Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Exp Med ; 220(6)2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-36920308

RESUMO

The hallmark of tuberculosis (TB) is the formation of immune cell-enriched aggregates called granulomas. While granulomas are pathologically diverse, their tissue-wide heterogeneity has not been spatially resolved at the single-cell level in human tissues. By spatially mapping individual immune cells in every lesion across entire tissue sections, we report that in addition to necrotizing granulomas, the human TB lung contains abundant non-necrotizing leukocyte aggregates surrounding areas of necrotizing tissue. These cellular lesions were more diverse in composition than necrotizing lesions and could be stratified into four general classes based on cellular composition and spatial distribution of B cells and macrophages. The cellular composition of non-necrotizing structures also correlates with their proximity to necrotizing lesions, indicating these are foci of distinct immune reactions adjacent to necrotizing granulomas. Together, we show that during TB, diseased lung tissue develops a histopathological superstructure comprising at least four different types of non-necrotizing cellular aggregates organized as satellites of necrotizing granulomas.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Humanos , Granuloma/patologia , Pulmão/patologia , Macrófagos
2.
Front Immunol ; 12: 634119, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33868254

RESUMO

Cystatin F encoded by CST7 is a cysteine peptidase inhibitor known to be expressed in natural killer (NK) and CD8+ T cells during steady-state conditions. However, little is known about its expression during inflammatory disease states in humans. We have developed an analytic approach capable of not only identifying previously poorly characterized disease-associated genes but also defining regulatory mechanisms controlling their expression. By exploring multiple cohorts of public transcriptome data comprising 43 individual datasets, we showed that CST7 is upregulated in the blood during a diverse set of infectious and non-infectious inflammatory conditions. Interestingly, this upregulation of CST7 was neutrophil-specific, as its expression was unchanged in NK and CD8+ T cells during sepsis. Further analysis demonstrated that known microbial products or cytokines commonly associated with inflammation failed to increase CST7 expression, suggesting that its expression in neutrophils is induced by an endogenous serum factor commonly present in human inflammatory conditions. Overall, through the identification of CST7 upregulation as a marker of acute inflammation in humans, our study demonstrates the value of publicly available transcriptome data in knowledge generation and potential biomarker discovery.


Assuntos
Biomarcadores Tumorais/genética , Cistatinas/genética , Perfilação da Expressão Gênica , Inflamação/genética , Neutrófilos/metabolismo , Sepse/genética , Transcriptoma , Doença Aguda , Biomarcadores Tumorais/sangue , Estudos de Casos e Controles , Cistatinas/sangue , Bases de Dados Genéticas , Humanos , Inflamação/sangue , Inflamação/imunologia , Neutrófilos/imunologia , Sepse/sangue , Sepse/imunologia , Regulação para Cima
3.
Mol Cancer Ther ; 19(1): 270-281, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31597714

RESUMO

Combinations of chemotherapy with immunotherapy have seen recent clinical success, including two approvals of anti-PD-1/L1 agents in combination with taxane-based chemotherapy in non-small cell lung cancer and triple-negative breast cancer. Here, we present a study on the combination activity and mechanistic rationale of a novel EphA2-targeted liposomal taxane (EphA2-ILs-DTXp) and anti-PD-1. This combination was highly active in mouse syngeneic tumor models, with complete responses observed in 3 of 5 models. In the EMT-6 tumor model, combination of EphA2-ILs-DTXp with anti-PD-1 resulted in a 60% complete response rate, with durable responses that were resistant to rechallenge. These responses were not observed in the absence of CD8+ T cells. Characterization of the immune infiltrates in EMT-6 tumors reveals increased CD8+ T cells, increased CD8+ IFNγ+ CTLs, and an increased CD8/regulatory T-cell (Treg) ratio. These immunomodulatory effects were not observed in mice treated with a combination of docetaxel and anti-PD-1. Pharmacokinetic analysis revealed that the AUC of docetaxel was increased 15 times, from 52.1 to 785 ng/mL/hour, when delivered by EphA2-ILs-DTXp. A dose reduction study of EphA2-ILs-DTXp showed a dose-response relationship for both tumor growth inhibition and the CD8/Treg ratio. Our data indicate that synergism between docetaxel and anti-PD-1 is achievable with nanoliposomal delivery.


Assuntos
Hidrocarbonetos Aromáticos com Pontes/uso terapêutico , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor EphA2/metabolismo , Taxoides/uso terapêutico , Animais , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Neoplasias/patologia , Taxoides/farmacologia
4.
Cell Rep ; 29(11): 3539-3550.e4, 2019 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-31825834

RESUMO

Interferons (IFN) are pleiotropic cytokines essential for defense against infection, but the identity and tissue distribution of IFN-responsive cells in vivo are poorly defined. In this study, we generate a mouse strain capable of reporting IFN-signaling activated by all three types of IFNs and investigate the spatio-temporal dynamics and identity of IFN-responding cells following IFN injection and influenza virus infection. Despite ubiquitous expression of IFN receptors, cellular responses to IFNs are highly heterogenous in vivo and are determined by anatomical site, cell type, cellular preference to individual IFNs, and activation status. Unexpectedly, type I and II pneumocytes, the primary target of influenza infection, exhibit striking differences in the strength and temporal dynamics of IFN signaling associated with differential susceptibility to the viral infection. Our findings suggest that time- and cell-type-dependent integration of distinct IFN signals govern the specificity and magnitude of IFN responses in vivo.


Assuntos
Interferons/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Transdução de Sinais , Células Epiteliais Alveolares/metabolismo , Animais , Células Cultivadas , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Nat Biomed Eng ; 3(4): 264-280, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30952988

RESUMO

Antibody-mediated tumour targeting and nanoparticle-mediated encapsulation can reduce the toxicity of antitumour drugs and improve their efficacy. Here, we describe the performance of a nanotherapeutic encapsulating a hydrolytically sensitive docetaxel prodrug and conjugated to an antibody specific for EphA2-a receptor overexpressed in many tumours. Administration of the nanotherapeutic in mice led to slow and sustained release of the prodrug, reduced exposure of active docetaxel in the circulation (compared with administration of the free drug) and maintenance of optimal exposure of the drug in tumour tissue. We also show that administration of the nanotherapeutic in rats and dogs resulted in minimal haematological toxicity, as well as the absence of neutropenia and improved overall tolerability in multiple rodent models. Targeting of the nanotherapeutic to EphA2 improved tumour penetration and resulted in markedly enhanced antitumour activity (compared with administration of free docetaxel and non-targeted nanotherapeutic controls) in multiple tumour-xenografted mice. This nanomedicine could become a potent and safe therapeutic alternative for cancer patients undergoing chemotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Nanopartículas/uso terapêutico , Receptor EphA2/metabolismo , Animais , Antineoplásicos/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Hidrocarbonetos Aromáticos com Pontes/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Docetaxel/sangue , Docetaxel/química , Docetaxel/farmacocinética , Docetaxel/uso terapêutico , Humanos , Lipossomos , Camundongos Endogâmicos NOD , Camundongos SCID , Taxoides/farmacologia , Taxoides/uso terapêutico , Distribuição Tecidual/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Sci Rep ; 6: 33277, 2016 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-27615159

RESUMO

Cell-cell fusion is fundamental to a multitude of biological processes ranging from cell differentiation and embryogenesis to cancer metastasis and biomaterial-tissue interactions. Fusogenic cells are exposed to biochemical and biophysical factors, which could potentially alter cell behavior. While biochemical inducers of fusion such as cytokines and kinases have been identified, little is known about the biophysical regulation of cell-cell fusion. Here, we designed experiments to examine cell-cell fusion using bulk metallic glass (BMG) nanorod arrays with varying biophysical cues, i.e. nanotopography and stiffness. Through independent variation of stiffness and topography, we found that nanotopography constitutes the primary biophysical cue that can override biochemical signals to attenuate fusion. Specifically, nanotopography restricts cytoskeletal remodeling-associated signaling, which leads to reduced fusion. This finding expands our fundamental understanding of the nanoscale biophysical regulation of cell fusion and can be exploited in biomaterials design to induce desirable biomaterial-tissue interactions.


Assuntos
Macrófagos/fisiologia , Nanoestruturas/ultraestrutura , Óxido de Alumínio/química , Animais , Técnicas de Cultura de Células , Fusão Celular , Linhagem Celular , Meios de Cultura , Citoesqueleto/metabolismo , Ativação Enzimática , Sistema de Sinalização das MAP Quinases , Camundongos , Nanoestruturas/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
J Control Release ; 232: 103-12, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27063424

RESUMO

Glioblastoma multiforme (GBM) is a fatal brain tumor characterized by infiltration beyond the margins of the main tumor mass and local recurrence after surgery. The blood-brain barrier (BBB) poses the most significant hurdle to brain tumor treatment. Convection-enhanced delivery (CED) allows for local administration of agents, overcoming the restrictions of the BBB. Recently, polymer nanoparticles have been demonstrated to penetrate readily through the healthy brain when delivered by CED, and size has been shown to be a critical factor for nanoparticle penetration. Because these brain-penetrating nanoparticles (BPNPs) have high potential for treatment of intracranial tumors since they offer the potential for cell targeting and controlled drug release after administration, here we investigated the intratumoral CED infusions of PLGA BPNPs in animals bearing either U87 or RG2 intracranial tumors. We demonstrate that the overall volume of distribution of these BPNPs was similar to that observed in healthy brains; however, the presence of tumors resulted in asymmetric and heterogeneous distribution patterns, with substantial leakage into the peritumoral tissue. Together, our results suggest that CED of BPNPs should be optimized by accounting for tumor geometry, in terms of location, size and presence of necrotic regions, to determine the ideal infusion site and parameters for individual tumors.


Assuntos
Neoplasias Encefálicas/metabolismo , Convecção , Sistemas de Liberação de Medicamentos , Ácido Láctico/administração & dosagem , Nanopartículas/administração & dosagem , Ácido Poliglicólico/administração & dosagem , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Proteínas de Fluorescência Verde/administração & dosagem , Proteínas de Fluorescência Verde/farmacocinética , Humanos , Masculino , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos Endogâmicos F344 , Ratos Nus , Ratos Sprague-Dawley , Carga Tumoral
8.
Biomaterials ; 89: 127-35, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26967647

RESUMO

The foreign body response (FBR) begins with injury acquired during implantation of a biomaterial (BM) and is detrimental due to the eventual encapsulation of the implant. Fusion of macrophages to form foreign body giant cells (FBGC), a hallmark of the FBR, is the consequence of a multistep mechanism induced by interleukin (IL)-4 that includes the acquisition of a fusion competent state and subsequent cytoskeletal rearrangements. However, the precise mechanism, regulation, and interplay among molecular mediators to generate FBGCs are insufficiently understood. Seeking novel mediators of fusion that might be regulated at the post-transcriptional level, we examined the role of microRNAs (miRs) in this process. A miR microarray was screened and identified miR-223 as a negative regulator of macrophage fusion. In addition, transfection of primary macrophages with a mir-223 mimic attenuated IL-4-induced fusion. Furthermore, miR-223 KO mice and mir-223 deficient cells displayed increased fusion in vivo and in vitro, respectively. Finally, we developed a method for in vivo delivery of miR-223 mimic utilizing PLGA nanoparticles, which inhibited FBGC formation in a biomaterial implant model. Our results identify miR-223 as a negative regulator of fusion and demonstrate miR-223 mimic-loaded nanoparticles as a therapeutic inhibitor of macrophage fusion.


Assuntos
Células Gigantes de Corpo Estranho/metabolismo , Macrófagos/metabolismo , MicroRNAs/genética , Animais , Fusão Celular , Células Cultivadas , Regulação da Expressão Gênica , Células Gigantes de Corpo Estranho/citologia , Ácido Láctico/química , Macrófagos/citologia , Camundongos , Camundongos Knockout , MicroRNAs/administração & dosagem , Nanopartículas/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
9.
Adv Drug Deliv Rev ; 97: 56-68, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26763408

RESUMO

Extracellular matrix is composed of a complex array of molecules that together provide structural and functional support to cells. These properties are mainly mediated by the activity of collagenous and elastic fibers, proteoglycans, and proteins such as fibronectin and laminin. ECM composition is tissue-specific and could include matricellular proteins whose primary role is to modulate cell-matrix interactions. In adults, matricellular proteins are primarily expressed during injury, inflammation and disease. Particularly, they are closely associated with the progression and prognosis of cardiovascular and fibrotic diseases, and cancer. This review aims to provide an overview of the potential use of matricellular proteins in drug delivery including the generation of therapeutic agents based on the properties and structures of these proteins as well as their utility as biomarkers for specific diseases.


Assuntos
Sistemas de Liberação de Medicamentos , Proteínas da Matriz Extracelular/uso terapêutico , Animais , Humanos
10.
Acta Biomater ; 11: 37-47, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25242651

RESUMO

Implantation of biomaterials elicits a foreign body response characterized by fusion of macrophages to form foreign body giant cells and fibrotic encapsulation. Studies of the macrophage polarization involved in this response have suggested that alternative (M2) activation is associated with more favorable outcomes. Here we investigated this process in vivo by implanting mixed cellulose ester filters or polydimethylsiloxane disks in the peritoneal cavity of wild-type (WT) and monocyte chemoattractant protein-1 (MCP-1) knockout mice. We analyzed classical (M1) and alternative (M2) gene expression via quantitative polymerase chain reaction, immunohistochemistry and enzyme-linked immunosorbent assay in both non-adherent cells isolated by lavage and implant-adherent cells. Our results show that macrophages undergo unique activation that displays features of both M1 and M2 polarization including induction of tumor necrosis factor α (TNF), which induces the expression and nuclear translocation of p50 and RelA determined by immunofluorescence and Western blot. Both processes were compromised in fusion-deficient MCP-1 KO macrophages in vitro and in vivo. Furthermore, inclusion of BAY 11-7028, an inhibitor of NFκB activation, reduced nuclear translocation of RelA and fusion in WT macrophages. Our studies suggest that peritoneal implants elicit a unique macrophage polarization phenotype leading to induction of TNF and activation of the NFκB pathway.


Assuntos
Núcleo Celular/metabolismo , Celulose/análogos & derivados , Quimiocina CCL2/metabolismo , Dimetilpolisiloxanos/toxicidade , Reação a Corpo Estranho/metabolismo , Macrófagos/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Fator de Transcrição RelA/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/genética , Animais , Núcleo Celular/genética , Núcleo Celular/patologia , Celulose/toxicidade , Quimiocina CCL2/genética , Reação a Corpo Estranho/induzido quimicamente , Reação a Corpo Estranho/genética , Reação a Corpo Estranho/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/genética , Nitrilas/farmacologia , Sulfonas/farmacologia , Fator de Transcrição RelA/genética
11.
Biomaterials ; 35(25): 6698-706, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24881026

RESUMO

Intracranial implants elicit neurodegeneration via the foreign body response (FBR) that includes BBB leakage, macrophage/microglia accumulation, and reactive astrogliosis, in addition to neuronal degradation that limit their useful lifespan. Previously, monocyte chemoattractant protein 1 (MCP-1, also CCL2), which plays an important role in monocyte recruitment and propagation of inflammation, was shown to be critical for various aspects of the FBR in a tissue-specific manner. However, participation of MCP-1 in the brain FBR has not been evaluated. Here we examined the FBR to intracortical silicon implants in MCP-1 KO mice at 1, 2, and 8 weeks after implantation. MCP-1 KO mice had a diminished FBR compared to WT mice, characterized by reductions in BBB leakage, macrophage/microglia accumulation, and astrogliosis, and an increased neuronal density. Moreover, pharmacological inhibition of MCP-1 in implant-bearing WT mice maintained the increased neuronal density. To elucidate the relative contribution of microglia and macrophages, bone marrow chimeras were generated between MCP-1 KO and WT mice. Increased neuronal density was observed only in MCP-1 knockout mice transplanted with MCP-1 knockout marrow, which indicates that resident cells in the brain are major contributors. We hypothesized that these improvements are the result of a phenotypic switch of the macrophages/microglia polarization state, which we confirmed using PCR for common activation markers. Our observations suggest that MCP-1 influences neuronal loss, which is integral to the progression of neurological disorders like Alzheimer's and Parkinson disease, via BBB leakage and macrophage polarization.


Assuntos
Quimiocina CCL2/metabolismo , Reação a Corpo Estranho/terapia , Doenças Neurodegenerativas/terapia , Neurônios/metabolismo , Animais , Benzoxazinas/farmacologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Doença Crônica , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Piperidinas/farmacologia , Próteses e Implantes , Receptores CCR2/antagonistas & inibidores , Receptores CCR2/metabolismo , Engenharia Tecidual
12.
ACS Nano ; 8(5): 4366-75, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24724817

RESUMO

Nanopatterning of biomaterials is rapidly emerging as a tool to engineer cell function. Bulk metallic glasses (BMGs), a class of biocompatible materials, are uniquely suited to study nanopattern-cell interactions as they allow for versatile fabrication of nanopatterns through thermoplastic forming. Work presented here employs nanopatterned BMG substrates to explore detection of nanopattern feature sizes by various cell types, including cells that are associated with foreign body response, pathology, and tissue repair. Fibroblasts decreased in cell area as the nanopattern feature size increased, and fibroblasts could detect nanopatterns as small as 55 nm in size. Macrophages failed to detect nanopatterns of 150 nm or smaller in size, but responded to a feature size of 200 nm, resulting in larger and more elongated cell morphology. Endothelial cells responded to nanopatterns of 100 nm or larger in size by a significant decrease in cell size and elongation. On the basis of these observations, nondimensional analysis was employed to correlate cellular morphology and substrate nanotopography. Analysis of the molecular pathways that induce cytoskeletal remodeling, in conjunction with quantifying cell traction forces with nanoscale precision using a unique FIB-SEM technique, enabled the characterization of underlying biomechanical cues. Nanopatterns altered serum protein adsorption and effective substrate stiffness, leading to changes in focal adhesion density and compromised activation of Rho-A GTPase in fibroblasts. As a consequence, cells displayed restricted cell spreading and decreased collagen production. These observations suggest that topography on the nanoscale can be designed to engineer cellular responses to biomaterials.


Assuntos
Materiais Biocompatíveis/química , Vidro/química , Metais/química , Nanotecnologia/métodos , Animais , Fenômenos Biomecânicos , Adesão Celular , Sobrevivência Celular , Colágeno/química , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Fibronectinas/química , Reação a Corpo Estranho , GTP Fosfo-Hidrolases/química , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Microscopia de Fluorescência , Células NIH 3T3 , Nanoestruturas/química , Engenharia de Proteínas , Proteína rhoA de Ligação ao GTP/química
13.
Int J Pharm ; 453(2): 651-5, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23727592

RESUMO

Management of skin wound infections presents a serious problem in the clinic, in the community, and in both civilian and military clinical treatment centers. Staphylococcus aureus is one of the most common microbial pathogens in cutaneous wounds. Peptide-morpholino oligomer (PMO) conjugates targeted to S. aureus gyrase A mRNA have shown the ability to reduce bacterial viability by direct site-specific mRNA cleavage via RNase P. As a treatment, these conjugates have the added advantages of not being susceptible to resistance due to genetic mutations and are effective against drug resistant strains. While this strategy has proven effective in liquid culture, it has yet to be evaluated in an animal model of infected surface wounds. In the present study, we combined PMO conjugates with a thermoresponsive gel delivery system to treat full-thickness mouse cutaneous wounds infected with S. aureus. Wounds treated with a single dose of PMO conjugate displayed improved healing that was associated with increased epithelialization, reduced bacterial load, and increased matrix deposition. Taken together, our findings demonstrate the efficacy and flexibility of the PMO conjugate drug delivery system and make it an attractive and novel topical antimicrobial agent.


Assuntos
Antibacterianos/administração & dosagem , DNA Girase/genética , Morfolinos/administração & dosagem , Peptídeos/administração & dosagem , Staphylococcus aureus , Infecção dos Ferimentos/terapia , Animais , Sistemas de Liberação de Medicamentos , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Pele/patologia , Cicatrização/efeitos dos fármacos , Infecção dos Ferimentos/patologia
14.
Biomaterials ; 34(15): 3891-901, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23453058

RESUMO

We developed a multi-functional construct capable of controlled delivery of bioactive substances that can improve wound repair by supporting the intrinsic ability of the skin to heal. We synthesized electrospun scaffolds-composed of a blend of the degradable polymers poly(l-lactide) (PLA) or polycaprolactone (PCL)-that produce highly efficient non-viral in vivo gene delivery to cells in the wound bed, provide a protective barrier during early wound healing, and support cell migration and growth. This multi-functional material was tested for its influence on wound healing: scaffolds were loaded with plasmids encoding keratinocyte growth factor (KGF) and applied to full-thickness wounds in mice. Compared to scaffolds with control plasmids, animals receiving the KGF plasmid-loaded scaffold produced significant enhancements in wound healing, which was quantified by improvements in the rate of wound re-epithelialization, keratinocyte proliferation, and granulation response. Further, we quantified the expression level of endogenous and plasmid-derived KGF in wound samples: qRT-PCR on wound sections revealed a correlation between the levels of plasmid-derived protein expression and histological analysis of wound healing, revealing an inverse relationship between the expression level of exogenous KGF and the size of the unhealed epithelial layer in wounds. Our findings suggest that engineered nanofiber PLA/PCL scaffolds are capable of highly efficient controlled DNA delivery and are promising materials for treatment of cutaneous wounds.


Assuntos
Técnicas de Transferência de Genes , Ácidos Nucleicos/administração & dosagem , Engenharia Tecidual , Alicerces Teciduais/química , Cicatrização , Células 3T3 , Animais , Proliferação de Células/efeitos dos fármacos , DNA/metabolismo , Epiderme/efeitos dos fármacos , Epiderme/patologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Fator 7 de Crescimento de Fibroblastos/genética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ácidos Nucleicos/genética , Ácidos Nucleicos/farmacologia , Plasmídeos/metabolismo , Poliésteres/química , Cicatrização/efeitos dos fármacos
15.
J Neural Eng ; 10(1): 016013, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23337399

RESUMO

OBJECTIVE: The brain foreign body response (FBR) is an important process that limits the functionality of electrodes that comprise the brain-machine interface. Associated events in this process include leakage of the blood-brain barrier (BBB), reactive astrogliosis, recruitment and activation of microglia, and neuronal degeneration. Proper BBB function is also integral to maintaining neuronal health and function. Previous attempts to characterize BBB integrity have shown homogeneous leakage of macromolecules up to 10 nm in size. In this study, we describe a new method of measuring BBB permeability during the foreign body response in a mouse model. APPROACH: Fluorescent nanoparticles were delivered via the tail vein into implant-bearing mice. Tissue sections were then analyzed using fluorescence microscopy to observe nanoparticles in the tissue. Gold nanoparticles were also used in conjunction with TEM to confirm the presence of nanoparticles in the brain parenchyma. MAIN RESULTS: By using polymer nanoparticle tracers, which are significantly larger than conventional macromolecular tracers, we show near-implant BBB gaps of up to 500 nm in size that persist for at least 4 weeks after implantation. Further characterization of the BBB illustrates that leakage during the brain FBR is heterogeneous with gaps between at least 10 and 500 nm. Moreover, electron microscopy was used to confirm that the nanoparticle tracers enter into the brain parenchyma near chronic brain implants. SIGNIFICANCE: Taken together, our findings demonstrate that the FBR-induced BBB leakage is characterized by larger gaps and is of longer duration than previously thought. This technique can be applied to examine the BBB in other disease states as well as during induced, transient, BBB opening.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Permeabilidade Capilar/fisiologia , Corantes Fluorescentes , Reação a Corpo Estranho/fisiopatologia , Nanopartículas , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/ultraestrutura , Reação a Corpo Estranho/diagnóstico , Reação a Corpo Estranho/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL
16.
Drug Deliv Transl Res ; 1(1): 34-42, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21691426

RESUMO

Direct delivery of chemotherapy agents to the brain via degradable polymer delivery systems-such as Gliadel®-is a clinically proven method for treatment of glioblastoma multiforme, but there are important limitations with the current technology-including the requirement for surgery, profound local tissue toxicity, and limitations in diffusional penetration of agents-that limit its application and effectiveness. Here, we demonstrate another technique for direct, controlled delivery of chemotherapy to the brain that provides therapeutic benefit with fewer limitations. In our new approach, camptothecin (CPT)-loaded poly(lacticco-glycolic acid) (PLGA) nanoparticles are infused via convection-enhanced delivery (CED) to a stereotactically defined location in the brain, allowing simultaneous control of location, spread, and duration of drug release. To test this approach, CPT-PLGA nanoparticles (~100 nm in diameter) were synthesized with 25% drug loading. When these nanoparticles were incubated in culture with 9L gliosarcoma cells, the IC50 of CPT-PLGA nanoparticles was 0.04 µM, compared to 0.3 µM for CPT alone. CPT-PLGA nanoparticles stereotactically delivered by CED improved survival in rats with intracranial 9L tumors: the median survival for rats treated with CPT-PLGA nanoparticles (22 days) was significantly longer than unloaded nanoparticles (15 days) and free CPT infusion (17 days). CPT-PLGA nanoparticle treatment also produced significantly more long-term survivors (30% of animals were free of disease at 60 days) than any other treatment. CPT was present in tissues harvested up to 53 days post-infusion, indicating prolonged residence at the local site of administration. These are the first results to demonstrate the effectiveness of combining polymer-controlled release nanoparticles with CED in treating fatal intracranial tumors.

17.
Brain Res ; 1180: 121-32, 2007 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-17920047

RESUMO

This study investigates methods of manipulating the brain extracellular matrix (ECM) to enhance the penetration of nanoparticle drug carriers in convection-enhanced delivery (CED). A probe was fabricated with two independent microfluidic channels to infuse, either simultaneously or sequentially, nanoparticles and ECM-modifying agents. Infusions were performed in the striatum of the normal rat brain. Monodisperse polystyrene particles with a diameter of 54 nm were used as a model nanoparticle system. Because the size of these particles is comparable to the effective pore size of the ECM, their transport may be significantly hindered compared with the transport of low molecular weight molecules. To enhance the transport of the infused nanoparticles, we attempted to increase the effective pore size of the ECM by two methods: dilating the extracellular space and degrading selected constituents of the ECM. Two methods of dilating the extracellular space were investigated: co-infusion of nanoparticles and a hyperosmolar solution of mannitol, and pre-infusion of an isotonic buffer solution followed by infusion of nanoparticles. These treatments resulted in an increase in the nanoparticle distribution volume of 51% and 123%, respectively. To degrade hyaluronan, a primary structural component of the brain ECM, a pre-infusion of hyaluronidase (20,000 U/mL) was followed after 30 min by infusion of nanoparticles. This treatment resulted in an increase in the nanoparticle distribution of 64%. Our results suggest that both dilation and enzymatic digestion can be incorporated into CED protocols to enhance nanoparticle penetration.


Assuntos
Dilatação/métodos , Sistemas de Liberação de Medicamentos/métodos , Líquido Extracelular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Nanopartículas/administração & dosagem , Neostriado/efeitos dos fármacos , Animais , Convecção , Sistemas de Liberação de Medicamentos/instrumentação , Líquido Extracelular/metabolismo , Matriz Extracelular/metabolismo , Hialuronoglucosaminidase/administração & dosagem , Hialuronoglucosaminidase/metabolismo , Masculino , Microeletrodos , Peso Molecular , Nanopartículas/química , Neostriado/metabolismo , Concentração Osmolar , Polímeros/administração & dosagem , Polímeros/farmacocinética , Poliestirenos/administração & dosagem , Poliestirenos/farmacocinética , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
18.
Yale J Biol Med ; 79(3-4): 141-52, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17940624

RESUMO

Despite advances in diagnostic imaging and drug discovery, primary malignant brain tumors remain fatal. Median survival for patients with the most severe forms is rarely past eight months. The severity of the disease and the lack of substantial improvement in patient survival demand that new approaches be explored in drug delivery to brain tumors. Recently, local delivery of chemotherapy to brain tumors has provided a way to circumvent the blood-brain barrier, allowing delivery of chemotherapy drugs directly to malignant cells in the brain. Two methods of local delivery have been developed: polymeric-controlled release and convection-enhanced delivery. Controlled release utilizes degradable or non-degradable polymers as carriers of chemotherapy; polymer implants or microparticles are implanted locally to introduce a sustained source of drug for periods of days or months. Convection-enhanced delivery employs the bulk flow of drugs dissolved in fluid, which is introduced intracranially using a catheter and pump. The convective fluid flow is capable of delivering drugs great distances within the brain, potentially treating invasive cells at a distance from the catheter infusion site. These two new delivery strategies are capable of delivering both standard chemotherapeutic drugs and new methods of anti-cancer therapy. Taken individually, or used in tandem, they represent a potential revolution in brain cancer treatment.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Glioma/tratamento farmacológico , Materiais Biocompatíveis/química , Ensaios Clínicos como Assunto , Portadores de Fármacos , Implantes de Medicamento , Humanos , Ácido Láctico/química , Microscopia Eletrônica de Varredura , Metástase Neoplásica , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Polímeros/química , Poliestirenos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA