Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38775127

RESUMEN

Primary, glioblastoma, and secondary brain tumors, from metastases outside the brain, are among the most aggressive and therapeutically resistant cancers. A physiological barrier protecting the brain, the blood-brain barrier (BBB), functions as a deterrent to effective therapies. To enhance cancer therapy, we developed a cancer terminator virus (CTV), a unique tropism-modified adenovirus consisting of serotype 3 fiber knob on an otherwise Ad5 capsid that replicates in a cancer-selective manner and simultaneously produces a potent therapeutic cytokine, melanoma differentiation-associated gene-7/interleukin-24 (MDA-7/IL-24). A limitation of the CTV and most other viruses, including adenoviruses, is an inability to deliver systemically to treat brain tumors because of the BBB, nonspecific virus trapping, and immune clearance. These obstacles to effective viral therapy of brain cancer have now been overcome using focused ultrasound with a dual microbubble treatment, the focused ultrasound-double microbubble (FUS-DMB) approach. Proof-of-principle is now provided indicating that the BBB can be safely and transiently opened, and the CTV can then be administered in a second set of complement-treated microbubbles and released in the brain using focused ultrasound. Moreover, the FUS-DMB can be used to deliver the CTV multiple times in animals with glioblastoma  growing in their brain thereby resulting in a further enhancement in survival. This strategy permits efficient therapy of primary and secondary brain tumors enhancing animal survival without promoting harmful toxic or behavioral side effects. Additionally, when combined with a standard of care therapy, Temozolomide, a further increase in survival is achieved. The FUS-DMB approach with the CTV highlights a noninvasive strategy to treat brain cancers without surgery. This innovative delivery scheme combined with the therapeutic efficacy of the CTV provides a novel potential translational therapeutic approach for brain cancers.

2.
Immunity ; 42(4): 679-91, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902482

RESUMEN

Mutations in MECP2, encoding the epigenetic regulator methyl-CpG-binding protein 2, are the predominant cause of Rett syndrome, a disease characterized by both neurological symptoms and systemic abnormalities. Microglial dysfunction is thought to contribute to disease pathogenesis, and here we found microglia become activated and subsequently lost with disease progression in Mecp2-null mice. Mecp2 was found to be expressed in peripheral macrophage and monocyte populations, several of which also became depleted in Mecp2-null mice. RNA-seq revealed increased expression of glucocorticoid- and hypoxia-induced transcripts in Mecp2-deficient microglia and peritoneal macrophages. Furthermore, Mecp2 was found to regulate inflammatory gene transcription in response to TNF stimulation. Postnatal re-expression of Mecp2 using Cx3cr1(creER) increased the lifespan of otherwise Mecp2-null mice. These data suggest that Mecp2 regulates microglia and macrophage responsiveness to environmental stimuli to promote homeostasis. Dysfunction of tissue-resident macrophages might contribute to the systemic pathologies observed in Rett syndrome.


Asunto(s)
Islas de CpG/inmunología , Epigénesis Genética , Macrófagos Peritoneales/inmunología , Proteína 2 de Unión a Metil-CpG/inmunología , Microglía/inmunología , Síndrome de Rett/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Metilación de ADN , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Homeostasis/inmunología , Humanos , Integrasas/genética , Integrasas/inmunología , Longevidad/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/patología , Masculino , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/patología , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Síndrome de Rett/genética , Síndrome de Rett/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología
3.
Proc Natl Acad Sci U S A ; 117(11): 5644-5654, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32123081

RESUMEN

Treatment of many pathologies of the brain could be improved markedly by the development of noninvasive therapeutic approaches that elicit robust, endothelial cell-selective gene expression in specific brain regions that are targeted under MR image guidance. While focused ultrasound (FUS) in conjunction with gas-filled microbubbles (MBs) has emerged as a noninvasive modality for MR image-guided gene delivery to the brain, it has been used exclusively to transiently disrupt the blood-brain barrier (BBB), which may induce a sterile inflammation response. Here, we introduce an MR image-guided FUS method that elicits endothelial-selective transfection of the cerebral vasculature (i.e., "sonoselective" transfection), without opening the BBB. We first determined that activating circulating, cationic plasmid-bearing MBs with pulsed low-pressure (0.1 MPa) 1.1-MHz FUS facilitates sonoselective gene delivery to the endothelium without MRI-detectable disruption of the BBB. The degree of endothelial selectivity varied inversely with the FUS pressure, with higher pressures (i.e., 0.3-MPa and 0.4-MPa FUS) consistently inducing BBB opening and extravascular transfection. Bulk RNA sequencing analyses revealed that the sonoselective low-pressure regimen does not up-regulate inflammatory or immune responses. Single-cell RNA sequencing indicated that the transcriptome of sonoselectively transfected brain endothelium was unaffected by the treatment. The approach developed here permits targeted gene delivery to blood vessels and could be used to promote angiogenesis, release endothelial cell-secreted factors to stimulate nerve regrowth, or recruit neural stem cells.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Transfección/métodos , Ondas Ultrasónicas , Animales , Barrera Hematoencefálica/efectos de la radiación , Imagen por Resonancia Magnética/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Microburbujas , Transcriptoma
4.
Nature ; 539(7630): 570-574, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27820945

RESUMEN

Professional phagocytes (such as macrophages) and non-professional phagocytes (such as epithelial cells) clear billions of apoptotic cells and particles on a daily basis. Although professional and non-professional macrophages reside in proximity in most tissues, whether they communicate with each other during cell clearance, and how this might affect inflammation, is not known. Here we show that macrophages, through the release of a soluble growth factor and microvesicles, alter the type of particles engulfed by non-professional phagocytes and influence their inflammatory response. During phagocytosis of apoptotic cells or in response to inflammation-associated cytokines, macrophages released insulin-like growth factor 1 (IGF-1). The binding of IGF-1 to its receptor on non-professional phagocytes redirected their phagocytosis, such that uptake of larger apoptotic cells was reduced whereas engulfment of microvesicles was increased. IGF-1 did not alter engulfment by macrophages. Macrophages also released microvesicles, whose uptake by epithelial cells was enhanced by IGF-1 and led to decreased inflammatory responses by epithelial cells. Consistent with these observations, deletion of IGF-1 receptor in airway epithelial cells led to exacerbated lung inflammation after allergen exposure. These genetic and functional studies reveal that IGF-1- and microvesicle-dependent communication between macrophages and epithelial cells can critically influence the magnitude of tissue inflammation in vivo.


Asunto(s)
Células Epiteliales/citología , Macrófagos/citología , Fagocitos/citología , Fagocitosis , Neumonía , Alérgenos/inmunología , Animales , Apoptosis , Comunicación Celular , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Fibroblastos/citología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Fagocitos/inmunología , Fagocitos/metabolismo , Neumonía/inmunología , Neumonía/metabolismo , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/metabolismo , Sistema Respiratorio/citología , Somatomedinas/metabolismo
5.
Int J Mol Sci ; 23(7)2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-35409274

RESUMEN

Malignant mesothelioma (MM) is a lethal tumor originating in the mesothelium with high chemotherapeutic resistance. Cancer stem cells (CSCs) persist in tumors and are critical targets responsible for tumor resistance and recurrence. The identification and characterization of CSCs may help develop effective treatment for MM. The objective of this study was to evaluate the therapeutic effect of molecular targeted radiotherapy by 177Lu-labeled immunoliposomes (177Lu-ILs) on CSCs of mesothelioma. MM CSCs were sorted based on CD26/CD24 expression level and their functional significances were established by small interference RNA. CSC potential of MM was evaluated for drug resistance, cell invasion, and cell growth rate in vitro. CSC metabolism was evaluated with the uptake of 18F-FDG. Therapeutic effects of 177Lu-labeled immunoliposomes targeting CD26 and CD24 were evaluated in vitro through proliferation and apoptotic assays. CSCs sorted from H28 cells exhibited significant drug resistance and enhanced proliferative activity as well as increased metabolism indicated by higher 18F-FDG uptake. Treatment with 177Lu-ILs, compared with 177Lu-CL and ILs, showed enhanced therapeutic effects on inhibition of proliferation, up-regulation of apoptosis, and suppression of CD26 and CD24 expression. Thus, our results suggest that molecular radiotherapy targeting both CD26 and CD24 could be a promising approach for CSC-targeting therapy for MM.


Asunto(s)
Mesotelioma Maligno , Mesotelioma , Línea Celular Tumoral , Dipeptidil Peptidasa 4/metabolismo , Fluorodesoxiglucosa F18/metabolismo , Humanos , Liposomas/metabolismo , Mesotelioma/tratamiento farmacológico , Mesotelioma/genética , Mesotelioma/radioterapia , Células Madre Neoplásicas/metabolismo
6.
Recent Results Cancer Res ; 216: 319-336, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32594391

RESUMEN

Gas-filled particles (microbubbles) can be prepared and stabilized for intravascular use as contrast agents in ultrasound imaging. Microbubbles are used in clinics as blood pool contrast materials for the past two decades. Shell of these bubbles is made of biocompatible and biodegradable lipids, proteins, and/or polymers. Gas core is air, or, lately, a perfluorinated gas, poorly soluble in water and blood. Making them useful for molecular targeting and molecular imaging in oncology is accomplished by decorating the shell of these particles with targeting ligands, that will selectively bind to the specific markers of tumor vasculature. In this review we discuss the formulation strategy for microbubble preparation, the logic of bubble shell selection, coupling tools that are used for the attachment of targeting ligands, and examples of the application of gas-filled bubbles for molecular imaging in oncology.


Asunto(s)
Medios de Contraste/química , Microburbujas , Imagen Molecular , Neoplasias/diagnóstico por imagen , Ultrasonografía , Humanos
7.
Langmuir ; 35(31): 10034-10041, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30509068

RESUMEN

For preparation of ligand-decorated microbubbles for targeted ultrasound contrast imaging, it is important to maximize the amount of ligand associated with the bubble shell. We describe optimization of the use of a biocompatible cosurfactant in the microbubble formulation media to maximize the incorporation of targeting ligand-lipid conjugate into the microbubble shell, and thus reduce the fraction of ligand not associated with microbubbles, following amalgamation preparation. The influence of the concentration of a helper cosurfactant propylene glycol (PG) on the efficacy of microbubble preparation by amalgamation and on the degree of association of fluorescent PEG-lipid with the microbubble shell was tested. Three sets of targeted bubbles were then prepared: with VCAM-1-targeting peptide VHPKQHRGGSK(FITC)GC-PEG-DSPE, cyclic RGDfK-PEG-DSPE, selective for αVß3, and control cRADfK-PEG-DSPE, without such affinity. Microbubbles were prepared by 45 s amalgamation, with DSPC and PEG stearate as the main components of the shell, with 15% PG in aqueous saline. In vitro microbubble targeting was assessed with a parallel plate flow chamber with a recombinant receptor coated surface. In vivo targeting was assessed in MC-38 tumor-bearing mice (subcutaneous tumor in hind leg), 10 min after intravenous bolus of microbubble contrast agent (20 million particles per injection). Ultrasound imaging of the tumor and control nontarget muscle tissue in a contralateral leg was performed with a clinical scanner. Amalgamation technique with PG cosurfactant produced microbubbles at concentrations exceeding 2 × 109 particles/mL, and ∼50-60% or more of the added fluorescein-PEG-DSPE or VCAM-1-targeted fluorescent peptide was associated with microbubbles, about 2 times higher than that in the absence of PG. After intravenous injection, peptide-targeted bubbles selectively accumulated in the tumor vasculature, with negligible accumulation in nontumor contralateral leg muscle, or with control nontargeted microbubbles (assessed by contrast ultrasound imaging). For comparison, administration of RGD-decorated microbubbles prepared by traditional sonication, and purified from free peptide-PEG-lipid by repeated centrifugation, resulted in the same accumulation pattern as for translatable amalgamated microbubbles. Following amalgamation in the presence of PG, efficient transfer of ligand-PEG-lipid to microbubble shell was achieved and quantified. Purification of microbubbles from free peptide-PEG-lipid was not necessary, as proven by in vitro and in vivo targeting studies, so PG cosurfactant amalgamation technique generated peptide-targeted microbubbles are amenable for bedside preparation and clinical translation. The pathway to clinical translation is simplified by the fact that most of the materials used in this study either are on the United States Food and Drug Administration GRAS list or can be procured as pharmaceutical grade substances.


Asunto(s)
Adenocarcinoma/diagnóstico por imagen , Neoplasias del Colon/diagnóstico por imagen , Medios de Contraste/química , Microburbujas , Péptidos Cíclicos/química , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Ratones , Ultrasonografía/métodos
8.
Nature ; 497(7448): 263-7, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23615608

RESUMEN

Skeletal muscle arises from the fusion of precursor myoblasts into multinucleated myofibres. Although conserved transcription factors and signalling proteins involved in myogenesis have been identified, upstream regulators are less well understood. Here we report an unexpected discovery that the membrane protein BAI1, previously linked to recognition of apoptotic cells by phagocytes, promotes myoblast fusion. Endogenous BAI1 expression increased during myoblast fusion, and BAI1 overexpression enhanced myoblast fusion by means of signalling through ELMO/Dock180/Rac1 proteins. During myoblast fusion, a fraction of myoblasts within the population underwent apoptosis and exposed phosphatidylserine, an established ligand for BAI1 (ref. 3). Blocking apoptosis potently impaired myoblast fusion, and adding back apoptotic myoblasts restored fusion. Furthermore, primary human myoblasts could be induced to form myotubes by adding apoptotic myoblasts, even under normal growth conditions. Mechanistically, apoptotic cells did not directly fuse with the healthy myoblasts, rather the apoptotic cells induced a contact-dependent signalling with neighbours to promote fusion among the healthy myoblasts. In vivo, myofibres from Bai1(-/-) mice are smaller than those from wild-type littermates. Muscle regeneration after injury was also impaired in Bai1(-/-)mice, highlighting a role for BAI1 in mammalian myogenesis. Collectively, these data identify apoptotic cells as a new type of cue that induces signalling via the phosphatidylserine receptor BAI1 to promote fusion of healthy myoblasts, with important implications for muscle development and repair.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Apoptosis/fisiología , Fusión Celular , Músculo Esquelético/citología , Mioblastos/citología , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Proteínas Angiogénicas/deficiencia , Proteínas Angiogénicas/genética , Animales , Apoptosis/efectos de los fármacos , Comunicación Celular , Diferenciación Celular , Línea Celular , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Desarrollo de Músculos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Fosfatidilserinas/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética
9.
Am J Physiol Heart Circ Physiol ; 315(2): H402-H414, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29631369

RESUMEN

Recent smooth muscle cell (SMC) lineage-tracing studies have revealed that SMCs undergo remarkable changes in phenotype during development of atherosclerosis. Of major interest, we demonstrated that Kruppel-like factor 4 (KLF4) in SMCs is detrimental for overall lesion pathogenesis, in that SMC-specific conditional knockout of the KLF4 gene ( Klf4) resulted in smaller, more-stable lesions that exhibited marked reductions in the numbers of SMC-derived macrophage- and mesenchymal stem cell-like cells. However, since the clinical consequences of atherosclerosis typically occur well after our reproductive years, we sought to identify beneficial KLF4-dependent SMC functions that were likely to be evolutionarily conserved. We tested the hypothesis that KLF4-dependent SMC transitions play an important role in the tissue injury-repair process. Using SMC-specific lineage-tracing mice positive and negative for simultaneous SMC-specific conditional knockout of Klf4, we demonstrate that SMCs in the remodeling heart after ischemia-reperfusion injury (IRI) express KLF4 and transition to a KLF4-dependent macrophage-like state and a KLF4-independent myofibroblast-like state. Moreover, heart failure after IRI was exacerbated in SMC Klf4 knockout mice. Surprisingly, we observed a significant cardiac dilation in SMC Klf4 knockout mice before IRI as well as a reduction in peripheral resistance. KLF4 chromatin immunoprecipitation-sequencing analysis on mesenteric vascular beds identified potential baseline SMC KLF4 target genes in numerous pathways, including PDGF and FGF. Moreover, microvascular tissue beds in SMC Klf4 knockout mice had gaps in lineage-traced SMC coverage along the resistance arteries and exhibited increased permeability. Together, these results provide novel evidence that Klf4 has a critical maintenance role within microvascular SMCs: it is required for normal SMC function and coverage of resistance arteries. NEW & NOTEWORTHY We report novel evidence that the Kruppel-like factor 4 gene ( Klf4) has a critical maintenance role within microvascular smooth muscle cells (SMCs). SMC-specific Klf4 knockout at baseline resulted in a loss of lineage-traced SMC coverage of resistance arteries, dilation of resistance arteries, increased blood flow, and cardiac dilation.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Microvasos/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Factores de Crecimiento de Fibroblastos/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Macrófagos/metabolismo , Ratones , Microvasos/citología , Miofibroblastos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Regeneración
10.
Clin Sci (Lond) ; 132(23): 2519-2545, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30442812

RESUMEN

Congenital obstructive nephropathy is a major cause of chronic kidney disease (CKD) in children. The contribution of changes in the identity of renal cells to the pathology of obstructive nephropathy is poorly understood. Using a partial unilateral ureteral obstruction (pUUO) model in genetically modified neonatal mice, we traced the fate of cells derived from the renal stroma, cap mesenchyme, ureteric bud (UB) epithelium, and podocytes using Foxd1Cre, Six2Cre, HoxB7Cre, and Podocyte.Cre mice respectively, crossed with double fluorescent reporter (membrane-targetted tandem dimer Tomato (mT)/membrane-targetted GFP (mG)) mice. Persistent obstruction leads to a significant loss of tubular epithelium, rarefaction of the renal vasculature, and decreased renal blood flow (RBF). In addition, Forkhead Box D1 (Foxd1)-derived pericytes significantly expanded in the interstitial space, acquiring a myofibroblast phenotype. Degeneration of Sine Oculis Homeobox Homolog 2 (Six2) and HoxB7-derived cells resulted in significant loss of glomeruli, nephron tubules, and collecting ducts. Surgical release of obstruction resulted in striking regeneration of tubules, arterioles, interstitium accompanied by an increase in blood flow to the level of sham animals. Contralateral kidneys with remarkable compensatory response to kidney injury showed an increase in density of arteriolar branches. Deciphering the mechanisms involved in kidney repair and regeneration post relief of obstruction has potential therapeutic implications for infants and children and the growing number of adults suffering from CKD.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Hidronefrosis/prevención & control , Riñón/cirugía , Regeneración , Obstrucción Ureteral/cirugía , Animales , Animales Recién Nacidos , Rastreo Celular/métodos , Modelos Animales de Enfermedad , Fibrosis , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hidronefrosis/genética , Hidronefrosis/metabolismo , Hidronefrosis/patología , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Ratones Transgénicos , Neovascularización Fisiológica , Estrés Oxidativo , Fenotipo , Circulación Renal , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Obstrucción Ureteral/genética , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología
11.
J Am Soc Nephrol ; 28(6): 1826-1837, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28062568

RESUMEN

Collectrin, encoded by the Tmem27 gene, is a transmembrane glycoprotein with approximately 50% homology with angiotensin converting enzyme 2, but without a catalytic domain. Collectrin is most abundantly expressed in the kidney proximal tubule and collecting duct epithelia, where it has an important role in amino acid transport. Collectrin is also expressed in endothelial cells throughout the vasculature, where it regulates L-arginine uptake. We previously reported that global deletion of collectrin leads to endothelial dysfunction, augmented salt sensitivity, and hypertension. Here, we performed kidney crosstransplants between wild-type (WT) and collectrin knockout (Tmem27Y/- ) mice to delineate the specific contribution of renal versus extrarenal collectrin on BP regulation and salt sensitivity. On a high-salt diet, WT mice with Tmem27Y/- kidneys had the highest systolic BP and were the only group to exhibit glomerular mesangial hypercellularity. Additional studies showed that, on a high-salt diet, Tmem27Y/- mice had lower renal blood flow, higher abundance of renal sodium-hydrogen antiporter 3, and lower lithium clearance than WT mice. In WT mice, administration of angiotensin II for 2 weeks downregulated collectrin expression in a type 1 angiotensin II receptor-dependent manner. This downregulation coincided with the onset of hypertension, such that WT and Tmem27Y/- mice had similar levels of hypertension after 2 weeks of angiotensin II administration. Altogether, these data suggest that salt sensitivity is determined by intrarenal collectrin, and increasing the abundance or activity of collectrin may have therapeutic benefits in the treatment of hypertension and salt sensitivity.


Asunto(s)
Angiotensina II/fisiología , Regulación hacia Abajo , Hipertensión/etiología , Glicoproteínas de Membrana/fisiología , Cloruro de Sodio Dietético/efectos adversos , Animales , Riñón/metabolismo , Masculino , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Noqueados
12.
Nano Lett ; 17(6): 3533-3542, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28511006

RESUMEN

Therapies capable of decelerating, or perhaps even halting, neurodegeneration in Parkinson's disease (PD) remain elusive. Clinical trials of PD gene therapy testing the delivery of neurotrophic factors, such as the glial cell-line derived neurotrophic factor (GDNF), have been largely ineffective due to poor vector distribution throughout the diseased regions in the brain. In addition, current delivery strategies involve invasive procedures that obviate the inclusion of early stage patients who are most likely to benefit from GDNF-based gene therapy. Here, we introduce a two-pronged treatment strategy, composed of MR image-guided focused ultrasound (FUS) and brain-penetrating nanoparticles (BPN), that provides widespread but targeted GDNF transgene expression in the brain following systemic administration. MR image-guided FUS allows circulating gene vectors to partition into the brain tissue by noninvasive and transient opening of the blood-brain barrier (BBB) within the areas where FUS is applied. Once beyond the BBB, BPN provide widespread and uniform GDNF expression throughout the targeted brain tissue. After only a single treatment, our strategy led to therapeutically relevant levels of GDNF protein content in the FUS-targeted regions in the striatum of the 6-OHDA-induced rat model of PD, which lasted at least up to 10 weeks. Importantly, our strategy restored both dopamine levels and dopaminergic neuron density and reversed behavioral indicators of PD-associated motor dysfunction with no evidence of local or systemic toxicity. Our combinatorial approach overcomes limitations of current delivery strategies, thereby potentially providing a novel means to treat PD.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Terapia Genética/métodos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Enfermedad de Parkinson/terapia , Animales , Transporte Biológico , Encéfalo/metabolismo , Dopamina/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Imagen por Resonancia Magnética , Nanopartículas/química , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Tamaño de la Partícula , Polietilenglicoles/química , Polietileneimina/química , Ratas , Ondas Ultrasónicas
13.
J Neurosci ; 36(10): 2881-93, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26961944

RESUMEN

Perinatal stroke leads to significant morbidity and long-term neurological and cognitive deficits. The pathophysiological mechanisms of brain damage depend on brain maturation at the time of stroke. To understand whether microglial cells limit injury after neonatal stroke by preserving neurovascular integrity, we subjected postnatal day 7 (P7) rats depleted of microglial cells, rats with inhibited microglial TGFbr2/ALK5 signaling, and corresponding controls, to transient middle cerebral artery occlusion (tMCAO). Microglial depletion by intracerebral injection of liposome-encapsulated clodronate at P5 significantly reduced vessel coverage and triggered hemorrhages in injured regions 24 h after tMCAO. Lack of microglia did not alter expression or intracellular redistribution of several tight junction proteins, did not affect degradation of collagen IV induced by the tMCAO, but altered cell types producing TGFß1 and the phosphorylation and intracellular distribution of SMAD2/3. Selective inhibition of TGFbr2/ALK5 signaling in microglia via intracerebral liposome-encapsulated SB-431542 delivery triggered hemorrhages after tMCAO, demonstrating that TGFß1/TGFbr2/ALK5 signaling in microglia protects from hemorrhages. Consistent with observations in neonatal rats, depletion of microglia before tMCAO in P9 Cx3cr1(GFP/+)/Ccr2(RFP/+) mice exacerbated injury and induced hemorrhages at 24 h. The effects were independent of infiltration of Ccr2(RFP/+) monocytes into injured regions. Cumulatively, in two species, we show that microglial cells protect neonatal brain from hemorrhage after acute ischemic stroke.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Infarto de la Arteria Cerebral Media/complicaciones , Hemorragias Intracraneales/etiología , Hemorragias Intracraneales/prevención & control , Microglía/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Benzamidas/farmacología , Conservadores de la Densidad Ósea/farmacología , Caspasa 3/metabolismo , Ácido Clodrónico/toxicidad , Dioxoles/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo
14.
Nanomedicine ; 13(8): 2565-2574, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28754465

RESUMEN

Liposome-based drug formulations represent an exciting avenue of research as they increase efficacy to toxicity ratios. Current formulations rely on passive accumulation to the disease site where drug is taken up by the cells. Ligand mediated targeting increases the net accumulation of liposomes, however, an unexplored benefit is to potentially refine pharmacodynamics (PD) of a drug specifically to different cell types within diseased tissue. As a model system, we engineered cardiomyocyte- (I-1) and endothelial-targeted (B-40) liposomes to carry a VEGFR2 inhibitor (PTK787), and examined the effect of cell type-specific delivery on both pharmacokinetics (PK) and PD. Neovascularization in post-myocardial infarction was significantly reduced by B-40 liposomes loaded with PTK787 as compared to animals injected with I-1 liposomes, and profoundly more as compared to free PTK787. This study thus shows that the intraorgan targeting of drugs through cell type-specific delivery holds substantial promise towards lowering the minimal efficacious dose administered systemically.


Asunto(s)
Liposomas/química , Péptidos/química , Ftalazinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridinas/administración & dosificación , Animales , Sistemas de Liberación de Medicamentos , Ratones , Infarto del Miocardio/complicaciones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/etiología , Biblioteca de Péptidos , Ftalazinas/farmacocinética , Ftalazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/farmacocinética , Piridinas/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
17.
Bioconjug Chem ; 27(2): 383-90, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26603129

RESUMEN

Magnetic resonance (MR) and photoacoustic (PA) imaging are currently being investigated as complementing strategies for applications requiring sensitive detection of cells in vivo. While combined MR/PAI detection of cells requires biocompatible cell labeling probes, water-based synthesis of dual-modality MR/PAI probes presents significant technical challenges. Here we describe facile synthesis and characterization of hybrid modular dextran-stabilized gold/iron oxide (Au-IO) multimetallic nanoparticles (NP) enabling multimodal imaging of cells. The stable association between the IO and gold NP was achieved by priming the surface of dextran-coated IO with silver NP resulting from silver(I) reduction by aldehyde groups, which are naturally present within the dextran coating of IO at the level of 19-23 groups/particle. The Au-IO NP formed in the presence of silver-primed Au-IO were stabilized by using partially thiolated MPEG5-gPLL graft copolymer carrying residual amino groups. This stabilizer served as a carrier of near-infrared fluorophores (e.g., IRDye 800RS) for multispectral PA imaging. Dual modality imaging experiments performed in capillary phantoms of purified Au-IO-800RS NPs showed that these NPs were detectible using 3T MRI at a concentration of 25 µM iron. PA imaging achieved approximately 2.5-times higher detection sensitivity due to strong PA signal emissions at 530 and 770 nm, corresponding to gold plasmons and IRDye integrated into the coating of the hybrid NPs, respectively, with no "bleaching" of PA signal. MDA-MB-231 cells prelabeled with Au-IO-800RS retained plasma membrane integrity and were detectable by using both MR and dual-wavelength PA at 49 ± 3 cells/imaging voxel. We believe that modular assembly of multimetallic NPs shows promise for imaging analysis of engineered cells and tissues with high resolution and sensitivity.


Asunto(s)
Medios de Contraste/química , Dextranos/química , Compuestos Férricos/química , Oro/química , Imagen por Resonancia Magnética/métodos , Nanopartículas/química , Técnicas Fotoacústicas/métodos , Línea Celular Tumoral , Medios de Contraste/farmacocinética , Dextranos/farmacocinética , Compuestos Férricos/farmacocinética , Oro/farmacocinética , Humanos , Nanopartículas del Metal , Imagen Multimodal/métodos
18.
Adv Exp Med Biol ; 880: 205-20, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26486340

RESUMEN

There are two alternative approaches to ultrasound-assisted drug delivery. First, the drug can be entrapped into or attached onto the ultrasound-responsive particles and administered in the vasculature, to achieve ultrasound-triggered drug release from the particles and localized tissue deposition in response to ultrasound treatment of the target zone. Second, the drug can be co-administered with the microbubbles or other sonosensitive particles. In this case, the action of ultrasound on the particles (which act as cavitation nuclei) results in the transient improvement of permeability of the physiological barriers, so that the circulating drug can exit the bloodstream and get into the target tissues and cells. We discuss and compare both of these approaches, their characteristic advantages and disadvantages for the specific drug delivery scenarios. Clearly, the system based on the off-label use of the existing approved microbubbles and drugs (or drug carriers) will have a chance of getting to clinical trials faster and with lesser resources spent. However, if a superior curative potential of a sonosensitive drug carrier is proven, and formulation stability problems are addressed properly, this approach may find its way to practical use, especially for nucleic acid delivery scenarios.


Asunto(s)
Sistemas de Liberación de Medicamentos , Microburbujas , Ultrasonido , Química Farmacéutica , Portadores de Fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Electricidad Estática
19.
Small ; 11(25): 3066-77, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25703465

RESUMEN

Microbubbles bearing plasmonic nanoparticles on their surface provide contrast enhancement for both photoacoustic and ultrasound imaging. In this work, the responses of microbubbles with surface-bound gold nanorods-termed AuMBs-to nanosecond pulsed laser excitation are studied using high-speed microscopy, photoacoustic imaging, and numerical modeling. In response to laser fluences below 5 mJ cm(-2) , AuMBs produce weak photoacoustic emissions and exhibit negligible microbubble wall motion. However, in reponse to fluences above 5 mJ cm(-2) , AuMBs undergo dramatically increased thermal expansion and emit nonlinear photoacoustic waves of over 10-fold greater amplitude than would be expected from freely dispersed gold nanorods. Numerical modeling suggests that AuMB photoacoustic responses to low laser fluences result from conductive heat transfer from the surface-bound nanorods to the microbubble gas core, whereas at higher fluences, explosive boiling may occur at the nanorod surface, producing vapor nanobubbles that contribute to rapid AuMB expansion. The results of this study indicate that AuMBs are capable of producing acoustic emissions of significantly higher amplitude than those produced by conventional sources of photoacoustic contrast. In vivo imaging performance of AuMBs in a murine kidney model suggests that AuMBs may be an effective alternative to existing contrast agents for noninvasive photoacoustic and ultrasound imaging applications.


Asunto(s)
Diagnóstico por Imagen de Elasticidad/métodos , Oro/química , Nanopartículas del Metal/química , Microburbujas , Técnicas Fotoacústicas/métodos , Resonancia por Plasmón de Superficie/métodos , Materiales Biocompatibles Revestidos/síntesis química , Medios de Contraste/síntesis química , Oro/efectos de la radiación , Aumento de la Imagen/métodos , Ensayo de Materiales , Nanopartículas del Metal/efectos de la radiación , Nanopartículas del Metal/ultraestructura , Oscilometría/métodos
20.
Biomed Microdevices ; 17(1): 23, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25663444

RESUMEN

Microfluidics-based production of stable microbubbles for ultrasound contrast enhancement or drug/gene delivery allows for precise control over microbubble diameter but at the cost of a low production rate. In situ microfluidic production of microbubbles directly in the vasculature may eliminate the necessity for high microbubble production rates, long stability, or small diameters. Towards this goal, we investigated whether microfluidic-produced microbubbles directly administered into a mouse tail vein could provide sufficient ultrasound contrast. Microbubbles composed of nitrogen gas and stabilized with 3 % bovine serum albumin and 10 % dextrose were injected for 10 seconds into wild type C57BL/6 mice, via a tail-vein catheter. Short-axis images of the right and left ventricle were acquired at 12.5 MHz and image intensity over time was analyzed. Microbubbles were produced on the order of 10(5) microbubbles/s and were observed in both the right and left ventricles. The median rise time, duration, and decay time within the right ventricle were 2.9, 21.3, and 14.3 s, respectively. All mice survived the procedure with no observable respiratory or heart rate distress despite microbubble diameters as large as 19 µm.


Asunto(s)
Medios de Contraste , Microburbujas , Técnicas Analíticas Microfluídicas/métodos , Ultrasonografía/métodos , Animales , Bovinos , Medios de Contraste/química , Medios de Contraste/farmacología , Ratones , Nitrógeno/química , Nitrógeno/farmacología , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA