Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nano Lett ; 23(3): 757-764, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36648291

RESUMO

Effective delivery of the CRISPR-Cas9 components is crucial to realizing the therapeutic potential. Although many delivery approaches have been developed for this application, oral delivery has not been explored due to the degradative nature of the gastrointestinal tract. For this issue, we developed a series of novel phenylboronic acid (PBA)-functionalized chitosan-polyethylenimine (CS-PEI) polymers for oral CRISPR delivery. PBA functionalization equipped the polyplex with higher stability, smooth transport across the mucus, and efficient endosomal escape and cytosolic unpackaging in the cells. From a library of 12 PBA-functionalized CS-PEI polyplexes, we identified a formulation that showed the most effective penetration in the intestinal mucosa after oral gavage to mice. The optimized formulation performed feasible CRISPR-mediated downregulation of the target protein and reduction in the downstream cholesterol. As the first oral CRISPR carrier, this study suggests the potential of addressing the needs of both local and systemic editing in a patient-compliant manner.


Assuntos
Ácidos Borônicos , Quitosana , Animais , Camundongos , Polímeros , Técnicas de Transferência de Genes
2.
Nano Lett ; 21(6): 2461-2469, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33686851

RESUMO

Circulating cell-free DNA (cfDNA) released by damaged cells causes inflammation and has been associated with the progression of sepsis. One proposed strategy to treat sepsis is to scavenge this inflammatory circulating cfDNA. Here, we develop a cfDNA-scavenging nanoparticle (NP) that consists of cationic polyethylenimine (PEI) of different molecular weight grafted to zeolitic imidazolate framework-8 (PEI-g-ZIF) in a simple one-pot process. PEI-g-ZIF NPs fabricated using PEI 1800 and PEI 25k but not PEI 600 suppressed cfDNA-induced TLR activation and subsequent nuclear factor kappa B pathway activity. PEI 1800-g-ZIF NPs showed greater inhibition of cfDNA-associated inflammation and multiple organ injury than naked PEI 1800 (lacking ZIF), and had greater therapeutic efficacy in treating sepsis. These results indicate that PEI-g-ZIF NPs acts as a "nanotrap" that improves upon naked PEI in scavenging circulating cfDNA, reducing inflammation, and reversing the progression of sepsis, thus providing a novel strategy for sepsis treatment.


Assuntos
Ácidos Nucleicos Livres , Estruturas Metalorgânicas , Nanopartículas , Sepse , Humanos , Polietilenoimina , Sepse/tratamento farmacológico
3.
Biomacromolecules ; 20(1): 528-538, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30537806

RESUMO

Oral drug delivery is a more favored mode of administration because of its ease of administration, high patient compliance, and low healthcare costs. However, no oral protein formulations are commercially available currently due to hostile gastrointestinal (GI) barriers resulting in insignificant oral bioavailability of macromolecular drugs. Herein, we used insulin as a model protein drug; insulin-loaded N-(2-hydroxy)-propyl-3-trimethylammonium chloride modified chitosan (HTCC)/sodium tripolyphosphate (TPP) nanocomplex (NC) as a nanocore was further encapsulated into enteric Eudragit L100-55 material, through a two-step flash nanocomplexation (FNC) process in a reliable and scalable manner, forming our NC-in-Eudragit composite particles (NE). Particle size and surface properties of our optimized NE were tailored to protect the loaded insulin from acidic degradation in the hostile stomach environment and to achieve intestinal site-specific drug release as well as the improvement of oral delivery efficiency of insulin. In addition, the oral administration of the optimized NE to type 1 diabetic rats could induce a very significant hypoglycemic effect with a relative oral bioavailability of 13.3%. Our results demonstrated that enteric encapsulation of nanotherapeutics using a FNC apparatus could cause drug formulations to possess better size controllability, batch-mode reproducibility, and homogeneous surface coating and then significantly enhance their oral bioavailability of insulin, indicating its great potential for clinical translation of oral protein therapeutics.


Assuntos
Absorção Gastrointestinal , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Nanocápsulas/química , Resinas Acrílicas/química , Administração Oral , Animais , Células CACO-2 , Quitosana/análogos & derivados , Células HT29 , Humanos , Hipoglicemiantes/farmacocinética , Insulina/farmacocinética , Masculino , Nanocápsulas/efeitos adversos , Polifosfatos/química , Ratos , Ratos Sprague-Dawley
4.
Nano Lett ; 18(1): 314-325, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29232130

RESUMO

We propose an effective siRNA delivery system by preparing poly(DAMA-HEMA)-multilayered gold nanoparticles using multiple surface-initiated atom transfer radical polymerization processes. The polymeric multilayer structure is characterized by transmission electron microscopy, matrix-associated laser desorption/ionization time-of-flight mass spectrometry, UV-vis spectroscopy, Fourier transform infrared spectroscopy, dynamic light scattering, and ζ-potential. The amount of siRNA electrostatically incorporated into the nanoparticle can be tuned by the number of polymeric shells, which in turn influences the cellular uptake and gene silencing effect. In a bioreductive environment, the interlayer disulfide bond breaks to release the siRNA from the degraded polymeric shells. Intravenously injected c-Myc siRNA-incorporated particles accumulate in the tumor site of a murine lung carcinoma model and significantly suppress the tumor growth. Therefore, the combination of a size-tunable AuNP core and an ATRP-functionalized shell offers control and versatility in the effective delivery of siRNA.


Assuntos
Ouro/química , Neoplasias Pulmonares/terapia , Nanopartículas Metálicas/química , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Terapêutica com RNAi , Células A549 , Animais , Cátions/química , Humanos , Neoplasias Pulmonares/genética , Nanopartículas Metálicas/ultraestrutura , Camundongos , Polimerização , Polímeros/química , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Propriedades de Superfície
5.
Nano Lett ; 18(5): 3007-3016, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29694053

RESUMO

Nanoparticulate vaccines can potentiate immune responses by site-specific drainage to lymph nodes (LNs). This approach may benefit from a nanoparticle engineering method with fine control over size and codelivery of antigen and adjuvant. Here, we applied the flash nanocomplexation (FNC) method to prepare nanovaccines via polyelectrolyte complexation of chitosan and heparin to coencapsulate the VP1 protein antigen from enterovirus 71, which causes hand-foot-mouth disease (HFMD), with tumor necrosis factor α (TNF) or CpG as adjuvants. FNC allows for reduction of the nanovaccine size to range from 90 to 130 nm with relatively narrower size distribution and a high payload capacity. These nanovaccines reached both proximal and distal LNs via subcutaneous injection and subsequently exhibited prolonged retention in the LNs. The codelivery induced strong immune activation toward a Th1 response in addition to a potent Th2 response, and conferred effective protection against lethal virus challenge comparable to that of an approved inactivated viral vaccine in mouse models of both passive and active immunization setting. In addition, these nanovaccines also elicited strong IgA titers, which may offer unique advantages for mucosal protection. This study addresses the issues of size control, antigen bioactivity retention, and biomanufacturing to demonstrate the translational potential of a subunit nanovaccine design.


Assuntos
Proteínas do Capsídeo/administração & dosagem , Sistemas de Liberação de Medicamentos , Enterovirus Humano A/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Nanopartículas/química , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas Virais/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacologia , Animais , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/uso terapêutico , Doença de Mão, Pé e Boca/imunologia , Doença de Mão, Pé e Boca/virologia , Humanos , Linfonodos/imunologia , Linfonodos/virologia , Camundongos Endogâmicos BALB C , Linfócitos T/imunologia , Linfócitos T/virologia , Vacinas de Subunidades Antigênicas/uso terapêutico , Vacinas Virais/imunologia , Vacinas Virais/uso terapêutico
6.
Angew Chem Int Ed Engl ; 58(13): 4254-4258, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30724436

RESUMO

Cell-free deoxyribonucleic acid (cfDNA) released from either dead or damaged cells serves as a key autoantigen in rheumatoid arthritis (RA). They can be recognized by nucleic acid (NA) sensors such as the toll-like receptor (TLR), leading to activation of the innate immune system and chronic inflammation. Developed here is a cationic molecular scavenger, by screening cationic dendronized polymers, which eliminates cfDNA and inhibits TLR recognition and nucleic-acid-induced inflammation. The structure-property study demonstrates that toxicity, NA binding capacity, and biodistribution could be balanced to achieve maximum therapeutic effect by exquisite control of the molecular structure. In addition, the optimized cationic polymer effectively inhibited joint swelling, synovial hyperplasia, and bone destruction in collagen-induced arthritis (CIA) rat models. The results offer support for synthetic polymers offering new paradigm in autoimmune disease treatment.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Cátions/química , Ácidos Nucleicos Livres/efeitos adversos , Inflamação/tratamento farmacológico , Polímeros/administração & dosagem , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/patologia , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/patologia , Modelos Animais de Doenças , Inflamação/etiologia , Inflamação/patologia , Polímeros/química , Polímeros/farmacocinética , Ratos , Distribuição Tecidual , Receptores Toll-Like/metabolismo
7.
Mol Pharm ; 14(11): 3644-3659, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-28994600

RESUMO

Although tumor-targeting nanovehicles for hepatocellular carcinoma (HCC) chemotherapy have attracted great research and clinic interest, the poor cancer penetration, inefficient cellular uptake, and slow intracellular drug release greatly compromise their therapeutic outcomes. In this work, a multifunctional mixed micellar system, consisting of glycyrrhetinic acid (GA) for specific liver-targeting, trans-activator of transcription (TAT) peptide for potent cell penetration, and pH-sensitive poly(ß-amino ester) polymers for acidic-triggered drug release, was developed to provide HCC-targeting delivery and pH-triggered release of doxorubicin (DOX). These micelles were hypothesized to efficaciously accumulate in HCC site by the guide of GA ligands, enter into cancer cells facilitated by the activated TAT peptide on the micellar surface, and finally rapidly release DOX in cytoplasm. To demonstrate this design, DOX was initially loaded in micelles modified with both GA and TAT (DOX/GA@TAT-M) with high drug loading efficiency and pH-sensitive drug release profiles. The HCC-targeting cellular uptake and synergetic anticancer efficacy were tested, indicating DOX/GA@TAT-M could be specifically and effectively internalized into HCC cells by the effect of GA targeting and TAT penetrating with enhanced cytotoxicity. In addition, the prolonged circulation time and enhanced accumulation in tumor facilitated its potent tumor growth inhibition activity in vivo. These results demonstrated that the cleavable multifunctional mixed micelles with tumor targeting, controlled TAT peptide activation, and sequential pH-sensitive drug release could be an efficient strategy for HCC treatment.


Assuntos
Micelas , Carcinoma Hepatocelular/tratamento farmacológico , Preparações de Ação Retardada , Doxorrubicina/química , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Ácido Glicirretínico/química , Humanos , Concentração de Íons de Hidrogênio , Neoplasias Hepáticas/tratamento farmacológico , Peptídeos/química , Polímeros/química
8.
Biomacromolecules ; 17(11): 3706-3713, 2016 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-27741396

RESUMO

Dying cells release nucleic acids (NA) and NA-containing complexes that activate inflammatory pathways of immune cells. Sustained activation of these pathways contributes to chronic inflammation frequently encountered in autoimmune and inflammatory diseases. In this study, grafting of cationic polymers onto a nanofibrous mesh enabled local scavenging of negatively charged pro-inflammatory molecules in the extracellular space. Nucleic acid scavenging nanofibers (NASFs) formed from poly(styrene-alt-maleic anhydride) conjugated with 1.8 kDa bPEI resulted in nanofibers of diameters 486 ± 9 nm. NASFs inhibited the NF-κB response stimulated by the negatively charged agonists, CpG and poly(I:C), in Ramos-blue cells but not Pam3CSK4, a nonanionic agonist. Moreover, NASFs significantly impeded NF-κB activation in cells stimulated with damage-associated molecular pattern molecules (DAMPs) released from doxorubicin killed cancer cells. In vivo application of NASFs to open wounds demonstrated nucleic acid scavenging in wounds of diabetic mice infected with Pseudomonas aeruginosa, suggesting the in vivo efficacy of NASFs. This simple technique of generating NASF results in effective localized anti-inflammation in vitro and local nucleic acid scavenging in vivo.


Assuntos
Inflamação/tratamento farmacológico , Maleatos/química , Nanofibras/química , Poliestirenos/química , Cicatrização/efeitos dos fármacos , Animais , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Humanos , Inflamação/microbiologia , Inflamação/patologia , Maleatos/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD , Nanofibras/administração & dosagem , Ácidos Nucleicos/química , Poliaminas/administração & dosagem , Poliaminas/química , Polieletrólitos , Poliestirenos/administração & dosagem , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade
9.
Annu Rev Biomed Eng ; 16: 347-70, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24905873

RESUMO

Inhibiting specific gene expression by short interfering RNA (siRNA) offers a new therapeutic strategy to tackle many diseases, including cancer, metabolic disorders, and viral infections, at the molecular level. The macromolecular and polar nature of siRNA hinders its cellular access to exert its effect. Nanoparticulate delivery systems can promote efficient intracellular delivery. Despite showing promise in many preclinical studies and potential in some clinical trials, siRNA has poor delivery efficiency, which continues to demand innovations, from carrier design to formulation, in order to overcome transport barriers. Previous findings for optimal plasmid DNA delivery cannot be generalized to siRNA delivery owing to significant discrepancy in size and subtle differences in chain flexibility between the two types of nucleic acids. In this review, we highlight the recent advances in improving the stability of siRNA nanoparticles, understanding their intracellular trafficking and release mechanisms, and applying judiciously the promising formulations to disease models.


Assuntos
Nanomedicina/métodos , Nanopartículas/química , Nanotecnologia/métodos , RNA Interferente Pequeno/metabolismo , Animais , Ensaios Clínicos como Assunto , DNA/metabolismo , Sistemas de Liberação de Medicamentos , Endossomos/metabolismo , Expressão Gênica , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lipídeos/química , Fígado/metabolismo , Ácidos Nucleicos/metabolismo , Plasmídeos/metabolismo , Polímeros/química , Interferência de RNA , RNA Mensageiro/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(34): 14055-60, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21844380

RESUMO

Dead and dying cells release nucleic acids. These extracellular RNAs and DNAs can be taken up by inflammatory cells and activate multiple nucleic acid-sensing toll-like receptors (TLR3, 7, 8, and 9). The inappropriate activation of these TLRs can engender a variety of inflammatory and autoimmune diseases. The redundancy of the TLR family encouraged us to seek materials that can neutralize the proinflammatory effects of any nucleic acid regardless of its sequence, structure or chemistry. Herein we demonstrate that certain nucleic acid-binding polymers can inhibit activation of all nucleic acid-sensing TLRs irrespective of whether they recognize ssRNA, dsRNA or hypomethylated DNA. Furthermore, systemic administration of such polymers can prevent fatal liver injury engendered by proinflammatory nucleic acids in an acute toxic shock model in mice. Therefore these polymers represent a novel class of anti-inflammatory agent that can act as molecular scavengers to neutralize the proinflammatory effects of various nucleic acids.


Assuntos
Anti-Inflamatórios/farmacologia , Ácidos Nucleicos/metabolismo , Polímeros/farmacologia , Animais , Anti-Inflamatórios/uso terapêutico , Cátions , Linhagem Celular , Endocitose/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Humanos , Imunidade/efeitos dos fármacos , Inflamação/patologia , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/imunologia , Ligantes , Hepatopatias/complicações , Hepatopatias/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Ácidos Nucleicos/toxicidade , Oligodesoxirribonucleotídeos/farmacologia , Polímeros/uso terapêutico , Choque Séptico/complicações , Choque Séptico/tratamento farmacológico , Receptores Toll-Like/imunologia
11.
ACS Nano ; 18(9): 7084-7097, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38377352

RESUMO

Severe airway inflammatory disorders impose a significant societal burden, and the available treatments are unsatisfactory. High levels of neutrophil extracellular trap (NET) and cell-free DNA (cfDNA) were detected in the inflammatory microenvironment of these diseases, which are closely associated with persistent uncontrolled neutrophilic inflammation. Although DNase has proven to be effective in mitigating neutrophilic airway inflammation in mice by reducing cfDNA and NET levels, its clinical use is hindered by severe side effects. Here, we synthesized polyglycerol-amine (PGA) with a series of hydroxyl/amine ratios and covered them with black phosphorus (BP) nanosheets. The BP nanosheets functionalized with polyglycerol-50% amine (BP-PGA50) efficiently lowered cfDNA levels, suppressed toll-like receptor 9 (TLR9) activation and inhibited NET formation in vitro. Importantly, BP-PGA50 nanosheets demonstrated substantial accumulation in inflamed airway tissues, excellent biocompatibility, and potent inflammation modulation ability in model mice. The 2D sheet-like structure of BP-PGA50 was identified as a crucial factor for the therapeutic efficacy, and the hydroxyl/amine ratio was revealed as a significant parameter to regulate the protein resistance, cfDNA-binding efficacy, and cytotoxicity. This study shows the promise of the BP-PGA50 nanosheet for tackling uncontrolled airway inflammation, which is also significant for the treatment of other neutrophilic inflammatory diseases. In addition, our work also highlights the importance of proper surface functionalization, such as hydroxyl/amine ratio, in therapeutic nanoplatform construction for inflammation modulation.


Assuntos
Ácidos Nucleicos Livres , Glicerol , Neutrófilos , Polímeros , Camundongos , Animais , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Aminas/farmacologia
12.
J Biomed Mater Res A ; 111(9): 1298-1308, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36951261

RESUMO

The field of biomaterials science is highly active, with a steadily increasing number of publications and new journals being founded. This article brings together contributions from the editors of six leading journals in the area of biomaterials science and engineering. Each contributor highlights specific advances, topics, and trends that have emerged through the publications in their respective journal in the calendar year 2022. It presents a global perspective on a wide range of material types, functionalities, and applications. The highlighted topics include a diversity of biomaterials; from proteins, polysaccharides, and lipids to ceramics, metals, advanced composites, and a variety of new forms of these materials. Important advances in dynamically functional materials are presented, including a range of fabrication techniques such as bioassembly, 3D bioprinting and microgel formation. Similarly, several applications are highlighted in drug and gene delivery, biological sensing, cell guidance, immunoengineering, electroconductivity, wound healing, infection resistance, tissue engineering, and treatment of cancer. The goal of this paper is to provide the reader with both a broad view of recent biomaterials research, as well as expert commentary on some of the key advances that will shape the future of biomaterials science and engineering.


Assuntos
Bioimpressão , Publicações Periódicas como Assunto , Materiais Biocompatíveis , Engenharia Tecidual/métodos , Proteínas , Impressão Tridimensional
13.
Acta Biomater ; 143: 1-25, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35202854

RESUMO

Conventional approaches to developing biomaterials and implants require intuitive tailoring of manufacturing protocols and biocompatibility assessment. This leads to longer development cycles, and high costs. To meet existing and unmet clinical needs, it is critical to accelerate the production of implantable biomaterials, implants and biomedical devices. Building on the Materials Genome Initiative, we define the concept 'biomaterialomics' as the integration of multi-omics data and high-dimensional analysis with artificial intelligence (AI) tools throughout the entire pipeline of biomaterials development. The Data Science-driven approach is envisioned to bring together on a single platform, the computational tools, databases, experimental methods, machine learning, and advanced manufacturing (e.g., 3D printing) to develop the fourth-generation biomaterials and implants, whose clinical performance will be predicted using 'digital twins'. While analysing the key elements of the concept of 'biomaterialomics', significant emphasis has been put forward to effectively utilize high-throughput biocompatibility data together with multiscale physics-based models, E-platform/online databases of clinical studies, data science approaches, including metadata management, AI/ Machine Learning (ML) algorithms and uncertainty predictions. Such integrated formulation will allow one to adopt cross-disciplinary approaches to establish processing-structure-property (PSP) linkages. A few published studies from the lead author's research group serve as representative examples to illustrate the formulation and relevance of the 'Biomaterialomics' approaches for three emerging research themes, i.e. patient-specific implants, additive manufacturing, and bioelectronic medicine. The increased adaptability of AI/ML tools in biomaterials science along with the training of the next generation researchers in data science are strongly recommended. STATEMENT OF SIGNIFICANCE: This leading opinion review paper emphasizes the need to integrate the concepts and algorithms of the data science with biomaterials science. Also, this paper emphasizes the need to establish a mathematically rigorous cross-disciplinary framework that will allow a systematic quantitative exploration and curation of critical biomaterials knowledge needed to drive objectively the innovation efforts within a suitable uncertainty quantification framework, as embodied in 'biomaterialomics' concept, which integrates multi-omics data and high-dimensional analysis with artificial intelligence (AI) tools, like machine learning. The formulation of this approach has been demonstrated for patient-specific implants, additive manufacturing, and bioelectronic medicine.


Assuntos
Inteligência Artificial , Materiais Biocompatíveis , Ciência de Dados , Humanos , Aprendizado de Máquina , Impressão Tridimensional
14.
Nat Commun ; 13(1): 5925, 2022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36207325

RESUMO

Periodontitis is a common type of inflammatory bone loss and a risk factor for systemic diseases. The pathogenesis of periodontitis involves inflammatory dysregulation, which represents a target for new therapeutic strategies to treat periodontitis. After establishing the correlation of cell-free DNA (cfDNA) level with periodontitis in patient samples, we test the hypothesis that the cfDNA-scavenging approach will benefit periodontitis treatment. We create a nanoparticulate cfDNA scavenger specific for periodontitis by coating selenium-doped hydroxyapatite nanoparticles (SeHANs) with cationic polyamidoamine dendrimers (PAMAM-G3), namely G3@SeHANs, and compare the activities of G3@SeHANs with those of soluble PAMAM-G3 polymer. Both G3@SeHANs and PAMAM-G3 inhibit periodontitis-related proinflammation in vitro by scavenging cfDNA and alleviate inflammatory bone loss in a mouse model of ligature-induced periodontitis. G3@SeHANs also regulate the mononuclear phagocyte system in a periodontitis environment, promoting the M2 over the M1 macrophage phenotype. G3@SeHANs show greater therapeutic effects than PAMAM-G3 in reducing proinflammation and alveolar bone loss in vivo. Our findings demonstrate the importance of cfDNA in periodontitis and the potential for using hydroxyapatite-based nanoparticulate cfDNA scavengers to ameliorate periodontitis.


Assuntos
Ácidos Nucleicos Livres , Dendrímeros , Periodontite , Selênio , Animais , Ácidos Nucleicos Livres/genética , Dendrímeros/farmacologia , Hidroxiapatitas , Camundongos , Periodontite/tratamento farmacológico
15.
Biomaterials ; 289: 121792, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36116170

RESUMO

Cell reprogramming can satisfy the demands of obtaining specific cell types for applications such as tissue regeneration and disease modeling. Here we report the reprogramming of human fibroblasts to produce chemically-induced osteogenic cells (ciOG), and explore the potential uses of ciOG in bone repair and disease treatment. A chemical cocktail of RepSox, forskolin, and phenamil was used for osteogenic induction of fibroblasts by activation of RUNX2 expression. Following a maturation, the cells differentiated toward an osteoblast phenotype that produced mineralized nodules. Bulk and single-cell RNA sequencing identified a distinct ciOG population. ciOG formed mineralized tissue in an ectopic site of immunodeficiency mice, unlike the original fibroblasts. Osteogenic reprogramming was modulated under engineered culture substrates. When generated on a nanofiber substrate ciOG accelerated bone matrix formation in a calvarial defect, indicating that the engineered biomaterial promotes the osteogenic capacity of ciOG in vivo. Furthermore, the ciOG platform recapitulated the genetic bone diseases Proteus syndrome and osteogenesis imperfecta, allowing candidate drug testing. The reprogramming of human fibroblasts into osteogenic cells with a chemical cocktail thus provides a source of specialized cells for use in bone tissue engineering and disease modeling.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core , Engenharia Tecidual , Animais , Materiais Biocompatíveis/metabolismo , Regeneração Óssea/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Colforsina/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Humanos , Camundongos , Osteoblastos , Osteogênese/fisiologia
16.
Sci Adv ; 8(4): eabj2372, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35089791

RESUMO

A therapeutic strategy that targets multiple proinflammatory factors in inflammatory bowel disease (IBD) with minimal systemic side effects would be attractive. Here, we develop a drug-free, biodegradable nanomedicine that acts against IBD by scavenging proinflammatory cell-free DNA (cfDNA) and reactive oxygen species (ROS). Polyethylenimine (PEI) was conjugated to antioxidative diselenide-bridged mesoporous organosilica nanoparticles (MONs) to formulate nanoparticles (MON-PEI) that exhibited high cfDNA binding affinity and ROS-responsive degradation. In ulcerative colitis and Crohn's disease mouse colitis models, orally administered MON-PEI accumulated preferentially in the inflamed colon and attenuated colonic and peritoneal inflammation by alleviating cfDNA- and ROS-mediated inflammatory responses, allowing a reduced dose frequency and ameliorating colitis even after delayed treatment. This work suggests a new nanomedicine strategy for IBD treatment.


Assuntos
Ácidos Nucleicos Livres , Colite , Doenças Inflamatórias Intestinais , Animais , Modelos Animais de Doenças , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/metabolismo , Camundongos , Polietilenoimina/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo
17.
Mol Pharm ; 8(5): 1662-8, 2011 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-21740056

RESUMO

Lipoplexes and polyplexes represent the two major nanocarrier systems for nucleic acid delivery. Previous studies examining their uptake and intracellular unpacking rely on organic fluorophores fraught with low signal intensity and photobleaching. In this work quantum dot mediated Förster resonance energy transfer (QD-FRET) was first used to study and compare the cellular uptake and the intracellular fate of oligodeoxynucelotide (ODN)-based lipoplexes and polyplexes. QD605-amine and Cy5-labeled ODN (Cy5-GTI2040) were chosen as the FRET pair. By adjusting the lipid/ODN ratio of lipoplexes and the nitrogen/phosphate (N/P) ratio of polyplexes, lipoplexes and polyplexes with comparable physical properties were produced. The biological activities of dual-labeled lipoplexes and polyplexes remained unaltered compared to their unlabeled counterparts as evidenced by their comparable antisense activities against protein R2 in KB cells. Flow cytometry and confocal microscopy revealed similar pattern of uptake for these two types of nanoparticles, although polyplexes had a higher dissociation rate than lipoplexes in KB cells. We demonstrate that QD-FRET is a sensitive tool to study the uptake and intracellular unpacking of lipoplexes and polyplexes, which may help optimize their formulations for various theranostics applications.


Assuntos
Antineoplásicos/metabolismo , Portadores de Fármacos/metabolismo , Lipídeos/química , Nanopartículas/química , Oligodesoxirribonucleotídeos Antissenso/metabolismo , Polietilenoimina/química , Pontos Quânticos , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Transporte Biológico , Sobrevivência Celular/efeitos dos fármacos , Fenômenos Químicos , Portadores de Fármacos/efeitos adversos , Portadores de Fármacos/química , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células KB , Microscopia Confocal , Nanopartículas/efeitos adversos , Nanopartículas/ultraestrutura , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/ultraestrutura , Oligodesoxirribonucleotídeos Antissenso/administração & dosagem , Oligodesoxirribonucleotídeos Antissenso/efeitos adversos , Oligodesoxirribonucleotídeos Antissenso/química , Tamanho da Partícula , RNA Mensageiro/metabolismo , Ribonucleotídeo Redutases/antagonistas & inibidores , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo , Propriedades de Superfície
18.
Biomaterials ; 230: 119627, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31767445

RESUMO

Cell therapy, the treatment of diseases using living cells, offers a promising clinical approach to treating refractory diseases. The global market for cell therapy is growing rapidly, and there is an increasing demand for automated methods that can produce large quantities of high quality therapeutic cells. Biomaterials can be used during cell production to establish a biomimetic microenvironment that promotes cell adhesion and proliferation while maintaining target cell genotype and phenotype. Here we review recent progress and emerging techniques in biomaterial-assisted cell production. The increasing use of auxiliary biomaterials and automated production methods provides an opportunity to improve quality control and increase production efficiency using standardized GMP-compliant procedures.


Assuntos
Materiais Biocompatíveis , Terapia Baseada em Transplante de Células e Tecidos , Adesão Celular
19.
Sci Adv ; 6(22): eaay7148, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32523983

RESUMO

Severe sepsis represents a common, expensive, and deadly health care issue with limited therapeutic options. Gaining insights into the inflammatory dysregulation that causes sepsis would help develop new therapeutic strategies against severe sepsis. In this study, we identified the crucial role of cell-free DNA (cfDNA) in the regulation of the Toll-like receptor 9-mediated proinflammatory pathway in severe sepsis progression. Hypothesizing that removing cfDNA would be beneficial for sepsis treatment, we used polyethylenimine (PEI) and synthesized PEI-functionalized, biodegradable mesoporous silica nanoparticles with different charge densities as cfDNA scavengers. These nucleic acid-binding nanoparticles (NABNs) showed superior performance compared with their nucleic acid-binding polymer counterparts on inhibition of cfDNA-induced inflammation and subsequent multiple organ injury caused by severe sepsis. Furthermore, NABNs exhibited enhanced accumulation and retention in the inflamed cecum, along with a more desirable in vivo safety profile. Together, our results revealed a key contribution of cfDNA in severe sepsis and shed a light on the development of NABN-based therapeutics for sepsis therapy, which currently remains intractable.


Assuntos
Ácidos Nucleicos Livres , Nanopartículas , Sepse , DNA/uso terapêutico , Humanos , Polietilenoimina/uso terapêutico , Sepse/etiologia , Sepse/genética
20.
ACS Appl Mater Interfaces ; 11(33): 29593-29603, 2019 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-31348859

RESUMO

Oral delivery of nucleic acid therapy is a promising strategy in treating various diseases because of its higher patient compliance and therapeutic efficiency compared to parenteral routes of administration. However, its success has been limited by the low transfection efficiency resulting from nucleic acid entrapment in the mucus layer and epithelial barrier of the gastrointestinal (GI) tract. Herein, we describe an approach to overcome this phenomenon and improve oral DNA delivery in the context of treating type II diabetes (T2D). Linear PEI (lPEI) was used as a carrier to form complexes with plasmid DNA encoding glucagon-like peptide 1 (GLP-1), a common target in T2D treatments. These nanoparticles were then coated with a mixture of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and 1,2-dimyristoyl-rac-glycero-3-methoxy poly(ethylene glycol)-2000 (DMG-PEG) to render the nanoparticle surface hydrophilic and electrostatically neutral. The surface-modified lPEI/DNA nanoparticles showed higher diffusivity and transport in the mucus layer of the GI tract and mediated high levels of transfection efficiency in vitro and in vivo. Moreover, these modified nanoparticles demonstrated high levels of GLP-1 expression for more than 24 h in the liver, lungs, and intestine in a T2D murine model after a single dose, as well as controlled blood glucose levels within a normal range for at least 18 h with repeatable therapeutic effects upon multiple dosages. Taken together, this work demonstrates the feasibility of an oral plasmid DNA delivery approach in the treatment of T2D through a facile surface modification to improve the mucus permeability and delivery efficiency of the nanoparticles.


Assuntos
DNA/química , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Nanopartículas/química , Células A549 , Animais , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Portadores de Fármacos/química , Peptídeo 1 Semelhante ao Glucagon/sangue , Peptídeo 1 Semelhante ao Glucagon/genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Imuno-Histoquímica , Insulina/sangue , Fígado/metabolismo , Pulmão/metabolismo , Camundongos , Polietilenoglicóis/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA