Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nano Lett ; 22(5): 2103-2111, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35166110

RESUMEN

Nanoparticle modification with poly(ethylene glycol) (PEG) is a widely used surface engineering strategy in nanomedicine. However, since the artificial PEG polymer may adversely impact nanomedicine safety and efficacy, alternative surface modifications are needed. Here, we explored the "self" polysaccharide heparosan (HEP) to prepare colloidally stable HEP-coated nanoparticles, including gold and silver nanoparticles and liposomes. We found that the HEP-coating reduced the nanoparticle protein corona formation as efficiently as PEG coatings upon serum incubation. Liquid chromatography-mass spectrometry revealed the protein corona profiles. Heparosan-coated nanoparticles exhibited up to 230-fold higher uptake in certain innate immune cells, but not in other tested cell types, than PEGylated nanoparticles. No noticeable cytotoxicity was observed. Serum proteins did not mediate the high cell uptake of HEP-coated nanoparticles. Our work suggests that HEP polymers may be an effective surface modification technology for nanomedicines to safely and efficiently target certain innate immune cells.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Corona de Proteínas , Adsorción , Proteínas Sanguíneas , Disacáridos , Nanopartículas/química , Polietilenglicoles/química , Polímeros , Polisacáridos , Plata
2.
EMBO Rep ; 21(10): e48483, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32851774

RESUMEN

MICU1 is a mitochondrial inner membrane protein that inhibits mitochondrial calcium entry; elevated MICU1 expression is characteristic of many cancers, including ovarian cancer. MICU1 induces both glycolysis and chemoresistance and is associated with poor clinical outcomes. However, there are currently no available interventions to normalize aberrant MICU1 expression. Here, we demonstrate that microRNA-195-5p (miR-195) directly targets the 3' UTR of the MICU1 mRNA and represses MICU1 expression. Additionally, miR-195 is under-expressed in ovarian cancer cell lines, and restoring miR-195 expression reestablishes native MICU1 levels and the associated phenotypes. Stable expression of miR-195 in a human xenograft model of ovarian cancer significantly reduces tumor growth, increases tumor doubling times, and enhances overall survival. In conclusion, miR-195 controls MICU1 levels in ovarian cancer and could be exploited to normalize aberrant MICU1 expression, thus reversing both glycolysis and chemoresistance and consequently improving patient outcomes.


Asunto(s)
Proteínas de Transporte de Catión , MicroARNs , Neoplasias Ováricas , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glucólisis/genética , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Neoplasias Ováricas/genética
3.
Bioconjug Chem ; 30(9): 2247-2263, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31408324

RESUMEN

Compared to normal tissues, the tumor microenvironment (TME) has a number of aberrant characteristics including hypoxia, acidosis, and vascular abnormalities. Many researchers have sought to exploit these anomalous features of the TME to develop anticancer therapies, and several nanoparticle-based cancer therapeutics have resulted. In this Review, we discuss the composition and pathophysiology of the TME, introduce nanoparticles (NPs) used in cancer therapy, and address the interaction between the TME and NPs. Finally, we outline both the potential problems that affect TME-based nanotherapy and potential strategies to overcome these challenges.


Asunto(s)
Nanopartículas , Microambiente Tumoral/efectos de los fármacos , Animales , Humanos , Inmunomodulación/efectos de los fármacos , Microambiente Tumoral/inmunología
4.
Bioconjug Chem ; 30(6): 1724-1733, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31067032

RESUMEN

It is currently recognized that perpetual cross talk among key players in tumor microenvironment such as cancer cells (CCs), cancer associated fibroblasts (CAFs), and endothelial cells (ECs) plays a critical role in tumor progression, metastasis, and therapy resistance. Disruption of the cross talk may be useful to improve the outcome of therapeutics for which limited options are available. In the current study we investigate the use of gold nanoparticles (AuNPs) as a therapeutic tool to disrupt the multicellular cross talk within the TME cells with an emphasis on inhibiting angiogenesis. We demonstrate here that AuNPs disrupt signal transduction from TME cells (CCs, CAFs, and ECs) to ECs and inhibit angiogenic phenotypes in vitro. We show that conditioned media (CM) from ovarian CCs, CAFs, or ECs themselves induce tube formation and migration of ECs in vitro. Migration of ECs is also induced when ECs are cocultured with CCs, CAFs, or ECs. In contrast, CM from the cells treated with AuNPs or cocultured cells pretreated with AuNPs demonstrate diminished effects on ECs tube formation and migration. Mechanistically, AuNPs deplete ∼95% VEGF165 from VEGF single-protein solution and remove up to ∼45% of VEGF165 from CM, which is reflected on reduced activation of VEGF-Receptor 2 (VEGFR2) as compared to control CM. These results demonstrate that AuNPs inhibit angiogenesis via blockade of VEGF-VEGFR2 signaling from TME cells to endothelial cells.


Asunto(s)
Oro/uso terapéutico , Nanopartículas del Metal/uso terapéutico , Neovascularización Patológica/terapia , Neoplasias Ováricas/terapia , Microambiente Tumoral , Movimiento Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Transducción de Señal , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Bioconjug Chem ; 29(6): 1793-1808, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29742344

RESUMEN

Nanoparticles, the building blocks of nanotechnology, have been widely utilized in various biomedical applications, such as detection, diagnosis, imaging, and therapy. However, another emerging, albeit under-represented, area is the employment of nanoparticles as tools to understand cellular processes (e.g., oxidative stress-induced signaling cascades). Such investigations have enormous potential to characterize a disease from a different perspective and unravel some new features that otherwise would have remained a mystery. In this review, we summarize the intrinsic biological properties of unmodified as well surface modified nanoparticles and discuss how such properties could be utilized to interrogate biological processes and provide a perspective for future evolution of this field.


Asunto(s)
Nanomedicina/métodos , Nanopartículas/metabolismo , Nanotecnología/métodos , Animales , Humanos , Nanopartículas/química , Nanopartículas/uso terapéutico , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
6.
Mol Ther ; 21(3): 533-41, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23295953

RESUMEN

Because the functional apoptosis-initiating protein, cytochrome C (CytC) is rapidly cleared from the circulation (t1/2 (half-life): 4 minutes), it cannot be used for in vivo therapy. We report herein on a hitherto unreported strategy for delivering exogenous CytC as a potential and safe antiobesity drug for preventing diet-induced obesity, the most common type of obesity in humans. The functional activity of CytC encapsulated in prohibitin (a white fat vessel-specific receptor)-targeted nanoparticles (PTNP) was evaluated quantitatively, as evidenced by the observations that CytC-loaded PTNP causes apoptosis in primary adipose endothelial cells in a dose-dependent manner, whereas CytC alone did not. The delivery of a single dose of CytC through PTNP into the circulation disrupted the vascular structure by the targeted apoptosis of adipose endothelial cells in vivo. Intravenous treatment of CytC-loaded PTNP resulted in a substantial reduction in obesity in high-fat diet (HFD) fed wild-type (wt) mice, as evidenced by the dose-dependent prevention of the percentage of increase in body weight and decrease in serum leptin levels. In addition, no detectable hepatotoxicity was found to be associated with this prevention. Thus, the finding highlights the promising potential of CytC for use as an antiobesity drug, when delivered through a nanosystem.


Asunto(s)
Fármacos Antiobesidad/farmacología , Citocromos c/farmacología , Células Endoteliales/efectos de los fármacos , Nanopartículas/química , Obesidad/terapia , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Animales , Peso Corporal , Colesterol/sangre , Dieta Alta en Grasa , Sistemas de Liberación de Medicamentos , Células Endoteliales/citología , Proteínas Inmovilizadas/química , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Prohibitinas , Proteínas Represoras/química , Triglicéridos/sangre
7.
Adv Sci (Weinh) ; 9(31): e2200491, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36104215

RESUMEN

By exploiting the self-therapeutic properties of gold nanoparticles (GNPs) a molecular axis that promotes the growth of high-grade serous ovarian cancer (HGSOC), one of the deadliest gynecologic malignancies with poorly understood underlying molecular mechanisms, has been identified. The biodistribution and toxicity of GNPs administered by intravenous or intraperitoneal injection, both as a single dose or by repeated dosing over two weeks are first assessed; no biochemical or histological toxicity to vital organs is found. Using an orthotopic patient-derived xenograft (PDX) model of HGSOC, the authors then show that GNP treatment robustly inhibits tumor growth. Investigating the molecular mechanisms underlying the GNP efficacy reveals that GNPs downregulate insulin growth factor binding protein 2 (IGFBP2) by disrupting its autoregulation via the IGFBP2/mTOR/PTEN axis. This mechanism is validated by treating a cell line-based human xenograft tumor with GNPs and an mTOR dual-kinase inhibitor (PI-103), either individually or in combination with GNPs; GNP and PI-103 combination therapy inhibit ovarian tumor growth similarly to GNPs alone. This report illustrates how the self-therapeutic properties of GNPs can be exploited as a discovery tool to identify a critical signaling axis responsible for poor prognosis in ovarian cancer and provides an opportunity to interrogate the axis to improve patient outcomes.


Asunto(s)
Nanopartículas del Metal , Neoplasias Ováricas , Femenino , Humanos , Oro/química , Insulina , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Fosfohidrolasa PTEN , Distribución Tisular , Serina-Treonina Quinasas TOR , Animales
8.
ACS Nano ; 15(2): 2099-2142, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33497197

RESUMEN

Inorganic/organic hybrid nanosystems have been increasingly developed for their versatility and efficacy at overcoming obstacles not readily surmounted by nonhybridized counterparts. Currently, hybrid nanosystems are implemented for gene therapy, drug delivery, and phototherapy in addition to tissue regeneration, vaccines, antibacterials, biomolecule detection, imaging probes, and theranostics. Though diverse, these nanosystems can be classified according to foundational inorganic/organic components, accessory moieties, and architecture of hybridization. Within this Review, we begin by providing a historical context for the development of biomedical hybrid nanosystems before describing the properties, synthesis, and characterization of their component building blocks. Afterward, we introduce the architectures of hybridization and highlight recent biomedical nanosystem developments by area of application, emphasizing hybrids of distinctive utility and innovation. Finally, we draw attention to ongoing clinical trials before recapping our discussion of hybrid nanosystems and providing a perspective on the future of the field.


Asunto(s)
Sistemas de Liberación de Medicamentos , Fototerapia
9.
Cancer Nanotechnol ; 12(1): 1, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33456622

RESUMEN

BACKGROUND: Ovarian cancer is one of the deadliest gynecological malignancies. While the overall survival of ovarian cancer patients has slightly improved in recent years in the developed world, it remains clinically challenging due to its frequent late diagnosis and the lack of reliable diagnostic and/or prognostic markers. The aim of this study was to identify potential new molecular target proteins (NMTPs) responsible for the poor outcomes. When nanoparticles (NP) are exposed to biological fluids, a protein coat, termed the protein corona (PC), forms around the NP, and the PC represents a tool to identify NMTPs. This study investigates the influence of pre-processing conditions, such as lysis conditions and serum/plasma treatment, on the PC composition and the resulting identification of NMTPs. RESULTS: Using gel electrophoresis, pre-processing conditions, including cell-lysis techniques and enrichment of low-abundance proteins (LAPs) by immunocentrifugation of serum/plasma, were shown to alter the relative amounts and compositions of proteins. PCs formed when 20 nm gold-NPs (GNPs) were incubated with lysate proteins from either RIPA- or urea lysis. Proteomic analysis of these PCs showed 2-22-fold enrichment of NMTPs in PCs from urea lysates as compared to RIPA lysates. Enriched NMTPs were then classified as cellular components, biological and molecular functions-associated proteins. The impact of enriched LAPs (eLAPs) on both PC composition and NMTP identification was shown by comparative proteomic analysis of original plasma, eLAPs, and PCs derived from eLAPs; eLAPs-PCs enhanced the abundance of NMTPs approximately 13%. Several NMTPs, including gasdermin-B, dermcidin, and kallistatin, were identified by this method demonstrating the potential use of this PC approach for molecular target discovery. CONCLUSION: The current study showed that the pre-processing conditions modulate PC composition and can be used to enhance identification of NMTPs.

10.
Bioact Mater ; 6(2): 326-332, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32954051

RESUMEN

Cancer-associated fibroblasts (CAFs) are a major constituent of the tumor microenvironment (TME) and an important contributor to cancer progression and therapeutic resistance. Regulation of CAF activation is a promising strategy to influence cancer outcomes. Here, we report that ovarian cancer cells (OCs) and TME cells promote the activation of ovarian CAFs, whereas gold nanoparticles (GNPs) of 20 nm in diameter inhibit the activation, as demonstrated by the changes in cell morphology, migration, and molecular markers. GNPs exert the effect by altering the levels of multiple fibroblast activation or inactivation proteins, such as TGF-ß1, PDGF, uPA and TSP1, secreted by OCs and TME cells. Thus, GNPs represent a potential tool to help understand multicellular communications existing in the TME as well as devise strategies to disrupt the communication.

11.
J Control Release ; 322: 122-136, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32165239

RESUMEN

When nanoparticles are introduced into biological systems, host proteins tend to associate on the particle surface to form a protein layer termed the "protein corona" (PC). Identifying the proteins that constitute the PC can yield useful information about nanoparticle processing, bio-distribution, toxicity and clearance. Similarly, characterizing and identifying proteins within the PC from patient samples provides opportunities to probe disease proteomes and identify molecules that influence the disease process. Thus, nanoparticles represent unique probing tools for discovery of molecular targets for diseases. Here, we report a first review on target identification using nanoparticles in biological samples based on analysing physico chemical interactions. We also summarize the evolution of the PC surrounding various nano-systems, comment on PC signature, address PC complexity in fluids, and outline challenges associated with analysing the PC. In addition, the influence on PC formation of various nanoparticle parameters is summarized; nanoparticle characteristics considered include size, charge, temperature, and surface modifications for both organic and inorganic nanomaterials. We also discuss the advantages of nanotechnology, over other more invasive and laborious methods, for identifying potential diagnostic and therapeutic targets.


Asunto(s)
Nanopartículas , Corona de Proteínas , Humanos , Cinética , Nanotecnología , Proteínas
12.
Int J Nanomedicine ; 15: 991-1003, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32103952

RESUMEN

INTRODUCTION AND OBJECTIVE: Pancreatic cancer (PC) is characterized by a robust desmoplastic environment, which limits the uptake of the standard first-line chemotherapeutic drug gemcitabine. Enhancing gemcitabine delivery to the complex tumor microenvironment (TME) is a major clinical challenge. Molecular crosstalk between pancreatic cancer cells (PCCs) and pancreatic stellate cells (PSCs) plays a critical role in desmoplastic reaction in PCs. Herein, we report the development of a targeted drug delivery system to inhibit the proliferation of PCCs and PSCs in vitro. Using gold nanoparticles as the delivery vehicle, the anti-EGFR antibody cetuximab (C225/C) as a targeting agent, gemcitabine as drug and polyethylene glycol (PEG) as a stealth molecule, we created a series of targeted drug delivery systems. METHODS: Fabricated nanoconjugates were characterized by various physicochemical techniques such as UV-Visible spectroscopy, transmission electron microscopy, HPLC and instrumental neutron activation analysis (INAA). RESULTS AND CONCLUSION: Targeted gemcitabine delivery systems containing mPEG-SH having molecular weights of 550 Da or 1000 Da demonstrated superior efficacy in reducing the viability of both PCCs and PSCs as compared to their non-targeted counterparts. EGFR-targeted pathway was further validated by pre-treating cells with C225 followed by determining cellular viability. Taken together, in our current study we have developed a PEGylated targeted nanoconjugate ACG44P1000 that showed enhanced selectivity towards pancreatic cancer cells and pancreatic stellate cells, among others, for gemcitabine delivery. We will investigate the ability of these optimized conjugates to inhibit desmoplasia and tumor growth in vivo in our future studies.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Nanoconjugados/química , Neoplasias Pancreáticas/tratamiento farmacológico , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cetuximab/administración & dosificación , Cetuximab/química , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Portadores de Fármacos/química , Oro/química , Humanos , Nanopartículas del Metal/administración & dosificación , Nanopartículas del Metal/química , Nanopartículas del Metal/toxicidad , Nanoconjugados/administración & dosificación , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Polietilenglicoles/química , Microambiente Tumoral/efectos de los fármacos , Gemcitabina
13.
Sci Adv ; 6(27)2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32937467

RESUMEN

The stringent expression of the hypoxia inducible factor-1α (HIF-1α) is critical to a variety of pathophysiological conditions. We reveal that, in normoxia, enzymatic action of cystathionine ß-synthase (CBS) produces H2S, which persulfidates prolyl hydroxylase 2 (PHD2) at residues Cys21 and Cys33 (zinc finger motif), augmenting prolyl hydroxylase activity. Depleting endogenous H2S either by hypoxia or by inhibiting CBS via chemical or genetic means reduces persulfidation of PHD2 and inhibits activity, preventing hydroxylation of HIF-1α, resulting in stabilization. Our in vitro findings are further supported by the depletion of CBS in the zebrafish model that exhibits axis defects and abnormal intersegmental vessels. Exogenous H2S supplementation rescues both in vitro and in vivo phenotypes. We have identified the persulfidated residues and defined their functional significance in regulating the activity of PHD2 via point mutations. Thus, the CBS/H2S/PHD2 axis may provide therapeutic opportunities for pathologies associated with HIF-1α dysregulation in chronic diseases.


Asunto(s)
Cistationina betasintasa , Subunidad alfa del Factor 1 Inducible por Hipoxia , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Animales , Cistationina betasintasa/metabolismo , Sulfuro de Hidrógeno , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Pez Cebra/metabolismo
14.
Sci Adv ; 6(30): eaba5379, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32743073

RESUMEN

Gene silencing using small-interfering RNA (siRNA) is a viable therapeutic approach; however, the lack of effective delivery systems limits its clinical translation. Herein, we doped conventional siRNA-liposomal formulations with gold nanoparticles to create "auroliposomes," which significantly enhanced gene silencing. We targeted MICU1, a novel glycolytic switch in ovarian cancer, and delivered MICU1-siRNA using three delivery systems-commercial transfection agents, conventional liposomes, and auroliposomes. Low-dose siRNA via transfection or conventional liposomes was ineffective for MICU1 silencing; however, in auroliposomes, the same dose gave >85% gene silencing. Efficacy was evident from both in vitro growth assays of ovarian cancer cells and in vivo tumor growth in human ovarian cell line-and patient-derived xenograft models. Incorporation of gold nanoparticles shifted intracellular uptake pathways such that liposomes avoided degradation within lysosomes. Auroliposomes were nontoxic to vital organs. Therefore, auroliposomes represent a novel siRNA delivery system with superior efficacy for multiple therapeutic applications.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Neoplasias Ováricas , Línea Celular Tumoral , Oro , Humanos , Liposomas , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
15.
ACS Appl Mater Interfaces ; 11(29): 26060-26068, 2019 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-31117437

RESUMEN

Activated cancer-associated fibroblasts (CAFs) play a major role in the poor outcome in many diseases including pancreatic cancer. Normally quiescent with high lipid content and low proliferative capacity, CAFs receiving cues from cancer cells in the tumor microenvironment become activated and transformed into a lipid-deprived and highly proliferative myofibroblast type phenotype. Therefore, reversal of activated fibroblasts to the quiescence state is an important area of investigation that may help the therapeutic management of a number of diseases including pancreatic cancer. Here, we describe a unique biological function of gold nanoparticles (GNPs) and demonstrate that GNPs may be used to transform activated CAFs to quiescence and provide insights into the underlying molecular mechanisms. Using immortalized and primary patient derived CAFs, we demonstrate that GNPs enhanced lipid content in the cells by inducing expression of lipogenesis genes such as FASN, SREBP2, and FABP3. Using pharmacological inhibitors of lipolysis, lipophagy, and fatty acid oxidation, we further demonstrate that CAFs utilized a GNP-induced endogenously synthesized lipid to maintain the quiescent phenotype. Consequently, treatment with GNP sensitizes CAF to FASN inhibitor or FASN siRNA. Hence, GNPs may be used as a tool to probe mechanisms of quiescence in CAFs and help device strategies to target the stromal compartment exploiting the mechanisms of lipid utilization.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Oro , Nanopartículas del Metal/química , Proteínas de Neoplasias/biosíntesis , Neoplasias Pancreáticas/metabolismo , Microambiente Tumoral/efectos de los fármacos , Fibroblastos Asociados al Cáncer/patología , Oro/química , Oro/farmacología , Humanos , Neoplasias Pancreáticas/patología
16.
Nat Commun ; 9(1): 3695, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30209303

RESUMEN

Protein/subunit vaccines often require external adjuvants to induce protective immunity. Due to the safety concern of chemical adjuvants, physical adjuvants were recently explored to boost vaccination. Physical adjuvants use physical energies rather than chemicals to stimulate tissue stress and endogenous danger signal release to boost vaccination. Here we present the safety and potency of non-invasive radiofrequency treatment to boost intradermal vaccination in murine models. We show non-invasive radiofrequency can increase protein antigen-induced humoral and cellular immune responses with adjuvant effects comparable to widely used chemical adjuvants. Radiofrequency adjuvant can also safely boost pandemic 2009 H1N1 influenza vaccination with adjuvant effects comparable to MF59-like AddaVax adjuvant. We find radiofrequency adjuvant induces heat shock protein 70 (HSP70) release and activates MyD88 to mediate the adjuvant effects. Physical radiofrequency can potentially be a safe and potent adjuvant to augment protein/subunit vaccine-induced humoral and cellular immune responses.


Asunto(s)
Inmunidad Celular/fisiología , Inmunidad Humoral/fisiología , Vacunas contra la Influenza/inmunología , Adyuvantes Inmunológicos , Animales , Western Blotting , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
17.
J Control Release ; 249: 94-102, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28132934

RESUMEN

Transdermal delivery of hydrophilic drugs is challenging. This study presents a novel sustained epidermal powder delivery technology (sEPD) for safe, efficient, and sustained delivery of hydrophilic drugs across the skin. sEPD is based on coating powder drugs into high-aspect-ratio, micro-coating channels (MCCs) followed by topical application of powder drug-coated array patches onto ablative fractional laser-generated skin MCs to deliver drugs into the skin. We found sEPD could efficiently deliver chemical drugs without excipients and biologics drugs in the presence of sugar excipients into the skin with a duration of ~12h. Interestingly the sEPD significantly improved zidovudine bioavailability by ~100% as compared to oral gavage delivery. sEPD of insulin was found to maintain blood glucose levels in normal range for at least 6h in chemical-induced diabetes mice, while subcutaneous injection failed to maintain blood glucose levels in normal range. sEPD of anti-programmed death-1 antibody showed more potent anti-tumor efficacy than intraperitoneal injection in B16F10 melanoma models. Tiny skin MCs and 'bulk' drug powder inside relatively deep MCCs are crucial to induce the sustained drug release. The improved bioavailability and functionality warrants further development of the novel sEPD for clinical use.


Asunto(s)
Sistemas de Liberación de Medicamentos/instrumentación , Epidermis/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Absorción Cutánea , Administración Cutánea , Animales , Fármacos Anti-VIH/administración & dosificación , Anticuerpos/administración & dosificación , Diseño de Equipo , Colorantes Fluorescentes/administración & dosificación , Interacciones Hidrofóbicas e Hidrofílicas , Inyecciones Subcutáneas , Insulina/administración & dosificación , Rayos Láser , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microinyecciones , Polvos/administración & dosificación , Rodaminas/administración & dosificación , Zidovudina/administración & dosificación
19.
ACS Nano ; 10(12): 10636-10651, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27758098

RESUMEN

Altered tumor microenvironment (TME) arising from a bidirectional crosstalk between the pancreatic cancer cells (PCCs) and the pancreatic stellate cells (PSCs) is implicated in the dismal prognosis in pancreatic ductal adenocarcinoma (PDAC), yet effective strategies to disrupt the crosstalk is lacking. Here, we demonstrate that gold nanoparticles (AuNPs) inhibit proliferation and migration of both PCCs and PSCs by disrupting the bidirectional communication via alteration of the cell secretome. Analyzing the key proteins identified from a functional network of AuNP-altered secretome in PCCs and PSCs, we demonstrate that AuNPs impair secretions of major hub node proteins in both cell types and transform activated PSCs toward a lipid-rich quiescent phenotype. By reducing activation of PSCs, AuNPs inhibit matrix deposition, enhance angiogenesis, and inhibit tumor growth in an orthotopic co-implantation model in vivo. Auto- and heteroregulations of secretory growth factors/cytokines are disrupted by AuNPs resulting in reprogramming of the TME. By utilizing a kinase dead mutant of IRE1-α, we demonstrate that AuNPs alter the cellular secretome through the ER-stress-regulated IRE1-dependent decay pathway (RIDD) and identify endostatin and matrix metalloproteinase 9 as putative RIDD targets. Thus, AuNPs could potentially be utilized as a tool to effectively interrogate bidirectional communications in the tumor microenvironment, reprogram it, and inhibit tumor growth by its therapeutic function.


Asunto(s)
Carcinoma Ductal Pancreático/terapia , Oro , Nanopartículas del Metal , Neoplasias Pancreáticas/terapia , Microambiente Tumoral , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas
20.
J Control Release ; 220(Pt A): 61-70, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26450668

RESUMEN

Current approaches to the diagnosis and therapy of atherosclerosis cannot target lesion-determinant cells in the artery wall. Intimal macrophage infiltration promotes atherosclerotic lesion development by facilitating the accumulation of oxidized low-density lipoproteins (oxLDL) and increasing inflammatory responses. The presence of these cells is positively associated with lesion progression, severity and destabilization. Hence, they are an important diagnostic and therapeutic target. The objective of this study was to noninvasively assess the distribution and accumulation of intimal macrophages using CD36-targeted nanovesicles. Soy phosphatidylcholine was used to synthesize liposome-like nanovesicles. 1-(Palmitoyl)-2-(5-keto-6-octene-dioyl) phosphatidylcholine was incorporated on their surface to target the CD36 receptor. All in vitro data demonstrate that these targeted nanovesicles had a high binding affinity for the oxLDL binding site of the CD36 receptor and participated in CD36-mediated recognition and uptake of nanovesicles by macrophages. Intravenous administration into LDL receptor null mice of targeted compared to non-targeted nanovesicles resulted in higher uptake in aortic lesions. The nanovesicles co-localized with macrophages and their CD36 receptors in aortic lesions. This molecular target approach may facilitate the in vivo noninvasive imaging of atherosclerotic lesions in terms of intimal macrophage accumulation and distribution and disclose lesion features related to inflammation and possibly vulnerability thereby facilitate early lesion detection and targeted delivery of therapeutic compounds to intimal macrophages.


Asunto(s)
Aterosclerosis/diagnóstico , Antígenos CD36/metabolismo , Macrófagos/metabolismo , Nanopartículas/química , Animales , Células Cultivadas , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de LDL/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda