Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 13(597)2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34108250

RESUMO

Continuous health monitoring and integrated diagnostic devices, worn on the body and used in the home, will help to identify and prevent early manifestations of disease. However, challenges lie ahead in validating new health monitoring technologies and in optimizing data analytics to extract actionable conclusions from continuously obtained health data.


Assuntos
Medicina de Precisão
3.
Nat Biomed Eng ; 4(6): 624-635, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32251391

RESUMO

Technologies for the longitudinal monitoring of a person's health are poorly integrated with clinical workflows, and have rarely produced actionable biometric data for healthcare providers. Here, we describe easily deployable hardware and software for the long-term analysis of a user's excreta through data collection and models of human health. The 'smart' toilet, which is self-contained and operates autonomously by leveraging pressure and motion sensors, analyses the user's urine using a standard-of-care colorimetric assay that traces red-green-blue values from images of urinalysis strips, calculates the flow rate and volume of urine using computer vision as a uroflowmeter, and classifies stool according to the Bristol stool form scale using deep learning, with performance that is comparable to the performance of trained medical personnel. Each user of the toilet is identified through their fingerprint and the distinctive features of their anoderm, and the data are securely stored and analysed in an encrypted cloud server. The toilet may find uses in the screening, diagnosis and longitudinal monitoring of specific patient populations.


Assuntos
Aparelho Sanitário , Desenho de Equipamento , Monitorização Fisiológica/instrumentação , Monitorização Fisiológica/métodos , Adulto , Aprendizado Profundo , Fezes/química , Feminino , Humanos , Masculino , Processamento de Sinais Assistido por Computador , Software , Urina/química , Interface Usuário-Computador
4.
Sci Transl Med ; 10(430)2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29491186

RESUMO

Health care systems primarily focus on patients after they present with disease, not before. The emerging field of precision health encourages disease prevention and earlier detection by monitoring health and disease based on an individual's risk. Active participation in health care can be encouraged with continuous health-monitoring devices, providing a higher-resolution picture of human health and disease. However, the development of monitoring technologies must prioritize the collection of actionable data and long-term user engagement.


Assuntos
Atenção à Saúde/métodos , Medicina de Precisão/métodos , Humanos
5.
Mol Imaging Biol ; 20(1): 55-64, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28631141

RESUMO

PURPOSE: The purposes of this study are to characterize magneto-endosymbiont (ME) labeling of mammalian cells and to discern the subcellular fate of these living contrast agents. MEs are novel magnetic resonance imaging (MRI) contrast agents that are being used for cell tracking studies. Understanding the fate of MEs in host cells is valuable for designing in vivo cell tracking experiments. PROCEDURES: The ME's surface epitopes, contrast-producing paramagnetic magnetosomal iron, and genome were studied using immunocytochemistry (ICC), Fe and MRI contrast measurements, and quantitative polymerase chain reaction (qPCR), respectively. These assays, coupled with other common assays, enabled validation of ME cell labeling and dissection of ME subcellular processing. RESULTS: The assays mentioned above provide qualitative and quantitative assessments of cell labeling, the subcellular localization and the fate of MEs. ICC results, with an ME-specific antibody, qualitatively shows homogenous labeling with MEs. The ferrozine assay shows that MEs have an average of 7 fg Fe/ME, ∼30 % of which contributes to MRI contrast and ME-labeled MDA-MB-231 (MDA-231) cells generally have 2.4 pg Fe/cell, implying ∼350 MEs/cell. Adjusting the concentration of Fe in the ME growth media reduces the concentration of non-MRI contrast-producing Fe. Results from the qPCR assay, which quantifies ME genomes in labeled cells, shows that processing of MEs begins within 24 h in MDA-231 cells. ICC results suggest this intracellular digestion of MEs occurs by the lysosomal degradation pathway. MEs coated with listeriolysin O (LLO) are able to escape the primary phagosome, but subsequently co-localize with LC3, an autophagy-associated molecule, and are processed for digestion. In embryos, where autophagy is transiently suppressed, MEs show an increased capacity for survival and even replication. Finally, transmission electron microscopy (TEM) of ME-labeled MDA-231 cells confirms that the magnetosomes (the MRI contrast-producing particles) remain intact and enable in vivo cell tracking. CONCLUSIONS: MEs are used to label mammalian cells for the purpose of cell tracking in vivo, with MRI. Various assays described herein (ICC, ferrozine, and qPCR) allow qualitative and quantitative assessments of labeling efficiency and provide a detailed understanding of subcellular processing of MEs. In some cell types, MEs are digested, but the MRI-producing particles remain. Coating with LLO allows MEs to escape the primary phagosome, enhances retention slightly, and confirms that MEs are ultimately processed by autophagy. Numerous intracellular bacteria and all endosymbiotically derived organelles have evolved molecular mechanisms to avoid intracellular clearance, and identification of the specific processes involved in ME clearance provides a framework on which to develop MEs with enhanced retention in mammalian cells.


Assuntos
Comunicação Celular , Rastreamento de Células , Nanopartículas de Magnetita/química , Coloração e Rotulagem , Simbiose , Animais , Autofagia , Linhagem Celular Tumoral , Meios de Contraste/química , Ferrozina/metabolismo , Humanos , Ferro/metabolismo , Nanopartículas de Magnetita/ultraestrutura , Camundongos Endogâmicos BALB C , Ratos , Reprodutibilidade dos Testes , Frações Subcelulares/metabolismo
6.
Mol Imaging Biol ; 20(1): 65-73, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28616842

RESUMO

PURPOSE: Magneto-endosymbionts (MEs) show promise as living magnetic resonance imaging (MRI) contrast agents for in vivo cell tracking. Here we characterize the biomedical imaging properties of ME contrast agents, in vitro and in vivo. PROCEDURES: By adapting and engineering magnetotactic bacteria to the intracellular niche, we are creating magneto-endosymbionts (MEs) that offer advantages relative to passive iron-based contrast agents (superparamagnetic iron oxides, SPIOs) for cell tracking. This work presents a biomedical imaging characterization of MEs including: MRI transverse relaxivity (r 2) for MEs and ME-labeled cells (compared to a commercially available iron oxide nanoparticle); microscopic validation of labeling efficiency and subcellular locations; and in vivo imaging of a MDA-MB-231BR (231BR) human breast cancer cells in a mouse brain. RESULTS: At 7T, r 2 relaxivity of bare MEs was higher (250 s-1 mM-1) than that of conventional SPIO (178 s-1 mM-1). Optimized in vitro loading of MEs into 231BR cells yielded 1-4 pg iron/cell (compared to 5-10 pg iron/cell for conventional SPIO). r 2 relaxivity dropped by a factor of ~3 upon loading into cells, and was on the same order of magnitude for ME-loaded cells compared to SPIO-loaded cells. In vivo, ME-labeled cells exhibited strong MR contrast, allowing as few as 100 cells to be detected in mice using an optimized 3D SPGR gradient-echo sequence. CONCLUSIONS: Our results demonstrate the potential of magneto-endosymbionts as living MR contrast agents. They have r 2 relaxivity values comparable to traditional iron oxide nanoparticle contrast agents, and provide strong MR contrast when loaded into cells and implanted in tissue.


Assuntos
Rastreamento de Células , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/química , Simbiose , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Humanos , Camundongos Nus
7.
Immunotherapy ; 9(10): 819-835, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28877626

RESUMO

The success of any given cancer immunotherapy relies on several key factors. In particular, success hinges on the ability to stimulate the immune system in a controlled and precise fashion, select the best treatment options and appropriate therapeutic agents, and use highly effective tools to accurately and efficiently assess the outcome of the immunotherapeutic intervention. Furthermore, a deep understanding and effective utilization of tumor-associated macrophages (TAMs), nanomedicine and biomedical imaging must be harmonized to improve treatment efficacy. Additionally, a keen appreciation of the dynamic interplay that occurs between immune cells and the tumor microenvironment (TME) is also essential. New advances toward the modulation of the immune TME have led to many novel translational research approaches focusing on the targeting of TAMs, enhanced drug and nucleic acid delivery, and the development of theranostic probes and nanoparticles for clinical trials. In this review, we discuss the key cogitations that influence TME, TAM modulations and immunotherapy in solid tumors as well as the methods and resources of tracking the tumor response. The vast array of current nanomedicine technologies can be readily modified to modulate immune function, target specific cell types, deliver therapeutic payloads and be monitored using several different imaging modalities. This allows for the development of more effective treatments, which can be specifically designed for particular types of cancer or on an individual basis. Our current capacities have allowed for greater use of theranostic probes and multimodal imaging strategies that have led to better image contrast, real-time imaging capabilities leveraging targeting moieties, tracer kinetics and enabling more detailed response profiles at the cellular and molecular levels. These novel capabilities along with new discoveries in cancer biology should drive innovation for improved biomarkers for efficient and individualized cancer therapy.


Assuntos
Imunoterapia/métodos , Macrófagos/imunologia , Nanomedicina , Neoplasias/terapia , Nanomedicina Teranóstica , Animais , Diagnóstico por Imagem , Humanos , Neoplasias/diagnóstico , Neoplasias/imunologia , Pesquisa Translacional Biomédica , Microambiente Tumoral
8.
Nat Nanotechnol ; 11(11): 986-994, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27668795

RESUMO

Until now, the Food and Drug Administration (FDA)-approved iron supplement ferumoxytol and other iron oxide nanoparticles have been used for treating iron deficiency, as contrast agents for magnetic resonance imaging and as drug carriers. Here, we show an intrinsic therapeutic effect of ferumoxytol on the growth of early mammary cancers, and lung cancer metastases in liver and lungs. In vitro, adenocarcinoma cells co-incubated with ferumoxytol and macrophages showed increased caspase-3 activity. Macrophages exposed to ferumoxytol displayed increased mRNA associated with pro-inflammatory Th1-type responses. In vivo, ferumoxytol significantly inhibited growth of subcutaneous adenocarcinomas in mice. In addition, intravenous ferumoxytol treatment before intravenous tumour cell challenge prevented development of liver metastasis. Fluorescence-activated cell sorting (FACS) and histopathology studies showed that the observed tumour growth inhibition was accompanied by increased presence of pro-inflammatory M1 macrophages in the tumour tissues. Our results suggest that ferumoxytol could be applied 'off label' to protect the liver from metastatic seeds and potentiate macrophage-modulating cancer immunotherapies.


Assuntos
Antineoplásicos/farmacologia , Óxido Ferroso-Férrico/farmacologia , Macrófagos/efeitos dos fármacos , Nanopartículas Metálicas/química , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , Feminino , Óxido Ferroso-Férrico/química , Humanos , Inflamação/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Nanopartículas Metálicas/uso terapêutico , Camundongos Endogâmicos NOD , Camundongos Endogâmicos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
Int J Biochem Cell Biol ; 75: 143-7, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26783938

RESUMO

In contact with biological fluids diverse type of biomolecules (e.g., proteins) adsorb onto nanoparticles forming protein corona. Surface properties of the coated nanoparticles, in terms of type and amount of associated proteins, dictate their interactions with biological systems and thus biological fate, therapeutic efficiency and toxicity. In this perspective, we will focus on the recent advances and pitfalls in the protein corona field.


Assuntos
Coroa de Proteína , Animais , Transporte Biológico , Liberação Controlada de Fármacos , Humanos , Nanopartículas/química , Coroa de Proteína/química
10.
J Biomed Opt ; 20(5): 051022, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25562608

RESUMO

Low level light therapy (LLLT) has numerous therapeutic benefits, including improving wound healing, but the precise mechanisms involved are not well established; in particular, the underlying role of cytochrome C oxidase (C-ox) as the primary photoacceptor and the associated biochemical mechanisms still require further investigation. We previously showed the nitric oxide (NO) donating drug nitrosyl-cobinamide (NO-Cbi) enhances wound healing through a cGMP/cGMP-dependent protein kinase/ERK1/2 mechanism. Here, we show that the combination of LLLT and NO-Cbi markedly improves wound healing compared to either treatment alone. LLLT-enhanced wound healing proceeded through an electron transport chain-C-ox-dependent mechanism with a reduction of reactive oxygen species and increased adenosine triphosphate production. C-ox was validated as the primary photoacceptor by three observations: increased oxygen consumption, reduced wound healing in the presence of sodium azide, and disassociation of cyanide, a known C-ox ligand, following LLLT. We conclude that LLLT and NO-Cbi accelerate wound healing through two independent mechanisms, the electron transport chain-C-ox pathway and cGMP signaling, respectively, with both resulting in ERK1/2 activation.


Assuntos
Cobamidas/química , Terapia com Luz de Baixa Intensidade , Cicatrização , Trifosfato de Adenosina/química , Linhagem Celular Tumoral , Proliferação de Células , Cianetos/química , GMP Cíclico/química , Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons/química , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Temperatura Alta , Humanos , Luz , Óxido Nítrico/química , Nitrogênio/química , Consumo de Oxigênio , Fotoquímica , Espécies Reativas de Oxigênio , Transdução de Sinais , Azida Sódica/química
11.
J Biomed Opt ; 19(3): 38001, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24638250

RESUMO

Low-level light therapy has been shown to improve in vitro wound healing. However, well-defined parameters of different light sources for this therapy are lacking. The goal of this study was (1) to determine if the wavelengths tested are effective for in vitro wound healing and (2) to compare a laser and a light-emitting diode (LED) source at similar wavelengths. We show four wavelengths, delivered by either a laser or LED array, improved in vitro wound healing in A549, U2OS, and PtK2 cells. Improved wound healing occurred through increased cell migration demonstrated through scratch wound and transwell assays. Cell proliferation was tested by the (3-(4,5-dimethylthiazol-2-yl)-5-(3-car-boxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay and was found generally not to be involved in the wound healing process. The laser and LED sources were found to be comparable when equal doses of light were applied. The biological response measured was similar in most cases. We conclude that the laser at 652 (5.57 mW/cm2, 10.02 J/cm2) and 806 nm (1.30 mW/cm2, 2.334 J/cm2) (full bandwidth 5 nm), and LED at 637 (5.57 mW/cm2, 10.02 J/cm2) and 901 nm (1.30 mW/cm2, 2.334 J/cm2) (full bandwidth 17 and 69 nm respectively) induce comparable levels of cell migration and wound closure.


Assuntos
Movimento Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Lasers , Fototerapia/métodos , Cicatrização/efeitos da radiação , Animais , Linhagem Celular , Humanos , Luz , Camundongos
12.
Mol Ther Nucleic Acids ; 2: e123, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-24045712

RESUMO

We previously identified short synthetic shRNAs (sshRNAs) that target a conserved hepatitis C virus (HCV) sequence within the internal ribosome entry site (IRES) of HCV and potently inhibit HCV IRES-linked gene expression. To assess in vivo liver delivery and activity, the HCV-directed sshRNA SG220 was formulated into lipid nanoparticles (LNP) and injected i.v. into mice whose livers supported stable HCV IRES-luciferase expression from a liver-specific promoter. After a single injection, RNase protection assays for the sshRNA and (3)H labeling of a lipid component of the nanoparticles showed efficient liver uptake of both components and long-lasting survival of a significant fraction of the sshRNA in the liver. In vivo imaging showed a dose-dependent inhibition of luciferase expression (>90% 1 day after injection of 2.5 mg/kg sshRNA) with t1/2 for recovery of about 3 weeks. These results demonstrate the ability of moderate levels of i.v.-injected, LNP-formulated sshRNAs to be taken up by liver hepatocytes at a level sufficient to substantially suppress gene expression. Suppression is rapid and durable, suggesting that sshRNAs may have promise as therapeutic agents for liver indications.Molecular Therapy-Nucleic Acids (2013) 2, e123; doi:10.1038/mtna.2013.50; published online 17 September 2013.

13.
Cell Signal ; 25(12): 2374-82, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23920342

RESUMO

Nitric oxide (NO) donors have been shown to improve wound healing, but the mechanism is not well defined. Here we show that the novel NO donor nitrosyl-cobinamide (NO-Cbi) improved in vitro wound healing in several cell types, including an established line of lung epithelial cells and primary human lung fibroblasts. On a molar basis, NO-Cbi was more effective than two other NO donors, with the effective NO-Cbi concentration ranging from 3 to 10µM, depending on the cell type. Improved wound healing was secondary to increased cell migration and not cell proliferation. The wound healing effect of NO-Cbi was mediated by cGMP, mainly through cGMP-dependent protein kinase type I (PKGI), as determined using pharmacological inhibitors and activators, and siRNAs targeting PKG type I and II. Moreover, we found that Src and ERK were two downstream mediators of NO-Cbi's effect. We conclude that NO-Cbi is a potent inducer of cell migration and wound closure, acting via cGMP, PKG, Src, and extracellular signal regulated kinase (ERK).


Assuntos
Movimento Celular/efeitos dos fármacos , Cobamidas/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxidos de Nitrogênio/farmacologia , Cicatrização/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Cobamidas/química , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Pulmão/citologia , Doadores de Óxido Nítrico/química , Óxidos de Nitrogênio/química , Transdução de Sinais/efeitos dos fármacos
14.
J Cell Sci ; 126(Pt 7): 1626-36, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23418348

RESUMO

The two isoforms of type I cGMP-dependent protein kinase (PKGIα and PKGIß) differ in their first ∼100 amino acids, giving each isoform unique dimerization and autoinhibitory domains. The dimerization domains form coiled-coil structures and serve as platforms for isoform-specific protein-protein interactions. Using the PKGIß dimerization domain as an affinity probe in a proteomic screen, we identified the actin/myosin-associated protein caldesmon (CaD) as a PKGIß-specific binding protein. PKGIß phosphorylated human CaD on serine 12 in vitro and in intact cells. Phosphorylation on serine 12 or mutation of serine 12 to glutamic acid (S12E) reduced the interaction between CaD and myosin IIA. Because CaD inhibits myosin ATPase activity and regulates cell motility, we examined the effects of PKGIß and CaD on cell migration and invasion. Inhibition of the NO/cGMP/PKG pathway reduced migration and invasion of human breast cancer cells, whereas PKG activation enhanced their motility and invasion. siRNA-mediated knockdown of endogenous CaD had pro-migratory and pro-invasive effects in human breast cancer cells. Reconstituting cells with wild-type CaD slowed migration and invasion; however, CaD containing a phospho-mimetic S12E mutation failed to reverse the pro-migratory and pro-invasive activity of CaD depletion. Our data suggest that PKGIß enhances breast cancer cell motility and invasive capacity, at least in part, by phosphorylating CaD. These findings identify a pro-migratory and pro-invasive function for PKGIß in human breast cancer cells, suggesting that PKGIß is a potential target for breast cancer treatment.


Assuntos
Actinas/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Miosinas/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Feminino , Humanos , Espectrometria de Massas
15.
J Am Chem Soc ; 133(25): 9855-62, 2011 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-21615158

RESUMO

Membrane proteins comprise a third of the human genome, yet present challenging targets for reverse chemical genetics. For example, although implicated in numerous diseases including multiple myeloma, the membrane protein caveolin-1 appears to offer a poor target for the discovery of synthetic ligands due to its largely unknown structure and insolubility. To break this impasse and identify new classes of caveolae controlling lead compounds, we applied phage-based, reverse chemical genetics for the discovery of caveolin-1 ligands derived from the anti-HIV therapeutic T20. Substitution of homologous residues into the T20 sequence used a process analogous to medicinal chemistry for the affinity maturation to bind caveolin. The resultant caveolin-1 ligands bound with >1000-fold higher affinity than wild-type T20. Two types of ELISAs and isothermal titration calorimetry (ITC) measurements demonstrated high affinity binding to caveolin by the T20 variants with K(d) values in the 150 nM range. Microscopy experiments with the highest affinity caveolin ligands confirmed colocalization of the ligands with endogenous caveolin in NIH 3T3 cells. The results establish the foundation for targeting caveolin and caveolae formation in living cells.


Assuntos
Caveolina 1/metabolismo , Evolução Molecular Direcionada/métodos , Descoberta de Drogas/métodos , Células 3T3 , Animais , Cavéolas , Caveolina 1/antagonistas & inibidores , Enfuvirtida , Proteína gp41 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/farmacologia , Inibidores da Fusão de HIV , Humanos , Ligantes , Proteínas de Membrana , Camundongos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Ligação Proteica
16.
Sci Rep ; 1: 158, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22355673

RESUMO

The accessibility of skin makes it an ideal target organ for nucleic acid-based therapeutics; however, effective patient-friendly delivery remains a major obstacle to clinical utility. A variety of limited and inefficient methods of delivering nucleic acids to keratinocytes have been demonstrated; further advances will require well-characterized reagents, rapid noninvasive assays of delivery, and well-developed skin model systems. Using intravital fluorescence and bioluminescence imaging and a standard set of reporter plasmids we demonstrate transfection of cells in mouse and human xenograft skin using intradermal injection and two microneedle array delivery systems. Reporter gene expression could be detected in individual keratinocytes, in real-time, in both mouse skin as well as human skin xenografts. These studies revealed that non-invasive intravital imaging can be used as a guide for developing gene delivery tools, establishing a benchmark for comparative testing of nucleic acid skin delivery technologies.


Assuntos
Queratinócitos/metabolismo , Plasmídeos/administração & dosagem , Animais , Sistemas de Liberação de Medicamentos , Epiderme/metabolismo , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Luciferases/genética , Luciferases/metabolismo , Medições Luminescentes , Camundongos , Microinjeções , Microscopia de Fluorescência , Plasmídeos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transplante de Pele , Transfecção , Transplante Heterólogo
17.
Invest Ophthalmol Vis Sci ; 52(3): 1780-7, 2011 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-21087969

RESUMO

PURPOSE: Subthreshold retinal phototherapy demonstrated clinical efficacy for the treatment of diabetic macular edema without visible signs of retinal damage. To assess the range of cellular responses to sublethal hyperthermia, expression of the gene encoding a 70 kDa heat shock protein (HSP70) was evaluated after laser irradiation using a transgenic reporter mouse. METHODS: One hundred millisecond, 532 nm laser exposures with 400 µm beam diameter were applied to the retina surrounding the optic nerve in 32 mice. Transcription from the HSP70 promoter was assessed relative to the control eye using a bioluminescence assay at 7 hours after laser application. The retinal pigmented epithelium (RPE) viability threshold was determined with a fluorescence assay. A computational model was developed to estimate temperature and the extent of cell damage. RESULTS: A significant increase in HSP70 transcription was found at exposures over 20 mW, half the threshold power for RPE cell death. Computational modeling estimated peak temperature T = 49°C at HSP70 expression threshold. At RPE viability threshold, T = 57°C. Similar temperatures and damage indices were calculated for clinical subvisible retinal treatment parameters. CONCLUSIONS: Beneficial effects of laser therapy have been previously shown to extend beyond those resulting from destruction of tissue. One hundred millisecond laser exposures at approximately half the threshold power of RPE damage induced transcription of HSP70, an indication of cellular response to sublethal thermal stress. A computational model of retinal hyperthermia can guide further optimization of laser parameters for nondamaging phototherapy.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas de Choque Térmico HSP70/genética , Fotocoagulação a Laser/efeitos adversos , Lasers de Estado Sólido/efeitos adversos , Retina/lesões , Animais , Sobrevivência Celular , Simulação por Computador , Análise de Elementos Finitos , Perfilação da Expressão Gênica , Genes Reporter , Medições Luminescentes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo
18.
J Pharm Sci ; 99(10): 4261-6, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20737633

RESUMO

A key challenge in developing RNAi-based therapeutics is efficient delivery of functional short interfering RNA (siRNA) to target cells. To address this need, we have used a supercritical CO(2) process to incorporate siRNA in biodegradable polymer nanoparticles (NPs) for in vivo sustained release. By this means we have obtained complete encapsulation of the siRNA with minimal initial burst effect from the surface of the NPs. The slow release of a fluorescently labeled siRNA mimic (siGLO Red) was observed for up to 80 days in vivo after intradermal injection into mouse footpads. In vivo gene silencing experiments were also performed, showing reduction of GFP signal in the epidermis of a reporter transgenic mouse model, which demonstrates that the siRNA retained activity following release from the polymer NPs.


Assuntos
Materiais Biocompatíveis , Nanopartículas , RNA Interferente Pequeno/administração & dosagem , Pele , Animais , Corantes Fluorescentes , Inativação Gênica , Genes Reporter , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Varredura
19.
Mol Ther ; 18(9): 1667-74, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20571543

RESUMO

Despite rapid progress in the development of potent and selective small interfering RNA (siRNA) agents for skin disorders, translation to the clinic has been hampered by the lack of effective, patient-friendly delivery technologies. The stratum corneum poses a formidable barrier to efficient delivery of large and/or charged macromolecules including siRNAs. Intradermal siRNA injection results in effective knockdown of targeted gene expression but is painful and the effects are localized to the injection site. The use of microneedle arrays represents a less painful delivery method and may have utility for the delivery of nucleic acids, including siRNAs. For this purpose, we developed a loadable, dissolvable protrusion array device (PAD) that allows skin barrier penetration. The PAD tips dissolve upon insertion, forming a gel-like plug that releases functional cargo. PAD-mediated delivery of siRNA (modified for enhanced stability and cellular uptake) resulted in effective silencing of reporter gene expression in a transgenic reporter mouse model. PAD delivery of luciferase reporter plasmids resulted in expression in cells of the ear, back, and footpad skin as assayed by intravital bioluminescence imaging. These results support the use of PADs for delivery of functional nucleic acids to cells in the skin with an efficiency that may support clinical translation.


Assuntos
Portadores de Fármacos , Genes Reporter/genética , Plasmídeos/genética , RNA Interferente Pequeno/fisiologia , Pele/metabolismo , Animais , Feminino , , Inativação Gênica/fisiologia , Camundongos , Microscopia de Fluorescência , RNA Interferente Pequeno/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...