Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PDA J Pharm Sci Technol ; 74(4): 377-393, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32179711

RESUMO

Cleaning validation acceptance criteria in multiproduct facilities are established using maximum allowable carryover calculations. Carryover calculations incorporate the shared equipment surface area between two products to ensure that an acceptable limit for residue from the previously manufactured product to the subsequent product is determined. The shared surface area can be limited to areas where carryover presents the highest risk to product quality or patient safety. In these cases, specifically for biologic drug substance manufacturing, the shared surface area is limited to equipment after the purification process based on the assumption that the purification process would remove potential product fragment residues from the previous product. Until now, this assumption has been based on empirical knowledge without experimental data quantifying the clearance or removal of potential residues. We present a three-part study that determined the effects of cleaning conditions on selected monoclonal antibodies (mAbs) and the generation of degraded fragments and evaluated the clearance of both the degraded mAb1 in a laboratory setting and the degraded fragments in the presence of a subsequent product, assessing the risk of co-purification. Several analytical techniques were used, including gel electrophoresis, capillary zone electrophoresis/laser-induced florescence detection, and liquid chromatography-mass spectrometry. Protein fragment generation was demonstrated for five different mAbs from different immunoglobulin G subclasses. The clearance of the degraded fragments in the absence and presence of the subsequent product was demonstrated by calculating fold clearance and log reduction value (LRV) for each chromatography step. The data showed that the fragments generated during cleaning could be removed by the purification process. The fold clearances were determined to be values of 5400 (3.7 LRV) in the absence of subsequent product and 4428 (3.6 LRV) in the presence of subsequent product. The results supported the removal of product residues from shared surface areas by the purification process in multiproduct biologic drug substance manufacturing facilities.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Contaminação de Medicamentos/prevenção & controle , Contaminação de Equipamentos/prevenção & controle , Tecnologia Farmacêutica , Anticorpos Monoclonais/efeitos adversos , Segurança do Paciente , Proteólise , Controle de Qualidade , Medição de Risco , Tecnologia Farmacêutica/instrumentação , Tecnologia Farmacêutica/normas
2.
Biomaterials ; 83: 12-22, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26773660

RESUMO

In this study, we used deoxyribozyme (DNAzyme) functionalized gold nanoparticles (AuNPs) to catalytically silence tumor necrosis factor-α (TNF-α) in vivo as a potential therapeutic for myocardial infarction (MI). Using primary macrophages as a model, we demonstrated 50% knockdown of TNF-α, which was not attainable using Lipofectamine-based approaches. Local injection of DNAzyme conjugated to gold particles (AuNPs) in the rat myocardium yielded TNF-α knockdown efficiencies of 50%, which resulted in significant anti-inflammatory effects and improvement in acute cardiac function following MI. Our results represent the first example showing the use of DNAzyme AuNP conjugates in vivo for viable delivery and gene regulation. This is significant as TNF-α is a multibillion dollar drug target implicated in many inflammatory-mediated disorders, thus underscoring the potential impact of DNAzyme-conjugated AuNPs.


Assuntos
Anti-Inflamatórios/uso terapêutico , DNA Catalítico/metabolismo , Técnicas de Silenciamento de Genes , Ouro/química , Nanopartículas Metálicas/química , Infarto do Miocárdio/tratamento farmacológico , Fator de Necrose Tumoral alfa/genética , Animais , Anti-Inflamatórios/farmacologia , Morte Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Fluorescência , Coração/efeitos dos fármacos , Coração/fisiopatologia , Testes de Função Cardíaca/efeitos dos fármacos , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Infarto do Miocárdio/complicações , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Células RAW 264.7 , Ratos Sprague-Dawley , Distribuição Tecidual/efeitos dos fármacos
3.
J Cardiovasc Pharmacol Ther ; 20(1): 93-103, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24831254

RESUMO

In the adult heart, catalase (CAT) activity increases appropriately with increasing levels of hydrogen peroxide, conferring cardioprotection. This mechanism is absent in the newborn for unknown reasons. In the present study, we examined how the posttranslational modification of CAT contributes to its activation during hypoxia/ischemia and the role of c-Abl tyrosine kinase in this process. Hypoxia studies were carried out using primary cardiomyocytes from adult (>8 weeks) and newborn rats. Following hypoxia, the ratio of phosphorylated to total CAT and c-Abl in isolated newborn rat myocytes did not increase and were significantly lower (1.3- and 4.2-fold, respectively; P < .05) than their adult counterparts. Similarly, there was a significant association (P < .0005) between c-Abl and CAT in adult cells following hypoxia (30.9 ± 8.2 to 70.7 ± 13.1 au) that was absent in newborn myocytes. Although ubiquitination of CAT was higher in newborns compared to adults following hypoxia, inhibition of this did not improve CAT activity. When a c-Abl activator (5-(1,3-diaryl-1H-pyrazol-4-yl)hydantoin [DPH], 200 µmol/L) was administered prior to hypoxia, not only CAT activity was significantly increased (P < .05) but also phosphorylation levels were also significantly improved (P < .01) in these newborn myocytes. Additionally, ischemia-reperfusion (IR) studies were performed using newborn (4-5 days) rabbit hearts perfused in a Langendorff method. The DPH given as an intracardiac injection into the right ventricle of newborn rabbit resulted in a significant improvement (P < .002) in the recovery of developed pressure after IR, a key indicator of cardiac function (from 74.6% ± 6.6% to 118.7% ± 10.9%). In addition, CAT activity was increased 3.92-fold (P < .02) in the same DPH-treated hearts. Addition of DPH to adult rabbits in contrast had no significant effect (from 71.3% ± 10.7% to 59.4% ± 12.1%). Therefore, in the newborn, decreased phosphorylation of CAT by c-Abl potentially mediates IR-induced dysfunction, and activation of c-Abl may be a strategy to prevent ischemic injury associated with surgical procedures.


Assuntos
Catalase/metabolismo , Genes abl/fisiologia , Miócitos Cardíacos/enzimologia , Proteínas Tirosina Quinases/fisiologia , Animais , Animais Recém-Nascidos , Hipóxia Celular/fisiologia , Ativação Enzimática/fisiologia , Coelhos , Ratos , Ratos Sprague-Dawley
4.
Biomaterials ; 35(28): 8103-12, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24974008

RESUMO

Myocardial infarction is the leading cause of death worldwide and phase I clinical trials utilizing cardiac progenitor cells (CPCs) have shown promising outcomes. Notch1 signaling plays a critical role in cardiac development and in the survival, cardiogenic lineage commitment, and differentiation of cardiac stem/progenitor cells. In this study, we functionalized self-assembling peptide (SAP) hydrogels with a peptide mimic of the Notch1 ligand Jagged1 (RJ) to evaluate the therapeutic benefit of CPC delivery in the hydrogels in a rat model of myocardial infarction. The behavior of CPCs cultured in the 3D hydrogels in vitro including gene expression, proliferation, and growth factor production was evaluated. Interestingly, we observed Notch1 activation to be dependent on hydrogel polymer density/stiffness with synergistic increase in presence of RJ. Our results show that RJ mediated Notch1 activation depending on hydrogel concentration differentially regulated cardiogenic gene expression, proliferation, and growth factor production in CPCs in vitro. In rats subjected to experimental myocardial infarction, improvement in acute retention and cardiac function was observed following cell therapy in RJ hydrogels compared to unmodified or scrambled peptide containing hydrogels. This study demonstrates the potential therapeutic benefit of functionalizing SAP hydrogels with RJ for CPC based cardiac repair.


Assuntos
Materiais Biocompatíveis/química , Hidrogéis/química , Infarto do Miocárdio/metabolismo , Receptor Notch1/metabolismo , Células-Tronco/citologia , Animais , Células CHO , Diferenciação Celular , Movimento Celular , Corantes/química , Cricetinae , Cricetulus , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Miocárdio/patologia , Miócitos Cardíacos/citologia , Peptídeos/química , Polímeros/química , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
5.
Int J Mol Sci ; 15(5): 9036-50, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24853285

RESUMO

Cardiovascular disease is the leading cause of death in the United States and new treatment options are greatly needed. Oxidative stress is increased following myocardial infarction and levels of antioxidants decrease, causing imbalance that leads to dysfunction. Therapy involving catalase, the endogenous scavenger of hydrogen peroxide (H2O2), has been met with mixed results. When over-expressed in cardiomyocytes from birth, catalase improves function following injury. When expressed in the same cells in an inducible manner, catalase showed a time-dependent response with no acute benefit, but a chronic benefit due to altered remodeling. In myeloid cells, catalase over-expression reduced angiogenesis during hindlimb ischemia and prevented monocyte migration. In the present study, due to the large inflammatory response following infarction, we examined myeloid-specific catalase over-expression on post-infarct healing. We found a significant increase in catalase levels following infarction that led to a decrease in H2O2 levels, leading to improved acute function. This increase in function could be attributed to reduced infarct size and improved angiogenesis. Despite these initial improvements, there was no improvement in chronic function, likely due to increased fibrosis. These data combined with what has been previously shown underscore the need for temporal, cell-specific catalase delivery as a potential therapeutic option.


Assuntos
Catalase/metabolismo , Células Mieloides/enzimologia , Infarto do Miocárdio/metabolismo , Animais , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Ecocardiografia , Fibrose/patologia , Peróxido de Hidrogênio/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/citologia , Células Mieloides/metabolismo , Infarto do Miocárdio/patologia , Neovascularização Fisiológica , Peroxidases/metabolismo
6.
Sci Rep ; 4: 4257, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24604065

RESUMO

There is a great need for the development of therapeutic strategies that can target biomolecules to damaged myocardium. Necrosis of myocardium during a myocardial infarction (MI) is characterized by extracellular release of DNA, which can serve as a potential target for ischemic tissue. Hoechst, a histological stain that binds to double-stranded DNA can be conjugated to a variety of molecules. Insulin-like growth factor-1 (IGF-1), a small protein/polypeptide with a short circulating-half life is cardioprotective following MI but its clinical use is limited by poor delivery, as intra-myocardial injections have poor retention and chronic systemic presence has adverse side effects. Here, we present a novel delivery vehicle for IGF-1, via its conjugation to Hoechst for targeting infarcted tissue. Using a mouse model of ischemia-reperfusion, we demonstrate that intravenous delivery of Hoechst-IGF-1 results in activation of Akt, a downstream target of IGF-1 and protects from cardiac fibrosis and dysfunction following MI.


Assuntos
DNA/metabolismo , Fator de Crescimento Insulin-Like I/administração & dosagem , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Espaço Extracelular/metabolismo , Fibrose , Humanos , Fator de Crescimento Insulin-Like I/química , Macrófagos/metabolismo , Masculino , Camundongos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Ligação Proteica , Transporte Proteico
7.
Stem Cells Dev ; 22(17): 2414-24, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23544670

RESUMO

There are a limited number of therapies available to prevent heart failure following myocardial infarction. One novel therapy that is currently being pursued is the implantation of cardiac progenitor cells (CPCs); however, their responses to oxidative stress during differentiation have yet to be elucidated. The objective of this study was to determine the effect of hydrogen peroxide (H2O2) treatment on CPC differentiation in vitro, as well as the effect of H2O2 preconditioning before implantation following ischemia-reperfusion (I/R) injury. CPCs were isolated and cloned from adult rat hearts, and then cultured in the absence or presence of H2O2 for 2 or 5 days. CPC survival was assessed with Annexin V, and cellular differentiation was evaluated through mRNA expression for cardiogenic genes. We found that 100 µM H2O2 decreased serum withdrawal-induced apoptosis by at least 45% following both 2 and 5 days of treatment. Moreover, 100 µM H2O2 treatment for 2 days significantly increased endothelial and smooth muscle markers compared to time-matched untreated CPCs. However, continued H2O2 treatment significantly decreased these markers. Left ventricular cardiac function was assessed 28 days after I/R and I/R with the implantation of Luciferase/GFP(+) CPCs, which were preconditioned with 100 µM H2O2 for 2 days. Hearts implanted with Luciferase/GFP(+) CPCs had significant improvement in both positive and negative dP/dT over I/R. Furthermore, cardiac fibrosis was significantly decreased in the preconditioned cells versus both I/R alone and I/R with control cells. We also observed a significant increase in endothelial cell density in the preconditioned CPC hearts compared to untreated CPC hearts, which also coincided with a higher density of Luciferase(+) vessels. These findings suggest that preconditioning of CPCs with H2O2 for 2 days stimulates neoangiogenesis in the peri-infarct area following I/R injury and could be a viable therapeutic option to prevent heart failure.


Assuntos
Insuficiência Cardíaca/prevenção & controle , Peróxido de Hidrogênio/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão/tratamento farmacológico , Células-Tronco/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Fibrose/tratamento farmacológico , Expressão Gênica , Insuficiência Cardíaca/tratamento farmacológico , Peróxido de Hidrogênio/metabolismo , Precondicionamento Isquêmico Miocárdico/métodos , Masculino , Contração Miocárdica/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Células-Tronco/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos
8.
Stem Cell Res Ther ; 4(2): 43, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23597145

RESUMO

INTRODUCTION: Administration of bone marrow-derived mesenchymal stem cells (MSCs) after myocardial infarction (MI) results in modest functional improvements. However; the effect of microenvironment changes after MI, such as elevated levels of oxidative stress on cardiogenic gene expression of MSCs, remains unclear. METHODS: MSCs were isolated from the bone marrow of adult rats and treated for 1 week with H2O2 (0.1 to 100 µM) or 48 hours with glucose oxidase (GOX; 0 to 5 mU/ml) to mimic long-term pulsed or short-term continuous levels of H2O2, respectively. RESULTS: In 100 µM H2O2 or 5 mU/ml GOX-treated MSCs, mRNA expression of selected endothelial genes (Flt1, vWF, PECAM1), and early cardiac marker (nkx2-5, αMHC) increased significantly, whereas early smooth muscle markers (smooth muscle α-actin and sm22α) and fibroblast marker vimentin decreased, as measured with real-time PCR. Interestingly, mRNA expression and activity of the cell-surface receptor Notch1 were significantly increased, as were its downstream targets, Hes5 and Hey1. Co-treatment of MSCs with 100 µM H2O2 and a γ-secretase inhibitor that prevents Notch signaling abrogated the increase in cardiac and endothelial genes, while augmenting the decrease in smooth muscle markers. Further, on GOX treatment, a significant increase in Wnt11, a downstream target of Notch1, was observed. Similar results were obtained with adult rat cardiac-derived progenitor cells. CONCLUSIONS: These data suggest that H2O2- or GOX-mediated oxidative stress upregulates Notch1 signaling, which promotes cardiogenic gene expression in adult stem/progenitor cells, possibly involving Wnt11. Modulating the balance between Notch activation and H2O2-mediated oxidative stress may lead to improved adult stem cell-based therapies for cardiac repair and regeneration.


Assuntos
Células-Tronco Mesenquimais/citologia , Estresse Oxidativo , Receptor Notch1/metabolismo , Actinas/metabolismo , Animais , Células da Medula Óssea/citologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Dipeptídeos/farmacologia , Expressão Gênica/efeitos dos fármacos , Glucose Oxidase/farmacologia , Peróxido de Hidrogênio/toxicidade , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/genética , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo
9.
Biomaterials ; 34(17): 4235-41, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23489923

RESUMO

Since the successful generation of induced pluripotent stem cells (iPSC) from adult somatic cells using integrating-viral methods, various methods have been tried for iPSC generation using non-viral and non-integrating technique for clinical applications. Recently, various non-viral approaches such as protein, mRNA, microRNA, and small molecule transduction were developed to avoid genomic integration and generate stem cell-like cells from mouse and human fibroblasts. Despite these successes, there has been no successful generation of iPSC from bone marrow (BM)-derived hematopoietic cells derived using non-viral methods to date. Previous reports demonstrate the ability of polymeric micro and nanoparticles made from polyketals to deliver various molecules to macrophages. MicroRNA-loaded nanoparticles were created using the polyketal polymer PK3 (PK3-miR) and delivered to somatic cells for 6 days, resulting in the formation of colonies. Isolated cells from these colonies were assayed and substantial induction of the pluripotency markers Oct4, Sox2, and Nanog were detected. Moreover, colonies transferred to feeder layers also stained positive for pluripotency markers including SSEA-1. Here, we demonstrate successful activation of pluripotency-associated genes in mouse BM-mononuclear cells using embryonic stem cell (ESC)-specific microRNAs encapsulated in the acid sensitive polyketal PK3. These reprogramming results demonstrate that a polyketal-microRNA delivery vehicle can be used to generate various reprogrammed cells without permanent genetic manipulation in an efficient manner.


Assuntos
Acetais/farmacologia , Células da Medula Óssea/citologia , Técnicas de Transferência de Genes , Células-Tronco Pluripotentes Induzidas/citologia , Leucócitos Mononucleares/citologia , MicroRNAs/metabolismo , Nanopartículas/química , Polímeros/farmacologia , Acetais/química , Adulto , Animais , Forma Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/metabolismo , Cinética , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Antígenos CD15/metabolismo , Camundongos , Nanopartículas/ultraestrutura , Fator 3 de Transcrição de Octâmero/metabolismo , Polímeros/química
10.
PLoS One ; 7(11): e50980, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226440

RESUMO

Acute myocardial infarction (MI) caused by ischemia and reperfusion (IR) is the most common cause of cardiac dysfunction due to local cell death and a temporally regulated inflammatory response. Current therapeutics are limited by delivery vehicles that do not address spatial and temporal aspects of healing. The aim of this study was to engineer biotherapeutic delivery materials to harness endogenous cell repair to enhance myocardial repair and function. We have previously engineered poly(ethylene glycol) (PEG)-based hydrogels to present cell adhesive motifs and deliver VEGF to promote vascularization in vivo. In the current study, bioactive hydrogels with a protease-degradable crosslinker were loaded with hepatocyte and vascular endothelial growth factors (HGF and VEGF, respectively) and delivered to the infarcted myocardium of rats. Release of both growth factors was accelerated in the presence of collagenase due to hydrogel degradation. When delivered to the border zones following ischemia-reperfusion injury, there was no acute effect on cardiac function as measured by echocardiography. Over time there was a significant increase in angiogenesis, stem cell recruitment, and a decrease in fibrosis in the dual growth factor delivery group that was significant compared with single growth factor therapy. This led to an improvement in chronic function as measured by both invasive hemodynamics and echocardiography. These data demonstrate that dual growth factor release of HGF and VEGF from a bioactive hydrogel has the capacity to significantly improve cardiac remodeling and function following IR injury.


Assuntos
Materiais Biocompatíveis/química , Sistemas de Liberação de Medicamentos/métodos , Coração/fisiopatologia , Fator de Crescimento de Hepatócito/administração & dosagem , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Peptídeo Hidrolases/metabolismo , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Animais , Movimento Celular/efeitos dos fármacos , Separação Celular , Fibrose , Coração/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/diagnóstico por imagem , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/fisiopatologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Ultrassonografia , Fator A de Crescimento do Endotélio Vascular/farmacologia
11.
Stem Cells Dev ; 21(17): 3136-46, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-22758933

RESUMO

Transplantation of cardiac progenitor cells (CPCs) is currently in early clinical testing as a potential therapeutic strategy. Superoxide is increased in the ischemic myocardium and poor survival of cells is one of the major limitations of cell transplantation therapy. Superoxide dismutase (SOD) levels were analyzed in c-kit-positive CPCs isolated from rat myocardium to identify their roles in protection against oxidative stress-induced apoptosis in vitro. CPCs were subjected to oxidative stress using xanthine/xanthine oxidase (XXO) and little apoptosis was detected. CPCs contained significantly higher levels of SOD1 and SOD2 as compared with adult cardiac cell types, both at the protein and activity levels. Both SOD1 and SOD2 were increased by XXO at the mRNA and protein level, suggesting compensatory adaptation. Only knockdown of SOD2 and not SOD1 with siRNA sensitized the cells to XXO-apoptosis, despite only accounting for 10% of total SOD levels. Finally, we found XXO activated Akt within 10 min, and this regulated both SOD2 gene expression and protection against apoptosis. Rat CPCs are resistant to superoxide-induced cell death, primarily through higher levels of SOD2 compared to adult cardiac-derived cells. Exposure to superoxide increases expression of SOD2 in an Akt-dependent manner and regulates CPC survival during oxidative stress.


Assuntos
Regulação Enzimológica da Expressão Gênica , Miocárdio/citologia , Miócitos Cardíacos/enzimologia , Células-Tronco/enzimologia , Superóxido Dismutase/metabolismo , Animais , Apoptose , Sobrevivência Celular , Ativação Enzimática , Ensaios Enzimáticos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Técnicas de Silenciamento de Genes , Miocárdio/enzimologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Fatores de Tempo , Transfecção , Xantina/efeitos adversos , Xantina Oxidase/efeitos adversos
12.
Bioorg Med Chem Lett ; 21(17): 4980-4, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21684742

RESUMO

Metabolic oligosaccharide engineering (MOE) provides a method to install novel chemical functional groups into the glycocalyx of living cells. In this Letter we use this technology to compare the impact of replacing natural sialic acid, GalNAc, and GlcNAc with their thiol-bearing counterparts in Jurkat and HL-60 cells. When incubated in the presence of gold-coated nanofibers, only Jurkat cells incubated with Ac(5)ManNTGc-an analogue that installs thiols into sialosides-experienced a distinctive 'spreading' morphology. The comparison of Ac(5)ManNTGc with Ac(5)GalNTGc and Ac(5)GlcNTGc in the two cell lines implicated sialosides of N-linked glycans as critical molecular mediators of the unusual responses evoked in the Jurkat line.


Assuntos
Carboidratos/química , Ouro/química , Nanofibras , Polissacarídeos/metabolismo , Compostos de Sulfidrila/química , Glicosilação , Humanos , Células Jurkat , Microscopia Eletrônica de Varredura , Polissacarídeos/química
13.
Biomaterials ; 32(23): 5427-37, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21549424

RESUMO

This study combines metabolic oligosaccharide engineering (MOE), a technology where the glycocalyx of living cells is endowed with chemical features not normally found in sugars, with custom-designed three-dimensional biomaterial substrates to enhance the adhesion of cancer cells and control their morphology and gene expression. Specifically, Ac(5)ManNTGc, a thiol-bearing analog of N-acetyl-d-mannosamine (ManNAc) was used to introduce thiolated sialic acids into the glycocalyx of human Jurkat T-lymphoma derived cells. In parallel 2D films and 3D electrospun nanofibrous scaffolds were prepared from polyethersulfone (PES) and (as controls) left unmodified or aminated. Alternately, the materials were malemided or gold-coated to provide bio-orthogonal binding partners for the thiol groups newly expressed on the cell surface. Cell attachment was modulated by both the topography of the substrate surface and by the chemical compatibility of the binding interface between the cell and the substrate; a substantial increase in binding for normally non-adhesive Jurkat line for 3D scaffold compared to 2D surfaces with an added degree of adhesion resulting from chemoselective binding to malemidede-derivatived or gold-coated surfaces. In addition, the morphology of the cells attached to the 3D scaffolds via MOE-mediated adhesion was dramatically altered and the expression of genes involved in cell adhesion changed in a time-dependent manner. This study showed that cell adhesion could be enhanced, gene expression modulated, and cell fate controlled by introducing the 3D topograhical cues into the growth substrate and by creating a glycoengineered binding interface where the chemistry of both the cell surface and biomaterials scaffold was controlled to facilitate a new mode of carbohydrate-mediated adhesion.


Assuntos
Materiais Biocompatíveis/química , Bioengenharia/métodos , Adesão Celular , Glicocálix/metabolismo , Neoplasias/patologia , Oligossacarídeos/metabolismo , Aminação , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/farmacologia , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Glicosaminoglicanos/metabolismo , Ouro/química , Hexosaminas/metabolismo , Humanos , Receptores de Hialuronatos/genética , Integrina beta1/genética , Células Jurkat , Maleimidas/química , Metaloproteinase 9 da Matriz/genética , Microscopia Eletrônica de Varredura , Neoplasias/metabolismo , Polímeros/química , Polímeros/farmacologia , Sulfonas/química , Sulfonas/farmacologia , Alicerces Teciduais/química
14.
PLoS One ; 5(11): e13883, 2010 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-21079735

RESUMO

BACKGROUND: This study was inspired by coalescing evidence that magnetic therapy may be a viable treatment option for certain diseases. This premise is based on the ability of moderate strength fields (i.e., 0.1 to 1 Tesla) to alter the biophysical properties of lipid bilayers and in turn modulate cellular signaling pathways. In particular, previous results from our laboratory (Wang et al., BMC Genomics, 10, 356 (2009)) established that moderate strength static magnetic field (SMF) exposure altered cellular endpoints associated with neuronal function and differentiation. Building on this background, the current paper investigated SMF by focusing on the adenosine A(2A) receptor (A(2A)R) in the PC12 rat adrenal pheochromocytoma cell line that displays metabolic features of Parkinson's disease (PD). METHODOLOGY AND PRINCIPAL FINDINGS: SMF reproduced several responses elicited by ZM241385, a selective A(2A)R antagonist, in PC12 cells including altered calcium flux, increased ATP levels, reduced cAMP levels, reduced nitric oxide production, reduced p44/42 MAPK phosphorylation, inhibited proliferation, and reduced iron uptake. SMF also counteracted several PD-relevant endpoints exacerbated by A(2A)R agonist CGS21680 in a manner similar to ZM241385; these include reduction of increased expression of A(2A)R, reversal of altered calcium efflux, dampening of increased adenosine production, reduction of enhanced proliferation and associated p44/42 MAPK phosphorylation, and inhibition of neurite outgrowth. CONCLUSIONS AND SIGNIFICANCE: When measured against multiple endpoints, SMF elicited qualitatively similar responses as ZM241385, a PD drug candidate. Provided that the in vitro results presented in this paper apply in vivo, SMF holds promise as an intriguing non-invasive approach to treat PD and potentially other neurological disorders.


Assuntos
Magnetismo , Receptor A2A de Adenosina/metabolismo , Triazinas/farmacologia , Triazóis/farmacologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Trifosfato de Adenosina/metabolismo , Análise de Variância , Animais , Western Blotting , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , AMP Cíclico/metabolismo , Ferro/metabolismo , Magnetoterapia/métodos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Células PC12 , Doença de Parkinson/metabolismo , Doença de Parkinson/terapia , Fenetilaminas/farmacologia , Fosforilação/efeitos dos fármacos , Ratos
15.
Curr Opin Chem Biol ; 13(5-6): 565-72, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19747874

RESUMO

Metabolic glycoengineering, a technique pioneered almost two decades ago wherein monosaccharide analogs are utilized to install non-natural sugars into the glycocalyx of mammalian cells, has undergone a recent flurry of advances spurred by efforts to make the methodology more efficient. This article describes the versatility of metabolic glycoengineering, which is a prime example of 'chemical glycobiology,' and gives an overview of its capability to endow complex carbohydrates in living cells and animals with interesting (and useful!) functionalities. Then an overview is provided describing how acylated monosaccharides, a class of molecules originally intended to be efficiently-used, membrane-permeable metabolic intermediates, have led to the discovery that a subset of these compounds (e.g. tributanoylated hexosamines) display unanticipated 'scaffold-dependent' activities; this finding establishes these molecules as a versatile platform for drug discovery.


Assuntos
Bioengenharia/métodos , Descoberta de Drogas/métodos , Hexosaminas/química , Hexosaminas/metabolismo , Animais , Produtos Biológicos/química , Produtos Biológicos/genética , Produtos Biológicos/metabolismo , Biopolímeros/química , Biopolímeros/genética , Biopolímeros/metabolismo , Hexosaminas/genética , Humanos
16.
BMC Genomics ; 10: 356, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19653909

RESUMO

BACKGROUND: Compelling evidence exists that magnetic fields modulate living systems. To date, however, rigorous studies have focused on identifying the molecular-level biosensor (e.g., radical ion pairs or membranes) or on the behavior of whole animals leaving a gap in understanding how molecular effects are translated into tissue-wide and organism-level responses. This study begins to bridge this gulf by investigating static magnetic fields (SMF) through global mRNA profiling in human embryonic cells coupled with software analysis to identify the affected signaling pathways. RESULTS: Software analysis of gene expression in cells exposed to 0.23-0.28 T SMF showed that nine signaling networks responded to SMF; of these, detailed biochemical validation was performed for the network linked to the inflammatory cytokine IL-6. We found the short-term (<24 h) activation of IL-6 involved the coordinate up-regulation of toll-like receptor-4 (TLR4) with complementary changes to NEU3 and ST3GAL5 that reduced ganglioside GM3 in a manner that augmented the activation of TLR4 and IL-6. Loss of GM3 also provided a plausible mechanism for the attenuation of cellular responses to SMF that occurred over longer exposure periods. Finally, SMF-mediated responses were manifest at the cellular level as morphological changes and biochemical markers indicative of pre-oligodendrocyte differentiation. CONCLUSION: This study provides a framework describing how magnetic exposure is transduced from a plausible molecular biosensor (lipid membranes) to cell-level responses that include differentiation toward neural lineages. In addition, SMF provided a stimulus that uncovered new relationships - that exist even in the absence of magnetic fields - between gangliosides, the time-dependent regulation of IL-6 signaling by these glycosphingolipids, and the fate of embryonic cells.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/citologia , Sistema de Sinalização das MAP Quinases , Magnetismo , Biomarcadores , Linhagem Celular , Gangliosídeos/metabolismo , Gangliosídeos/farmacologia , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , RNA Mensageiro/metabolismo , Software , Receptor 4 Toll-Like/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA