Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Mol Pharm ; 17(9): 3633-3642, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32786959

RESUMO

Therapeutic nucleic acids hold great promise for the treatment of genetic diseases, yet the delivery of this highly charged macromolecular drug remains a challenge in the field. Peptides are promising agents to mediate nucleic acid delivery because they can encode a biological function to overcome the trafficking barriers. Electrostatic nanocomplexes of nucleic acid and peptides can achieve effective delivery, but the balance between their stability and biological function must be finely tuned. In this work, we explore two peptide building blocks that have been studied in the literature: targeting ligands and intracellular trafficking peptides. We grafted these peptides on a polyethylene glycol (PEG) backbone with eight sites for substitution to create so-called "peptide spiders". These conjugates achieve stability via the well-known hydrophilic shielding effect of PEG. In addition, the coordination of peptide building blocks into multimers may create new biological properties, such as the well-known phenomena of increased binding avidity with multivalent ligands. In this work, we linked two trafficking peptides to the PEG backbone using either nonreducible or reducible chemistries and investigated the ability of these materials to carry silencing RNAs into mammalian cells. We then investigated these nanomaterials for their pharmacokinetic properties and silencing of undruggable targets in a mouse model of cancer. While reducible linkages were more potent at silencing in vitro, this effect was reversed when applied in the context of living animals. This work offers an insight into peptide-based delivery materials and investigates peptide-polymer linkages.


Assuntos
Ácidos Nucleicos/química , Peptídeos/química , Polímeros/química , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Ligantes , Substâncias Macromoleculares/química , Camundongos , Camundongos Nus , Nanopartículas/química , Neoplasias/metabolismo , Polietilenoglicóis/química , RNA Interferente Pequeno/química , Células U937
2.
Nano Lett ; 16(10): 6303-6310, 2016 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-27622711

RESUMO

Targeted cancer therapies require a precise determination of the underlying biological processes driving tumorigenesis within the complex tumor microenvironment. Therefore, new diagnostic tools that capture the molecular activity at the disease site in vivo are needed to better understand tumor behavior and ultimately maximize therapeutic responses. Matrix metalloproteinases (MMPs) drive multiple aspects of tumorigenesis, and their activity can be monitored using engineered peptide substrates as protease-specific probes. To identify tumor specific activity profiles, local sampling of the tumor microenvironment is necessary, such as through remote control of probes, which are only activated at the tumor site. Alternating magnetic fields (AMFs) provide an attractive option to remotely apply local triggering signals because they penetrate deep into the body and are not likely to interfere with biological processes due to the weak magnetic properties of tissue. Here, we report the design and evaluation of a protease-activity nanosensor that can be remotely activated at the site of disease via an AMF at 515 kHz and 15 kA/m. Our nanosensor was composed of thermosensitive liposomes containing functionalized protease substrates that were unveiled at the target site by remotely triggered heat dissipation of coencapsulated magnetic nanoparticles (MNPs). This nanosensor was combined with a unique detection assay to quantify the amount of cleaved substrates in the urine. We applied this spatiotemporally controlled system to determine tumor protease activity in vivo and identified differences in substrate cleavage profiles between two mouse models of human colorectal cancer.


Assuntos
Neoplasias Colorretais/epidemiologia , Campos Magnéticos , Nanopartículas de Magnetita , Metaloproteinases da Matriz/metabolismo , Animais , Linhagem Celular Tumoral , Compostos Férricos , Humanos , Lipossomos , Camundongos , Peptídeos
3.
Bioconjug Chem ; 27(10): 2323-2331, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27583545

RESUMO

Nanoparticulate systems have shown great promise in overcoming the considerable trafficking barriers associated with systemic nucleic acid delivery, which must be addressed to unlock the full potential of technologies such as RNAi and gene editing in vivo. In addition to mediating the cytoplasmic delivery of nucleic cargo and shielding it from nuclease degradation and immunostimulation, nucleic-acid-containing nanomaterials delivered intravenously must also be stable in the bloodstream after administration to avoid toxicity and off-target delivery. To this end, the hydrophilic molecule polyethylene glycol (PEG) has been deployed in many different nanoparticle systems to prevent aggregation and recognition by the reticuloendothelial system. However, the optimal strategy for incorporating PEG into self-assembled nucleic acid delivery systems to obtain nanoparticle stability while retaining important functions such as receptor targeting and cargo activity remains unclear. In this work, we develop substantially improved formulations of tumor-penetrating nanocomplexes (TPNs), targeted self-assembled nanoparticles formulated with peptide carriers and siRNA that have been shown to mitigate tumor burden in an orthotopic model of ovarian cancer. We specifically sought to tailor TPNs for intravenous delivery by systematically comparing formulations with three different classes of modular PEG incorporation (namely PEG graft polymers, PEG lipids, and PEGylated peptide), each synthesized using straightforward bioconjugation techniques. We found that the addition of PEG lipids or PEGylated peptide carriers led to the formation of small and stable nanoparticles, but only nanoparticles formulated with PEGylated peptide carriers retained substantial activity in a gene silencing assay. In vivo, this formulation significantly decreased accumulation in off-target organs and improved initial availability in circulation compared to results from the original non-PEGylated particles. Thus, from among a set of candidate strategies, we identified TPNs with admixed PEGylated peptide carriers as the optimal formulation for systemic administration of siRNA on the basis of their performance in a battery of physicochemical and biological assays. Moreover, this optimized formulation confers pharmacologic advantages that may enable further translational development of tumor-penetrating nanocomplexes, highlighting the preclinical value of comparing formulation strategies and the relevance of this systematic approach for the development of other self-assembled nanomaterials.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Nanopartículas/química , Peptídeos/química , Polietilenoglicóis/química , RNA Interferente Pequeno/química , Animais , Sistemas de Liberação de Medicamentos , Difusão Dinâmica da Luz , Feminino , Inativação Gênica , Humanos , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão , Nanopartículas/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacocinética , Distribuição Tecidual
4.
Nat Mater ; 11(9): 768-74, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22751181

RESUMO

In the absence of perfusable vascular networks, three-dimensional (3D) engineered tissues densely populated with cells quickly develop a necrotic core. Yet the lack of a general approach to rapidly construct such networks remains a major challenge for 3D tissue culture. Here, we printed rigid 3D filament networks of carbohydrate glass, and used them as a cytocompatible sacrificial template in engineered tissues containing living cells to generate cylindrical networks that could be lined with endothelial cells and perfused with blood under high-pressure pulsatile flow. Because this simple vascular casting approach allows independent control of network geometry, endothelialization and extravascular tissue, it is compatible with a wide variety of cell types, synthetic and natural extracellular matrices, and crosslinking strategies. We also demonstrated that the perfused vascular channels sustained the metabolic function of primary rat hepatocytes in engineered tissue constructs that otherwise exhibited suppressed function in their core.


Assuntos
Vasos Sanguíneos/citologia , Perfusão , Impressão/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Materiais Biocompatíveis/farmacologia , Carboidratos/química , Vidro/química , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Ratos , Fatores de Tempo
5.
Adv Sci (Weinh) ; 9(3): e2102698, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34786888

RESUMO

Patients with Alagille syndrome carry monogenic mutations in the Notch signaling pathway and face complications such as jaundice and cholestasis. Given the presence of intrahepatic ductopenia in these patients, Notch2 receptor signaling is implicated in driving normal biliary development and downstream branching morphogenesis. As a result, in vitro model systems of liver epithelium are needed to further mechanistic insight of biliary tissue assembly. Here, primary human intrahepatic cholangiocytes as a candidate population for such a platform are systematically evaluated, and conditions that direct their branching morphogenesis are described. It is found that extracellular matrix presentation, coupled with mitogen stimulation, promotes biliary branching in a Notch-dependent manner. These results demonstrate the utility of using 3D scaffolds for mechanistic investigation of cholangiocyte branching and provide a gateway to integrate biliary architecture in additional in vitro models of liver tissue.


Assuntos
Síndrome de Alagille/metabolismo , Materiais Biocompatíveis/metabolismo , Fígado/metabolismo , Organoides/metabolismo , Alicerces Teciduais , Adulto , Sistema Biliar/metabolismo , Células Cultivadas , Células Epiteliais/metabolismo , Humanos , Morfogênese , Transdução de Sinais
6.
J Am Chem Soc ; 133(49): 19582-5, 2011 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21981330

RESUMO

Mesoporous silica nanoparticles (MSNPs) have garnered a great deal of attention as potential carriers for therapeutic payloads. However, achieving triggered drug release from MSNPs in vivo has been challenging. Here, we describe the synthesis of stimulus-responsive polymer-coated MSNPs and the loading of therapeutics into both the core and shell domains. We characterize MSNP drug-eluting properties in vitro and demonstrate that the polymer-coated MSNPs release doxorubicin in response to proteases present at a tumor site in vivo, resulting in cellular apoptosis. These results demonstrate the utility of polymer-coated nanoparticles in specifically delivering an antitumor payload.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Preparações de Ação Retardada/química , Doxorrubicina/administração & dosagem , Nanopartículas/química , Dióxido de Silício/química , Antibióticos Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada/metabolismo , Doxorrubicina/farmacologia , Células HeLa , Humanos , Nanopartículas/ultraestrutura , Neoplasias/tratamento farmacológico , Peptídeo Hidrolases/metabolismo , Polímeros/química , Polímeros/metabolismo , Porosidade , Dióxido de Silício/metabolismo
7.
Nat Mater ; 8(4): 331-6, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19234444

RESUMO

Nanomaterials that can circulate in the body hold great potential to diagnose and treat disease. For such applications, it is important that the nanomaterials be harmlessly eliminated from the body in a reasonable period of time after they carry out their diagnostic or therapeutic function. Despite efforts to improve their targeting efficiency, significant quantities of systemically administered nanomaterials are cleared by the mononuclear phagocytic system before finding their targets, increasing the likelihood of unintended acute or chronic toxicity. However, there has been little effort to engineer the self-destruction of errant nanoparticles into non-toxic, systemically eliminated products. Here, we present luminescent porous silicon nanoparticles (LPSiNPs) that can carry a drug payload and of which the intrinsic near-infrared photoluminescence enables monitoring of both accumulation and degradation in vivo. Furthermore, in contrast to most optically active nanomaterials (carbon nanotubes, gold nanoparticles and quantum dots), LPSiNPs self-destruct in a mouse model into renally cleared components in a relatively short period of time with no evidence of toxicity. As a preliminary in vivo application, we demonstrate tumour imaging using dextran-coated LPSiNPs (D-LPSiNPs). These results demonstrate a new type of multifunctional nanostructure with a low-toxicity degradation pathway for in vivo applications.


Assuntos
Portadores de Fármacos , Nanopartículas , Silício , Animais , Antineoplásicos/administração & dosagem , Materiais Biocompatíveis , Doxorrubicina/administração & dosagem , Células HeLa , Humanos , Luminescência , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Varredura
8.
Nat Nanotechnol ; 15(9): 792-800, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32690884

RESUMO

Human breath contains many volatile metabolites. However, few breath tests are currently used in the clinic to monitor disease due to bottlenecks in biomarker identification. Here we engineered breath biomarkers for respiratory disease by local delivery of protease-sensing nanoparticles to the lungs. The nanosensors shed volatile reporters upon cleavage by neutrophil elastase, an inflammation-associated protease with elevated activity in lung diseases such as bacterial infection and alpha-1 antitrypsin deficiency. After intrapulmonary delivery into mouse models with acute lung inflammation, the volatile reporters are released and expelled in breath at levels detectable by mass spectrometry. These breath signals can identify diseased mice with high sensitivity as early as 10 min after nanosensor administration. Using these nanosensors, we performed serial breath tests to monitor dynamic changes in neutrophil elastase activity during lung infection and to assess the efficacy of a protease inhibitor therapy targeting neutrophil elastase for the treatment of alpha-1 antitrypsin deficiency.


Assuntos
Biomarcadores/análise , Testes Respiratórios/métodos , Elastase de Leucócito/metabolismo , Deficiência de alfa 1-Antitripsina/enzimologia , Animais , Testes Respiratórios/instrumentação , Simulação por Computador , Relação Dose-Resposta a Droga , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Feminino , Glicina/análogos & derivados , Glicina/farmacologia , Humanos , Elastase de Leucócito/antagonistas & inibidores , Pneumopatias/enzimologia , Pneumopatias/microbiologia , Espectrometria de Massas , Camundongos Endogâmicos , Camundongos Knockout , Nanoestruturas/química , Polietilenoglicóis/química , Infecções por Pseudomonas/enzimologia , Sulfonamidas/farmacologia , Compostos Orgânicos Voláteis/análise , Compostos Orgânicos Voláteis/química , Compostos Orgânicos Voláteis/metabolismo , Deficiência de alfa 1-Antitripsina/tratamento farmacológico , Deficiência de alfa 1-Antitripsina/genética
9.
Bioconjug Chem ; 19(8): 1570-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18611045

RESUMO

The in vivo fate of nanomaterials strongly determines their biomedical efficacy. Accordingly, much effort has been invested into the development of library screening methods to select targeting ligands for a diversity of sites in vivo. Still, broad application of chemical and biological screens to the in vivo targeting of nanomaterials requires ligand attachment chemistries that are generalizable, efficient, covalent, orthogonal to diverse biochemical libraries, applicable under aqueous conditions, and stable in in vivo environments. To date, the copper(I)-catalyzed Huisgen 1,3-dipolar cycloaddition or "click" reaction has shown considerable promise as a method for developing targeted nanomaterials in vitro. Here, we investigate the utility of "click" chemistry for the in vivo targeting of inorganic nanoparticles to tumors. We find that "click" chemistry allows cyclic LyP-1 targeting peptides to be specifically linked to azido-nanoparticles and to direct their binding to p32-expressing tumor cells in vitro. Moreover, "click" nanoparticles are able to stably circulate for hours in vivo following intravenous administration (>5 h circulation time), extravasate into tumors, and penetrate the tumor interstitium to specifically bind p32-expressing cells in tumors. In the future, in vivo use of "click" nanomaterials should expedite the progression from ligand discovery to in vivo evaluation and diversify approaches toward multifunctional nanoparticle development.


Assuntos
Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Peptídeos Cíclicos/química , Peptídeos Cíclicos/uso terapêutico , Alcinos/química , Aminas/química , Sequência de Aminoácidos , Animais , Azidas/química , Proteínas de Transporte , Bovinos , Linhagem Celular Tumoral , Fluorescência , Regulação da Expressão Gênica , Humanos , Magnetismo , Camundongos , Proteínas Mitocondriais/metabolismo , Peptídeos Cíclicos/metabolismo , Polímeros/química , Sensibilidade e Especificidade , Compostos de Sulfidrila/química
10.
Mol Cancer Ther ; 17(11): 2377-2388, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30097486

RESUMO

Pancreatic cancer is one of the leading causes of cancer-related death, with 5-year survival of 8.5%. The lack of significant progress in improving therapy reflects our inability to overcome the desmoplastic stromal barrier in pancreatic ductal adenocarcinoma (PDAC) as well as a paucity of new approaches targeting its genetic underpinnings. RNA interference holds promise in targeting key mutations driving PDAC; however, a nucleic acid delivery vehicle that homes to PDAC and breaches the stroma does not yet exist. Noting that the cyclic peptide iRGD mediates tumor targeting and penetration through interactions with αvß3/5 integrins and neuropilin-1, we hypothesized that "tandem" peptides combining a cell-penetrating peptide and iRGD can encapsulate siRNA to form tumor-penetrating nanocomplexes (TPN) capable of delivering siRNA to PDAC. The use of directly conjugated iRGD is justified by receptor expression patterns in human PDAC biopsies. In this work, we optimize iRGD TPNs with polyethylene glycol (PEG)-peptide conjugates for systemic delivery to sites of disease. We show that TPNs effectively knockdown siRNA targets in PDAC cell lines and in an immunocompetent genetically engineered mouse model of PDAC. Furthermore, we validate their tumor-penetrating ability in three-dimensional organoids and autochthonous tumors. In murine therapeutic trials, TPNs delivering anti-Kras siRNA significantly delay tumor growth. Thus, iRGD TPNs hold promise in treating PDAC by not only overcoming physical barriers to therapy, but by leveraging the stroma to achieve knockdown of the gold-standard genetic target. Moreover, the modular construction of this delivery platform allows for facile adaptation to future genetic target candidates in pancreatic cancer. Mol Cancer Ther; 17(11); 2377-88. ©2018 AACR.


Assuntos
Peptídeos Penetradores de Células/química , Nanopartículas/química , Oligopeptídeos/química , Neoplasias Pancreáticas/terapia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Peptídeos Penetradores de Células/farmacocinética , Camundongos Nus , Modelos Biológicos , Nanopartículas/ultraestrutura , Organoides/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/ultraestrutura , Polietilenoglicóis/química , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
11.
Lab Chip ; 7(6): 702-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17538711

RESUMO

Tissues formed by cells encapsulated in hydrogels have uses in biotechnology, cell-based assays, and tissue engineering. We have previously presented a 3D micropatterning technique that rapidly localizes live cells within hydrogels using dielectrophoretic (DEP) forces, and have demonstrated the ability to modulate tissue function through the control of microscale cell architecture. A limitation of this method is the requirement that a single biomaterial must simultaneously harbor biological properties that support cell survival and function and material properties that permit efficient dielectrophoretic patterning. Here, we resolve this issue by forming multiphase tissues consisting of microscale tissue sub-units in a 'local phase' biomaterial, which, in turn, are organized by DEP forces in a separate, mechanically supportive 'bulk phase' material. We first define the effects of medium conductivity on the speed and quality of DEP cell patterning. As a case study, we then produce multiphase tissues with microscale architecture that combine high local hydrogel conductivity for enhanced survival of sensitive liver progenitor cells with low bulk conductivity required for efficient DEP micropatterning. This approach enables an expanded range of studies examining the influence of 3D cellular architecture on diverse cell types, and in the future may improve the biological function of inhomogeneous tissues assembled from a variety of modular tissue sub-units.


Assuntos
Materiais Biocompatíveis/química , Técnicas de Cultura/métodos , Campos Eletromagnéticos , Eletroforese/métodos , Hidrogéis/química , Engenharia Tecidual/métodos , Células 3T3 , Animais , Adesão Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Fibroblastos/citologia , Teste de Materiais , Camundongos
12.
Biomaterials ; 28(2): 256-70, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16979755

RESUMO

Tissue-engineered therapies for liver failure offer the potential to augment or replace whole organ transplantation; however, fabrication of hepatic tissue poses unique challenges largely stemming from the complexity of liver structure and function. In this study, we illustrate the utility of highly-tunable, photopolymerizable poly(ethylene glycol) (PEG) hydrogels for 3D encapsulation of hepatic cells and highlight a range of techniques important for examining hepatocellular function in this platform. Owing to our long-term interest in incorporating proliferative progenitor cell types (e.g. hepatoblasts, oval cells, or cells derived from embryonic stem cells) and maintaining the phenotype of differentiated cells, we explored the behavior of bipotential mouse embryonic liver (BMEL) cells as a model progenitor cell and mature, fully differentiated, primary hepatocytes in this biomaterial system. We demonstrated the importance of cell-cell and cell-matrix interactions in the survival and function of these cell types, and the capacity to influence encapsulated cell phenotypes through modulation of hydrogel characteristics or gene silencing. Additionally, we demonstrated imaging techniques critical for the in situ assessment of encapsulated hepatocyte function combined with the ability to control cellular organization and overall architecture through microscale patterning technologies. Further analysis of liver progenitor as well as mature hepatocyte processes within the versatile PEG hydrogel platform will aid in the development of tissue engineered implantable liver systems.


Assuntos
Materiais Biocompatíveis/química , Técnicas de Cultura/métodos , Hepatócitos/fisiologia , Hidrogéis/química , Polietilenoglicóis/química , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Teste de Materiais , Ratos , Engenharia Tecidual/métodos
13.
Curr Opin Biotechnol ; 17(5): 524-31, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16978857

RESUMO

Engineered tissues can be employed for studies on the fundamental mechanisms of embryology and adult physiology and for investigating the evolution of disease processes. They also provide platforms to evaluate the behavior of new chemical entities in drug development. The recent development of three specific technologies has greatly facilitated the engineering of tissues for in vitro applications: the microfabrication tools that serve to both define the cellular microenvironment and enable parallelization of cell-based assays; synthetic, tunable hydrogels to create three-dimensional microenvironments; and bioreactors to control nutrient transport and fluid shear stress. Furthermore, convergence of these tools is providing investigators with the opportunity to construct and study tissues in vitro with unprecedented levels of sophistication.


Assuntos
Proliferação de Células , Engenharia Tecidual/métodos , Animais , Reatores Biológicos , Técnicas de Cultura de Células/métodos , Fenômenos Fisiológicos Celulares , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Hidrogel de Polietilenoglicol-Dimetacrilato/metabolismo
14.
Sci Transl Med ; 9(399)2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28724577

RESUMO

Control of both tissue architecture and scale is a fundamental translational roadblock in tissue engineering. An experimental framework that enables investigation into how architecture and scaling may be coupled is needed. We fabricated a structurally organized engineered tissue unit that expanded in response to regenerative cues after implantation into mice with liver injury. Specifically, we found that tissues containing patterned human primary hepatocytes, endothelial cells, and stromal cells in a degradable hydrogel expanded more than 50-fold over the course of 11 weeks in mice with injured livers. There was a concomitant increase in graft function as indicated by the production of multiple human liver proteins. Histologically, we observed the emergence of characteristic liver stereotypical microstructures mediated by coordinated growth of hepatocytes in close juxtaposition with a perfused vasculature. We demonstrated the utility of this system for probing the impact of multicellular geometric architecture on tissue expansion in response to liver injury. This approach is a hybrid strategy that harnesses both biology and engineering to more efficiently deploy a limited cell mass after implantation.


Assuntos
Hepatopatias/cirurgia , Fígado/citologia , Albuminas/metabolismo , Animais , Hepatócitos/citologia , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato , Fígado/patologia , Engenharia Tecidual/métodos , Alicerces Teciduais , Transferrina/metabolismo
15.
Lab Chip ; 5(1): 111-8, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15616749

RESUMO

Living cells have the potential to serve as sensors, naturally integrating the response to stimuli to generate predictions about cell fate (e.g., differentiation, migration, proliferation, apoptosis). Miniaturized arrays of living cells further offer the capability to interrogate many cells in parallel and thereby enable high-throughput and/or combinatorial assays. However, the interface between living cells and synthetic chip platforms is a critical one wherein the cellular phenotype must be preserved to generate useful signals. While some cell types retain tissue-specific features on a flat (2-D) surface, it has become increasingly apparent that a 3-D physical environment will be required for others. In this paper, we present two independent methods for creating living cell arrays that are encapsulated within a poly(ethylene glycol)-based hydrogel to create a local 3-D microenvironment. First, 'photopatterning' selectively crosslinks hydrogel microstructures containing living cells with approximately 100 microm feature size. Second, 'electropatterning' utilizes dielectrophoretic forces to position cells within a prepolymer solution prior to crosslinking, forming cell patterns with micron resolution. We further combine these methods to obtain hierarchical control of cell positioning over length scales ranging from microns to centimeters. This level of microenvironmental control should enable the fabrication of next-generation cellular microarrays in which robust 3-D cultures of cells are presented with appropriate physical and chemical cues and, consequently, report on cellular responses that resemble in vivo behavior.


Assuntos
Técnicas Biossensoriais , Hidrogéis/química , Polietilenoglicóis/química , Células 3T3 , Animais , Técnicas Biossensoriais/instrumentação , Técnicas Biossensoriais/métodos , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Campos Eletromagnéticos , Camundongos , Raios Ultravioleta
16.
J Biomed Mater Res A ; 103(10): 3331-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25851120

RESUMO

Engineered tissue constructs have the potential to augment or replace whole organ transplantation for the treatment of liver failure. Poly(ethylene glycol) (PEG)-based systems are particularly promising for the construction of engineered liver tissue due to their biocompatibility and amenability to modular addition of bioactive factors. To date, primary hepatocytes have been successfully encapsulated in non-degradable hydrogels based on PEG-diacrylate (PEGDA). In this study, we describe a hydrogel system based on PEG-diacrylamide (PEGDAAm) containing matrix-metalloproteinase sensitive (MMP-sensitive) peptide in the hydrogel backbone that is suitable for hepatocyte culture both in vitro and after implantation. By replacing hydrolytically unstable esters in PEGDA with amides in PEGDAAm, resultant hydrogels resisted non-specific hydrolysis, while still allowing for MMP-mediated hydrogel degradation. Optimization of polymerization conditions, hepatocellular density, and multicellular tissue composition modulated both the magnitude and longevity of hepatic function in vitro. Importantly, hepatic PEGDAAm-based tissues survived and functioned for over 3 weeks after implantation ectopically in the intraperitoneal (IP) space of nude mice. Together, these studies suggest that MMP-sensitive PEGDAAm-based hydrogels may be a useful material system for applications in tissue engineering and regenerative medicine. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 103A: 3331-3338, 2015.


Assuntos
Hepatócitos/metabolismo , Hidrogéis/química , Fígado Artificial , Fígado , Polietilenoglicóis/química , Engenharia Tecidual , Animais , Células Cultivadas , Masculino , Camundongos , Ratos , Ratos Endogâmicos Lew
17.
Tissue Eng Part C Methods ; 21(5): 509-17, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25390971

RESUMO

The ultimate design of functionally therapeutic engineered tissues and organs will rely on our ability to engineer vasculature that can meet tissue-specific metabolic needs. We recently introduced an approach for patterning the formation of functional spatially organized vascular architectures within engineered tissues in vivo. Here, we now explore the design parameters of this approach and how they impact the vascularization of an engineered tissue construct after implantation. We used micropatterning techniques to organize endothelial cells (ECs) into geometrically defined "cords," which in turn acted as a template after implantation for the guided formation of patterned capillaries integrated with the host tissue. We demonstrated that the diameter of the cords before implantation impacts the location and density of the resultant capillary network. Inclusion of mural cells to the vascularization response appears primarily to impact the dynamics of vascularization. We established that clinically relevant endothelial sources such as induced pluripotent stem cell-derived ECs and human microvascular endothelial cells can drive vascularization within this system. Finally, we demonstrated the ability to control the juxtaposition of parenchyma with perfused vasculature by implanting cords containing a mixture of both a parenchymal cell type (hepatocytes) and ECs. These findings define important characteristics that will ultimately impact the design of vasculature structures that meet tissue-specific needs.


Assuntos
Células Endoteliais/citologia , Engenharia Tecidual/métodos , Animais , Padronização Corporal , Capilares , Colágeno/química , Dimetilpolisiloxanos/química , Feminino , Hepatócitos/citologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Nus , Microcirculação , Neovascularização Fisiológica , Oxigênio/química , Ratos
18.
Adv Drug Deliv Rev ; 56(11): 1635-47, 2004 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-15350293

RESUMO

In recent years, advances in fabrication technologies have brought a new dimension to the field of tissue engineering. Using manufacturing-based methods and hydrogel chemistries, researchers have been able to fabricate tissue engineering scaffolds with complex 3-D architectures and customized chemistries that mimic the in vivo tissue environment. These techniques may be useful in developing therapies for replacing lost tissue function, as in vitro models of living tissue, and also for further enabling fundamental studies of structure/function relationships in three dimensional contexts. Here, we present an overview of 3-D tissue fabrication techniques based on methods for: scaffold fabrication, cellular assembly, and hybrid hydrogel/cell methods and review their potential utility for tissue engineering.


Assuntos
Imageamento Tridimensional/métodos , Engenharia Tecidual/métodos , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/normas , Hidrogéis/síntese química , Hidrogéis/normas , Imageamento Tridimensional/normas , Engenharia Tecidual/normas
19.
ACS Nano ; 8(9): 8776-85, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25119520

RESUMO

Antithrombotic therapy is a critical portion of the treatment regime for a number of life-threatening conditions, including cardiovascular disease, stroke, and cancer; yet, proper clinical management of anticoagulation remains a challenge because existing agents increase the propensity for bleeding in patients. Here, we describe the development of a bioresponsive peptide-polysaccharide nanocomplex that utilizes a negative feedback mechanism to self-titrate the release of anticoagulant in response to varying levels of coagulation activity. This nanoscale self-titrating activatable therapeutic, or nanoSTAT, consists of a cationic thrombin-cleavable peptide and heparin, an anionic polysaccharide and widely used clinical anticoagulant. Under nonthrombotic conditions, nanoSTATs circulate inactively, neither releasing anticoagulant nor significantly prolonging bleeding time. However, in response to life-threatening pulmonary embolism, nanoSTATs locally release their drug payload and prevent thrombosis. This autonomous negative feedback regulator may improve antithrombotic therapy by increasing the therapeutic window and decreasing the bleeding risk of anticoagulants.


Assuntos
Anticoagulantes/química , Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Nanoestruturas/química , Animais , Bovinos , Heparina/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Polietilenoglicóis/química , Embolia Pulmonar/prevenção & controle , Trombose/prevenção & controle
20.
Tissue Eng Part A ; 20(15-16): 2200-12, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24498910

RESUMO

Drug-induced liver injury is a major cause of drug development failures and postmarket withdrawals. In vitro models that incorporate primary hepatocytes have been shown to be more predictive than model systems which rely on liver microsomes or hepatocellular carcinoma cell lines. Methods to phenotypically stabilize primary hepatocytes ex vivo often rely on mimicry of hepatic microenvironmental cues such as cell-cell interactions and cell-matrix interactions. In this work, we sought to incorporate phenotypically stable hepatocytes into three-dimensional (3D) microtissues, which, in turn, could be deployed in drug-screening platforms such as multiwell plates and diverse organ-on-a-chip devices. We first utilize micropatterning on collagen I to specify cell-cell interactions in two-dimensions, followed by collagenase digestion to produce well-controlled aggregates for 3D encapsulation in polyethylene glycol (PEG) diacrylate. Using this approach, we examined the influence of homotypic hepatocyte interactions and composition of the encapsulating hydrogel, and achieved the maintenance of liver-specific function for over 50 days. Optimally preaggregated structures were subsequently encapsulated using a microfluidic droplet-generator to produce 3D microtissues. Interactions of engineered hepatic microtissues with drugs was characterized by flow cytometry, and yielded both induction of P450 enzymes in response to prototypic small molecules and drug-drug interactions that give rise to hepatotoxicity. Collectively, this study establishes a pipeline for the manufacturing of 3D hepatic microtissues that exhibit stabilized liver-specific functions and can be incorporated into a wide array of emerging drug development platforms.


Assuntos
Comunicação Celular/efeitos dos fármacos , Hepatócitos/citologia , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Engenharia Tecidual/métodos , Células 3T3 , Acetaminofen/toxicidade , Albuminas/metabolismo , Animais , Células Cultivadas , Células Imobilizadas/citologia , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/metabolismo , Colágeno/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Camundongos , Microfluídica , Ratos Endogâmicos Lew
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA