Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Clin Oral Implants Res ; 27(2): e15-24, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25359716

RESUMO

OBJECTIVE: Our objective was to study the role of Collagen type-I (Col-I) coating on Magnesium-Zirconia (Mg-Zr) alloys, containing different quantities of Strontium (Sr), in enhancing the in vitro bioactivity and in vivo bone-forming and mineralisation properties of the implants. MATERIALS AND METHODS: MC3T3-E1 osteoblast cell line was used to analyse the in vitro properties of Col-I coated and uncoated alloys. Cell viability analysis was performed by MTT assay; cell attachment on alloy surfaces was studied by scanning electron microscopy (SEM); and gene profiling of bone-specific markers in cells plated on uncoated alloys was performed by Quantitative RT-PCR. In vivo studies were performed by implanting 2-mm-sized cylindrical pins of uncoated and coated alloys in male New Zealand white rabbits (n = 33). Bone formation and mineralisation was studied by Dual Energy X-ray Absorptiometry (DXA) and histological analysis at one and three months post-implantation. RESULTS: Our results clearly showed that Sr content and Col-I coating of Mg-Zr-Sr alloys significantly improved their bone inducing activity in vitro and in vivo. Osteoblasts on coated alloys showed better viability and surface binding than those on uncoated alloys. Sr inclusion in the alloys enhanced their bone-specific gene expression. The in vivo activity of implants with higher Sr and Col-I coating was superior to uncoated and other coated alloys as they showed faster bone induction and higher mineral content in the newly formed bone. CONCLUSION: Our results indicate that bone-forming and mineralising activity of Mg-Zr-Sr implants can be significantly improved by controlling their Sr content and coating their surface with Col-I.


Assuntos
Reabsorção Óssea/induzido quimicamente , Materiais Revestidos Biocompatíveis/farmacologia , Colágeno Tipo I/farmacologia , Magnésio/farmacologia , Osteogênese/efeitos dos fármacos , Estrôncio/farmacologia , Zircônio/farmacologia , Absorciometria de Fóton , Animais , Linhagem Celular , Sobrevivência Celular , Microscopia Eletrônica de Varredura , Osteoblastos , Coelhos , Reação em Cadeia da Polimerase em Tempo Real
2.
Eur J Clin Invest ; 44(12): 1169-76, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25315426

RESUMO

BACKGROUND: Angiotensin II (Ang II) and high-fat diet are implicated in causing pathological changes in the vascular endothelium, brain, kidney and liver. The association of aneurysm leading to histopathological changes in the splenic compartment remains elusive. Further, the salubrious credentials of antioxidants, especially α-tocopherol and ß-carotene in the resolution of splenic pathology have not been investigated. METHODS: Four-month-old Apoe(-/-) mice were used in the induction of aneurysm by infusing Ang II, and subsequently were orally administered with α-tocopherol and ß-carotene-enriched diet for 60 days. RESULTS: We observed splenomegaly in Ang II-infused aneurysm and high-fat diet-supplemented mice as compared to normal mice. These observations were further confirmed through histopathological investigations, demonstrating splenic follicular hypertrophy. We observed a remarkable decrease in the size of spleen in α-tocopherol and ß-carotene-treated Apoe(-/-) mice as compared with Ang II-treated animals. Furthermore, no marked changes in the histopathological splenic sections were seen in the ß-carotene-treated group. However, hyperplasia and proliferation of immature lymphocytes in the follicles were observed in the α-tocopherol-treated animals. We found that CD4+ T-cell levels were increased in the high-fat diet group relative to the control group and were decreased in the ß-carotene-treated animals. CONCLUSIONS: Our study provides evidence that Ang II infusion and high-fat supplementation induces abdominal aortic aneurysm that has pathological implications to the spleen. The use of ß-carotene but not α-tocopherol as an antioxidant markedly ameliorates the pathological changes in spleen.


Assuntos
Angiotensina II/toxicidade , Aneurisma da Aorta Abdominal/etiologia , Dieta Hiperlipídica/efeitos adversos , Esplenomegalia/etiologia , Vasoconstritores/toxicidade , Animais , Antioxidantes/farmacologia , Apolipoproteínas E/deficiência , Suplementos Nutricionais/efeitos adversos , Masculino , Camundongos Knockout , Linfócitos T/fisiologia , alfa-Tocoferol/farmacologia , beta Caroteno/farmacologia
3.
Mol Ther ; 17(4): 623-31, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19223869

RESUMO

Glucocorticoid receptors (GRs) are ubiquitous, nuclear hormone receptors residing in cell types of both cancer and noncancerous origin. It is not known whether cancer cell-associated GR alone can be selectively manipulated for delivery of exogenous genes to its nucleus for eliciting anticancer effect. We find that GR ligand, dexamethasone (Dex) in association with cationic lipoplex (termed as targeted lipoplex) could selectively manipulate GR in cancer cells alone for the delivery of transgenes in the nucleus, a phenomenon that remained unobserved in normal cells. The targeted lipoplex (i) showed GR-targeted transfections in all cancer cells experimented (P < 0.01), (ii) significantly diminished transfection in cancer cells when GR is downregulated (P < 0.01), and (iii) elicited specific nuclear translocation of targeted lipoplex in cancer cells, followed by upregulated transactivation of glucocorticoid response element (GRE)- promoted gene. Using anticancer gene, targeted lipoplex induced significant tumor growth retardation in mice in comparison to different control groups (P < 0.05). Interestingly, cell surface-associated Hsp90 in cancer cells assisted the intracellular uptake of GR-targeted lipoplex. Moreover, selective inhibition of Hsp90 in noncancer cells resulted in cancer cell-like, aberrant, GR activation. The current study discovers a therapeutically important, unique property of cancer cell associated-GR that may be linked to a compromised role of Hsp90.Molecular Therapy (2009) 17 4, 623-631 doi:10.1038/mt.2009.4.


Assuntos
Neoplasias/metabolismo , Receptores de Glucocorticoides/metabolismo , Linhagem Celular Tumoral , Proteínas de Choque Térmico HSP90/fisiologia , Humanos , Neoplasias/patologia , Receptores de Glucocorticoides/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ativação Transcricional , Transfecção , Transgenes
4.
Sci Rep ; 7(1): 6602, 2017 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-28747713

RESUMO

Targeted delivery of drugs to the brain is challenging due to the restricted permeability across the blood brain barrier (BBB). Gliomas are devastating cancers and their positive treatment outcome using Temozolomide (TMZ) is limited due to its short plasma half-life, systemic toxicity and limited access through the blood-brain barrier (BBB). Nanoparticles made of Lactoferrin (Lf) protein, have been shown to enhance the pharmacological properties of drugs. Here, we report the specific ability of Lf nanoparticles to cross BBB and target over-expressed Lf receptors on glioma for enhanced TMZ delivery. TMZ-loaded Lf nanoparticles (TMZ-LfNPs) were prepared by our previously reported sol-oil method. While the Lf protein in the NP matrix aids in transcytosis across the BBB and preferential tumor cell uptake, the pH responsiveness leads to TMZ release exclusively in the tumor microenvironment. Delivery through LfNPs results in an enhanced and sustained intracellular concentration of TMZ in GL261 cells in vitro along with improving its in vivo pharmacokinetics and brain accumulation. TMZ-LfNPs treatment results in a significant reduction of tumor volume, higher tumor cell apoptosis and improved median survival in glioma bearing mice. These results demonstrate that LfNPs present an efficient TMZ delivery platform for an effective treatment of gliomas.


Assuntos
Antineoplásicos Alquilantes/farmacocinética , Barreira Hematoencefálica/metabolismo , Portadores de Fármacos/farmacocinética , Glioma/tratamento farmacológico , Lactoferrina/farmacocinética , Temozolomida/farmacocinética , Animais , Antineoplásicos Alquilantes/administração & dosagem , Linhagem Celular Tumoral , Portadores de Fármacos/administração & dosagem , Lactoferrina/administração & dosagem , Camundongos , Nanopartículas/administração & dosagem , Temozolomida/administração & dosagem , Resultado do Tratamento
5.
Front Microbiol ; 4: 12, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23386847

RESUMO

Noroviruses cause most cases of acute viral gastroenteritis worldwide. The lack of a cell culture infection model for human norovirus necessitates the use of molecular methods and/or viral surrogate models amenable to cell culture to predict norovirus inactivation. Murine norovirus (MNV) may be used to construct a small animal model for studying the biology and pathogenesis of noroviruses because MNV is the only norovirus that replicates in cell culture and a small animal model. However, recent studies have shown that natural MNV infection is widespread in laboratory mouse colonies. We investigated MNV infection in both conventional and specific pathogen-free (SPF) genetically modified mice from Japan and the US, and commercial mice from several animal breeders in Japan, using serological and molecular techniques. MNV antibodies were detected in 67.3% of conventional mice and 39.1% of SPF mice from Japan and 62.5% of conventional mice from the US. MNV antibodies were also found in 20% of commercial SPF C57BL/6 mice from one of three breeders. Partial gene amplification of fecal isolates from infected animals showed that the isolates were homologous to reported MNV sequences. These results suggest that both conventional and SPF laboratory mice, including commercial mice, are widely infected with MNV, which might require considerable attention as an animal model of human disease.

6.
PLoS One ; 8(6): e67098, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23826202

RESUMO

Abdominal aortic aneurysm (AAA) is a common chronic degenerative disease characterized by progressive aortic dilation and rupture. The mechanisms underlying the role of α-tocopherol and ß-carotene on AAA have not been comprehensively assessed. We investigated if α-tocopherol and ß-carotene supplementation could attenuate AAA, and studied the underlying mechanisms utilized by the antioxidants to alleviate AAA. Four-months-old Apoe(-/-) mice were used in the induction of aneurysm by infusion of angiotensin II (Ang II), and were orally administered with α-tocopherol and ß-carotene enriched diet for 60 days. Significant increase of LDL, cholesterol, triglycerides and circulating inflammatory cells was observed in the Ang II-treated animals, and gene expression studies showed that ICAM-1, VCAM-1, MCP-1, M-CSF, MMP-2, MMP-9 and MMP-12 were upregulated in the aorta of aneurysm-induced mice. Extensive plaques, aneurysm and diffusion of inflammatory cells into the tunica intima were also noticed. The size of aorta was significantly (P = 0.0002) increased (2.24±0.20 mm) in the aneurysm-induced animals as compared to control mice (1.17±0.06 mm). Interestingly, ß-carotene dramatically controlled the diffusion of macrophages into the aortic tunica intima, and circulation. It also dissolved the formation of atheromatous plaque. Further, ß-carotene significantly decreased the aortic diameter (1.33±0.12 mm) in the aneurysm-induced mice (ß-carotene, P = 0.0002). It also downregulated ICAM-1, VCAM-1, MCP-1, M-CSF, MMP-2, MMP-9, MMP-12, PPAR-α and PPAR-γ following treatment. Hence, dietary supplementation of ß-carotene may have a protective function against Ang II-induced AAA by ameliorating macrophage recruitment in Apoe(-/-) mice.


Assuntos
Aneurisma da Aorta Abdominal/dietoterapia , Apolipoproteínas E/deficiência , Suplementos Nutricionais , Macrófagos/metabolismo , alfa-Tocoferol/administração & dosagem , beta Caroteno/administração & dosagem , Angiotensina II , Animais , Antioxidantes/administração & dosagem , Aorta Abdominal/imunologia , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/patologia , Apolipoproteínas E/genética , LDL-Colesterol/metabolismo , Modelos Animais de Doenças , Linfócitos/metabolismo , Linfócitos/patologia , Macrófagos/patologia , Masculino , Camundongos Knockout , Tamanho do Órgão , Placa Aterosclerótica/dietoterapia , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia
7.
Biomaterials ; 33(8): 2570-82, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22192536

RESUMO

Cell-penetrating peptide (CPP)-based delivery systems represent a strategy that facilitates DNA import efficiently and non-specifically into cells. To introduce specificity, we devised an approach that combines a cell-penetrating peptide, TAT-Mu (TM) and a targeting ligand, an HER2 antibody mimetic-affibody (AF), designated as TMAF to deliver nucleic acids into the cells. In this study, we synthesized TMAF protein and its truncated versions, i.e. MAF and AF, by expressing the corresponding plasmids in Escherichia coli BL21(DE3)pLysS cells. Purified TMAF binds DNA efficiently and protects plasmid DNA from DNaseI action. Transfection of HER2+ breast cancer cell lines MDA-MB-453, SK-OV-3, SK-BR-3 and an ovarian cancer cell line with plasmid DNA pCMVß-gal, resulted in enhanced ß-galactosidase activity when compared to control MDA-MB-231 cells. Maximal activity observed in MDA-MB-453 cells at DNA:TMAF:Protamine sulphate (PS) corresponding to 1:8:2 charge ratios. Further the observed gene transfection was resistant to serum, sensitive to the presence of free AF and non-toxic. Variants of TMAF although non-toxic, were far less efficient indicating the effective role of the TAT and Mu domains. The observed DNA uptake and reporter gene activity mediated by TMAFin vitro could be linked with the cell-surface density of tyrosine kinase receptor HER2 (ErbB2) levels estimated by Western blot. Further, we confirmed the efficacy of DNA transfer by TMAF protein in xenograft mouse models using MDA-MB-453 cells. Expression of ß-galactosidase as the reporter gene, upon intratumoral injection of DNA, in complex with TMAF, lends credence to specific DNA import and distribution within the tumor tissue that was attributed to high HER2 receptor overexpression in MDA-MB-453 cells. Through delivery of anti-TF hshRNA: TMAF: PS complex, we demonstrate specific knockdown of tissue factor (TF) in MDA-MB-453 cells in vitro. Most importantly, in a xenograft mouse model, we observe significant (P<0.05) and specific reduction of tumor volume when anti-TF hshRNA: TMAF: PS complex was injected intratumorally. Collectively our data indicate that AF-based chimeric peptides with nucleic acid binding properties may provide an effective tumor specific strategy to deliver therapeutic nucleic acids.


Assuntos
Materiais Biocompatíveis/farmacologia , Peptídeos Penetradores de Células/farmacologia , Terapia de Alvo Molecular/métodos , Receptor ErbB-2/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Desoxirribonuclease I/metabolismo , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Técnicas de Transferência de Genes , Humanos , Ligantes , Camundongos , Camundongos SCID , Plasmídeos/metabolismo , Protaminas , Estabilidade Proteica/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Receptor ErbB-2/metabolismo , Soro/metabolismo , Tromboplastina/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa