expressions of IFN-γ and pSTAT4 in lung tissue were detected with immunohistochemistry and quantified by Image-Pro Plus 7.0. RESULTS: HE staining showed that the lung tissue of silicious group were damaged seriously than Sup ODN group. Compared with the normal control group (serum: (280.1±41.3) pg/ml, lung tissue: (0.249±0.373), IFN-γ increased in silicious group (serum: (886.3±81.7) pg/ml, lung tissue: (0.270±0.300) (P < 0.05). Compared with the normal control group and Con ODN group [(894.5±91.6) pg/ml], IFN-γ in the serum of Sup ODN group decreased significantly (P < 0.01). Compared with the silicious group , IFN-γ in lung tissue decreased in Sup ODN group (0.241±0.250) (P < 0.05). Compared with the normal control group (0.279±0.353), pSTAT4 in lung tissue increased significantly in silicious group (0.313±0.231) (P < 0.01). Compared with the silicious group, pSTAT4 in lung tissue decreased significantly in Sup ODN group (0.269±0.523) (P < 0.01). CONCLUSION: Sup ODN attained protective effect on Silica treated mice by suppressing expression of IFN-γ and pSTAT4.


Assuntos
Interferon gama/metabolismo , Pulmão/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Fator de Transcrição STAT4/metabolismo , Dióxido de Silício/toxicidade , Animais , Feminino , Inflamação/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação
14.
Int J Oncol ; 62(4)2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36866758

RESUMO

Subsequently to the publication of the above article, an interested reader drew to the authors' attention that two pairs of data panels in Fig. 7D on p. 1008, showing the results from Transwell invasion assay experiments, contained overlapping sections such that these panels were likely to have been derived from the same original sources where they were intended to show the results from differently performed experiments. After having consulted their original data, the authors were able to identify that two of the data panels in Fig. 7D were inadvertently selected incorrectly; specifically, the 'GST+SB203580' and 'GST­hS100A9+PD98059' panels in this figure. The revised version of Fig. 7, showing the correct data panels for the 'GST+SB203580' and 'GST­hS100A9+PD98059' panels in Fig. 7D, is shown on the next page. The authors confirm that the errors made during the assembly of Fig. 7 did not grossly affect the major conclusions presented in this paper, and are grateful to the Editor of International Journal of Oncology for allowing them this opportunity to publish a Corrigendum. They also apologize to the readership for any inconvenience caused. [International Journal of Oncology 42: 1001-1010, 2013; DOI: 10.3892/ijo.2013.1796].

16.
Artigo em Chinês | MEDLINE | ID: mdl-22804932

RESUMO

OBJECTIVE: To explore the correlation between the exposure levels and serum protein fingerprints in population exposed to silica. METHODS: Liquid chip time-of-flight mass spectrometry technology was used to investigate the serum profiles in control group (30 cases), group exposed to silica (30 cases), silicosis group (I stage, 25 cases) and suspected silicosis group (30 cases), and screen the differential expression proteins. The correlation between the levels of the differential expression proteins and the exposure levels was performed. RESULTS: Five differential expression proteins were found among 4 groups, the expression of 5081 and 5066 proteins was upregulated, and the expression of 3954, 2021 and 1777 proteins was downregulated. There was no the correlation between the exposure levels and the peak with M/Z among those proteins. CONCLUSION: the results of present investigation indicated there was no correlation between the exposure levels and protein/peptide peak.


Assuntos
Proteínas Sanguíneas/análise , Poeira/análise , Exposição Ocupacional/análise , Dióxido de Silício/toxicidade , Silicose/sangue , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Mapeamento de Peptídeos , Proteômica
17.
Vascul Pharmacol ; 144: 106986, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35318164

RESUMO

AIMS: Calcified aortic valve disease (CAVD) is a cardiovascular disease with increasing morbidity and mortality. The pathogenetic cellular mechanism is the phenotypic transition of aortic valve interstitial cells (VICs). Here, we explored the effect of berberine (BBR) on the phenotypic transition of VICs and elucidated the underlying molecular mechanisms, providing a theoretical basis in finding novel clinical treatments for CAVD. METHODS AND RESULTS: Calcific aortic valves and normal controls were collected for western blot and the results demonstrated that osteogenic and inflammatory markers were significantly up-regulated in calcific aortic valves. BBR inhibited inflammation and osteogenic differentiation of VICs under osteogenic conditions, as well as alkaline phosphatase activity and calcified nodule formation. Mechanistically, BBR could inhibit the activation of Smad1/5/8 and NF-κB pathways under OM conditions. LDN193189 and BAY11-7082, the inhibitor of Smad1/5/8 and NF-κB respectively, were added for further verification. Similarly, the osteogenic and fibrotic markers of VICs induced by osteogenic induction medium were decreased by LDN193189 and BAY11-7082. Western blot was used to examine upstream receptors of Smad1/5/8, the results showed that BBR inhibited the activation of Smad1/5/8 by downregulating ALK2 and ALK3. CONCLUSION: BBR decreased the inflammatory factors and suppressed the osteogenic differentiation of VICs, which might be associated with the inhibition of Smad1/5/8 and NF-κB signaling pathways.


Assuntos
Valva Aórtica , Berberina , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Estenose da Valva Aórtica , Berberina/farmacologia , Biomarcadores/metabolismo , Calcinose , Células Cultivadas , NF-kappa B/metabolismo , Osteogênese
18.
Bioeng Transl Med ; 7(3): e10306, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36176604

RESUMO

Effective and safe liver-directed gene therapy has great promise in treating a broad range of liver diseases. While adenoviral (Ad) vectors have been widely used for efficacious in vivo gene delivery, their translational utilities are severely limited due to the short duration of transgene expression and solicitation of host immune response. Used as a promising polymeric vehicle for drug release and nucleic acid delivery, carboxymethyl chitosan (CMC) is biocompatible, biodegradable, anti-microbial, inexpensive, and easy accessible. Here, by exploiting its biocompatibility, controlled release capability and anti-inflammatory activity, we investigated whether CMC can overcome the shortcomings of Ad-mediated gene delivery, hence improving the prospect of Ad applications in gene therapy. We demonstrated that in the presence of optimal concentrations of CMC, Ad-mediated transgene expression lasted up to 50 days after subcutaneous injection, and at least 7 days after intrahepatic injection. Histologic evaluation and immunohistochemical analysis revealed that CMC effectively alleviated Ad-induced host immune response. In our proof-of-principle experiment using the CCl4-induced experimental mouse model of chronic liver damage, we demonstrated that repeated intrahepatic administrations of Ad-IL10 mixed with CMC effectively mitigated the development of hepatic fibrosis. Collectively, these results indicate that CMC can improve the prospect of Ad-mediated gene therapy by diminishing the host immune response while allowing readministration and sustained transgene expression.

19.
Mol Hum Reprod ; 17(1): 14-21, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20643875

RESUMO

During early development of the human embryo, chromosomal imbalance and instability may cause spontaneous miscarriages. In this study, we observe aberrant chromosome numbers in nearly half of spontaneous miscarriage embryo samples, most of which show abnormalities in karotype. We also detect significantly reduced expression of two important mitotic checkpoint proteins, Mad2 and Bub1. To further investigate the role of Bub1 and Mad2 in chromosome mis-segregation, in embryogenesis, and in errors leading to spontaneous miscarriages, we used RNA interference technology to knockdown Bub1 and Mad2 genes. We examined the effect of reduced expression of Mad2 and Bub1 on chromosome number, cell proliferation and cell cycle progression. Significant suppression of cell proliferation and increased abnormal chromosome numbers were detected. M phase arrest was observed in cultured villus cell lines with depleted Mad2 or Bub1 mRNA by RNAi technique. The results from the in vitro RNAi-mediated silencing model may provide an explanation for the observations in clinical samples of spontaneous miscarriages. Thus, our findings strongly suggest that the loss of spindle assembly checkpoint proteins, such as Bub1 and Mad2, may cause spontaneous miscarriages.


Assuntos
Aborto Espontâneo/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Aborto Espontâneo/genética , Western Blotting , Proteínas de Ligação ao Cálcio/genética , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Humanos , Técnicas In Vitro , Cariotipagem , Proteínas Mad2 , Proteínas Serina-Treonina Quinases/genética , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trofoblastos/citologia , Trofoblastos/metabolismo
20.
Pathol Res Pract ; 223: 152824, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34000674

RESUMO

Triple-negative breast cancer (TNBC) is a type of malignant and heterogeneous tumor in premenopausal females with ineffective therapeutic targets. IL-8 is one of the earliest discovered chemotaxis cytokines which expression is closely related to the progress of various cancers. Previous studies show that IL-8 determines the prognosis of TNBC patients, nevertheless how IL-8 influence the progress of TNBC is unclear. In our studies, we discovered that overexpression of IL-8 promotes TNBC cells (TNBCs) migration and tumor growth via the PI3K-Akt and MAPK signaling pathway. Cell-cycle of TNBCs arrest at S phase by overexpression of IL-8, however, there is no significant difference on the cell viability and cell apoptosis of TNBCs. Besides, overexpression of IL-8 result in the downregulation of E-cadherin and the upregulation of Cyclin B1 in MDA-MB-231 cells. Taken together, our results suggest that IL-8 plays a crucial role in the progress of TNBC, and it could be a novel therapeutic target of TNBC.


Assuntos
Transição Epitelial-Mesenquimal , Interleucina-8/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias de Mama Triplo Negativas/enzimologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-8/genética , Camundongos Nus , Invasividade Neoplásica , Pontos de Checagem da Fase S do Ciclo Celular , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Carga Tumoral , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa