Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Nano ; 16(10): 16118-16132, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36214219

RESUMO

The lack of drugs that target both disease progression and tissue preservation makes it difficult to effectively manage rheumatoid arthritis (RA). Here, we report a porous silicon-based nanomedicine that efficiently delivers an antirheumatic drug to inflamed synovium while degrading into bone-remodeling products. Methotrexate (MTX) is loaded into the porous silicon nanoparticles using a calcium silicate based condenser chemistry. The calcium silicate-porous silicon nanoparticle constructs (pCaSiNPs) degrade and release the drug preferentially in an inflammatory environment. The biodegradation products of the pCaSiNP drug carrier are orthosilicic acid and calcium ions, which exhibit immunomodulatory and antiresorptive effects. In a mouse model of collagen-induced arthritis, systemically administered MTX-loaded pCaSiNPs accumulate in the inflamed joints and ameliorate the progression of RA at both early and established stages of the disease. The disease state readouts show that the combination is more effective than the monotherapies.


Assuntos
Antirreumáticos , Artrite Reumatoide , Camundongos , Animais , Metotrexato/farmacologia , Metotrexato/uso terapêutico , Nanomedicina , Silício , Porosidade , Cálcio , Artrite Reumatoide/tratamento farmacológico , Antirreumáticos/uso terapêutico , Portadores de Fármacos/uso terapêutico , Inflamação/tratamento farmacológico
2.
Bioconjug Chem ; 33(9): 1685-1697, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36017941

RESUMO

Traumatic brain injury (TBI) is a major cause of disability and death among children and young adults in the United States, yet there are currently no treatments that improve the long-term brain health of patients. One promising therapeutic for TBI is brain-derived neurotrophic factor (BDNF), a protein that promotes neurogenesis and neuron survival. However, outstanding challenges to the systemic delivery of BDNF are its instability in blood, poor transport into the brain, and short half-life in circulation and brain tissue. Here, BDNF is encapsulated into an engineered, biodegradable porous silicon nanoparticle (pSiNP) in order to deliver bioactive BDNF to injured brain tissue after TBI. The pSiNP carrier is modified with the targeting ligand CAQK, a peptide that binds to extracellular matrix components upregulated after TBI. The protein cargo retains bioactivity after release from the pSiNP carrier, and systemic administration of the CAQK-modified pSiNPs results in effective delivery of the protein cargo to injured brain regions in a mouse model of TBI. When administered after injury, the CAQK-targeted pSiNP delivery system for BDNF reduces lesion volumes compared to free BDNF, supporting the hypothesis that pSiNPs mediate therapeutic protein delivery after systemic administration to improve outcomes in TBI.


Assuntos
Lesões Encefálicas Traumáticas , Nanopartículas , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Matriz Extracelular , Ligantes , Camundongos , Peptídeos/uso terapêutico , Porosidade , Silício
3.
Nat Biomed Eng ; 6(9): 1057-1073, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36038771

RESUMO

Many crowded biomolecular structures in cells and tissues are inaccessible to labelling antibodies. To understand how proteins within these structures are arranged with nanoscale precision therefore requires that these structures be decrowded before labelling. Here we show that an iterative variant of expansion microscopy (the permeation of cells and tissues by a swellable hydrogel followed by isotropic hydrogel expansion, to allow for enhanced imaging resolution with ordinary microscopes) enables the imaging of nanostructures in expanded yet otherwise intact tissues at a resolution of about 20 nm. The method, which we named 'expansion revealing' and validated with DNA-probe-based super-resolution microscopy, involves gel-anchoring reagents and the embedding, expansion and re-embedding of the sample in homogeneous swellable hydrogels. Expansion revealing enabled us to use confocal microscopy to image the alignment of pre-synaptic calcium channels with post-synaptic scaffolding proteins in intact brain circuits, and to uncover periodic amyloid nanoclusters containing ion-channel proteins in brain tissue from a mouse model of Alzheimer's disease. Expansion revealing will enable the further discovery of previously unseen nanostructures within cells and tissues.


Assuntos
Microscopia , Nanoestruturas , Animais , Encéfalo/metabolismo , Canais de Cálcio/metabolismo , DNA/metabolismo , Hidrogéis , Camundongos , Microscopia/métodos , Proteínas/metabolismo
4.
ACS Biomater Sci Eng ; 8(10): 4123-4131, 2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-34468123

RESUMO

Peptide nucleic acids (PNAs) are a class of artificial oligonucleotide mimics that have garnered much attention as precision biotherapeutics for their efficient hybridization properties and their exceptional biological and chemical stability. However, the poor cellular uptake of PNA is a limiting factor to its more extensive use in biomedicine; encapsulation in nanoparticle carriers has therefore emerged as a strategy for internalization and delivery of PNA in cells. In this study, we demonstrate that PNA can be readily loaded into porous silicon nanoparticles (pSiNPs) following a simple salt-based trapping procedure thus far employed only for negatively charged synthetic oligonucleotides. We show that the ease and versatility of PNA chemistry also allows for producing PNAs with different net charge, from positive to negative, and that the use of differently charged PNAs enables optimization of loading into pSiNPs. Differently charged PNA payloads determine different release kinetics and allow modulation of the temporal profile of the delivery process. In vitro silencing of a set of specific microRNAs using a pSiNP-PNA delivery platform demonstrates the potential for biomedical applications.


Assuntos
MicroRNAs , Nanopartículas , Ácidos Nucleicos Peptídicos , MicroRNAs/genética , Nanopartículas/química , Oligonucleotídeos , Ácidos Nucleicos Peptídicos/química , Porosidade , Silício/química
5.
Environ Technol ; 42(3): 429-443, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31230556

RESUMO

This study aimed to fully recover ammonia contained at a high concentration in anaerobic digestate of piggery wastewater (ADPW) by forming struvite. As magnesium and phosphorus sources, ferronickel slag (FNS) and K2HPO4 were used, respectively. By leaching 200 g L-1 of FNS with 3.0 M H2SO4, 10,309 mg L-1 of magnesium ions were extracted, and this acid-leachate of FNS (FNSL) also contained 5965 mg L-1 of total iron. In order to simultaneously remove both high concentrations of organic matters in ADPW and iron in FNSL which were known to hinder struvite formation, the mixture of ADPW and FNSL was added with H2O2 at the H2O2/Fe molar ratio of 0.75 and pH 4.0. After Fenton reaction, removal efficiencies of COD and total iron reached 77.36% and 99.89%, respectively. Then COD and an iron-reduced mixture of ADPW and FNSL were added with K2HPO4 satisfying Mg:N:P molar ratio of 1.2:1:1.15 at pH 9.5 to produce struvite for 1 h. From 1 L of ADPW (2.21 g NH3-N), 0.65 L of FNSL (4.65 g Mg2+), and 5.63 g of PO4 3-P, 46.7 g of precipitates were obtained. Overall removal efficiencies of magnesium, NH3-N, and phosphorus were 98.59%, 94.25%, and 99.97%, respectively. Obtained precipitates were analysed by using XRD, XRF, SEM-EDX and found to be struvite with impurities of potassium and metals. Additionally, the economic feasibility of FNS was assessed by estimating chemical costs of various magnesium sources.


Assuntos
Magnésio , Águas Residuárias , Anaerobiose , Precipitação Química , Peróxido de Hidrogênio , Ferro , Níquel , Fosfatos , Fósforo , Estruvita
6.
Adv Funct Mater ; 30(25)2020 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-32982626

RESUMO

Scaffolds made from biocompatible polymers provide physical cues to direct the extension of neurites and to encourage repair of damaged nerves. The inclusion of neurotrophic payloads in these scaffolds can substantially enhance regrowth and repair processes. However, many promising neurotrophic candidates are excluded from this approach due to incompatibilities with the polymer or with the polymer processing conditions. This work provides one solution to this problem by incorporating porous silicon nanoparticles (pSiNPs) that are pre-loaded with the therapeutic into a polymer scaffold during fabrication. The nanoparticle-drug-polymer hybrids are prepared in the form of oriented poly(lactic-co-glycolic acid) nanofiber scaffolds. We test three different therapeutic payloads: bpV(HOpic), a small molecule inhibitor of phosphatase and tensin homolog (PTEN); an RNA aptamer specific to tropomyosin-related kinase receptor type B (TrkB); and the protein nerve growth factor (NGF). Each therapeutic is loaded using a loading chemistry that is optimized to slow the rate of release of these water-soluble payloads. The drug-loaded pSiNP-nanofiber hybrids release approximately half of their TrkB aptamer, bpV(HOpic), or NGF payload in 2, 10, and >40 days, respectively. The nanofiber hybrids increase neurite extension relative to drug-free control nanofibers in a dorsal root ganglion explant assay.

7.
Nanoscale ; 12(4): 2333-2339, 2020 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-31930266

RESUMO

Synthetic DNA-based oligonucleotides are loaded into porous silicon nanoparticles (pSiNPs) and incorporated into nanofibers of poly(lactide-co-glycolide) (PLGA), poly-l-lactic acid (PLA), or polycaprolactone (PCL). The resulting hybrid nanofibers are characterized for their ability to release the functional oligonucleotide payload under physiologic conditions. Under temperature and pH conditions mimicking physiological values, the PLGA-based nanofibers release >80% of their DNA cargo within 5 days, whereas the PLA and PCL-based fibers require 15 days to release >80% of their cargo. The quantity of DNA released scales with the quantity of DNA-loaded pSiNPs embedded in the nanofibers; mass loadings of between 2.4 and 9.1% (based on mass of DNA-pSiNP construct relative to mass of polymer composite) are investigated. When a responsive DNA-based nanodevice (i.e. molecular beacon) is used as a payload, it retains its functionality during the release period, independent of the polymer used for the formation of the nanofibers.


Assuntos
DNA/química , Nanopartículas/química , Oligonucleotídeos/química , Polímeros/química , Silício/química , Preparações de Ação Retardada/química , Concentração de Íons de Hidrogênio , Teste de Materiais , Nanofibras , Poliésteres/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Porosidade , Medicina Regenerativa , Razão Sinal-Ruído
9.
Am J Med Sci ; 358(2): 143-148, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31200920

RESUMO

BACKGROUND: The influence of time to defibrillation in patients with shockable in-hospital cardiac arrest (IHCA) has not been fully assessed. This study investigated the association between time to defibrillation and neurologic outcome in shockable IHCA survivors. MATERIALS AND METHODS: A 7-year retrospective cohort study was conducted using a prospectively collected registry of adult IHCA patients. Patients whose first documented rhythm was pulseless ventricular tachycardia or ventricular fibrillation and who received defibrillation within 5 minutes were included. RESULTS: Among 1,683 IHCA patients, 261 patients were included. At 28 days, a good neurologic outcome (Cerebral Performance Category score 1 or 2) according to time to defibrillation was seen in 49.0%, 21.1%, 13.4% and 16.5% of patients treated at <2 minutes (n = 128), 2-3 minutes (n = 55), 3-4 minutes (n = 35) and 4-5 minutes (n = 43) after IHCA, respectively. After adjusting for clinical characteristics, a graded inverse association was found after 3 minutes. CONCLUSIONS: A graded inverse association between time to defibrillation and neurologic outcome was observed beyond 3 minutes following cardiac arrest. A target time to defibrillation of <3 minutes may be a practical target goal in resource-limited hospitals.


Assuntos
Reanimação Cardiopulmonar , Estado de Consciência/fisiologia , Cardioversão Elétrica , Parada Cardíaca/terapia , Hospitalização , Idoso , Reanimação Cardiopulmonar/métodos , Reanimação Cardiopulmonar/estatística & dados numéricos , Estudos de Coortes , Cardioversão Elétrica/métodos , Cardioversão Elétrica/estatística & dados numéricos , Feminino , Parada Cardíaca/epidemiologia , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Centros de Atenção Terciária , Fatores de Tempo
10.
ACS Appl Mater Interfaces ; 11(27): 23926-23937, 2019 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-31251556

RESUMO

Silencing of aberrantly expressed microRNAs (miRNAs or miRs) has emerged as one of the strategies for molecular targeted cancer therapeutics. In particular, miR-21 is an oncogenic miRNA overexpressed in many tumors, including ovarian cancer. To achieve efficient administration of anti-miR therapeutics, delivery systems are needed that can ensure local accumulation in the tumor environment, low systemic toxicity, and reduced adverse side effects. In order to develop an improved anti-miR therapeutic agent for the treatment of ovarian cancer, a nanoformulation is engineered that leverages biodegradable porous silicon nanoparticles (pSiNPs) encapsulating an anti-miR-21 locked nucleic acid payload and displaying a tumor-homing peptide for targeted distribution. Targeting efficacy, miR-21 silencing, and anticancer activity are optimized in vitro on a panel of ovarian cancer cell lines, and a formulation of anti-miR-21 in a pSiNP displaying the targeting peptide CGKRK is identified for in vivo evaluation. When this nanoparticulate agent is delivered to mice bearing tumor xenografts, a substantial inhibition of tumor growth is achieved through silencing of miR-21. This study presents the first successful application of tumor-targeted anti-miR porous silicon nanoparticles for the treatment of ovarian cancer in a mouse xenograft model.


Assuntos
Portadores de Fármacos , MicroRNAs , Nanopartículas , Neoplasias Ovarianas , Silício , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Feminino , Humanos , Camundongos , Camundongos Nus , MicroRNAs/química , MicroRNAs/genética , MicroRNAs/farmacologia , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Porosidade , Silício/química , Silício/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Control Release ; 301: 42-53, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-30871996

RESUMO

Macrophages play important and diverse roles during cancer progression. However, cancer therapies based on macrophage modulation are lacking in tools that can recognize and deliver therapeutic payloads to macrophages in a tumor-specific manner. As a result, treatments tend to interfere with normal macrophage functions in healthy organs. We previously identified a macrophage-binding peptide, termed CRV. Here, we show that upon systemic administration into tumor-bearing mice, CRV selectively homes to tumors, extravasates, and preferentially binds to macrophages within. CRV exhibits a higher affinity for tumor macrophages than for other cells in tumors or for other macrophage types elsewhere in the body. We further identified and validated retinoid X receptor beta (RXRB) as the CRV receptor. Intriguingly, although it is known as a nuclear receptor, RXRB shows a prominent cell surface localization that is largely restricted to tumor macrophages. Systemic administration of anti-RXRB antibodies also results in tumor-selective binding to macrophages similar to CRV. Lastly, we demonstrate the ability of CRV to improve the delivery of nano-carriers into solid tumors and macrophages within. In summary, we describe here a novel cell surface marker and targeting tools for tumor macrophages that may aid in future development of macrophage-modulatory cancer therapies.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Portadores de Fármacos/administração & dosagem , Macrófagos/metabolismo , Neoplasias/metabolismo , Peptídeos/administração & dosagem , Animais , Anticorpos/administração & dosagem , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/imunologia , Portadores de Fármacos/farmacocinética , Feminino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Peptídeos/farmacocinética
12.
Materials (Basel) ; 12(4)2019 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-30769924

RESUMO

Porous silicon nanoparticles (pSiNPs) have been utilized within a wide spectrum of biological studies, as well as in chemistry, chemical biology, and biomedical fields. Recently, pSiNPs have been constantly coming under the spotlight, mostly in biomedical applications, due to their advantages, such as controlled-release drug delivery in vivo by hydrolysis-induced degradation, self-reporting property through long life-time photoluminescence, high loading efficiency of substrate into pore, and the homing to specific cells/organ/bacteria by surface functionalization. However, the systematic degradation rate analysis of surface-functionalized pSiNPs in different biological media has not been conducted yet. In this paper, we prepared four different surface-functionalized pSiNPs samples and analyzed the degradation rate in six different media (DI H2O (deionized water), PBS (phosphate-buffered saline), HS (human serum), DMEM (Dulbecco's modified Eagle's medium), LB (lysogeny broth), and BHI (brain heart infusion)). The obtained results will now contribute to understanding the correlation between surface functionalization in the pSiNPs and the degradation rate in different biological media. The characterized data with the author's suggestions will provide useful insights in designing the new pSiNPs formulation for biomedical applications.

13.
Adv Mater ; 30(35): e1802878, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30003620

RESUMO

A nanoparticle system for systemic delivery of therapeutics is described, which incorporates a means of tracking the fate of the nanocarrier and its residual drug payload in vivo by photoluminescence (PL). Porous silicon nanoparticles (PSiNPs) containing the proapoptotic antimicrobial peptide payload, D [KLAKLAK]2 , are monitored by measurement of the intrinsic PL intensity and the PL lifetime of the nanoparticles. The PL lifetime of the PSiNPs is on the order of microseconds, substantially longer than the nanosecond lifetimes typically exhibited by conventional fluorescent tags or by autofluorescence from cells and tissues; thus, emission from the nanoparticles is readily discerned in the time-resolved PL spectrum. It is found that the luminescence lifetime of the PSiNP host decreases as the nanoparticle dissolves in phosphate-buffered saline solution (37 °C), and this correlates with the extent of release of the peptide payload. The time-resolved PL measurement allows tracking of the in vivo fate of PSiNPs injected (via tail vein) into mice. Clearance of the nanoparticles through the liver, kidneys, and lungs of the animals is observed. The luminescence lifetime of the PSiNPs decreases with increasing residence time in the mice, providing a measure of half-life for degradation of the drug nanocarriers.


Assuntos
Nanopartículas , Animais , Luminescência , Camundongos , Peptídeos , Porosidade , Silício
14.
Nat Biomed Eng ; 2(2): 95-103, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29955439

RESUMO

Bacterial resistance to antibiotics has made it necessary to resort to antibiotics that have considerable toxicities. Here, we show that the cyclic 9-amino acid peptide CARGGLKSC (CARG), identified via phage display on Staphylococcus aureus (S. aureus) bacteria and through in vivo screening in mice with S. aureus-induced lung infections, increases the antibacterial activity of CARG-conjugated vancomycin-loaded nanoparticles in S. aureus-infected tissues and reduces the needed overall systemic dose, minimizing side effects. CARG binds specifically to S. aureus bacteria but not Pseudomonas bacteria in vitro, selectively accumulates in S. aureus-infected lungs and skin of mice but not in non-infected tissue and Pseudomonas-infected tissue, and significantly enhances the accumulation of intravenously injected vancomycin-loaded porous silicon nanoparticles bearing the peptide in S. aureus-infected mouse lung tissue. The targeted nanoparticles more effectively suppress staphylococcal infections in vivo relative to equivalent doses of untargeted vancomycin nanoparticles or of free vancomycin. The therapeutic delivery of antibiotic-carrying nanoparticles bearing peptides targeting infected tissue may help combat difficult-to-treat infections.

15.
Nat Commun ; 9(1): 1969, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773788

RESUMO

The incidence of adverse effects and pathogen resistance encountered with small molecule antibiotics is increasing. As such, there is mounting focus on immunogene therapy to augment the immune system's response to infection and accelerate healing. A major obstacle to in vivo gene delivery is that the primary uptake pathway, cellular endocytosis, results in extracellular excretion and lysosomal degradation of genetic material. Here we show a nanosystem that bypasses endocytosis and achieves potent gene knockdown efficacy. Porous silicon nanoparticles containing an outer sheath of homing peptides and fusogenic liposome selectively target macrophages and directly introduce an oligonucleotide payload into the cytosol. Highly effective knockdown of the proinflammatory macrophage marker IRF5 enhances the clearance capability of macrophages and improves survival in a mouse model of Staphyloccocus aureus pneumonia.


Assuntos
Antibacterianos/farmacologia , Terapia Genética/métodos , Macrófagos/efeitos dos fármacos , Pneumonia Estafilocócica/terapia , Staphylococcus aureus/fisiologia , Animais , Antibacterianos/uso terapêutico , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Farmacorresistência Bacteriana/genética , Técnicas de Silenciamento de Genes , Terapia Genética/efeitos adversos , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Lipossomos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Nanopartículas/química , Peptídeos Cíclicos/administração & dosagem , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/mortalidade , Células RAW 264.7 , Interferência de RNA/imunologia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Staphylococcus aureus/efeitos dos fármacos , Análise de Sobrevida , Resultado do Tratamento
16.
Adv Mater ; 30(27): e1800512, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29782671

RESUMO

Photoacoustic (PA) imaging allows visualization of the physiology and pathology of tissues with good spatial resolution and relatively deep tissue penetration. The method converts near-infrared (NIR) laser excitation into thermal expansion, generating pressure transients that are detected with an acoustic transducer. Here, we find that the response of the PA contrast agent indocyanine green (ICG) can be enhanced 17-fold when it is sealed within a rigid nanoparticle. ICG encapsulated in particles composed of porous silicon (pSiNP), porous silica, or calcium silicate all show greater PA contrast relative to equivalent quantities of free ICG, with the pSiNPs showing the strongest enhancement. A liposomal formulation of ICG performs similar to free ICG, suggesting that a rigid host nanostructure is necessary to enhance ICG performance. The improved response of the nanoparticle formulations is attributed to the low thermal conductivity of the porous inorganic hosts and their ability to protect the ICG payload from photolytic and/or thermal degradation. The translational potential of ICG-loaded pSiNPs as photoacoustic probes is demonstrated via imaging of a whole mouse brain.

17.
Adv Mater ; 30(12): e1706785, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29363828

RESUMO

Oriented composite nanofibers consisting of porous silicon nanoparticles (pSiNPs) embedded in a polycaprolactone or poly(lactide-co-glycolide) matrix are prepared by spray nebulization from chloroform solutions using an airbrush. The nanofibers can be oriented by an appropriate positioning of the airbrush nozzle, and they can direct growth of neurites from rat dorsal root ganglion neurons. When loaded with the model protein lysozyme, the pSiNPs allow the generation of nanofiber scaffolds that carry and deliver the protein under physiologic conditions (phosphate-buffered saline (PBS), at 37 °C) for up to 60 d, retaining 75% of the enzymatic activity over this time period. The mass loading of protein in the pSiNPs is 36%, and in the resulting polymer/pSiNP scaffolds it is 3.6%. The use of pSiNPs that display intrinsic photoluminescence (from the quantum-confined Si nanostructure) allows the polymer/pSiNP composites to be definitively identified and tracked by time-gated photoluminescence imaging. The remarkable ability of the pSiNPs to protect the protein payload from denaturation, both during processing and for the duration of the long-term aqueous release study, establishes a model for the generation of biodegradable nanofiber scaffolds that can load and deliver sensitive biologics.


Assuntos
Nanofibras , Animais , Nanopartículas , Polímeros , Porosidade , Ratos , Silício , Engenharia Tecidual , Alicerces Teciduais
18.
Environ Technol ; 39(7): 831-842, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28355496

RESUMO

Anaerobic digestate of piggery wastewater (ADPW) contains high concentrations of ammonia and phosphorus with unbalanced molar ratio. Thus, ammonia remains at a high level even after phosphorus is completely removed through struvite formation. In this study, both ammonia and phosphorus were recovered by adding leachate of sewage sludge ash (SSA) into ADPW. It was demonstrated that 11,600 mg L-1 of total phosphorus and 7266.7 mg L-1 of [Formula: see text]-P were extracted from SSA by using sulfuric acid at the H2SO4/SSA mass ratio of 0.35. ADPW and the leachate of SSA were mixed at the volumetric ratio of 1:1.29, and then struvite was formed at the molar ratio of 1.2 (Mg2+):1.0 ([Formula: see text]-P):1.0 (NH3-N). Removal efficiencies of ammonia and phosphorus were 91.95% and 99.65%, respectively. The obtained struvite was analyzed by various methods and was found to meet the Korean fertilizer standards, except for copper.


Assuntos
Amônia/química , Esgotos , Estruvita/química , Fosfatos , Fósforo , Águas Residuárias
19.
Adv Mater ; 29(39)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28833739

RESUMO

A major obstacle in luminescence imaging is the limited penetration of visible light into tissues and interference associated with light scattering and autofluorescence. Near-infrared (NIR) emitters that can also be excited with NIR radiation via two-photon processes can mitigate these factors somewhat because they operate at wavelengths of 650-1000 nm where tissues are more transparent, light scattering is less efficient, and endogenous fluorophores are less likely to absorb. This study presents photolytically stable, NIR photoluminescent, porous silicon nanoparticles with a relatively high two-photon-absorption cross-section and a large emission quantum yield. Their ability to be targeted to tumor tissues in vivo using the iRGD targeting peptide is demonstrated, and the distribution of the nanoparticles with high spatial resolution is visualized.

20.
J Am Chem Soc ; 138(46): 15106-15109, 2016 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-27933884

RESUMO

Heterocyclic silanes containing Si-N or Si-S bonds in the ring undergo a ring opening reaction with -OH groups at the surface of porous Si nanostructures to generate -SH or -NH functional surfaces, grafted via O-Si bonds. The reaction is substantially faster (0.5-2 h at 25 °C) and more efficient than hydrolytic condensation of trialkoxysilanes on similar hydroxy-terminated surfaces, and the reaction retains the open pore structure and photoluminescence of the quantum-confined silicon nanostructures. The chemistry is sufficiently mild to allow trapping of the test protein lysozyme, which retains its enzymatic activity upon release from the modified porous nanostructure.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...