Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 468
Filtrar
1.
Med Sci Monit ; 24: 4121-4127, 2018 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-29908129

RESUMO

BACKGROUND The role of miR-181a in the development of cardiac disease and in particular, myocardial fibrosis following myocardial infarction (MI) remains unknown. The aim of this study was to explore the role of miR-181a in myocardial fibrosis in a rat model of MI and the expression of TGF-ß receptor III (TßRIII). MATERIAL AND METHODS Forty adult male Wistar rats were randomly divided into an MI model group (n=30) and a control group with (n=10). The rat MI model involved ligating the left anterior descending (LAD) coronary artery in the model group; the control group was treated with a sham operation. Cardiac function was assessed using cardiac ultrasound. Myocardial fibroblasts were extracted from the rat hearts and transfected with a miR-mimic or miR-inhibitor, and cell growth was measured using an MTT assay. The level of miR-181a expression was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blots. RESULTS miR-181a expression was significantly increased during the progression of MI (P<0.05). Over-expression of miR-181a was associated with increased deposition of extracellular matrix (ECM) components, collagen I and fibronectin. This effect was reversed with the use of a miR-181a inhibitor (P<0.05). Upregulation of miR-181a suppressed the expression of TGF-ß receptor III (TßRIII) by binding with 3'-UTR. CONCLUSIONS In this rat model of MI, the findings were that miR-181a had a role in the progression of myocardial fibrosis. The findings require further studies to determine whether miR-181a might provide a novel therapeutic target to limit myocardial fibrosis following MI.


Assuntos
Fibrose Endomiocárdica/genética , MicroRNAs/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Animais , Proliferação de Células/fisiologia , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Fibrose Endomiocárdica/metabolismo , Fibrose Endomiocárdica/patologia , Masculino , MicroRNAs/biossíntese , MicroRNAs/genética , Infarto do Miocárdio/metabolismo , Proteoglicanas/biossíntese , Proteoglicanas/genética , Ratos , Ratos Wistar , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/genética
2.
J Enzyme Inhib Med Chem ; 33(1): 920-935, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29768059

RESUMO

Pentabromopseudilin (PBrP) is a marine antibiotic isolated from the marine bacteria Pseudomonas bromoutilis and Alteromonas luteoviolaceus. PBrP exhibits antimicrobial, anti-tumour, and phytotoxic activities. In mammalian cells, PBrP is known to act as a reversible and allosteric inhibitor of myosin Va (MyoVa). In this study, we report that PBrP is a potent inhibitor of transforming growth factor-ß (TGF-ß) activity. PBrP inhibits TGF-ß-stimulated Smad2/3 phosphorylation, plasminogen activator inhibitor-1 (PAI-1) protein production and blocks TGF-ß-induced epithelial-mesenchymal transition in epithelial cells. PBrP inhibits TGF-ß signalling by reducing the cell-surface expression of type II TGF-ß receptor (TßRII) and promotes receptor degradation. Gene silencing approaches suggest that MyoVa plays a crucial role in PBrP-induced TßRII turnover and the subsequent reduction of TGF-ß signalling. Because, TGF-ß signalling is crucial in the regulation of diverse pathophysiological processes such as tissue fibrosis and cancer development, PBrP should be further explored for its therapeutic role in treating fibrotic diseases and cancer.


Assuntos
Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Miosina Tipo V/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirróis/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/antagonistas & inibidores , Alteromonas/química , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Células HEK293 , Células Hep G2 , Humanos , Vison , Estrutura Molecular , Miosina Tipo V/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Pseudomonas/química , Pirróis/química , Pirróis/isolamento & purificação , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Relação Estrutura-Atividade , Fator de Crescimento Transformador beta/metabolismo
3.
Am J Med Sci ; 355(2): 183-190, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29406047

RESUMO

BACKGROUND: Although excess iron induces oxidative stress in the liver, it is unclear whether it directly activates the hepatic stellate cells (HSC). MATERIALS AND METHODS: We evaluated the effects of excess iron on fibrogenesis and transforming growth factor beta (TGF-ß) signaling in murine HSC. Cells were treated with holotransferrin (0.005-5g/L) for 24 hours, with or without the iron chelator deferoxamine (10µM). Gene expressions (α-SMA, Col1-α1, Serpine-1, TGF-ß, Hif1-α, Tfrc and Slc40a1) were analyzed by quantitative real time-polymerase chain reaction, whereas TfR1, ferroportin, ferritin, vimentin, collagen, TGF-ß RII and phospho-Smad2 proteins were evaluated by immunofluorescence, Western blot and enzyme-linked immunosorbent assay. RESULTS: HSC expressed the iron-uptake protein transferrin receptor 1 (TfR1) and the iron-export protein ferroportin. Holotransferrin upregulated TfR1 expression by 1.8-fold (P < 0.03) and ferritin accumulation (iron storage) by 2-fold (P < 0.01), and activated HSC with 2-fold elevations (P < 0.03) in α-SMA messenger RNA and collagen secretion, and a 1.6-fold increase (P < 0.01) in vimentin protein. Moreover, holotransferrin activated the TGF-ß pathway with TGF-ß messenger RNA elevated 1.6-fold (P = 0.05), and protein levels of TGF-ß RII and phospho-Smad2 increased by 1.8-fold (P < 0.01) and 1.6-fold (P < 0.01), respectively. In contrast, iron chelation decreased ferritin levels by 30% (P < 0.03), inhibited collagen secretion by 60% (P < 0.01), repressed fibrogenic genes α-SMA (0.2-fold; P < 0.05) and TGF-ß (0.4-fold; P < 0.01) and reduced levels of TGF-ß RII and phospho-Smad2 proteins. CONCLUSIONS: HSC express iron-transport proteins. Holotransferrin (iron) activates HSC fibrogenesis and the TGF-ß pathway, whereas iron depletion by chelation reverses this, suggesting that this could be a useful adjunct therapy for patients with fibrosis. Further studies in primary human HSC and animal models are necessary to confirm this.


Assuntos
Regulação da Expressão Gênica , Células Estreladas do Fígado/metabolismo , Ferro/metabolismo , Cirrose Hepática/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Ferritinas/biossíntese , Células Estreladas do Fígado/patologia , Cirrose Hepática/patologia , Camundongos , Proteínas Serina-Treonina Quinases/biossíntese , RNA Mensageiro/biossíntese , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores da Transferrina/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Proteína Smad2/metabolismo
4.
Cardiovasc Pathol ; 32: 44-49, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29198452

RESUMO

BACKGROUND: Marfan syndrome (MFS), a congenital connective tissue disorder leading to aortic aneurysm development, is caused by fibrillin-1 (FBN1) gene mutations. Transforming growth factor beta (TGF-ß) might play a role in the pathogenesis. It is still a matter of discussion if and how TGF-ß up-regulates the intracellular downstream pathway, although TGF-ß receptor 3 (TGFBR3 or Betaglycan) is thought to be involved. We aimed to elucidate the role of TGFBR3 protein in TGF-ß signaling in Marfan patients. METHODS: Dermal fibroblasts of MFS patients with haploinsufficient (HI; n=9) or dominant negative (DN; n=4) FBN1 gene mutations, leading to insufficient or malfunctioning fibrillin-1, respectively, were used. Control cells (n=10) were from healthy volunteers. We quantified TGFBR3 protein expression by immunofluorescence microscopy and gene expression of FBN1, TGFB1, its receptors, and downstream transcriptional target genes by quantitative polymerase chain reaction. RESULTS: Betaglycan protein expression in FBN1 mutants pooled was higher than in controls (P=.004) and in DN higher than in HI (P=.015). In DN, significantly higher mRNA expression of FBN1 (P=.014), SMAD7 (P=.019), HSP47 (P=.023), and SERPINE1 (P=.008), but a lower HSPA5 expression (P=.029), was observed than in HI. A pattern of higher expression was noted for TGFB1 (P=.059), FN1 (P=.089), and COL1A1 (P=.089) in DN as compared to HI. TGFBR3 protein expression in cells, both presence in the endoplasmic reticulum and amount of vesicles per cell, correlated positively with TGFB1 mRNA expression (Rs=0.60, P=.017; Rs=0.55, P=.029; respectively). TGFBR3 gene expression did not differ between groups. CONCLUSION: We demonstrated that activation of TGF-ß signaling is higher in patients with a DN than an HI FBN1 gene mutation. Also, TGFBR3 protein expression is increased in the DN group and correlates positively with TGFB1 expression in groups pooled. We suggest that TGFBR3 protein expression is involved in up-regulated TGF-ß signaling in MFS patients with a DN FBN1 gene mutation.


Assuntos
Fibroblastos/metabolismo , Síndrome de Marfan/metabolismo , Proteoglicanas/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Adolescente , Adulto , Células Cultivadas , Criança , Chaperona BiP do Retículo Endoplasmático , Feminino , Humanos , Masculino , Síndrome de Marfan/patologia , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia , Regulação para Cima , Adulto Jovem
5.
Pediatr Hematol Oncol ; 34(4): 238-253, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-29065267

RESUMO

Fascin plays a role in tumor metastasis under the influence of TGF-ß, each potentiating the effect of the other. We retrospectively investigated whether there was a prognostic relationship between TGF-ß and fascin, and disease stage, local recurrence, metastasis tendency, and response to treatment. Twelve neuroblastomas, 17 osteosarcomas, 14 Ewing's sarcomas, 15 rhabdomyosarcoma cases, and 8 rare solid tumors were included. Serum TGF-ß levels were high at the time of diagnosis in all groups (p = .015) and decreased significantly during remission (p = .008). Serum TGF-ß values in the relapse period rarely reached high levels at the time of diagnosis and even stayed under the control group values (p = .017). When TGF-ß receptor expression in tumor tissues was evaluated, the association of TGF-ß receptor positivity with metastatic disease and advanced stage was striking. We found that 88% of rhabdomyosarcoma cases with alveolar histopathology expressed the TGF-ß receptor, and the association between TGF-ß receptor positivity and alveolar histopathology seemed to be a negative prognostic marker. When fascin levels were evaluated in childhood solid tumor tissue, the risk of relapse increased when the fascin total score at diagnosis was >4. This is one of the few studies including prognostic markers such as serum TGF-ß, tissue TGF-ß, TGF-ß receptor, and fascin in pediatric solid tumors. Considering the poor prognosis of advanced stage pediatric solid tumors and the need for biomarkers to predict which patient might need more intensive therapy or warrant closer follow-up afterward, we think that TGF-ß, TGF-ß receptor, and fascin expression have an important prognostic role.


Assuntos
Proteínas de Transporte/biossíntese , Proteínas dos Microfilamentos/biossíntese , Proteínas de Neoplasias/biossíntese , Neoplasias , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Adolescente , Criança , Pré-Escolar , Intervalo Livre de Doença , Feminino , Humanos , Lactente , Masculino , Neoplasias/metabolismo , Neoplasias/mortalidade , Neoplasias/patologia , Taxa de Sobrevida
6.
Life Sci ; 187: 22-30, 2017 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-28823564

RESUMO

AIM: To understand the contribution of miR-142-3p in the activation of hepatic stellate cells (HSCs) and liver fibrosis, and the underlying mechanism. MATERIALS AND METHODS: We detected microRNAs expression profiles in quiescent and activated HSCs by microRNA-array, and performed qRT-PCR to validate these data in HSCs and plasma of cirrhosis patients. In vitro, the 3rd-5th passage HSCs was transfected with mir-142-3p mimics or stimulated with TGF ß. The markers of HSCs activation (i.e. FN and α-SMA) were examined by qRT-PCR and western blotting, and cell viability was detected by MTT, colony formation assays respectively. KEY FINDING: In our study, we identified miR-142-3p as a novel regulator of HSCs activation and indicator of hepatic cirrhosis. We found that miR-142-3p was significantly reduced in activated HSCs, while TGFßRI was distinctly up-regulated in activated HSCs. Ectopic expression of miR-142-3p in activated HSCs inhibited cell viability as well as cell growth, and blocked HSCs activation, concomitant with decreased transdifferentiation markers (i.e. FN and α-SMA). Further, we confirmed that miR-142-3p was reduced upon TGF-ß exposure, while diminishing TGF-ß-Smad signaling pathway in turn by reducing TGFßRI expression in HSCs. Besides, the plasma level of miR-142-3p declined significantly in patients with hepatic cirrhosis. SIGNIFICANCE: In conclusion, we demonstrated that miR-142-3p repressed TGF-ß-Smad signaling pathway to prevent HSCs activation through directly targeting TGFßRI in HSCs.


Assuntos
Células Estreladas do Fígado/metabolismo , MicroRNAs/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Estudos de Casos e Controles , Proliferação de Células/efeitos dos fármacos , Transdiferenciação Celular/efeitos dos fármacos , Humanos , Cirrose Hepática/sangue , Cirrose Hepática/metabolismo , Masculino , MicroRNAs/sangue , MicroRNAs/metabolismo , Cultura Primária de Células , Proteínas Serina-Treonina Quinases/biossíntese , Ratos , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/farmacologia , Regulação para Cima
7.
Curr Eye Res ; 42(7): 1018-1028, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28632033

RESUMO

PURPOSE: To investigate the involvement of activin receptor-like kinase 5 (ALK5) in human Tenon's capsule fibroblasts (HTFs) transdifferentiation and fibrosis. METHODS: (1) Cultured HTFs were treated with transforming growth factor beta 1 (TGF-ß1) at different concentrations for different durations, mRNA expression of ALK5 and plasminogen activator inhibitor-1 (PAI-1) was measured by quantitative polymerase chain reaction (PCR) while protein expression of ALK5, α-smooth muscle actin (α-SMA), and extracellular matrix deposition including fibronectin (FN) and collagen I (Col1) was assessed by western blot. HTFs with or without TGF-ß1 were also treated with an ALK5 activity inhibitor, SB-431542, and fibrosis-related genes were assessed. (2) HTFs were transduced with ALK5 lentivirus (ALK5-OE group) or empty lentivirus (NC-OE) with or without the treatment of SB-431542. Protein expression of ALK5, α-SMA, FN, and Col1 was evaluated. (3) HTFs in the ALK5-OE group and NC-OE group were subjected to a scratch-wound assay and their migratory activities assessed. RESULTS: (1) TGF-ß1, in a concentration-dependent manner, upregulated ALK5 and PAI-1 expressions in the HTFs, which peaked between 24 and 36 h. These changes were associated with increases in protein levels of FN, Col1, and α-SMA. These TGF-ß1 effects were blocked by the ALK5 inhibitor SB-431542. (2) Similarly, overexpression of ALK5 by lentiviral vector significantly increased protein expression of α-SMA, FN, and Col1. Addition of TGF-ß1 to the ALK5-OE cells did not produce additional expression of any of the marker proteins. The upregulation of extracellular matrix and α-SMA can be reduced by SB-431542. (3) In ALK5-OE group, HTFs migration was significantly increased compared with normal control and TGF-ß1 could still promote ALK5-OE cells migration. CONCLUSIONS: Our findings suggest that ALK5 is an important mediator of HTFs fibrosis. ALK5 is a potential therapeutic target to suppress scar formation after filtration surgery.


Assuntos
Fibroblastos/patologia , Regulação da Expressão Gênica , Glaucoma/genética , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Cápsula de Tenon/patologia , Adulto , Western Blotting , Transdiferenciação Celular , Células Cultivadas , Fibroblastos/metabolismo , Fibrose/genética , Fibrose/metabolismo , Fibrose/patologia , Glaucoma/metabolismo , Glaucoma/patologia , Humanos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases/biossíntese , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Cápsula de Tenon/metabolismo
8.
Bioengineered ; 8(5): 600-612, 2017 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-28281868

RESUMO

Transforming growth factor-ß1 (TGF-ß1) signaling is involved in cell metabolism, growth, differentiation, carcinoma invasion and fibrosis development, which suggests TGF-ß1 can be treated as a therapeutic target extensively. Because TGF-ß1 receptor type α(TGFBR2) is the directed and essential mediator for TGF-ß1 signals, the extracellular domain of TGFBR2 (eTGFBR2), binding partner for TGF-ß1, has been produced in a series of expression systems to inhibit TGF-ß1 signaling. However, eTGFBR2 is unstable with a short half-life predominantly because of enzymatic degradation and kidney clearance. In this study, a fusion protein consisting of human eTGFBR2 fused at the C-terminal of human serum albumin (HSA) was stably and highly expressed in Chinese Hamster Ovary (CHO) cells. The high and stable expression sub-clones with Ig kappa signal peptide were selected by Western blot analysis and used for suspension culture. After fed-batch culture over 8 d, the expression level of HSA-eTGFBR2 reached 180 mg/L. The fusion protein was then purified from culture medium using a 2-step chromatographic procedure that resulted in 39% recovery rate. The TGF-ß1 binding assay revealed that HSA-eTGFBR2 could bind to TGF-ß1 with the affinity constant (KD of 1.42 × 10-8 M) as determined by the ForteBio Octet System. In addition, our data suggested that HSA-eTGFBR2 exhibited a TGF-ß1 neutralizing activity and maintained a long-term activity more than eTGFBR2. It concluded that the overexpressing CHO cell line supplied sufficient recombinant human HSA-eTGFBR2 for further research and other applications.


Assuntos
Engenharia de Proteínas/métodos , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/química , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/química , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Albumina Sérica/biossíntese , Albumina Sérica/química , Animais , Sítios de Ligação , Células CHO , Cricetulus , Melhoramento Genético/métodos , Humanos , Ligação Proteica , Receptor do Fator de Crescimento Transformador beta Tipo II , Proteínas Recombinantes de Fusão/genética , Albumina Sérica/genética
9.
Cancer Sci ; 108(3): 419-426, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28064471

RESUMO

Vasohibin-2 (VASH2) is a homolog of VASH1, an endothelium-derived angiogenesis inhibitor. Vasohibin-2 is mainly expressed in cancer cells, and has been implicated in the progression of cancer by inducing angiogenesis and tumor growth. Although VASH2 has been recently reported to be involved in epithelial-mesenchymal transition (EMT), its precise roles are obscure. The aim of the present study was to clarify the role of VASH2 in the EMT of cancer cells in relation to transforming growth factor-ß (TGF-ß) signaling, which is a major stimulator of EMT. Decreased expression of VASH2 in ovarian cancer cells significantly repressed the expression of TGF-ß type I receptor, namely activin receptor-like kinase 5. Transforming growth factor-ß1-induced phosphorylation of Smad2 and Smad3 was markedly decreased in VASH2 knockdown cells while the expression of Smad2 and Smad3 was unchanged. Accordingly, the responses to TGF-ß1 shown by promoter assay and plasminogen activator inhibitor type 1 expression were significantly attenuated in VASH2 knockdown cells. Furthermore, knockdown of VASH2 in cancer cells abrogated the TGF-ß1-induced reduced expression of epithelial markers including E-cadherin, and the elevated expression of mesenchymal markers including fibronectin, ZEB2, and Snail2, suggesting that endogenous VASH2 is required for TGF-ß1-induced EMT. In accordance with these results, the effects of TGF-ß1 on cell morphology, migration, invasion, and MMP2 expression were also abrogated when VASH2 was knocked down. These results indicate that VASH2 played a significant role in the EMT by modulating the TGF-ß signaling. We propose that VASH2 would be a novel molecular target for the prevention of EMT in cancers.


Assuntos
Proteínas Angiogênicas/genética , Transição Epitelial-Mesenquimal/genética , Neoplasias Ovarianas/patologia , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo , Caderinas/biossíntese , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Fibronectinas/biossíntese , Proteínas de Homeodomínio/biossíntese , Humanos , Metaloproteinase 2 da Matriz/biossíntese , Neovascularização Patológica/genética , Neoplasias Ovarianas/genética , Fosforilação/genética , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Proteínas Repressoras/biossíntese , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fatores de Transcrição da Família Snail/biossíntese , Homeobox 2 de Ligação a E-box com Dedos de Zinco
10.
Phlebology ; 32(5): 334-341, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27329669

RESUMO

Objectives Transforming growth factor-beta1 (TGF-ß1) may participate in local chronic inflammatory processes in varicose veins and in venous wall structure modifications through regulation of matrix metalloproteinases (MMP) and their inhibitors (tissue inhibitor of metalloproteinase (TIMP)). The aim of this study was to analyze the effect of TGF-ß1 in the vein wall, namely on the gene expression of selected MMP, TIMP and TGF-ß1 receptors. Methods Healthy vein samples were harvested from eight subjects who underwent coronary bypass graft surgery with great saphenous vein. Each vein sample was divided into two segments, which were cultivated separately in vitro (one of the segments had TGF-ß1 added) and then submitted to gene expression analysis. Results In the TGF-ß1 supplemented group, there was a general increase in the mean gene expression. Specifically, expression of MMP9, MMP12, TIMP1 and TIMP2 were statistically significant. Conclusion The results of this study demonstrate that the gene expression of MMP9, MMP12, TIMP1 and TIMP2 was influenced by the addition of TGF-ß1. These results may be translated to chronic venous insufficiency framework and suggest involvement of TGF-ß1 in the vein wall pathology.


Assuntos
Regulação da Expressão Gênica , Metaloproteinase 12 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Veia Safena/metabolismo , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Inibidor Tecidual de Metaloproteinase-2/biossíntese , Fator de Crescimento Transformador beta1/biossíntese , Insuficiência Venosa/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença Crônica , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Veia Safena/patologia , Insuficiência Venosa/patologia
11.
J Cell Biochem ; 118(1): 172-181, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27292615

RESUMO

For decades stem cells have proven to be invaluable to the study of tissue development. More recently, mesenchymal stem cells (MSCs) derived from embryonic stem cells (ESCs) (ESC-MSCs) have emerged as a cell source with great potential for the future of biomedical research due to their enhanced proliferative capability compared to adult tissue-derived MSCs and effectiveness of musculoskeletal lineage-specific cell differentiation compared to ESCs. We have previously compared the properties and differentiation potential of ESC-MSCs to bone marrow-derived MSCs. In this study, we evaluated the potential of TGFß1 and BMP7 to induce chondrogenic differentiation of ESC-MSCs compared to that of TGFß1 alone and further investigated the cellular phenotype and intracellular signaling in response to these induction conditions. Our results showed that the expression of cartilage-associated markers in ESC-MSCs induced by the TGFß1 and BMP7 combination was increased compared to induction with TGFß1 alone. The TGFß1 and BMP7 combination upregulated the expression of TGFß receptor and the production of endogenous TGFßs compared to TGFß1 induction. The growth factor combination also increasingly activated both of the TGF and BMP signaling pathways, and inhibition of the signaling pathways led to reduced chondrogenesis of ESC-MSCs. Our findings suggest that by adding BMP7 to TGFß1-supplemented induction medium, ESC-MSC chondrogenesis is upregulated through increased production of endogenous TGFß and activities of TGFß and BMP signaling. J. Cell. Biochem. 118: 172-181, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Proteína Morfogenética Óssea 7/farmacologia , Condrogênese/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Fator de Crescimento Transformador beta1 , Linhagem Celular , Células-Tronco Embrionárias Humanas/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
12.
J Recept Signal Transduct Res ; 37(2): 189-199, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27401208

RESUMO

PURPOSE: The transforming growth factor-beta (TGF-ß) pathway is an important in the initiation and progression of cancer. Due to a strong association between an elevated colorectal cancer risk and increase fecal excretion of cholest-4-en-3-one, we aim to determine the effects of cholest-4-en-3-one on TGF-ß signaling in the mink lung epithelial cells (Mv1Lu) and colorectal cancer cells (HT29) in vitro. METHODS: The inhibitory effects of cholest-4-en-3-one on TGF-ß-induced Smad signaling, cell growth inhibition, and the subcellular localization of TGF-ß receptors were investigated in epithelial cells using a Western blot analysis, luciferase reporter assays, DNA synthesis assay, confocal microscopy, and subcellular fractionation. RESULTS: Cholest-4-en-3-one attenuated TGF-ß signaling in Mv1Lu cells and HT29 cells, as judged by a TGF-ß-specific reporter gene assay of plasminogen activator inhibitor-1 (PAI-1), Smad2/3 phosphorylation and nuclear translocation. We also discovered that cholest-4-en-3-one suppresses TGF-ß responsiveness by increasing lipid raft and/or caveolae accumulation of TGF-ß receptors and facilitating rapid degradation of TGF-ß and thus suppressing TGF-ß-induced signaling. CONCLUSIONS: Our results suggest that cholest-4-en-3-one inhibits TGF-ß signaling may be due, in part to the translocation of TGF-ß receptor from non-lipid raft to lipid raft microdomain in plasma membranes. Our findings also implicate that cholest-4-en-3-one may be further explored for its potential role in colorectal cancer correlate to TGF-ß deficiency.


Assuntos
Neoplasias Colorretais/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta1/genética , Animais , Colestenonas/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Células Epiteliais/patologia , Células HT29 , Humanos , Pulmão/patologia , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/genética , Microdomínios da Membrana/metabolismo , Vison/genética , Fosforilação , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Proteólise/efeitos dos fármacos , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/biossíntese
13.
J Asthma ; 54(5): 467-475, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27905842

RESUMO

OBJECTIVE: This study aimed to elucidate the role of Transforming growth factor (TGF)-ß1 signaling in the proliferation of airway smooth muscle cells (ASMCs). BACKGROUND: TGF-ß1 is an important cytokine in airway remodeling in asthma. However, results of studies focusing on the effect of TGFß1 on proliferation of ASMCs are controversial. METHODS: An allergic model that mimics airway remodeling in chronic asthma was established and primary ASMCs were cultured. Cell proliferation was detected by viable cell counting and Cell Counting Kit (CCK)-8 analysis. Expression and phosphorylation of Smad3, type 1 TGFß receptor (TGFßRI), type 2 TGFß receptor (TGFßRII), extracellular signal-regulated kinase (ERK)-1/2, p38 mitogen-activated protein kinase (MAPK), C-Jun N-terminal kinase (JNK) and AKT were detected by western blot. siRNAs were used to knock down Smad3 and TGFßRII. RESULTS: Smad3 and TGFßRII were up-regulated in primary ASMCs isolated from ovalbumin (OVA)-sensitized mice as compared with ASMCs isolated from unsensitized control mice, which persisted for at least four passages. TGFß1 stimulated proliferation of ASMCs isolated from OVA-sensitized mice, which was inhibited by specific siRNA targeting Smad3 or TGFßRII. However ASMCs from control mice showed no proliferative response to TGFß1. TGFß1-induced proliferation of ASMCs from OVA-sensitized mice was markedly attenuated by PD-98059, a specific ERK1/2 inhibitor. TGFß1 induced ERK1/2 phosphorylation within 15 minute, which was partially blocked by specific inhibitor of Smad3 (SIS3). CONCLUSIONS: ASMCs isolated from OVA-sensitized mice showed hyper-proliferation upon TGFß1 stimulation. This might have been associated with up-regulated Smad3 and TGFßRII and mediated by ERK1/2 downstream to Smad3.


Assuntos
Remodelação das Vias Aéreas/fisiologia , Asma/fisiopatologia , Miócitos de Músculo Liso/patologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/farmacologia , RNA Interferente Pequeno/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Proteína Smad3/biossíntese , Regulação para Cima
14.
Clin Exp Metastasis ; 34(1): 37-49, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27770373

RESUMO

ECM1 overexpression is an independent predictor of poor prognosis in primary breast carcinomas, however the mechanisms by which ECM1 affects tumor progression have not been completely elucidated. ECM1 was silenced in the triple-negative breast cancer cell lines Hs578T and MDAMB231 using siRNA and the cells were evaluated for changes in morphology, migration, invasion and adhesion. Actin cytoskeleton alterations were evaluated by fluorescent staining and levels of activated Rho GTPases by pull down assays. ECM1 downregulation led to significantly diminished cell migration (p = 0.0005 for Hs578T and p = 0.02 for MDAMB231) and cell adhesion (p < 0.001 for Hs578T and p = 0.01 for MDAMB231). Cell invasion (matrigel) was reduced only in the Hs578T cells (p < 0.01). Silencing decreased the expression of the prometastatic molecules S100A4 and TGFßR2 in both cell lines and CD44 in Hs578T cells. ECM1-silenced cells also exhibited alterations in cell shape and showed bundles of F-actin across the cell (stress fibers) whereas NT-siRNA treated cells showed peripheral membrane ruffling. Downregulation of ECM1 was also associated with an increased F/G actin ratio, when compared to the cells transfected with NT siRNA (p < 0.001 for Hs578T and p < 0.00035 for MDAMB231) and a concomitant decline of activated Rho A in the Hs578T cells. Re-expression of S100A4 in ECM1-silenced cells rescued the phenotype in the Hs578T cells but not the MDAMB231 cells. We conclude that ECM1 is a key player in the metastatic process and regulates the actin cytoskeletal architecture of aggressive breast cancer cells at least in part via alterations in S100A4 and Rho A.


Assuntos
Proteínas da Matriz Extracelular/genética , Proteínas Serina-Treonina Quinases/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Proteína A4 de Ligação a Cálcio da Família S100/biossíntese , Neoplasias de Mama Triplo Negativas/genética , Citoesqueleto de Actina/genética , Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Colágeno , Combinação de Medicamentos , Matriz Extracelular/genética , Proteínas da Matriz Extracelular/biossíntese , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Hialuronatos/genética , Laminina , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteínas Serina-Treonina Quinases/genética , Proteoglicanas , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteína A4 de Ligação a Cálcio da Família S100/genética , Neoplasias de Mama Triplo Negativas/patologia , Proteínas rho de Ligação ao GTP/genética
15.
Clin Cancer Res ; 23(3): 804-813, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27756784

RESUMO

PURPOSE: Immunotherapy of high-risk neuroblastoma using the anti-GD2 antibody dinutuximab induces antibody-dependent cell-mediated cytotoxicity (ADCC). Galunisertib, an inhibitor of TGFßR1, was examined for its ability to enhance the efficacy of dinutuximab in combination with human ex vivo activated NK (aNK) cells against neuroblastoma. EXPERIMENTAL DESIGN: TGFB1 and TGFBR1 mRNA expression was determined for 249 primary neuroblastoma tumors by microarray analysis. The ability of galunisertib to inhibit SMAD activity induced by neuroblastoma patient blood and bone marrow plasmas in neuroblastoma cells was tested. The impact of galunisertib on TGFß1-induced inhibition of aNK cytotoxicity and ADCC in vitro and on anti-neuroblastoma activity in NOD-scid gamma (NSG) mice was determined. RESULTS: Neuroblastomas express TGFB1 and TGFBR1 mRNA. Galunisertib suppressed SMAD activation in neuroblastoma cells induced by exogenous TGFß1 or by patient blood and bone marrow plasma, and suppressed SMAD2 phosphorylation in human neuroblastoma cells growing in NSG mice. In NK cells treated in vitro with exogenous TGFß1, galunisertib suppressed SMAD2 phosphorylation and restored the expression of DNAM-1, NKp30, and NKG2D cytotoxicity receptors and the TRAIL death ligand, the release of perforin and granzyme A, and the direct cytotoxicity and ADCC of aNK cells against neuroblastoma cells. Addition of galunisertib to adoptive cell therapy with aNK cells plus dinutuximab reduced tumor growth and increased survival of mice injected with two neuroblastoma cell lines or a patient-derived xenograft. CONCLUSIONS: Galunisertib suppresses activation of SMAD2 in neuroblastomas and aNK cells, restores NK cytotoxic mechanisms, and increases the efficacy of dinutuximab with aNK cells against neuroblastoma tumors. Clin Cancer Res; 23(3); 804-13. ©2016 AACRSee related commentary by Zenarruzabeitia et al., p. 615.


Assuntos
Anticorpos Monoclonais/farmacologia , Células Matadoras Naturais/transplante , Proteínas de Neoplasias/antagonistas & inibidores , Neuroblastoma/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirazóis/farmacologia , Quinolinas/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Fator de Crescimento Transformador beta1/fisiologia , Animais , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Sinergismo Farmacológico , Feminino , Perfilação da Expressão Gênica , Humanos , Imunoterapia Adotiva , Masculino , Camundongos , Camundongos Endogâmicos NOD , Proteínas de Neoplasias/fisiologia , Neuroblastoma/metabolismo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Proteína Smad2/antagonistas & inibidores , Proteína Smad2/metabolismo , Organismos Livres de Patógenos Específicos , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/genética , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Mol Cell Biochem ; 424(1-2): 99-110, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27761847

RESUMO

Fibrosis is the dramatic consequence of a dysregulated reparative process in which activated fibroblasts (myofibroblasts) and Transforming Growth Factor ß1 (TGFß1) play a central role. When exposed to TGFß1, fibroblast and epithelial cells differentiate in myofibroblasts; in addition, endothelial cells may undergo endothelial-to-mesenchymal transition (EndoMT) and actively participate to the progression of fibrosis. Recently, the role of αv integrins, which recognize the Arg-Gly-Asp (RGD) tripeptide, in the release and signal transduction activation of TGFß1 became evident. In this study, we present a class of triazole-derived RGD antagonists that interact with αvß3 integrin. Above different compounds, the RGD-2 specifically interferes with integrin-dependent TGFß1 EndoMT in Endothelial Colony-Forming Cells (ECPCs) derived from circulating Endothelial Precursor Cells (ECPCs). The RGD-2 decreases the amount of membrane-associated TGFß1, and reduces both ALK5/TGFß1 type I receptor expression and Smad2 phosphorylation in ECPCs. We found that RGD-2 antagonist reverts EndoMT, reducing α-smooth muscle actin (α-SMA) and vimentin expression in differentiated ECPCs. Our results outline the critical role of integrin in fibrosis progression and account for the opportunity of using integrins as target for anti-fibrotic therapeutic treatment.


Assuntos
Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal , Oligopeptídeos/antagonistas & inibidores , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Células Endoteliais/citologia , Humanos , Integrina alfaVbeta3/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Proteína Smad2/biossíntese , Células-Tronco/citologia , Triazóis/química
17.
Sci Rep ; 6: 34504, 2016 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-27703260

RESUMO

Deregulated expression of microRNAs has been associated with angiogenesis. Studying the miRNome of locally advanced breast tumors we unsuspectedly found a dramatically repression of miR-204, a small non-coding RNA with no previous involvement in tumor angiogenesis. Downregulation of miR-204 was confirmed in an independent cohort of patients and breast cancer cell lines. Gain-of-function analysis indicates that ectopic expression of miR-204 impairs cell proliferation, anchorage-independent growth, migration, invasion, and the formation of 3D capillary networks in vitro. Likewise, in vivo vascularization and angiogenesis were suppressed by miR-204 in a nu/nu mice model. Genome-wide profiling of MDA-MB-231 cells expressing miR-204 revealed changes in the expression of hundred cancer-related genes. Of these, we focused on the study of pro-angiogenic ANGPT1 and TGFßR2. Functional analysis using luciferase reporter and rescue assays confirmed that ANGPT1 and TGFßR2 are novel effectors downstream of miR-204. Accordingly, an inverse correlation between miR-204 and ANGPT1/TGFßR2 expression was found in breast tumors. Knockdown of TGFßR2, but not ANGPT1, impairs cell proliferation and migration whereas inhibition of both genes inhibits angiogenesis. Taken altogether, our findings reveal a novel role for miR-204/ANGPT1/TGFßR2 axis in tumor angiogenesis. We propose that therapeutic manipulation of miR-204 levels may represent a promising approach in breast cancer.


Assuntos
Angiopoietina-1/biossíntese , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , MicroRNAs/biossíntese , Proteínas de Neoplasias/biossíntese , Neovascularização Patológica/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , RNA Neoplásico/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Angiopoietina-1/genética , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Feminino , Humanos , Células MCF-7 , MicroRNAs/genética , Proteínas de Neoplasias/genética , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , Proteínas Serina-Treonina Quinases/genética , RNA Neoplásico/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética
18.
J Assist Reprod Genet ; 33(12): 1665-1675, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27638727

RESUMO

PURPOSE: The main goals of this study were to investigate the expression of anti-Müllerian hormone (AMH) and its receptor (AMHR2) during follicular development in primates, and to evaluate the potential of AMH as a biomarker for follicle growth and oocyte maturation in vitro. METHODS: The mRNA and protein expression of AMH and AMHR2 were determined using isolated follicles and ovarian sections from rhesus macaques (n = 4) by real-time PCR and immunohistochemistry, respectively. Isolated secondary follicles were cultured individually. Follicle growth and media AMH concentrations were assessed by ELISA. The mRNA expression profiles, obtained from RNA sequencing, of in vitro- and in vivo-developed antral follicles were compared. Secondary follicles from additional animals (n = 35) were cultured. Follicle growth, oocyte maturation, and media AMH concentrations were evaluated for forecasting follicular development in vitro by AMH levels. RESULTS: AMH immunostaining was heterogeneous in the population of preantral follicles that were also stained for AMHR2. The mRNA expression profiles were comparable between in vivo- and in vitro-developed follicles. AMH levels produced by growing follicles were higher than those of nongrowing follicles in culture. With a cutoff value of 1.40 ng/ml, 85 % of nongrowing follicles could be identified while eliminating only 5 % of growing follicles. Growing follicles that generated metaphase II-stage oocytes secreted greater amounts of AMH than did those yielding immature germinal vesicle-stage oocytes. CONCLUSIONS: AMH, co-expressed with AMHR2, was produced heterogeneously by preantral follicles in macaques with levels correlated positively with follicle growth and oocyte maturation. AMH may serve as a biomarker for primate follicular development in vitro.


Assuntos
Hormônio Antimülleriano/biossíntese , Técnicas de Maturação in Vitro de Oócitos , Folículo Ovariano/metabolismo , Receptores de Peptídeos/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Animais , Hormônio Antimülleriano/genética , Biomarcadores/metabolismo , Estradiol/metabolismo , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/genética , Humanos , Macaca mulatta , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Oogênese/genética , Folículo Ovariano/crescimento & desenvolvimento , Progesterona/metabolismo , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética
19.
Gene ; 595(1): 1-8, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-27637515

RESUMO

Transforming growth factor beta (TGF-ß) is an important cytokine with pleiotropic regulatory functions in the immune system and in the responses against viral infections. TGF-ß acts on a variety of immune cells through the cell surface TGF-ß receptor (University of Duisburg-EssenTGFBR). The woodchuck has been used as a biomedical model for studies of obesity and energy balance, endocrine and metabolic function, cardiovascular, cerebrovascular and neoplastic disease. Woodchucks infected with woodchuck hepatitis virus (WHV) represent an informative animal model to study hepatitis B virus (HBV) infection. In this study, the cDNA sequences of woodchuck TGF-ß1, TGF-ß2, TGFBR1 and TGFBR2 were cloned, sequenced and characterized. The full-length TGFBR1 cDNA sequence consisted of 1305bp coding sequence (CDS) that encoded 434 amino acids with a molecular weight of 48.9kDa. The phylogenetic tree analysis revealed that the woodchuck TGF-ß family genes had a closer genetic relationship with Ictidomys tridecemlineatus. One antibody with cross-reactivity to woodchuck TGFBR1 was identified by flow cytometry. Moreover, the expression of these genes were analyzed at the transcriptional level. The quantitative PCR analysis showed that the TGF-ß family transcripts were constitutively expressed in many tissues tested. Altered expression levels of the TGF-ß family transcripts in the liver of WHV infected woodchucks were observed. These results serve as a foundation for further insight into the role of the TGF-ß family in viral hepatitis in woodchuck model. Our work also possesses the potential value for characterizing the TGF-ß family in other related diseases, such as obesity-related diseases, metabolic disorder, cardiovascular disease and cancer.


Assuntos
Clonagem Molecular , Regulação da Expressão Gênica/fisiologia , Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta2 , Animais , Marmota/genética , Marmota/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta2/biossíntese , Fator de Crescimento Transformador beta2/genética
20.
Tissue Cell ; 48(4): 305-11, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27344285

RESUMO

BACKGROUND: Umbilical cord blood (UCB) is an important source of hematopoietic stem cells (HSCs). However, low number of HSCs in UCB has been an obstacle for adult hematopoietic stem cell transplantation. The expansion of HSCs in culture is one approach to overcome this problem. In this study, we investigated the expansion of UCB-HSCs by using human bone marrow mesenchymal stromal cells (MSCs) as feeder layer as well as inhibiting the TGF-ß signaling pathway through reduction of TGF-ßRII expression. MATERIALS AND METHODS: CD34(+) cells were isolated from UCB and transfected by SiRNA targeting TGF-ßRII mRNA. CD34(+) cells were expanded in four culture media with different conditions, including 1) expansion of CD34(+) cells in serum free medium containing growth factors, 2) expansion of cells transfected with SiRNA targeting TGF-ßRII in medium containing growth factors, 3) expansion of cells in presence of growth factors and MSCs, 4) expansion of cells transfected with SiRNA targeting TGF-ßRII on MSCs feeder layer in medium containing growth factors. These culture conditions were evaluated for the number of total nucleated cells (TNCs), CD34 surface marker as well as using CFU assay on 8th day after culture. RESULTS: The fold increase in CD34(+) cells, TNCs, and colony numbers (71.8±6.9, 93.2±10.2 and 128±10, respectively) was observed to be highest in fourth culture medium compared to other culture conditions. The difference between number of cells in four culture media in 8th day compared to unexpanded cells (0day) before expansion was statistically significant (P<0.05). CONCLUSION: The results showed that transfection of CD34(+) cells with SiRNA targeting TGF-ßRII and their co-culture with MSCs could considerably increase the number of progenitors. Therefore, this method could be useful for UCB-HSCs expansion.


Assuntos
Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , Proteínas Serina-Treonina Quinases/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Adulto , Antígenos CD34/genética , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular/genética , Proliferação de Células/genética , Técnicas de Cocultura , Células Alimentadoras , Sangue Fetal/metabolismo , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...