Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 1541, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33750829

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by marked desmoplasia and drug resistance due, in part, to poor drug delivery to extravascular tumor tissue. Here, we report that carcinoma-associated fibroblasts (CAFs) induce ß5 integrin expression in tumor cells in a TGF-ß dependent manner, making them an efficient drug delivery target for the tumor-penetrating peptide iRGD. The capacity of iRGD to deliver conjugated and co-injected payloads is markedly suppressed when ß5 integrins are knocked out in the tumor cells. Of note, ß5 integrin knock-out in tumor cells leads to reduced disease burden and prolonged survival of the mice, demonstrating its contribution to PDAC progression. iRGD significantly potentiates co-injected chemotherapy in KPC mice with high ß5 integrin expression and may be a powerful strategy to target an aggressive PDAC subpopulation.


Asunto(s)
Cadenas beta de Integrinas/genética , Cadenas beta de Integrinas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Sistemas de Liberación de Medicamentos , Quimioterapia , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Oligopéptidos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
2.
Nat Biomed Eng ; 2(2): 95-103, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29955439

RESUMEN

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.

3.
Sci Adv ; 4(5): eaat0351, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29795786

RESUMEN

Noninvasive stimulation of cells is crucial for the accurate examination and control of their function both at the cellular and the system levels. To address this need, we present a pioneering optical stimulation platform that does not require genetic modification of cells but instead capitalizes on unique optoelectronic properties of graphene, including its ability to efficiently convert light into electricity. We report the first studies of optical stimulation of cardiomyocytes via graphene-based biointerfaces (G-biointerfaces) in substrate-based and dispersible configurations. The efficiency of stimulation via G-biointerfaces is independent of light wavelength but can be tuned by changing the light intensity. We demonstrate that an all-optical evaluation of use-dependent drug effects in vitro can be enabled using substrate-based G-biointerfaces. Furthermore, using dispersible G-biointerfaces in vivo, we perform optical modulation of the heart activity in zebrafish embryos. Our discovery is expected to empower numerous fundamental and translational biomedical studies.


Asunto(s)
Grafito/química , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/efectos de la radiación , Nanoestructuras , Estimulación Luminosa , Animales , Fenómenos Biofísicos , Células Cultivadas , Concentración de Iones de Hidrógeno , Luz , Ratas , Temperatura , Pez Cebra
4.
Nanomedicine ; 14(4): 1279-1287, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29597048

RESUMEN

Canine lower urinary tract neoplasia is a clinically important disease process that has high mortality due to late stage diagnosis and poorly durable response to treatment. Non-invasive diagnostic techniques (e.g. dipstick test, urine cytology) currently have poor diagnostic value, while more invasive tests (e.g. cystoscopy and biopsy) are costly and often require general anesthesia. We have developed and herein describe a quantitative cytological analysis method based on the use of surface-enhanced Raman spectroscopy (SERS), for identifying cancerous transitional cells in urine using SERS biotags (SBTs) carrying the peptide PLZ4 (amino acid sequence cQDGRMGFc) that targets malignant transitional cells. By analyzing the ratio of the PLZ4-SBTs to an on board control we were able to show that transitional cells had significantly higher ratios (P < 0.05) in patients diagnosed with transitional cell carcinoma (TCC) than in healthy samples.


Asunto(s)
Carcinoma de Células Transicionales/diagnóstico , Espectrometría Raman/métodos , Animales , Biomarcadores de Tumor/orina , Biopsia/métodos , Carcinoma de Células Transicionales/orina , Cistoscopía/métodos , Perros , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/orina
5.
Nat Commun ; 9(1): 1070, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29523838

RESUMEN

The original version of the Supplementary Information associated with this Article inadvertently omitted Supplementary Table 1. The HTML has now been updated to include a corrected version of the Supplementary Information.

6.
Nat Commun ; 8(1): 1403, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29123083

RESUMEN

Cerebrovascular changes occur in Alzheimer's disease (AD). Using in vivo phage display, we searched for molecular markers of the neurovascular unit, including endothelial cells and astrocytes, in mouse models of AD. We identified a cyclic peptide, CDAGRKQKC (DAG), that accumulates in the hippocampus of hAPP-J20 mice at different ages. Intravenously injected DAG peptide homes to neurovascular unit endothelial cells and to reactive astrocytes in mouse models of AD. We identified connective tissue growth factor (CTGF), a matricellular protein that is highly expressed in the brain of individuals with AD and in mouse models, as the target of the DAG peptide. We also showed that exogenously delivered DAG homes to the brain in mouse models of glioblastoma, traumatic brain injury, and Parkinson's disease. DAG may potentially be used as a tool to enhance delivery of therapeutics and imaging agents to sites of vascular changes and astrogliosis in diseases associated with neuroinflammation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos Cíclicos/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/patología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Modelos Animales de Enfermedad , Hipocampo/irrigación sanguínea , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Biblioteca de Péptidos , Péptidos Cíclicos/química , Unión Proteica
7.
Nat Commun ; 8(1): 343, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28839238

RESUMEN

In vivo tumor imaging with nanoprobes suffers from poor tumor specificity. Here, we introduce a nanosystem, which allows selective background quenching to gain exceptionally tumor-specific signals. The system uses near-infrared quantum dots and a membrane-impermeable etchant, which serves as a cation donor. The etchant rapidly quenches the quantum dots through cation exchange (ionic etching), and facilitates renal clearance of metal ions released from the quantum dots. The quantum dots are intravenously delivered into orthotopic breast and pancreas tumors in mice by using the tumor-penetrating iRGD peptide. Subsequent etching quenches excess quantum dots, leaving a highly tumor-specific signal provided by the intact quantum dots remaining in the extravascular tumor cells and fibroblasts. No toxicity is noted. The system also facilitates the detection of peritoneal tumors with high specificity upon intraperitoneal tumor targeting and selective etching of excess untargeted quantum dots. In vivo cation exchange may be a promising strategy to enhance specificity of tumor imaging.The imaging of tumors in vivo using nanoprobes has been challenging due to the lack of sufficient tumor specificity. Here, the authors develop a tumor-specific quantum dot system that permits in vivo cation exchange to achieve selective background quenching and high tumor-specific imaging.


Asunto(s)
Cationes/química , Diagnóstico por Imagen/métodos , Neoplasias/diagnóstico por imagen , Puntos Cuánticos , Animales , Línea Celular Tumoral , Humanos , Ratones , Nanomedicina/métodos , Neoplasias/patología , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Trasplante Heterólogo
8.
Nanoscale ; 9(28): 10094-10100, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28695222

RESUMEN

Attaching affinity ligands to nanoparticles (NPs) increases selectivity for targeting cells and tissues, and can result in improved sensitivity and reduced off-target toxicity in diagnostic and therapeutic systems. The decision over key features - NP size, shape, coating strategies and targeting ligands for clinical translation is often hampered by a lack of quantitative in vivo NP homing assays. Sensitive, internally controlled assays are needed which allow for quantitative comparisons (auditions) among various formulations of targeted NPs. We recently reported the development of peptide-functionalized, isotopically-barcoded silver NPs (AgNPs) for ultrasensitive studies centered on measuring relative ratios of NP internalization into cultured cells. Here we evaluated the application of this technology for NP homing studies in live mice using peptides with previously described tissue tropism; one peptide that favors vascular beds of the normal lungs (RPARPAR; receptor neuropilin-1, or NRP-1) and another that is selective for central nervous system vessels (CAGALCY). Equimolar mixtures of the peptide-targeted Ag107-NPs and Ag109 control particles were mixed and injected intravenously. Distribution profiles of Ag107 and Ag109 in tissue extracts were determined simultaneously through inductively coupled plasma mass spectrometry (ICP-MS). Compared to non-targeted particles up to ∼9-fold increased lung accumulation was seen for RPARPAR-OH AgNPs (but not for AgNPs functionalized with RPARPAR-NH2, which does not bind to NRP-1). Similarly, AgNPs functionalized with the brain-homing CAGALCY peptide were overrepresented in brain extracts. Spatial distribution (mapping) analysis by laser ablation ICP-MS (LA-ICP-MS) was used to determine the ratio Ag107/Ag109 in tissue cryosections. The mapping demonstrated preferential accumulation of the RPARPAR-AgNPs in the perivascular areas around pulmonary veins, and CAGALCY AgNPs accumulated in discrete areas of the brain (e.g. in the vessels of cerebellar fibrillary tracts). Based on these results, the internally controlled ratiometric AgNP system is suitable for quantitative studies of the effect of targeting ligands on NP biodistribution, at average tissue concentration and distribution at the microscopic level. The platform might be particularly relevant for target sites with high local variability in uptake, such as tumors.


Asunto(s)
Nanopartículas del Metal , Terapia Molecular Dirigida , Plata/farmacocinética , Animales , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Péptidos/administración & dosificación , Distribución Tisular
9.
ACS Appl Mater Interfaces ; 8(44): 30449-30457, 2016 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-27754645

RESUMEN

A theranostic nanoparticle with biochemically triggered antibacterial activity is demonstrated. Metallic silver is deposited onto porous silicon nanoparticles (pSiNPs) by galvanic displacement. When aqueous diaminesilver ([Ag(NH3)2]+) is used as a silver source, the pSiNPs template the crystalline silver as small (mean diameter 13 nm) and well-dispersed nanoparticles embedded within and on the larger (100 nm) pSiNPs. The silver nanoparticles (AgNPs) quench intrinsic photoluminescence (PL) from the porous silicon (pSi) matrix. When exposed to an aqueous oxidant, the AgNPs are preferentially etched, Ag+ is released into solution, and PL from the pSi carrier is recovered. The released Ag+ results in 90% killing of (Gram-negative) Pseudomonas aeruginosa and (Gram-positive) Staphylococcus aureus within 3 h. When conjugated with the TAT peptide (sequence RKKRRQRRR), the silver-deposited porous silicon (pSi-Ag) nanocomposite shows distinct targeting toward S. aureus bacteria in vitro. Intravenously injected TAT-conjugated pSi-Ag nanoparticles accumulate in the liver, spleen, and lungs of mice, and the in vivo release of Ag+ and recovery of PL from pSi are demonstrated by the subsequent intraperitoneal administration of a hexacyanoferrate solution. The released Ag+ leads to a significant bacterial count reduction in liver tissue relative to the control. The data demonstrate the feasibility of the targeted and triggered delivery of antibacterial Ag+ ion in vivo, using a self-reporting and nontoxic nanocarrier.


Asunto(s)
Nanopartículas del Metal , Animales , Antibacterianos , Ratones , Pruebas de Sensibilidad Microbiana , Silicio , Plata , Staphylococcus aureus , Nanomedicina Teranóstica
10.
Nano Lett ; 16(9): 5495-502, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27490923

RESUMEN

Glutamatergic cytotoxicity mediated by overactivation of N-methyl-d-aspartate receptors (NMDARs) is implicated in numerous neurological disorders. To be therapeutically viable, NMDAR antagonists must preserve physiological role of synaptic NMDARs (sNMDARs) in synaptic transmission and block only excessive pathological activation of NMDARs. Here we present a novel NMDAR antagonist that satisfies this two-fold requirement by exploiting spatial differences in NMDAR subcellular locations. Specifically, we designed a hybrid nanodrug (AuM) to be larger than the synaptic cleft by attaching memantine, NMDAR antagonist, via polymer linkers to a gold nanoparticle. We show that AuM efficiently and selectively inhibited extrasynaptic NMDARs (eNMDARs), while having no effect on sNMDARs and synaptic transmission. AuM exhibited neuroprotective properties both in vitro and ex vivo during such neurotoxic insults as NMDAR-mediated cytotoxicity in cerebrocortical cell culture and oxygen-glucose deprivation in acute hippocampal slices. Furthermore, AuM prevented dendritic spine loss triggered by Aß oligomers in organotypic hippocampal slices and was more effective than free memantine. Using a novel rational design strategy, we demonstrate a proof of concept for a new class of neuroprotective drugs that might be beneficial for treatment of several neurological disorders.


Asunto(s)
Nanopartículas del Metal , Neuronas/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Transmisión Sináptica , Animales , Células Cultivadas , Corteza Cerebral/citología , Oro , Memantina/farmacología , Fármacos Neuroprotectores/farmacología , Ratas Sprague-Dawley , Sinapsis
11.
Adv Funct Mater ; 26(2): 267-276, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-27441036

RESUMEN

The rapid development of fluorescence imaging technologies requires concurrent improvements in the performance of fluorescent probes. Quantum dots have been extensively used as an imaging probe in various research areas because of their inherent advantages based on unique optical and electronic properties. However, their clinical translation has been limited by the potential toxicity especially from cadmium. Here, a versatile bioimaging probe is developed by using highly luminescent cadmium-free CuInSe2/ZnS core/shell quantum dots conjugated with CGKRK (Cys-Gly-Lys-Arg-Lys) tumor-targeting peptides. This probe exhibits excellent photostability, reasonably long circulation time, minimal toxicity, and strong tumor-specific homing property. The most important feature of this probe is that it shows distinctive versatility in tumor-targeted multimodal imaging including near-infrared, time-gated, and two-photon imaging in different tumor models. In a glioblastoma mouse model, the targeted probe clearly denotes tumor boundaries and positively labels a population of diffusely infiltrating tumor cells, suggesting its utility in precise tumor detection during surgery. This work lays a foundation for potential clinical translation of the probe.

12.
Nat Commun ; 7: 11980, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27351915

RESUMEN

Traumatic brain injury (TBI) is a major health and socio-economic problem, but no pharmacological agent is currently approved for the treatment of acute TBI. Thus, there is a great need for advances in this field. Here, we describe a short peptide (sequence CAQK) identified by in vivo phage display screening in mice with acute brain injury. The CAQK peptide selectively binds to injured mouse and human brain, and systemically injected CAQK specifically homes to sites of brain injury in mouse models. The CAQK target is a proteoglycan complex upregulated in brain injuries. Coupling to CAQK increased injury site accumulation of systemically administered molecules ranging from a drug-sized molecule to nanoparticles. CAQK-coated nanoparticles containing silencing oligonucleotides provided the first evidence of gene silencing in injured brain parenchyma by systemically administered siRNA. These findings present an effective targeting strategy for the delivery of therapeutics in clinical management of acute brain injuries.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Sistemas de Liberación de Medicamentos , Péptidos , Anciano , Animales , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Matriz Extracelular/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad
13.
J Control Release ; 232: 188-95, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27106816

RESUMEN

Tumor penetrating peptides contain a cryptic (R/K)XX(R/K) CendR element that must be C-terminally exposed to trigger neuropilin-1 (NRP-1) binding, cellular internalization and malignant tissue penetration. The specific proteases that are involved in processing of tumor penetrating peptides identified using phage display are not known. Here we design de novo a tumor-penetrating peptide based on consensus cleavage motif of urokinase-type plasminogen activator (uPA). We expressed the peptide, uCendR (RPARSGR↓SAGGSVA, ↓ shows cleavage site), on phage or coated it onto silver nanoparticles and showed that it is cleaved by uPA, and that the cleavage triggers binding to recombinant NRP-1 and to NPR-1-expressing cells. Upon systemic administration to mice bearing uPA-overexpressing breast tumors, FAM-labeled uCendR peptide and uCendR-coated nanoparticles preferentially accumulated in tumor tissue. We also show that uCendR phage internalization into cultured cancer cells and its penetration in explants of murine tumors and clinical tumor explants can be potentiated by combining the uCendR peptide with tumor-homing module, CRGDC. Our work demonstrates the feasibility of designing tumor-penetrating peptides that are activated by a specific tumor protease. As upregulation of protease expression is one of the hallmarks of cancer, and numerous tumor proteases have substrate specificities compatible with proteolytic unmasking of cryptic CendR motifs, the strategy described here may provide a generic approach for designing proteolytically-actuated peptides for tumor-penetrative payload delivery.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Neoplasias Mamarias Animales/metabolismo , Péptidos/administración & dosificación , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Bacteriófago T7 , Línea Celular Tumoral , Portadores de Fármacos/farmacocinética , Humanos , Nanopartículas del Metal/administración & dosificación , Ratones Endogámicos BALB C , Péptidos/farmacocinética , Plata/administración & dosificación , Plata/farmacocinética
14.
Chembiochem ; 17(7): 570-5, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26895508

RESUMEN

Cell surface p32, the target of LyP-1 homing peptide, is upregulated in tumors and atherosclerotic plaques and has been widely used as a receptor for systemic delivery of payloads. Here, we identified an improved LyP-1-mimicking peptide (TT1, CKRGARSTC). We used this peptide in a fluorescence polarization-based high-throughput screening of a 50,000-compound chemical library and identified a panel of compounds that bind p32 with low micromolar affinity. Among the hits identified in the screen, two compounds were shown to specifically bind to p32 in multiple assays. One of these compounds was chosen for an in vivo study. Nanoparticles surface-functionalized with this compound specifically adhered to surfaces coated with recombinant p32 and, when injected intravenously, homed to p32-expressing breast tumors in mice. This compound provides a lead for the development of p32-targeted affinity ligands that circumvent some of the limitations of peptide-based probes in guided drug delivery.


Asunto(s)
Aminopiridinas/química , Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Etilenodiaminas/química , Proteínas Mitocondriales/administración & dosificación , Péptidos Cíclicos/administración & dosificación , Aminopiridinas/farmacología , Animales , Antineoplásicos/administración & dosificación , Proteínas Portadoras , Línea Celular Tumoral , Etilenodiaminas/farmacología , Femenino , Humanos , Ligandos , Ratones , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Nanopartículas/química
15.
Mol Cancer Ther ; 15(4): 670-9, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26880267

RESUMEN

Peritoneal carcinomatosis is present in more than 60% of gastric cancer, 40% of ovarian cancer, and 35% of colon cancer patients. It is the second most common cause of cancer-related mortality, with a median survival of 1 to 3 months. Cytoreductive surgery combined with intraperitoneal chemotherapy is the current clinical treatment, but achieving curative drug accumulation and penetration in peritoneal carcinomatosis lesions remains an unresolved challenge. Here, we used flexible and pH-sensitive polymersomes for payload delivery to peritoneal gastric (MKN-45P) and colon (CT26) carcinoma in mice. Polymersomes were loaded with paclitaxel and in vitro drug release was studied as a function of pH and time. Paclitaxel-loaded polymersomes remained stable in aqueous solution at neutral pH for up to 4 months. In cell viability assay on cultured cancer cell lines (MKN-45P, SKOV3, CT26), paclitaxel-loaded polymersomes were more toxic than free drug or albumin-bound paclitaxel (Abraxane). Intraperitoneally administered fluorescent polymersomes accumulated in malignant lesions, and immunofluorescence revealed an intense signal inside tumors with no detectable signal in control organs. A dual targeting of tumors was observed: direct (circulation-independent) penetration, and systemic, blood vessel-associated accumulation. Finally, we evaluated preclinical antitumor efficacy of paclitaxel-polymersomes in the treatment of MKN-45P disseminated gastric carcinoma using a total dose of 7 mg/kg. Experimental therapy with paclitaxel-polymersomes improved the therapeutic index of drug over free paclitaxel and Abraxane, as evaluated by intraperitoneal tumor burden and number of metastatic nodules. Our findings underline the potential utility of the polymersome platform for delivery of drugs and imaging agents to peritoneal carcinomatosis lesions. Mol Cancer Ther; 15(4); 670-9. ©2016 AACR.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Portadores de Fármacos , Paclitaxel/administración & dosificación , Polímeros , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Composición de Medicamentos , Liberación de Fármacos , Estabilidad de Medicamentos , Humanos , Inyecciones Intraperitoneales , Ratones , Paclitaxel/química , Paclitaxel/farmacocinética , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Peritoneales/patología , Polímeros/química , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nanoscale ; 8(17): 9096-101, 2016 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-26646247

RESUMEN

Affinity targeting is used to deliver nanoparticles to cells and tissues. For efficient targeting, it is critical to consider the expression and accessibility of the relevant receptors in the target cells. Here, we describe isotopically barcoded silver nanoparticles (AgNPs) as a tool for auditing affinity ligand receptors in cells. Tumor penetrating peptide RPARPAR (receptor: NRP-1) and tumor homing peptide GKRK (receptor: p32) were used as affinity ligands on the AgNPs. The binding and uptake of the peptide-functionalized AgNPs by cultured PPC-1 prostate cancer and M21 melanoma cells was dependent on the cell surface expression of the cognate peptide receptors. Barcoded peptide-functionalized AgNPs were synthesized from silver and palladium isotopes. The cells were incubated with a cocktail of the barcoded nanoparticles [RPARPAR (R), GKRK (K), and control], and cellular binding and internalization of each type of nanoparticle was assessed by inductively coupled plasma mass spectrometry. The results of isotopic analysis were in agreement with data obtained using optical methods. Using ratiometric measurements, we were able to classify the PPC-1 cell line as mainly NRP-1-positive, with 75 ± 5% R-AgNP uptake, and the M21 cell line as only p32-positive, with 89 ± 9% K-AgNP uptake. The isotopically barcoded multiplexed AgNPs are useful as an in vitro ratiometric phenotyping tool and have potential uses in functional evaluation of the expression of accessible homing peptide receptors in vivo.


Asunto(s)
Técnicas Citológicas , Nanopartículas del Metal , Plata , Línea Celular Tumoral , Humanos , Isótopos , Masculino , Péptidos , Fenotipo
17.
Sci Adv ; 1(10): e1500821, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26601141

RESUMEN

Cell-penetrating peptides (CPPs) have been widely used to deliver nanomaterials and other types of macromolecules into mammalian cells for therapeutic and diagnostic use. Cationic CPPs that bind to heparan sulfate (HS) proteoglycans on the cell surface induce potent endocytosis; however, the role of other surface receptors in this process is unclear. We describe the convergence of an HS-dependent pathway with the C-end rule (CendR) mechanism that enables peptide ligation with neuropilin-1 (NRP1), a cell surface receptor known to be involved in angiogenesis and vascular permeability. NRP1 binds peptides carrying a positive residue at the carboxyl terminus, a feature that is compatible with cationic CPPs, either intact or after proteolytic processing. We used CPP and CendR peptides, as well as HS- and NRP1-binding motifs from semaphorins, to explore the commonalities and differences of the HS and NRP1 pathways. We show that the CendR-NRP1 interaction determines the ability of CPPs to induce vascular permeability. We also show at the ultrastructural level, using a novel cell entry synchronization method, that both the HS and NRP1 pathways can initiate a macropinocytosis-like process and visualize these CPP-cargo complexes going through various endosomal compartments. Our results provide new insights into how CPPs exploit multiple surface receptor pathways for intracellular delivery.

18.
Nano Lett ; 15(10): 6745-50, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26317146

RESUMEN

Surface enhanced Raman spectroscopy (SERS) is a powerful analytical technique that has been proposed as a substitute for fluorescence for biological imaging and detection but is not yet commercially utilized. The reason lies primarily in the lower intensity and poor reproducibility of most metal nanoparticle-based tags as compared to their fluorescence-based counterparts. Here, using a technique that scrupulously preserves the same number of dye molecules in both the SERS and fluorescence measurements, we show that SERS-based biotags (SBTs) with highly reproducible optical properties can be nanoengineered such that their brightness is at least equal to that of fluorescence-based tags.

19.
J Control Release ; 212: 59-69, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26071630

RESUMEN

Peritoneal carcinomatosis is a major source of morbidity and mortality in patients with advanced abdominal neoplasms. Intraperitoneal chemotherapy (IPC) is an area of intense interest given its efficacy in ovarian cancer. However, IPC suffers from poor drug penetration into peritoneal tumors. As such, extensive cytoreductive surgery is required prior to IPC. Here, we explore the utility of iRGD, a tumor-penetrating peptide, for improved tumor-specific penetration of intraperitoneal compounds and enhanced IPC in mice. Intraperitoneally administered iRGD significantly enhanced penetration of an attached fluorescein into disseminated peritoneal tumor nodules. The penetration was tumor-specific, circulation-independent, and mediated by the neuropilin-binding RXXK tissue-penetration peptide motif of iRGD. Q-iRGD, which fluoresces upon cleavage, including the one that leads to RXXK activation, specifically labeled peritoneal metastases displaying different growth patterns in mice. Importantly, iRGD enhanced intratumoral entry of intraperitoneally co-injected dextran to approximately 300% and doxorubicin to 250%. Intraperitoneal iRGD/doxorubicin combination therapy inhibited the growth of bulky peritoneal tumors and reduced systemic drug toxicity. iRGD delivered attached fluorescein and co-applied nanoparticles deep into fresh human peritoneal metastasis explants. These results indicate that intraperitoneal iRGD co-administration serves as a simple and effective strategy to facilitate tumor detection and improve the therapeutic index of IPC for peritoneal carcinomatosis.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma/tratamiento farmacológico , Péptidos de Penetración Celular/administración & dosificación , Doxorrubicina/administración & dosificación , Oligopéptidos/administración & dosificación , Neoplasias Peritoneales/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Péptidos de Penetración Celular/uso terapéutico , Doxorrubicina/uso terapéutico , Humanos , Ratones Desnudos , Oligopéptidos/uso terapéutico , Flujo Sanguíneo Regional
20.
Biomaterials ; 63: 70-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26086448

RESUMEN

We describe a near infrared (NIR) light-activated gene silencing method in undifferentiated human embryonic stem cell (hESC) using a plasmonic hollow gold nanoshell (HGN) as the siRNA carrier. Our modular biotin-streptavidin coupling strategy enables positively charged TAT-peptide to coat oligonucleotides-saturated nanoparticles as a stable colloid formation. TAT-peptide coated nanoparticles with dense siRNA loading show efficient penetration into a wide variety of hESC cell lines. The siRNA is freed from the nanoparticles and delivered to the cytosol by femtosecond pulses of NIR light with potentially exquisite spatial and temporal control. The effectiveness of this approach is shown by targeting GFP and Oct4 genes in undifferentiated hESC (H9). The accelerated expression of differentiation markers for all three germ layers resulting from Oct4 knockdown confirms that this method has no detectable adverse effects that limit the range of differentiation. This biocompatible and NIR laser-activated patterning method makes possible single cell resolution of siRNA delivery for diverse studies in stem cell biology, tissue engineering and regenerative medicine.


Asunto(s)
Oro/química , Células Madre Embrionarias Humanas/metabolismo , Nanocápsulas/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Biotina/química , Línea Celular , Productos del Gen tat/química , Proteínas Fluorescentes Verdes/genética , Humanos , Luz , Factor 3 de Transcripción de Unión a Octámeros/genética , Fragmentos de Péptidos/química , ARN Interferente Pequeño/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...