Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Curr Oncol ; 29(4): 2650-2661, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35448191

RESUMO

Radiation-induced fibrosis (RIF) is a severe side effect related with soft tissues sarcomas (STS) radiotherapy. RIF is a multicellular process initiated primarily by TGF-ß1 that is increased in irradiated tissue, whose signaling leads to intracellular Smad2/3 phosphorylation and further induction of profibrotic target genes. P144 (Disetertide©) is a peptide inhibitor of TGF-ß1 and is proposed as a candidate compound for reducing RIF associated wound healing problems and muscle fibrosis in STS. METHODS: A treatment and control group of WNZ rabbits were employed to implement a brachytherapy animal model, through catheter implantation at the lower limb. Two days after implantation, animals received 20 Gy isodosis, intended to induce a high RIF grade. The treatment group received intravenous P144 administration following a brachytherapy session, repeated at 24-72 h post-radiation, while the control group received placebo. Four weeks later, affected muscular tissues underwent histological processing for collagen quantification and P-Smad2/3 immunohistochemistry through image analysis. RESULTS: High isodosis Brachytherapy produced remarkable fibrosis in this experimental model. Results showed retained macro and microscopical morphology of muscle in the P144 treated group, with reduced extracellular matrix fibrosis, with a lower area of collagen deposition measured through Masson's trichrome staining. Intravenous P144 also induced a significant reduction in Smad2/3 phosphorylation levels compared with the placebo group. CONCLUSIONS: P144 administration clearly reduces RIF and opens a new potential co-treatment approach to reduce complications in soft tissue sarcoma (STS) radiotherapy. Further studies are required to establish whether the dosage and timing optimization of P144 administration, in different RIF phases, might entirely avoid fibrosis associated with STS brachytherapy.


Assuntos
Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta , Animais , Fibrose , Humanos , Peptídeos/farmacologia , Coelhos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
2.
Methods Enzymol ; 636: 129-172, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32178817

RESUMO

Transforming growth factor beta (TGF-ß) is a potent pleiotropic polypeptide cytokine, with a complex and context dependent control of its activation, signaling and effector functions. This cytokine is pivotal in the regulation of immunological responses, tumor initiation and development, stromal homeostasis and all their intricate related interactions. Last decade advances in cancer immunotherapy have reactivated the clinical interest on potential drug with TGF-ß inhibition effect, combined with immunomodulating enhancer drugs. The correct quantification of the in vitro and in vivo biological activity of this cytokine is essential to understand the intrinsic underlying biological mechanisms and TGF-ß role in the immune system, tumor and stromal codevelopment, modulation and interactions. There is a wide variety of available procedures to quantify TGF-ß activity, which includes different methodological approximations like ELISA, Bioassays including reporter gene assays, Flow cytometry (FC), Western blotting (WB), immunochemical/fluorescence microscopy, among others. Here, we detail available methods for TGF-ß biological activity analysis, together with their applicability and suitability for each experimental setting, in order to get a complete analytical perspective and more comprehensive information along the development and design of combined antitumor immunotherapies, which include the inhibition of TGF-ß biological activity.


Assuntos
Neoplasias , Fator de Crescimento Transformador beta , Citocinas , Humanos , Neoplasias/tratamento farmacológico , Transdução de Sinais
3.
J Mater Sci Mater Med ; 29(6): 73, 2018 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-29796827

RESUMO

Osseointegration of implants is conversely related to the generation of a fibrous tissue capsule around the implant by the host environment. Although TGF-ß1 plays many roles in regeneration processes, it is the cytokine to be mostly associated to the production of fibrotic tissue and thus, its inhibition has demonstrated to be beneficial to prevent several fibrotic reactions. Surface biofunctionalization enables the immobilization of biologically active molecules on an implant surface to tailor the biological response of the host. Here, we studied in vitro biological effects of biofunctionalized CP-Ti surfaces with a TGF-ß1 inhibitor peptide, P144. A reliable biofunctionalization process that tethers P144 peptides to commercially pure titanium was developed. Differentiation of human mesenchymal stem cells, osteoblasts and fibroblasts on P144-functionalized and control surfaces was assessed at the gene expression and protein production levels. Results showed that P144-functionalized surfaces reduced expression and production of fibrotic differentiation markers and increased osteoblastic differentiation markers. Therefore, biofunctionalization of surfaces with TGF-ß1 inhibitor peptides are an alternative promising strategy for inducing osseointegration around medical devices and implants.


Assuntos
Materiais Revestidos Biocompatíveis/química , Células-Tronco Mesenquimais/citologia , Peptídeos/química , Fator de Crescimento Transformador beta1/química , Fosfatase Alcalina/metabolismo , Animais , Diferenciação Celular , Citocinas/metabolismo , Fibroblastos/metabolismo , Humanos , Teste de Materiais , Camundongos , Células NIH 3T3 , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Espectrometria por Raios X , Propriedades de Superfície , Titânio/química
4.
J Immunol Methods ; 457: 1-5, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29522776

RESUMO

Since its first description, Western blot has been widely used in molecular labs. It constitutes a multistep method that allows the detection and/or quantification of proteins from simple to complex protein mixtures. Western blot quantification method constitutes a critical step in order to obtain accurate and reproducible results. Due to the technical knowledge required for densitometry analysis together with the resources availability, standard office scanners are often used for the imaging acquisition of developed Western blot films. Furthermore, the use of semi-quantitative software as ImageJ (Java-based image-processing and analysis software) is clearly increasing in different scientific fields. In this work, we describe the use of office scanner coupled with the ImageJ software together with a new image background subtraction method for accurate Western blot quantification. The proposed method represents an affordable, accurate and reproducible approximation that could be used in the presence of limited resources availability.


Assuntos
Western Blotting , Densitometria , Processamento de Imagem Assistida por Computador/métodos , Proteínas/análise , Reprodutibilidade dos Testes , Software
5.
Cancer Lett ; 381(1): 67-75, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27473823

RESUMO

Glioblastoma (GBM) is the most prevalent malignant primary brain tumor, accounting for 60-70% of all gliomas. Current median patient survival time is 14-16 months after diagnosis. Numerous efforts in therapy have not significantly altered the nearly uniform lethality of this malignancy. The Transforming Growth Factor beta (TGF-ß) signaling pathway plays a key role in GBM and is implicated in proliferation, invasion and therapy resistance. Several inhibitors of the TGF-ß pathway have entered clinical trials or are under development. In this work, the therapeutic potential of P144, a TGF-ß inhibitor peptide, was analyzed. P144 decreased proliferation, migration, invasiveness, and tumorigenicity in vitro, whereas apoptosis and anoikis were significantly increased for GBM cell lines. SMAD2 phosphorylation was reduced, together with a downregulation of SKI and an upregulation of SMAD7 at both transcriptional and translational levels. Additionally, P144 was able to impair tumor growth and increase survival in an in vivo flank model. Our findings suggest a potential effect of P144 in vitro and in vivo that is mediated by regulation of transcriptional target genes of the TGF-ß pathway, suggesting a therapeutic potential of P144 for GBM treatment.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Proteínas de Ligação a DNA/metabolismo , Glioblastoma/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Anoikis/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Relação Dose-Resposta a Droga , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos Nus , Invasividade Neoplásica , Fosforilação , Proteínas Proto-Oncogênicas/genética , Receptores de Fatores de Crescimento Transformadores beta , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad7/genética , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Carga Tumoral/efeitos dos fármacos
6.
J Pathol ; 239(1): 48-59, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27071481

RESUMO

Peritoneal adhesions (PAs) are fibrotic bands formed between bowel loops, solid organs, and the parietal peritoneum, which may appear following surgery, infection or endometriosis. They represent an important health problem with no effective treatment. Mesothelial cells (MCs) line the peritoneal cavity and undergo a mesothelial-to-mesenchymal transition (MMT) under pathological conditions, transforming into myofibroblasts, which are abundant in peritoneal fibrotic tissue. The aim of this study was to investigate if peritoneal MCs undergo a MMT contributing to the formation of post-surgical adhesions. Biopsies from patients with PAs were analysed by immunohistochemistry, immunofluorescence, and quantitative RT-PCR. A mouse model of PAs based on ischaemic buttons was used to modulate MMT by blocking the transforming growth factor-beta (TGF-ß) pathway. The severity of adhesions and MMT-related marker expression were studied. We observed myofibroblasts derived from the conversion of MCs in submesothelial areas of patients with PAs. In addition, MMT-related markers were dysregulated in adhesion zones when compared to distant normal peritoneal tissue of the same patient. In animal experiments, blockage of TGF-ß resulted in molecular reprogramming of markers related to the mesenchymal conversion of MCs and in a significant decrease in the severity of the adhesions. These data indicate for the first time that MMT is involved in PA pathogenesis. This finding opens new therapeutic strategies to interfere with adhesion formation by modulating MMT with a wide range of pharmacological agents.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Aderências Teciduais/etiologia , Actinas/metabolismo , Adulto , Idoso , Animais , Calbindina 2/metabolismo , Feminino , Fibroblastos/fisiologia , Humanos , Queratinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fragmentos de Peptídeos/farmacologia , Peritônio , Receptores de Fatores de Crescimento Transformadores beta , Proteína Smad3/metabolismo , Aderências Teciduais/patologia , Adulto Jovem
7.
J Biomed Mater Res B Appl Biomater ; 104(2): 385-94, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25826572

RESUMO

TGF-ß1 is the most related cytokine with the production of fibrotic tissue. It plays an important role on the production of collagen by fibroblasts and other types of cells. The inhibition of this cytokine reduces fibrosis in various types of tissue. Biofunctionalization of dental and orthopedic implants with biomolecules enables modification of the physical, chemical and biochemical properties of their surfaces to improve its biological and clinical performance. Our objective was to develop a reliable method to immobilize oligopeptides on Ti surfaces to obtain a surface with TGF-ß1 inhibitory activity that will potentially minimize fibrotic encapsulation of implants during the process of osseointegration. We covalently immobilized TGF-ß1 inhibitor P17-peptides on Ti surfaces and assessed by characterizing each step of the process that we successfully biofunctionalized the implant surfaces. High amounts of peptides were anchored and homogeneously distributed on the surfaces with mechanical and thermochemical stability after in vitro simulated challenges. Notably, the immobilized peptides retained their TGF-ß1 inhibitory activity in vitro. Thus, these biofunctional coatings are potential candidates for inducing a fast and reliable osseointegration in vivo.


Assuntos
Materiais Revestidos Biocompatíveis/química , Implantes Dentários , Implantes Experimentais , Peptídeos/química , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Linhagem Celular , Humanos , Proteínas Imobilizadas/química , Camundongos
8.
Cell Tissue Res ; 359(3): 829-39, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25557989

RESUMO

Colorectal cancer (CRC) cells often metastatize to the liver. Cancer-associated fibroblasts (CAFs) enhance metastasis by providing cytokines that create a favorable microenvironment and by inducing co-dissemination with tumor cells. However, the mechanisms of co-metastatization remain elusive. The aim of this study is to assess the role of TGFß1 in CRC cell-CAFs attachment and its impact on liver metastasis. CAFs were obtained after xenotransplantation of Mc38 cells into EGFP-C57BL/6 mice. Attachment experiments with CRC cells and CAFs (with or without TGFß1 and the inhibitory peptide P17) were carried out, as well as in vivo liver metastasis assays. TGFß1 induced adhesion of CRC cells to CAFs, whereas exposure to P17 abrogated this effect. Co-injection of Mc38 cells with CAFs intrasplenically increased liver metastasis, as compared to injection of tumor cells alone. Pretreatment of Mc38 cells with TGFß1 enhanced the metastatic burden, in comparison to untreated Mc38 + CAFs. TGFß1-pretreated Mc38 cells co-metastatized with CAFs to the liver in a highly efficient way. Importantly, the metastatic burden was significantly reduced (p < 0.001) when P17 was administered in mice. The number of PCNA+ and CD-31+ cells was also reduced by P17 in these animals, indicating a decrease in proliferation and angiogenesis upon TGFß1 signaling blockade. Through microarray analysis, we identified potential TGFß1-regulated genes that may mediate cancer cell-stroma interactions to increase metastasis. In conclusion, TGFß1 promotes co-travelling of CRC cells and CAFs to the liver to enhance metastasis. Our results strongly support the use of TGFß1 targeted drugs as a novel strategy to reduce liver metastasis in CRC patients.


Assuntos
Movimento Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Fibroblastos/patologia , Neoplasias Hepáticas/secundário , Fator de Crescimento Transformador beta1/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Imunofluorescência , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos Endogâmicos C57BL
9.
PLoS One ; 10(12): e0144489, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26720517

RESUMO

BACKGROUND: Hypertrophic scars are one of the most important complications in surgery due to their cosmetic and functional impairments. Previous studies in tissue fibrotic disorders have shown promising results by inhibiting the biological activity effect of Transforming Growth Factor-beta 1 (TGF-ß1). The aim of the current study was to determine the clinical effect of the inhibition of TGF-ß1 signaling in human hypertrophic scars implanted in nude mice by topical application of an inhibitor of TGF-ß1 (P144®). MATERIAL AND METHODS: A total of 30 human hypertrophic scars were implanted in 60 nude mice. The animals were divided in two groups, group A (placebo) and group B (treatment). Group C (basal) was considered as the preimplanted scar samples and they were not implanted in the nude mice. After the shedding period, topical application of a lipogel containing placebo (group A) or P144 (group B) was daily administered during two weeks. The animals were sacrificed upon completion of the study. Total area, thickness and collagen fibers area were measure and compared across all groups. Immunohistochemistry was also performed in order to quantify collagen type I and type III and elastic fiber expressions present in the dermis. RESULTS: Successful shedding was achieved in 83,3% of the xenografts. The mean time for shedding was 35±5.4 days. Statistically significant differences were found in the total area, collagen fibers area and thickness between the groups. Increased elastic fibers and decreased collagen I were found in the P144-treated group compared to the basal group. CONCLUSION: Topical application of an inhibitor of TGF-ß1 may promote scar maturation and clinical improvement of hypertrophic scar morphology features in an "in vivo" model in nude mice after two weeks of treatment.


Assuntos
Anticorpos Monoclonais/farmacologia , Cicatriz Hipertrófica/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Biomarcadores , Cicatriz Hipertrófica/tratamento farmacológico , Cicatriz Hipertrófica/etiologia , Cicatriz Hipertrófica/patologia , Colágeno/metabolismo , Modelos Animais de Doenças , Fibrilinas , Xenoenxertos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Proteínas dos Microfilamentos/metabolismo
10.
J Am Soc Nephrol ; 26(1): 173-82, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25012164

RESUMO

In patients undergoing peritoneal dialysis (PD), chronic exposure to nonphysiologic PD fluids elicits low-grade peritoneal inflammation, leading to fibrosis and angiogenesis. Phenotype conversion of mesothelial cells into myofibroblasts, the so-called mesothelial-to-mesenchymal transition (MMT), significantly contributes to the peritoneal dysfunction related to PD. A number of factors have been described to induce MMT in vitro and in vivo, of which TGF-ß1 is probably the most important. The vasoconstrictor peptide endothelin-1 (ET-1) is a transcriptional target of TGF-ß1 and mediates excessive scarring and fibrosis in several tissues. This work studied the contribution of ET-1 to the development of peritoneal damage and failure in a mouse model of PD. ET-1 and its receptors were expressed in the peritoneal membrane and upregulated on PD fluid exposure. Administration of an ET receptor antagonist, either bosentan or macitentan, markedly attenuated PD-induced MMT, fibrosis, angiogenesis, and peritoneal functional decline. Adenovirus-mediated overexpression of ET-1 induced MMT in human mesothelial cells in vitro and promoted the early cellular events associated with peritoneal dysfunction in vivo. Notably, TGF-ß1-blocking peptides prevented these actions of ET-1. Furthermore, a positive reciprocal relationship was observed between ET-1 expression and TGF-ß1 expression in human mesothelial cells. These results strongly support a role for an ET-1/TGF-ß1 axis as an inducer of MMT and subsequent peritoneal damage and fibrosis, and they highlight ET-1 as a potential therapeutic target in the treatment of PD-associated dysfunction.


Assuntos
Endotelina-1/fisiologia , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/patologia , Adenoviridae/genética , Animais , Células Cultivadas , Endotelina-1/metabolismo , Células Epiteliais/metabolismo , Epitélio/metabolismo , Feminino , Fibrose/metabolismo , Técnicas de Transferência de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Peritônio/metabolismo , Peritônio/patologia , Fenótipo , Fator de Crescimento Transformador beta1/metabolismo
11.
Cell Mol Biol Lett ; 19(4): 659-74, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25424912

RESUMO

We aim to demonstrate the role of Alk receptors in the response of hydrogel expansion. Chondrocytes from rat knees were cultured onto plastic and hydrogel surfaces. Alk-1 and Alk-5 were overexpressed or silenced and the effects on cells during expansion were tested and confirmed using peptide inhibitors for TGFß. Overexpression of Alk-5 and silencing of Alk-1 led to a loss of the chondrocyte phenotype, proving that they are key regulators of chondrocyte mechanosensing. An analysis of the gene expression profile during the expansion of these modified cartilage cells in plastic showed a better maintenance of the chondrocyte phenotype, at least during the first passages. These passages were also assayed in a mouse model of intramuscular chondrogenesis. Our findings indicate that these two receptors are important mediators in the response of chondrocytes to changes in the mechanical environment, making them suitable targets for modulating chondrogenesis. Inhibition of TGFß could also be effective in improving chondrocyte activity in aged or expanded cells that overexpress Alk-1.


Assuntos
Receptores de Ativinas/fisiologia , Condrócitos/fisiologia , Mecanotransdução Celular , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Animais , Proliferação de Células , Células Cultivadas , Condrócitos/transplante , Meios de Cultura/química , Metilação de DNA , Expressão Gênica , Hidrogéis/química , Camundongos Nus , Ratos Wistar , Receptor do Fator de Crescimento Transformador beta Tipo I , Fator de Crescimento Transformador beta/fisiologia
12.
Mol Biol Rep ; 41(10): 6335-41, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24990701

RESUMO

The determination of cell invasion by matrigel assay is usually evaluated by counting cells able to pass through a porous membrane and attach themselves to the other side, or by an indirect quantification of eluted specific cell staining dye by means of optical density measurement. This paper describes a quantitative analytical imaging approach for determining the invasiveness of tumor cells using a simple method, based on images processing with the public domain software, ImageJ. Images obtained by direct capture are split into the red channel, and the generated image is used to measure the area that cells cover in the picture. To overcome the several disadvantages that classical cell invasion determinations present, we propose this method because it generates more accurate and sensitive determinations, and it could be a reasonable option for improving the quality of the results. The cost-effective alternative method proposed is based on this simple and robust software that is worldwide affordable.


Assuntos
Movimento Celular , Rastreamento de Células/métodos , Colágeno , Técnicas In Vitro , Laminina , Proteoglicanas , Software , Linhagem Celular Tumoral , Combinação de Medicamentos , Humanos
13.
J Bone Joint Surg Am ; 96(13): 1109-1117, 2014 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-24990976

RESUMO

BACKGROUND: The use of autologous chondrocytes in cartilage repair is limited because of loss of the cartilage phenotype during expansion. The mechanosensing capacity of chondrocytes suggests evaluating the use of soft substrates for in vitro expansion. Our aim was to test the expansion of chondrocytes on collagen hydrogels to improve their capacity for chondrogenesis after a number of passages. METHODS: Rat cartilage cells were expanded on collagen hydrogels and on plastic, and the preservation of their chondrogenic capacity was tested both in vitro and in vivo. The expression of relevant markers during expansion on each surface was measured by real-time PCR (polymerase chain reaction). Expanded cells were then implanted in focal lesions in the medial femoral condyle of healthy sheep, and the newly formed tissue was analyzed by histomorphometry. RESULTS: Compared with cells cultured on plastic, cells cultured on hydrogels had better maintenance of the expression of the Sox9, Col2 (type-II collagen), FGFR3, and Alk-5 genes and decreased expression of Alk-1 and BMP-2. Pellets also showed increased expression of the cartilage marker genes aggrecan, Sox9, and Col2, and decreased expression of Col1 and Col10 (type-I and type-X collagen). ELISA (enzyme-linked immunosorbent assay) also showed a higher ratio of type-II to type-I collagen in pellets formed from cells expanded on hydrogels. When sheep chondrocytes were expanded and implanted in cartilage lesions in the femoral condyle of healthy sheep, hydrogel-expanded cells produced histologically better tissue compared with plastic-expanded cells. CONCLUSIONS: The expansion of chondrocytes on collagen hydrogels yielded cells with an improved chondrogenic capacity compared with cells expanded on plastic. CLINICAL RELEVANCE: The study results favor the use of hydrogel-expanded cells over the traditional plastic-expanded cells for autologous chondrocyte implantation.


Assuntos
Cartilagem Articular/citologia , Técnicas de Cultura de Células/métodos , Condrócitos/citologia , Condrogênese/fisiologia , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Animais , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Articulação do Joelho , Plásticos , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Ovinos
14.
Mol Cancer ; 13: 112, 2014 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-24884715

RESUMO

BACKGROUND: Transforming Growth Factor beta (TGF-ß) acts as a tumor suppressor early in carcinogenesis but turns into tumor promoter in later disease stages. In fact, TGF-ß is a known inducer of integrin expression by tumor cells which contributes to cancer metastatic spread and TGF-ß inhibition has been shown to attenuate metastasis in mouse models. However, carcinoma cells often become refractory to TGF-ß-mediated growth inhibition. Therefore identifying patients that may benefit from anti-TGF-ß therapy requires careful selection. METHODS: We performed in vitro analysis of the effects of exposure to TGF-ß in NSCLC cell chemotaxis and adhesion to lymphatic endothelial cells. We also studied in an orthotopic model of NSCLC the incidence of metastases to the lymph nodes after inhibition of TGF-ß signaling, ß3 integrin expression or both. RESULTS: We offer evidences of increased ß3-integrin dependent NSCLC adhesion to lymphatic endothelium after TGF-ß exposure. In vivo experiments show that targeting of TGF-ß and ß3 integrin significantly reduces the incidence of lymph node metastasis. Even more, blockade of ß3 integrin expression in tumors that did not respond to TGF-ß inhibition severely impaired the ability of the tumor to metastasize towards the lymph nodes. CONCLUSION: These findings suggest that lung cancer tumors refractory to TGF-ß monotherapy can be effectively treated using dual therapy that combines the inhibition of tumor cell adhesion to lymphatic vessels with stromal TGF-ß inhibition.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/terapia , Regulação Neoplásica da Expressão Gênica , Integrina beta3/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Fator de Crescimento Transformador beta1/genética , Animais , Anticorpos Monoclonais/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Técnicas de Cocultura , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Integrina beta3/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Metástase Linfática , Camundongos , Terapia de Alvo Molecular , Molécula L1 de Adesão de Célula Nervosa/antagonistas & inibidores , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
PLoS One ; 9(5): e96799, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24797128

RESUMO

Transforming growth factor ß (TGF-ß) is a powerful promoter of cancer progression and a key target for antitumor therapy. As cancer cells exhibit active cholesterol metabolism, high density lipoproteins (HDLs) appear as an attractive delivery system for anticancer TGFß-inhibitory molecules. We constructed a plasmid encoding a potent TGF-ß-blocking peptide (P144) linked to apolipoprotein A-I (ApoA-I) through a flexible linker (pApoLinkerP144). The ApoLinkerP144 sequence was then incorporated into a hepatotropic adeno-associated vector (AAVApoLinkerP144). The aim was to induce hepatocytes to produce HDLs containing a modified ApoA-I capable of blocking TGF-ß. We observed that transduction of the murine liver with pApoLinkerP144 led to the appearance of a fraction of circulating HDL containing the fusion protein. These HDLs were able to attenuate TGF-ß signaling in the liver and to enhance IL-12 -mediated IFN-γ production. Treatment of liver metastasis of MC38 colorectal cancer with AAVApoLinkerP144 resulted in a significant reduction of tumor growth and enhanced expression of IFN-γ and GM-CSF in cancerous tissue. ApoLinkerP144 also delayed MC38 liver metastasis in Rag2-/-IL2rγ-/- immunodeficient mice. This effect was associated with downregulation of TGF-ß target genes essential for metastatic niche conditioning. Finally, in a subset of ret transgenic mice, a model of aggressive spontaneous metastatic melanoma, AAVApoLinkerP144 delayed tumor growth in association with increased CD8+ T cell numbers in regional lymph nodes. In conclusion, modification of HDLs to transport TGF-ß-blocking molecules is a novel and promising approach to inhibit the growth of liver metastases by immunological and non-immunological mechanisms.


Assuntos
Neoplasias Colorretais/patologia , Lipoproteínas HDL/farmacologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Terapia de Alvo Molecular , Fator de Crescimento Transformador beta1/metabolismo , Animais , Complexo CD3/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Feminino , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Hepatócitos/metabolismo , Interferon gama/metabolismo , Fígado/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Metástase Neoplásica/prevenção & controle , Plasmídeos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Fator de Crescimento Transformador beta1/antagonistas & inibidores
16.
Am J Respir Crit Care Med ; 189(10): 1250-9, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24749506

RESUMO

RATIONALE: Nasopharyngeal carriage of Streptococcus pneumoniae is a prerequisite for invasive disease, but the majority of carriage episodes are asymptomatic and self-resolving. Interactions determining the development of carriage versus invasive disease are poorly understood but will influence the effectiveness of vaccines or therapeutics that disrupt nasal colonization. OBJECTIVES: We sought to elucidate immunological mechanisms underlying noninvasive pneumococcal nasopharyngeal carriage. METHODS: Pneumococcal interactions with human nasopharyngeal and bronchial fibroblasts and epithelial cells were investigated in vitro. A murine model of nasopharyngeal carriage and an experimental human pneumococcal challenge model were used to characterize immune responses in the airways during carriage. MEASUREMENTS AND MAIN RESULTS: We describe the previously unknown immunological basis of noninvasive carriage and highlight mechanisms whose perturbation may lead to invasive disease. We identify the induction of active transforming growth factor (TGF)-ß1 by S. pneumoniae in human host cells and highlight the key role for TGF-ß1 and T regulatory cells in the establishment and maintenance of nasopharyngeal carriage in mice and humans. We identify the ability of pneumococci to drive TGF-ß1 production from nasopharyngeal cells in vivo and show that an immune tolerance profile, characterized by elevated TGF-ß1 and high nasopharyngeal T regulatory cell numbers, is crucial for prolonged carriage of pneumococci. Blockade of TGF-ß1 signaling prevents prolonged carriage and leads to clearance of pneumococci from the nasopharynx. CONCLUSIONS: These data explain the mechanisms by which S. pneumoniae colonize the human nasopharynx without inducing damaging host inflammation and provide insight into the role of bacterial and host constituents that allow and maintain carriage.


Assuntos
Portador Sadio/imunologia , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/imunologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/imunologia , Animais , Biomarcadores/sangue , Portador Sadio/microbiologia , Portador Sadio/prevenção & controle , Humanos , Técnicas In Vitro , Camundongos , Nasofaringe/imunologia , Infecções Pneumocócicas/microbiologia , Infecções Pneumocócicas/prevenção & controle , Fatores de Tempo
17.
J Pathol ; 231(4): 517-31, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24114721

RESUMO

Peritoneal dissemination is a frequent metastatic route for cancers of the ovary and gastrointestinal tract. Tumour cells metastasize by attaching to and invading through the mesothelial cell (MC) monolayer that lines the peritoneal cavity. Metastases are influenced by carcinoma-associated fibroblasts (CAFs), a cell population that derives from different sources. Hence, we investigated whether MCs, through mesothelial-mesenchymal transition (MMT), were a source of CAFs during peritoneal carcinomatosis and whether MMT affected the adhesion and invasion of tumour cells. Biopsies from patients with peritoneal dissemination revealed the presence of myofibroblasts expressing mesothelial markers in the proximity of carcinoma implants. Prominent new vessel formation was observed in the peritoneal areas harbouring tumour cells when compared with tumour-free regions. The use of a mouse model of peritoneal dissemination confirmed the myofibroblast conversion of MCs and the increase in angiogenesis at places of tumour implants. Treatment of omentum MCs with conditioned media from carcinoma cell cultures resulted in phenotype changes reminiscent of MMT. Adhesion experiments demonstrated that MMT enhanced the binding of cancer cells to MCs in a ß1-integrin-dependent manner. Scanning electron microscopy imaging showed that the enhanced adhesion was mostly due to increased cell-cell interaction and not to a mere matrix exposure. Invasion assays suggested a reciprocal stimulation of the invasive capacity of tumour cells and MCs. Our results demonstrate that CAFs can derive from mesothelial cells during peritoneal metastasis. We suggest that MMT renders the peritoneum more receptive for tumour cell attachment/invasion and contributes to secondary tumour growth by promoting its vascularization.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Fibroblastos/patologia , Neoplasias Peritoneais/secundário , Animais , Biópsia , Adesão Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Meios de Cultivo Condicionados/farmacologia , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Células Epiteliais/ultraestrutura , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Fibroblastos/fisiologia , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Microscopia Eletrônica de Varredura , Invasividade Neoplásica , Transplante de Neoplasias , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/irrigação sanguínea , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/ultraestrutura
18.
PLoS One ; 8(5): e65434, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23741494

RESUMO

The purpose of this study was to assess the effects of transforming growth factor beta (TGF-ß) inhibitor peptides (P17 & P144) on early laser-induced choroidal neovascularization (LI-CNV) lesions in rats, two weeks after laser CNV induction. Seventy-one Long Evans rats underwent diode laser application in an established LI-CNV model. Baseline fluorescein angiography (FA) was performed 14 days following laser procedure, and treatments were administered 16 days post-laser application via different administration routes. Intravenous groups included control (IV-Control), P17 (IV-17), and P144 (IV-144) groups, whereas intravitreal groups included P17 (IVT-17), P144 (IVT-144), and a mixture of both peptides (IVT-17+144) (with fellow eyes receiving vehicle alone). CNV evolution was assessed using FA performed weekly for four weeks after treatment. Following sacrifice, VEGF, TGF-ß, COX-2, IGF-1, PAI-1, IL-6, MMP-2, MMP-9, and TNF-α gene expression was assessed using RT-PCR. VEGF and p-SMAD2 protein levels were also assessed by western-blot, while MMP-2 activity was assessed with gelatin zymography. Regarding the FA analysis, the mean CNV area was lower from the 3(rd) week in IVT-17 and IVT-144 groups, and also from the 2(nd) week in IVT-17+144. Biochemical analysis revealed that gene expression was lower for VEGF and COX-2 genes in IV-17 and IV-144 groups, VEGF gene in IVT-17+144 group and MMP-2 gene in IVT-17 and IVT-144 groups. VEGF protein expression was also decreased in IV-17, IV-144, IVT-17 and IVT-144, whereas pSMAD-2 levels were lower in IV-17, IV-144 and IVT-17+144 groups. Zymogram analysis revealed decreased MMP-2 activity in IV-17, IV-144, IVT-17 and IVT-144 groups. These data suggest that the use of TGF-ß inhibitor peptides (P17 & P144) decrease the development of early CNV lesions by targeting different mediators than those typically affected using current anti-angiogenic therapies. Its potential role in the treatment of early CNV appears promising as a single therapy or adjuvant to anti-VEGF drugs.


Assuntos
Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Neovascularização de Coroide/tratamento farmacológico , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Progressão da Doença , Angiofluoresceinografia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/administração & dosagem , Fosforilação/efeitos dos fármacos , Ratos , Receptores de Fatores de Crescimento Transformadores beta/administração & dosagem , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Antioxid Redox Signal ; 19(14): 1607-18, 2013 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-23350688

RESUMO

AIMS: The NADPH oxidases constitute a major source of superoxide anion (·O2(-)) in hypertension. Several studies suggest an important role of NADPH oxidases in different effects mediated by transforming growth factor-ß1 (TGF-ß1). We investigated whether a chronic treatment with P144, a peptide synthesized from type III TGF-ß1 receptor, inhibited NADPH oxidases in the renal cortex of spontaneously hypertensive rats (SHR). RESULTS: Here, we show that chronic administration of P144 significantly reduced the NADPH oxidase expression and activity as well as the oxidative stress observed in control vehicle-treated SHR (V-SHR). In addition, P144 was also able to reduce the significant increase in the renal fibrosis and in mRNA expression of different components of collagen metabolism, as well as in the levels of connective tissue growth factor observed in V-SHR. Finally, TGF-ß1-stimulated NRK52E exhibited a significant increase in NADPH oxidase expression and activity as well as a TGF-ß1-dependent intracellular pathway that were inhibited in the presence of P144. INNOVATION: Our experimental evidence suggests that reversing oxidative stress may be therapeutically useful in preventing fibrosis-associated renal damage. We show here that (i) the TGF-ß1-NADPH oxidases axis is crucial in the development of fibrosis in an experimental hypertensive renal disease animal model, and (ii) the use of P144 reverses TGF-ß1-dependent NADPH oxidase activity; thus, P144 may be considered a novel therapeutic tool in kidney disease associated with hypertension. CONCLUSION: We demonstrate that P144 inhibits NADPH oxidases and prevents oxidative stress in kidneys from hypertensive rats. Our data also suggest that these effects are associated with the renal antifibrotic effect of P144.


Assuntos
Rim/efeitos dos fármacos , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Proteoglicanas/química , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Animais , Fibrose/metabolismo , Fibrose/prevenção & controle , Rim/metabolismo , Rim/patologia , Masculino , Ratos , Ratos Endogâmicos SHR , Transdução de Sinais/efeitos dos fármacos
20.
Arthritis Rheum ; 65(1): 258-69, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22972461

RESUMO

OBJECTIVE: Clinical evidence suggests that the vascular abnormalities of systemic sclerosis (SSc) precede the onset of fibrosis, but molecular cues accounting for a possible vascular connection of SSc fibrosis have been elusive, although they have been searched for intensively. Since we had previously shown that connective tissue growth factor (CCN2), endowed with fibroblast-oriented activities, was overexpressed by endothelial cells (ECs) from SSc patients, we undertook this study to investigate its role and mechanisms in fibroblast activation. METHODS: Normal fibroblasts were challenged with conditioned medium of normal microvascular ECs (MVECs) and MVECs obtained from SSc patients with the diffuse form of the disease. Fibroblast invasion was studied using the Boyden chamber Matrigel assay. Fibroblast activation was evaluated by Western blotting and immunofluorescence of α-smooth muscle actin (α-SMA), vimentin, and type I collagen. Matrix metalloproteinase (MMP) production was evaluated by zymography and reverse transcription-polymerase chain reaction. Signal transduction and activation of the small GTPases RhoA and Rac1 were studied by Western blotting. Inhibition of SSc MVEC conditioned medium-dependent fibroblast activation was obtained by anti-CCN2 antibodies and the transforming growth factor ß (TGFß) antagonist peptide p17. RESULTS: SSc MVEC CCN2 stimulated fibroblast activation and invasion. Such activities depended on CCN2-induced overexpression of TGFß and its type I, II, and III receptors combined with overproduction of MMP-2 and MMP-9 gelatinases. All of these effects were reversed by the TGFß antagonist peptide p17. Motility increase required Rac1 activation and RhoA inhibition and was inhibited by an MMP inhibitor. These features connoted a mesenchymal style of cell invasion. Since fibroblast activation also fostered overexpression of α-SMA, vimentin, and type I collagen, the CCN2-dependent increase in fibroblast activities recapitulated the characteristics of a mesenchymal-to-mesenchymal transition. CONCLUSION: SSc MVECs recruit and activate dermal fibroblasts by induction of a CCN2/TGFß-dependent mesenchymal-to-mesenchymal transition.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Células Endoteliais/metabolismo , Fibroblastos/efeitos dos fármacos , Escleroderma Sistêmico/metabolismo , Pele/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Western Blotting , Colágeno , Combinação de Medicamentos , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Imunofluorescência , Humanos , Laminina , Masculino , Mesoderma/patologia , Proteoglicanas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Escleroderma Sistêmico/patologia , Transdução de Sinais , Pele/patologia , Fator de Crescimento Transformador beta/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...