Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Int J Mol Sci ; 20(19)2019 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-31569732

RESUMEN

Bronchopulmonary dysplasia (BPD), caused by hyperoxia in newborns and infants, results in lung damage and abnormal pulmonary function. However, the current treatments for BPD are steroidal and pharmacological therapies, which cause neurodevelopmental impairment. Treatment with umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) is an efficient alternative approach. To prevent pulmonary inflammation in BPD, this study investigated the hypothesis that a key regulator was secreted by MSCs to polarize inflammatory macrophages into anti-inflammatory macrophages at inflammation sites. Lipopolysaccharide-induced macrophages co-cultured with MSCs secreted low levels of the inflammatory cytokines, IL-8 and IL-6, but high levels of the anti-inflammatory cytokine, IL-10. Silencing decorin in MSCs suppressed the expression of CD44, which mediates anti-inflammatory activity in macrophages. The effects of MSCs were examined in a rat model of hyperoxic lung damage. Macrophage polarization differed depending on the levels of decorin secreted by MSCs. Moreover, intratracheal injection of decorin-silenced MSCs or MSCs secreting low levels of decorin confirmed impaired alveolarization of damaged lung tissues by down-regulation of decorin. In tissues, a decrease in the anti-inflammatory macrophage marker, CD163, was observed via CD44. Thus, we identified decorin as a key paracrine factor, inducing macrophage polarization via CD44, a master immunoregulator in mesenchymal stem cells.


Asunto(s)
Decorina/biosíntesis , Sangre Fetal/citología , Receptores de Hialuranos/sangre , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Hiperoxia/complicaciones , Lesión Pulmonar/diagnóstico , Lesión Pulmonar/etiología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/terapia , Ratas
2.
Med Oncol ; 36(12): 96, 2019 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-31659495

RESUMEN

Oncolytic adenovirus is an emerging alternative to current therapeutics. The adenoviral E1A, the first protein expressed upon oncolytic adenoviral infection, has been identified as an antitumor agent, but the mechanisms of its tumor inhibition ability are unclear enough. Decorin is ubiquitous in the extracellular matrix (ECM), which regulates multiple functions through interaction with ECM. Here, we intended to explore the effects of adenoviral E1A on the tumor extracellular matrix during gene therapy. We demonstrated that reduced decorin expression was found in patients with lung cancer. The adenoviral E1A or a mutant adenoviral E1A with Rb-binding ability absent (E1A 30-60aa, 120-127aa deletion) could increase the expression of decorin and down-regulate VEGF, two members of tumor ECM, involved in both vasculogenesis and angiogenesis. E1A/mE1A-mediated suppressing the migration and invasion ability of tumor cells was depended on decorin. E1A interacted with decorin directly and induced the proteasomal degradation of VEGF. In addition, E1A or mE1A can inhibit tumor growth in a subcutaneous lung cancer xenograft model. It suggested that decorin might be a crucial mediator among ECM components for adenoviral E1A-mediated antitumor activities. These studies on adenovirus E1A provide a new mechanism for the emerging therapies of tumor gene therapy.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Decorina/metabolismo , Neoplasias Pulmonares/terapia , Viroterapia Oncolítica/métodos , Células A549 , Proteínas E1A de Adenovirus/genética , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Decorina/biosíntesis , Decorina/genética , Femenino , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Lett ; 459: 15-29, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31150821

RESUMEN

Pancreatic cancer is a highly lethal disease. Excessive accumulation of tumor extracellular matrix (ECM) and epithelial-to-mesenchymal transition (EMT) phenotype are two main contributors to drug resistance in desmoplastic pancreatic tumors. To overcome desmoplasia and chemoresistance of pancreatic cancer, we utilized an oncolytic adenovirus (Ad) co-expressing decorin and soluble Wnt decoy receptor (HEmT-DCN/sLRP6). An orthotopic pancreatic xenograft tumor model was established in athymic nude mice using Mia PaCa-2 cells, and the antimetastatic and antitumor efficacy of systemically administered HEmT-DCN/sLRP6 was evaluated. Immunohistochemical analysis of tumor tissues was performed to assess ECM degradation, induction of apoptosis, viral dispersion, and inhibition of the Wnt/ß-catenin signaling pathway. HEmT-DCN/sLRP6 effectively degraded tumor ECM and inhibited EMT, leading to enhanced viral distribution, induction of apoptosis, and attenuation of tumor cell proliferation in tumor tissue. HEmT-DCN/sLRP6 prevented metastasis of pancreatic cancer. Importantly, HEmT-DCN/sLRP6 sensitized pancreatic tumor to gemcitabine treatment. Furthermore, HEmT-DCN/sLRP6 augmented drug penetration and dispersion within pancreatic tumor xenografts and patient-derived tumor spheroids. Collectively, these results illustrate that HEmT-DCN/sLRP6 can enhance the dispersion of both oncolytic Ad and a chemotherapeutic agent in chemoresistant and desmoplastic pancreatic tumor, effectively overcoming the preexisting limitations of standard treatments.


Asunto(s)
Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/virología , Células A549 , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Animales , Línea Celular Tumoral , Decorina/biosíntesis , Decorina/genética , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Células HEK293 , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/biosíntesis , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Distribución Aleatoria , Receptores Wnt/antagonistas & inhibidores , Receptores Wnt/metabolismo , Vía de Señalización Wnt , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Clin Transl Oncol ; 21(2): 220-231, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29956073

RESUMEN

PURPOSE: The aim of this study is to explore the roles of ß-catenin, decorin, septin-7, and S100A10 expression in colorectal cancer development. METHODS: Twenty-five BALB/c mice were divided into five groups; four groups were administrated N,N-dimethylhydrazine for 0, 10, 15, and 20 weeks, and one group was administrated normal saline for 20 weeks. The colons were collected for histopathological analysis. Protein samples prepared from the frozen colon tissues of mice treated with N,N-dimethylhydrazine for the different time points were evaluated using the isobaric tags for relative and absolute quantification (iTRAQ) labeling technique coupled with the 2D liquid chromatography-tandem mass spectrometry analysis. Based on the proteomic analysis results, immunohistochemical staining of ß-catenin, decorin, septin-7, and S100A10 was performed in paraffin-embedded mice colorectal tissue, and 53 cases of human hereditary polyposis colorectal cancer samples. RESULTS: Colorectal cancer was observed in mice treated with N,N-dimethylhydrazine for 20 weeks, and adenomas were observed in mice subjected to the 10-, and 15-week treatments. Seventy-two differentially expressed proteins were involved in the development of cancer as per the iTRAQ and spectrometry analysis. In normal epithelium, adenoma, and cancer from human hereditary polyposis colorectal cancer, S100A10 expression (c2 = 100.989, P = 0.000) was highest in cancer, whereas decorin (c2 = 12.852, P = 0.002) and septin-7 (c2 = 66.519, P = 0.002) expressions were highest in the normal epithelium, which was confirmed via immunohistochemical staining. CONCLUSIONS: The subcellular localization of ß-catenin and decorin, septin-7, and S100A10 expressions are associated with the development of colorectal cancer in mice after N,N-dimethylhydrazine treatment and in human hereditary polyposis colorectal cancers.


Asunto(s)
Poliposis Adenomatosa del Colon/patología , Biomarcadores de Tumor/análisis , Neoplasias Colorrectales/patología , Adulto , Animales , Anexina A2/análisis , Anexina A2/biosíntesis , Carcinógenos/toxicidad , Neoplasias Colorrectales/inducido químicamente , Decorina/análisis , Decorina/biosíntesis , Dimetilhidrazinas/toxicidad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Proteómica/métodos , Proteínas S100/análisis , Proteínas S100/biosíntesis , Septinas/análisis , Septinas/biosíntesis , beta Catenina/análisis , beta Catenina/biosíntesis
5.
Hum Gene Ther ; 30(2): 197-210, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30032645

RESUMEN

The majority of advanced breast cancer patients develop distal metastasis, including lung and bone metastasis. However, effective therapeutic strategies to prevent metastasis are still lacking. Decorin is a natural inhibitor of transforming growth factor ß, which plays a pivotal role in tumor metastasis. An oncolytic adenovirus expressing decorin, rAd.DCN, has been developed previously. In an immune-competent breast tumor (4T1) model, intratumoral (i.t.) as well as intravenous (i.v.) delivery of rAd.DCN inhibited growth of orthotopic tumors and spontaneous lung metastasis. It was shown that i.t. delivery of rAd.DCN produced higher levels of transgene expression and evoked stronger oncolysis of the tumors compared to i.v. delivery. However, i.v. delivery resulted in higher amount of virus accumulation in the lungs and produced stronger responses to prevent tumor lung metastasis. Oncolytic adenovirus-mediated decorin expression in the tumors downregulated the decorin target genes and decreased epithelial mesenchymal transition markers. Decorin expression in lung tissues also increased Th1 cytokine expression, such as interleukin (IL)-2, IL-12, and tumor necrosis factor α, and decreased Th2 cytokines, such as transforming growth factor ß and IL-6. Moreover, rAd.DCN treatment induced strong systemic inflammatory responses and upregulated CD8+ T lymphocytes. In conclusion, rAd.DCN inhibits tumor growth and lung metastasis of breast cancer via regulating wnt/ß-catenin, vascular endothelial growth factor (VEGF), and Met pathways, and modulating the antitumor inflammatory and immune responses. Considering that i.v. delivery was much more effective in preventing lung metastasis, systemic delivery of rAd.DCN might be a promising strategy to treat breast cancer lung metastasis.


Asunto(s)
Adenoviridae , Neoplasias de la Mama , Decorina , Neoplasias Pulmonares , Viroterapia Oncolítica , Virus Oncolíticos , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Decorina/biosíntesis , Decorina/genética , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Metástasis de la Neoplasia , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Eur J Prev Cardiol ; 25(1_suppl): 51-58, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29708036

RESUMEN

Background Sporadic non-syndromic thoracic aortic aneurysms (SNSTAAs) are less well understood than familial non-syndromic or syndromic ones. Here, we focused on morphologic and molecular changes of the extracellular matrix of the tunica media of SNSTAAs. Design Single centre design. Methods Surgical media samples from seven SNSTAAs and seven controls underwent quantitative polymerase chain reaction, proteomics-bioinformatics, immunoblotting, histology and immunohistochemistry analysis. Results A down-regulation of Decorin mRNA with unchanged protein levels associated with a remarkable increase of collagen fibres. A reduced and distorted network of elastic fibres partnered with an attenuated expression of microfibril-associated glycoprotein1 despite the rise of MFAP2 gene-encoded mRNA levels. An increasingly proteolysed paxillin (55 kDa PXN), a focal adhesion protein, combined with an upregulated 62 kDa PXN holoprotein, without changes in amount and phosphorylation of focal adhesion kinase (pp125FAK). The upregulation of SPOCK2-encoded Testican2 proteoglycan and of ectodysplasin (EDA) protein was coupled with a down-regulation of EDA2 receptor (EDA2R). Conclusions Several tunica media extracellular matrix-related changes favour SNSTAA development. A steady level of decorin and a microfibril-associated glycoprotein1 protein shortage cause the assembly of structurally defective collagen and elastic fibres. Up-regulation of PXN holoproteins perturbs PXN/pp125FAK interaction and focal adhesion functioning. Testican2 up-regulation suppresses the membrane-type matrix metalloproteinase inhibiting activities of other SPOCK family members thus enhancing extracellular matrix proteolysis. Finally, the altered EDA•EDA2R signalling would impact on the remodelling of SNSTAA tunica media. Altogether, our results pave the way to a deeper molecular understanding of SNSTAAs necessary to identify their early diagnostic biochemical markers.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , Decorina/genética , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Regulación de la Expresión Génica , Proteoglicanos/genética , Receptor Xedar/genética , Aneurisma de la Aorta Torácica/metabolismo , Aneurisma de la Aorta Torácica/patología , Decorina/biosíntesis , Matriz Extracelular/patología , Humanos , Immunoblotting , Inmunohistoquímica , Reacción en Cadena de la Polimerasa , Proteoglicanos/biosíntesis , ARN/genética , Receptor Xedar/biosíntesis
7.
Eur Rev Med Pharmacol Sci ; 22(8): 2483-2489, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29762850

RESUMEN

OBJECTIVE: To explore a new approach for treating renal insufficiency with gene therapy by implanting decorin (DCN)-expressing fibroblasts within the renal tissue of rats with renal failure to neutralize TGF-ß1 activity. MATERIALS AND METHODS: The 5/6 kidney of the selected male SD rats were removed under aseptic conditions. The rats were grouped randomly after the establishment of the model. There were 10 rats in the sham-operated group (Group A), 10 in the operation control group (without treatment, Group B), 10 in the blank control group [treated with empty vector-transfected fibroblasts (FB (LXSN) cells), Group C], and 10 in the treatment group [treated with FB (LDCNSN) cells, Group D]. The pathological changes of rats including body weight, blood lipids, renal function, and renal histology, were observed. The expression of TGF-ß1 and DCN in renal tissue was detected by immunohistochemistry. RESULTS: There were no significant differences in body weight and blood lipids between the groups at 4 weeks after treatment. The levels of blood urea nitrogen and serum creatinine in rats in Group D were significantly decreased compared with those in Group C (p < 0.05). Although the differences were not statistically significant, the levels of those pathological indicators are higher than baseline values. The expression of DCN in renal tissue increased significantly after 4 weeks in rats of Group D and the differences were significant compared with the other groups. There were no significant differences in TGF-ß1 expression between any two groups of Group D, B, and C. Furthermore, pathological damage to the renal interstitium of rats in Group D was significantly decreased compared with that of Group B and C. CONCLUSIONS: DCN can alleviate fibrosis and delay the progression of renal failure.


Asunto(s)
Decorina/administración & dosificación , Decorina/biosíntesis , Terapia Genética/métodos , Insuficiencia Renal/metabolismo , Insuficiencia Renal/terapia , Animales , Nitrógeno de la Urea Sanguínea , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Ratas , Ratas Sprague-Dawley , Insuficiencia Renal/patología , Factor de Crecimiento Transformador beta1/biosíntesis , Resultado del Tratamiento
8.
Sci Rep ; 7(1): 8225, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28811645

RESUMEN

Acute kidney injury (AKI) is a public health problem worldwide. Several therapeutic strategies have been made to accelerate recovery and improve renal survival. Recent studies have shown that human adult renal progenitor cells (ARPCs) participate in kidney repair processes, and may be used as a possible treatment to promote regeneration in acute kidney injury. Here, we show that human tubular ARPCs (tARPCs) protect physically injured or chemically damaged renal proximal tubular epithelial cells (RPTECs) by preventing cisplatin-induced apoptosis and enhancing proliferation of survived cells. tARPCs without toll-like receptor 2 (TLR2) expression or TLR2 blocking completely abrogated this regenerative effect. Only tARPCs, and not glomerular ARPCs, were able to induce tubular cell regeneration process and it occurred only after damage detection. Moreover, we have found that ARPCs secreted inhibin-A and decorin following the RPTEC damage and that these secreted factors were directly involved in cell regeneration process. Polysaccharide synthetic vesicles containing these molecules were constructed and co-cultured with cisplatin damaged RPTECs. These synthetic vesicles were not only incorporated into the cells, but they were also able to induce a substantial increase in cell number and viability. The findings of this study increase the knowledge of renal repair processes and may be the first step in the development of new specific therapeutic strategies for renal repair.


Asunto(s)
Células Madre Adultas/metabolismo , Decorina/biosíntesis , Células Epiteliales/metabolismo , Inhibinas/biosíntesis , Túbulos Renales/metabolismo , Regeneración , Receptor Toll-Like 2/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Adulto , Células Madre Adultas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Biomarcadores , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cisplatino/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Humanos
9.
Biomed Res Int ; 2017: 1038984, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28567416

RESUMEN

Aim. To demonstrate the regulatory role of miRNA in colorectal carcinoma (CRC) and reveal the transcript markers that may be associated with CRC clinical outcomes. Method. Herein, we analyzed both mRNA and miRNA gene expression profiles of 255 CRC tumor samples from The Cancer Genome Atlas project to reveal the regulatory association between miRNA and mRNA. Also, the potential role of gene coexpression network in CRC has been explored. Results. The negative correlation between miR-200c and DCN (Decorin) was calculated in CRC, indicating that DCN could be a potential target of miR-200c. Clinical features indicated that colon polyp history and overall survival were significantly related to the expression level of miR-200c. Three coexpression networks have been constructed, and genes involved in the networks are related to cell cycle, NOTCH, and mTOR signaling pathways. Conclusion. Our result provides a new insight into cancer related mRNA coexpression network in CRC research.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Decorina/biosíntesis , Regulación Neoplásica de la Expresión Génica , MicroARNs/biosíntesis , Proteínas de Neoplasias/biosíntesis , ARN Neoplásico/biosíntesis , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Decorina/genética , Femenino , Humanos , Masculino , MicroARNs/genética , Proteínas de Neoplasias/genética , ARN Neoplásico/genética
10.
J Invest Dermatol ; 137(8): 1774-1783, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28454729

RESUMEN

In this study, we report that TIP39, a parathyroid hormone ligand family member that was recently identified to be expressed in the skin, can induce decorin expression and enhance wound repair. Topical treatment of mice with TIP39 accelerated wound repair, whereas TIP39-deficient mice had delayed repair that was associated with formation of abnormal collagen bundles. To study the potential mechanism responsible for the action of TIP39 in the dermis, fibroblasts were cultured in three-dimensional collagen gels, a process that results in enhanced decorin expression unless activated to differentiate to adipocytes, whereupon these cells reduce expression of several proteoglycans, including decorin. Small interfering RNA-mediated silencing of parathyroid hormone 2 receptor (PTH2R), the receptor for TIP39, suppressed the expression of extracellular matrix-related genes, including decorin, collagens, fibronectin, and matrix metalloproteases. Skin wounds in TIP39-/- mice had decreased decorin expression, and addition of TIP39 to cultured fibroblasts induced decorin and increased phosphorylation and nuclear translocation of CREB. Fibroblasts differentiated to adipocytes and treated with TIP39 also showed increased decorin and production of chondroitin sulfate. Furthermore, the skin of PTH2R-/- mice showed abnormal extracellular matrix structure, decreased decorin expression, and skin hardness. Thus, the TIP39-PTH2R system appears to be a previously unrecognized mechanism for regulation of extracellular matrix formation and wound repair.


Asunto(s)
Decorina/genética , Regulación de la Expresión Génica , Proteínas Nucleares/farmacología , ARN/genética , Receptor de Hormona Paratiroídea Tipo 2/genética , Proteínas de Transporte Vesicular/farmacología , Cicatrización de Heridas/fisiología , Heridas y Lesiones/genética , Animales , Diferenciación Celular , Células Cultivadas , Decorina/biosíntesis , Modelos Animales de Enfermedad , Femenino , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Factores de Empalme de ARN , Proteínas de Unión al ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Hormona Paratiroídea Tipo 2/biosíntesis , Transducción de Señal , Piel/lesiones , Piel/metabolismo , Piel/patología , Cicatrización de Heridas/efectos de los fármacos , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología
11.
Biochim Biophys Acta ; 1861(9 Pt A): 1121-1131, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27317982

RESUMEN

Visceral and subcutaneous adipose tissue depots have distinct features and contribute differentially to the development of metabolic dysfunction. We show here that adipocyte differentiation in subcutaneous stromal-vascular fraction (SVF) is increased compared to visceral SVF, however this increased differentiation capacity seems not to be due to changes in the number of adipocyte precursor cells. Rather, we demonstrate that secreted heat-sensitive factors from the SVF can inhibit adipocyte differentiation and that this effect is higher in visceral than in subcutaneous SVF, suggesting that visceral SVF is a source of secreted factors that can inhibit adipocyte formation. In order to explore secreted proteins that potentially inhibit differentiation in visceral preadipocytes we analyzed the secretome of both SVFs which led to the identification of 113 secreted proteins with an overlap of 42%. Further expression analysis in both depots revealed 16 candidates that were subsequently analyzed in a differentiation screen using an adenoviral knockdown system. From this analysis we were able to identify two potential inhibitory candidates, namely decorin (Dcn) and Sparc-like 1 (Sparcl1). We could show that ablation of either candidate enhanced adipogenesis in visceral preadipocytes, while treatment of primary cultures with recombinant Sparcl1 and Dcn blocked adipogenesis in a dose dependent manner. In conclusion, our data suggests that the differences in adipogenesis between depots might be due to paracrine and autocrine feedback mechanisms which could in turn contribute to metabolic homeostasis.


Asunto(s)
Adipogénesis/genética , Proteínas de Unión al Calcio/biosíntesis , Decorina/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , Obesidad/genética , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Diferenciación Celular/genética , Decorina/genética , Proteínas de la Matriz Extracelular/genética , Humanos , Grasa Intraabdominal/crecimiento & desarrollo , Grasa Intraabdominal/metabolismo , Ratones , Obesidad/metabolismo , Comunicación Paracrina/genética , Grasa Subcutánea/crecimiento & desarrollo , Grasa Subcutánea/metabolismo
12.
Tumour Biol ; 37(4): 5171-83, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26547587

RESUMEN

Decorin (DCN), as an important component of the extracellular matrix (ECM), is a small leucine-rich proteoglycan and synthesized by fibroblasts. Although DCN is dysregulated in numerous cancer types, limited data are available on the expression level and important role of DCN proteins in renal cell carcinoma (RCC). In our study, we examined the expression patterns of DCN messenger RNA (mRNA) in RCCs through the Oncomine database and DCN protein in 94 RCC specimens by immunohistochemistry (IHC). The results revealed that DCN expression was decreased in cancerous tissues compared to adjacent noncancerous tissues and was highly correlated to tumor size. Then, via gain-of-function analyses, DCN overexpression could inhibit RCC cell proliferation and metastasis in vitro and vivo. At the mechanism level, we found that an ectopic expression of DCN significantly upregulated P21 and E-cadherin expression. Altogether, these results revealed that DCN is a tumor suppressor in RCC, and it could serve as a potential therapeutic target in patients with RCC.


Asunto(s)
Cadherinas/biosíntesis , Carcinoma de Células Renales/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Decorina/biosíntesis , Anciano , Animales , Cadherinas/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Decorina/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , ARN Mensajero/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Reprod Sci ; 23(3): 302-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26423601

RESUMEN

Uterine leiomyoma are a common benign pelvic tumors composed of modified smooth muscle cells and a large amount of extracellular matrix (ECM). The proteoglycan composition of the leiomyoma ECM is thought to affect pathophysiology of the disease. To test this hypothesis, we examined the abundance (by immunoblotting) and expression (by quantitative real-time polymerase chain reaction) of the proteoglycans biglycan, decorin, and versican in leiomyoma and normal myometrium and determined whether expression is affected by steroid hormones and menstrual phase. Leiomyoma and normal myometrium were collected from women (n = 17) undergoing hysterectomy or myomectomy. In vitro studies were performed on immortalized leiomyoma (UtLM) and normal myometrial (hTERT-HM) cells with and without exposure to estradiol and progesterone. In leiomyoma tissue, abundance of decorin messenger RNA (mRNA) and protein were 2.6-fold and 1.4-fold lower, respectively, compared with normal myometrium. Abundance of versican mRNA was not different between matched samples, whereas versican protein was increased 1.8-fold in leiomyoma compared with myometrium. Decorin mRNA was 2.4-fold lower in secretory phase leiomyoma compared with proliferative phase tissue. In UtLM cells, progesterone decreased the abundance of decorin mRNA by 1.3-fold. Lower decorin expression in leiomyoma compared with myometrium may contribute to disease growth and progression. As decorin inhibits the activity of specific growth factors, its reduced level in the leiomyoma cell microenvironment may promote cell proliferation and ECM deposition. Our data suggest that decorin expression in leiomyoma is inhibited by progesterone, which may be a mechanism by which the ovarian steroids affect leiomyoma growth and disease progression.


Asunto(s)
Decorina/biosíntesis , Leiomioma/metabolismo , Miometrio/metabolismo , Proteoglicanos/biosíntesis , Neoplasias Uterinas/metabolismo , Adulto , Línea Celular Transformada , Línea Celular Tumoral , Decorina/antagonistas & inhibidores , Estradiol/farmacología , Femenino , Humanos , Leiomioma/fisiopatología , Persona de Mediana Edad , Miometrio/efectos de los fármacos , Miometrio/fisiopatología , Progesterona/farmacología , Promegestona/farmacología , Proteoglicanos/antagonistas & inhibidores , Neoplasias Uterinas/fisiopatología
14.
Biomed Res Int ; 2015: 654765, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26697491

RESUMEN

Decorin (DCN) is the best characterized member of the extracellular small leucine-rich proteoglycan family present in connective tissues, typically in association with or "decorating" collagen fibrils. It has substantial interest to clinical medicine owing to its antifibrotic, anti-inflammatory, and anticancer effects. Studies on DCN knockout mice have established that a lack of DCN is permissive for tumor development and it is regarded as a tumor suppressor gene. A reduced expression or a total disappearance of DCN has been reported to take place in various forms of human cancers during tumor progression. Furthermore, when used as a therapeutic molecule, DCN has been shown to inhibit tumor progression and metastases in experimental cancer models. DCN affects the biology of various types of cancer by targeting a number of crucial signaling molecules involved in cell growth, survival, metastasis, and angiogenesis. The active sites for the neutralization of different growth factors all reside in different parts of the DCN molecule. An emerging concept that multiple proteases, especially those produced by inflammatory cells, are capable of cleaving DCN suggests that native DCN could be inactivated in a number of pathological inflammatory conditions. In this paper, we review the role of DCN in cancer.


Asunto(s)
Carcinogénesis/genética , Proliferación Celular/genética , Decorina/biosíntesis , Neoplasias/genética , Animales , Decorina/genética , Matriz Extracelular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Metástasis de la Neoplasia , Neoplasias/patología
15.
Sci Rep ; 5: 15676, 2015 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-26511275

RESUMEN

The mammalian kidney develops from reciprocal interactions between the metanephric mesenchyme and ureteric bud, the former of which contains nephron progenitors. The third lineage, the stroma, fills up the interstitial space and is derived from distinct progenitors that express the transcription factor Foxd1. We showed previously that deletion of the nuclear factor Sall1 in nephron progenitors leads to their depletion in mice. However, Sall1 is expressed not only in nephron progenitors but also in stromal progenitors. Here we report that specific Sall1 deletion in stromal progenitors leads to aberrant expansion of nephron progenitors, which is in sharp contrast with a nephron progenitor-specific deletion. The mutant mice also exhibited cystic kidneys after birth and died before adulthood. We found that Decorin, which inhibits Bmp-mediated nephron differentiation, was upregulated in the mutant stroma. In contrast, the expression of Fat4, which restricts nephron progenitor expansion, was reduced mildly. Furthermore, the Sall1 protein binds to many stroma-related gene loci, including Decorin and Fat4. Thus, the expression of Sall1 in stromal progenitors restricts the excessive expansion of nephron progenitors in a non-cell autonomous manner, and Sall1-mediated regulation of Decorin and Fat4 might at least partially underlie the pathogenesis.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Nefronas/metabolismo , Células Madre/metabolismo , Factores de Transcripción/biosíntesis , Animales , Cadherinas/biosíntesis , Cadherinas/genética , Decorina/biosíntesis , Decorina/genética , Ratones , Ratones Mutantes , Nefronas/patología , Células Madre/patología , Factores de Transcripción/genética
16.
J Control Release ; 220(Pt B): 766-82, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26471393

RESUMEN

Pancreatic cancer is highly aggressive, malignant, and notoriously difficult to cure using conventional cancer therapies. These conventional therapies have significant limitations due to excessive extracellular matrix (ECM) of pancreatic cancer and poor cancer specificity. The excess ECM prevents infiltration of drugs into the inner layer of the solid tumor. Therefore, novel treatment modalities that can specifically target the tumor and degrade the ECM are required for effective therapy. In the present study, we used ECM-degrading and Wnt signal-disrupting oncolytic adenovirus (oAd/DCN/LRP) to achieve a desirable therapeutic outcome against pancreatic cancer. In addition, to overcome the limitations in systemic delivery of oncolytic Ad (oAd) and to specifically target pancreatic cancer, neurotensin peptide (NT)-conjugated polyethylene glycol (PEG) was chemically crosslinked to the surface of Ad, generating a systemically injectable hybrid system, oAd/DCN/LRP-PEG-NT. We tested the targeting and therapeutic efficacy of oAd/DCN/LRP-PEG-NT toward neurotensin receptor 1 (NTR)-overexpressing pancreatic cancer cells, both in vitro and in vivo. The oAd/DCN/LRP-PEG-NT elicited increased NTR-selective cancer cell killing and transduction efficiency when compared with a cognate control lacking NT (oAd/DCN/LRP-PEG). Furthermore, systemic administration of oAd/DCN/LRP-PEG-NT significantly decreased induction of innate and adaptive immune responses against Ad, and blood retention time was markedly prolonged by PEGylation. Moreover, NTR-targeting oAd elicited greater in vivo tumor growth suppression when compared with naked oAd and 9.5 × 10(6)-fold increased tumor-to-liver ratio. This significantly enhanced antitumor effect of oAd/DCN/LRP-PEG-NT was mediated by active viral replication and viral spreading, which was facilitated by ECM degradation and inhibition of Wnt signaling-related factors (Wnt, ß-catenin, and/or vimentin) in the tumor tissues. Taken together, these results demonstrate that oAd/DCN/LRP-PEG-NT has strong therapeutic potential for systemic treatment of NTR-overexpressing pancreatic cancer due to its NTR-targeting ability, enhanced therapeutic efficacy, and safety.


Asunto(s)
Adenoviridae/genética , Decorina/genética , Terapia Genética/métodos , Neurotensina/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Neoplasias Pancreáticas/terapia , Receptores de Neurotensina/metabolismo , Carga Tumoral/efectos de los fármacos , Vía de Señalización Wnt/genética , Inmunidad Adaptativa , Adenoviridae/crecimiento & desarrollo , Adenoviridae/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Decorina/biosíntesis , Regulación Viral de la Expresión Génica , Terapia Genética/efectos adversos , Humanos , Inmunidad Innata , Masculino , Ratones Desnudos , Neurotensina/biosíntesis , Neurotensina/inmunología , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Virus Oncolíticos/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/virología , Polietilenglicoles/química , Factores de Tiempo , Transducción Genética , Carga Viral , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Growth Factors ; 33(5-6): 326-36, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26431359

RESUMEN

This study aimed to investigate the synergy between transforming growth factor beta 3 (TGF-ß3) and insulin-like growth factor 1 (IGF-1) on nucleus pulposus-derived mesenchymal stem cells (NP-MSCs) and the underlying mechanism using a serum-free culture system. NP-MSC proliferation and viability were measured using a CCK-8 assay and annexin V-FITC/propidium iodide, respectively. NP-MSCs in micromasses were investigated for differentiation towards nucleus pulposus cells (NPCs). SOX-9, collagen-I, collagen-II, aggrecan and decorin expressions were detected by RT-PCR and immunoblotting. Matrix deposition was assessed by sulfated glycosaminoglycan (sGAG) analysis. Novel chondrogenic and nucleus pulposus (NP) genes were detected to distinguish differentiated cell types. MAPK/ERK and TGF/Smad signaling pathways were also examined. As a result, the synergy between TGF-ß3 and IGF-1 enhanced NP-MSC viability, extracellular matrix (ECM) biosynthesis and differentiation towards NPCs, partly through the activation of the MAPK/ERK signaling pathway. Therefore, the synergy between TGF-ß3 and IGF-1 ameliorates NP-MSC viability, differentiation and promotes intervertebral disc regeneration.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Disco Intervertebral/crecimiento & desarrollo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Factor de Crecimiento Transformador beta3/farmacología , Agrecanos/biosíntesis , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/biosíntesis , Colágeno Tipo II/biosíntesis , Decorina/biosíntesis , Sinergismo Farmacológico , Matriz Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Disco Intervertebral/citología , Masculino , Ratas , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos , Factor de Transcripción SOX9/biosíntesis , Proteínas Smad/metabolismo
18.
Lasers Med Sci ; 30(7): 1931-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26188855

RESUMEN

Intense pulsed light (IPL) devices have been shown to be highly effective for the skin rejuvenation. In our study, we try to elucidate effects of IPL in fibroblast proliferation, in gene expression, and in extracellular matrix protein production. 1BR3G human skin fibroblasts were used to test the effects of an IPL device (MiniSilk FT, Deka®). Fibroblasts were divided into three groups: group 1 was irradiated with filter 800-1200 nm (frequency 10 Hz, 15 s, fluence 60.1 J/cm) twice; group 2 was irradiated with filter 550-1200 nm (double pulse 5 ms + 5 ms, delay 10 ms, fluence 13 J/cm2) twice; and group 3 was irradiated with filter 550-1200 nm (frequency 10 Hz, 15 s, fluence 60.1 J/cm2) twice. To determine changes in gene expression, messenger RNA (mRNA) levels for collagen types I and III and metalloproteinase 1 (MMP-1) were performed 48 h after irradiation. To determine changes in hyaluronic acid, versican, and decorin, mRNA and ELISA tests were performed after 48 h of treatment. In addition to this, a Picro-Sirius red staining for collagen was made. The study showed an increase of mRNA and hyaluronic acid, decorin, and versican production. With RT-PCR assays, an increase mRNA for collagen type I, type III, and MMP-1 was observed. Collagen and hyaluronic synthesis was increased in all groups with no differences among them, while decorin and versican synthesis was higher in those groups irradiated with 550-1200-nm filters with no dependence of type pulse or total energy dose. IPL applied in vitro cultured cells increases fibroblasts activity. Synthesis of extracellular proteins seems to be produced more specifically in determined wavelengths, which could demonstrate a biochemical mechanism light depending.


Asunto(s)
Colágeno Tipo III/metabolismo , Colágeno Tipo I/metabolismo , Rayos Láser , Metaloproteinasa 1 de la Matriz/metabolismo , Activación Transcripcional/efectos de la radiación , Línea Celular , Proliferación Celular , Colágeno Tipo I/genética , Colágeno Tipo III/genética , Decorina/biosíntesis , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Expresión Génica , Humanos , Ácido Hialurónico/biosíntesis , Tratamiento de Luz Pulsada Intensa , Metaloproteinasa 1 de la Matriz/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Piel/citología , Versicanos/biosíntesis
19.
Virchows Arch ; 467(2): 211-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25940995

RESUMEN

The histologic differential diagnosis between intramuscular myxoma and low-grade myxofibrosarcoma can be quite difficult in some cases. To identify a diagnostic immunohistochemical marker, we compared the staining profiles of 19 different antigens, including cell cycle proteins, apoptosis proteins, and proliferative markers, and selected other signaling and structural proteins in these two tumors. Ten cases each of intramuscular myxoma and low-grade myxofibrosarcoma were stained with antibodies directed against apoptosis regulatory proteins (Bcl2, activated caspase-3, phospho-H2A.X, and cleaved PARP), cell cycle regulatory proteins (Rb1, Cyclin-A, CDKN1B, and Cdt1), proliferative markers (KI67, MCM2, phospho-histone H3, and geminin), cell signalling molecules (c-Myc, EGF, EGFR, PLA2G4A, and HSP90), a dendritic cell marker (CD209), and the extracellular matrix proteoglycan decorin. Staining patterns of myxoma and myxofibrosarcoma were compared using Fisher's exact test and the Mann-Whitney test. For each potential diagnostic marker studied, the proportions of cases scored as positive on both dichotomous or ordinal scales were not significantly different between myxoma and myxofibrosarcoma. Myxoma and myxofibrosarcoma share a common immunophenotype for each of the markers studied. Distinction between these tumors is still predominantly based on morphologic criteria.


Asunto(s)
Biomarcadores de Tumor/análisis , Fibrosarcoma/diagnóstico , Mixoma/diagnóstico , Adulto , Anciano , Proteínas Reguladoras de la Apoptosis/análisis , Proteínas Reguladoras de la Apoptosis/biosíntesis , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/biosíntesis , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/biosíntesis , Decorina/análisis , Decorina/biosíntesis , Diagnóstico Diferencial , Femenino , Humanos , Inmunohistoquímica , Lectinas Tipo C/análisis , Lectinas Tipo C/biosíntesis , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/biosíntesis
20.
PLoS One ; 10(4): e0123054, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25837671

RESUMEN

Hypertrophic scarring is a frequent fibroproliferative complication following deep dermal burns leading to impaired function and lifelong disfigurement. Decorin reduces fibrosis and induces regeneration in many tissues, and is significantly downregulated in hypertrophic scar and normal deep dermal fibroblasts. It was hypothesized that microRNAs in these fibroblasts downregulate decorin and blocking them would increase decorin and may prevent hypertrophic scarring. Lower decorin levels were found in hypertrophic scar as compared to normal skin, and in deep as compared to superficial dermis. A decorin 3' un-translated region reporter assay demonstrated microRNA decreased decorin in deep dermal fibroblasts, and microRNA screening predicted miR- 24, 181b, 421, 526b, or 543 as candidates. After finding increased levels of mir-181b in deep dermal fibroblasts, it was demonstrated that TGF-ß1 stimulation decreased miR-24 but increased miR-181b and that hypertrophic scar and deep dermis contained increased levels of miR-181b. By blocking miR-181b with an antagomiR, it was possible to increase decorin protein expression in dermal fibroblasts. This suggests miR-181b is involved in the differential expression of decorin in skin and wound healing. Furthermore, blocking miR-181b reversed TGF-ß1 induced decorin downregulation and myofibroblast differentiation in hypertrophic scar fibroblasts, suggesting a potential therapy for hypertrophic scar.


Asunto(s)
Cicatriz Hipertrófica/terapia , Decorina/biosíntesis , MicroARNs/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas/genética , Regiones no Traducidas 3'/genética , Quemaduras/patología , Células Cultivadas , Dermis/citología , Dermis/metabolismo , Regulación hacia Abajo/genética , Fibroblastos/metabolismo , Humanos , MicroARNs/genética , Oligonucleótidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...