Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
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
2.
bioRxiv ; 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37732205

RESUMEN

Background: Boiling histotripsy (BH), a mechanical focused ultrasound ablation strategy, can elicit intriguing signatures of anti-tumor immunity. However, the influence of BH on dendritic cell function is unknown, compromising our ability to optimally combine BH with immunotherapies to control metastatic disease. Methods: BH was applied using a sparse scan (1 mm spacing between sonications) protocol to B16F10-ZsGreen melanoma in bilateral and unilateral settings. Ipsilateral and contralateral tumor growth was measured. Flow cytometry was used to track ZsGreen antigen and assess how BH drives dendritic cell behavior. Results: BH monotherapy elicited ipsilateral and abscopal tumor control in this highly aggressive model. Tumor antigen presence in immune cells in the tumor-draining lymph nodes (TDLNs) was ~3-fold greater at 24h after BH, but this abated by 96h. B cells, macrophages, monocytes, granulocytes, and both conventional dendritic cell subsets (i.e. cDC1s and cDC2s) acquired markedly more antigen with BH. BH drove activation of both cDC subsets, with activation being dependent upon tumor antigen acquisition. Our data also suggest that BH-liberated tumor antigen is complexed with damage-associated molecular patterns (DAMPs) and that cDCs do not traffic to the TDLN with antigen. Rather, they acquire antigen as it flows through afferent lymph vessels into the TDLN. Conclusion: When applied with a sparse scan protocol, BH monotherapy elicits abscopal melanoma control and shapes dendritic cell function through several previously unappreciated mechanisms. These results offer new insight into how to best combine BH with immunotherapies for the treatment of metastatic melanoma.

3.
Theranostics ; 14(4): 1647-1661, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38389838

RESUMEN

Background: Boiling histotripsy (BH), a mechanical focused ultrasound ablation strategy, can elicit intriguing signatures of anti-tumor immunity. However, the influence of BH on dendritic cell function is unknown, compromising our ability to optimally combine BH with immunotherapies to control metastatic disease. Methods: BH was applied using a sparse scan (1 mm spacing between sonications) protocol to B16F10-ZsGreen melanoma in bilateral and unilateral settings. Ipsilateral and contralateral tumor growth was measured. Flow cytometry was used to track ZsGreen antigen and assess how BH drives dendritic cell behavior. Results: BH monotherapy elicited ipsilateral and abscopal tumor control in this highly aggressive model. Tumor antigen presence in immune cells in the tumor-draining lymph nodes (TDLNs) was ~3-fold greater at 24h after BH, but this abated by 96h. B cells, macrophages, monocytes, granulocytes, and both conventional dendritic cell subsets (i.e. cDC1s and cDC2s) acquired markedly more antigen with BH. BH drove activation of both cDC subsets, with activation being dependent upon tumor antigen acquisition. Our data also suggest that BH-liberated tumor antigen is complexed with damage-associated molecular patterns (DAMPs) and that cDCs do not traffic to the TDLN with antigen. Rather, they acquire antigen as it flows through afferent lymph vessels into the TDLN. Conclusion: When applied with a sparse scan protocol, BH monotherapy elicits abscopal melanoma control and shapes dendritic cell function through several previously unappreciated mechanisms. These results offer new insight into how to best combine BH with immunotherapies for the treatment of metastatic melanoma.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación , Melanoma , Humanos , Melanoma/terapia , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Antígenos de Neoplasias , Células Dendríticas
4.
Theranostics ; 11(20): 9847-9858, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34815790

RESUMEN

Background: Focused ultrasound (FUS) blood brain barrier disruption (BBBD) permits the noninvasive, targeted, and repeatable delivery of drugs to the brain. FUS BBBD also elicits secondary responses capable of augmenting immunotherapies, clearing amyloid-ß and hyperphosphorylated tau, and driving neurogenesis. Leveraging these secondary effects will benefit from an understanding of how they correlate to the magnitude of FUS BBBD and are differentially affected by the mechanical and biochemical stimuli imparted during FUS BBBD. Methods: We aggregated 75 murine transcriptomes in a multiple regression framework to identify genes expressed in proportion to biochemical (i.e. contrast MR image enhancement (CE)) or mechanical (i.e. harmonic acoustic emissions from MB-activation (MBA)) stimuli associated with FUS BBBD. Models were constructed to control for potential confounders, such as sex, anesthesia, and sequencing batch. Results: MBA and CE differentially predicted expression of 1,124 genes 6 h or 24 h later. While there existed overlap in the transcripts correlated with MBA vs CE, MBA was principally predictive of expression of genes associated with endothelial reactivity while CE chiefly predicted sterile inflammation gene sets. Over-representation analysis identified transcripts not previously linked to BBBD, including actin filament organization, which is likely important for BBB recovery. Transcripts and pathways associated with neurogenesis, microglial activation, and amyloid-ß clearance were significantly correlated to BBBD metrics. Conclusions: The secondary effects of BBBD may have the potential to be tuned by modulating FUS parameters during BBBD, and MBA and CE may serve as independent predictors of transcriptional reactions in the brain.


Asunto(s)
Barrera Hematoencefálica/fisiología , Sistemas de Liberación de Medicamentos/métodos , Terapia por Ultrasonido/métodos , Animales , Fenómenos Biomecánicos/fisiología , Encéfalo/metabolismo , Medios de Contraste/farmacología , Bases de Datos Genéticas , Expresión Génica/genética , Ratones , Microburbujas , Análisis Multivariante , Análisis de Regresión , Transcriptoma/genética
5.
Bioeng Transl Med ; 6(2): e10198, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34027087

RESUMEN

Focused ultrasound (FUS) mediated blood-brain barrier disruption (BBBD) targets the delivery of systemically-administered therapeutics to the central nervous system. Preclinical investigations of BBBD have been performed on different anesthetic backgrounds; however, the influence of the choice of anesthetic on the molecular response to BBBD is unknown, despite its potential to critically affect interpretation of experimental therapeutic outcomes. Here, using bulk RNA sequencing, we comprehensively examined the transcriptomic response of both normal brain tissue and brain tissue exposed to FUS-induced BBBD in mice anesthetized with either isoflurane with medical air (Iso) or ketamine/dexmedetomidine (KD). In normal murine brain tissue, Iso alone elicited minimal differential gene expression (DGE) and repressed pathways associated with neuronal signaling. KD alone, however, led to massive DGE and enrichment of pathways associated with protein synthesis. In brain tissue exposed to BBBD (1 MHz, 0.5 Hz pulse repetition frequency, 0.4 MPa peak-negative pressure), we systematically evaluated the relative effects of anesthesia, microbubbles, and FUS on the transcriptome. Of particular interest, we observed that gene sets associated with sterile inflammatory responses and cell-cell junctional activity were induced by BBBD, regardless of the choice of anesthesia. Meanwhile, gene sets associated with metabolism, platelet activity, tissue repair, and signaling pathways, were differentially affected by BBBD, with a strong dependence on the anesthetic. We conclude that the underlying transcriptomic response to FUS-mediated BBBD may be powerfully influenced by anesthesia. These findings raise considerations for the translation of FUS-BBBD delivery approaches that impact, in particular, metabolism, tissue repair, and intracellular signaling.

6.
Theranostics ; 10(16): 7436-7447, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32642004

RESUMEN

Background: Increasing evidence points to the critical role of extracellular vesicles (EVs) as molecular parcels that carry a diverse array of bioactive payloads for coordination of complex intracellular signaling. Focused ultrasound (FUS) hyperthermia is a technique for non-invasive, non-ionizing sublethal heating of cells in a near-instantaneous manner; while it has been shown to improve drug delivery and immunological recognition of tumors, its impact on EVs has not been explored to date. The goal of this study was to determine whether FUS impacts the release, proteomic profile, and immune-activating properties of tumor-derived EVs. Methods: Monolayered murine glioma cells were seeded within acoustically transparent cell culture chambers, and FUS hyperthermia was applied to achieve complete coverage of the chamber. Glioma-derived EVs (GEVs) were isolated for characterization by Nanoparticle Tracking Analysis, cryo-electron microscopy and mass spectrometry. An in vitro experimental setup was designed to further dissect the impact of GEVs on innate inflammation; immortalized murine dendritic cells (DCs) were pulsed with GEVs (either naïve or FUS hyperthermia-exposed) and assayed for production of IL-12p70, an important regulator of DC maturation and T helper cell polarization toward the interferon-γ-producing type 1 phenotype. Results: We confirmed that FUS hyperthermia significantly augments GEV release (by ~46%) as well as shifts the proteomic profile of these GEVs. Such shifts included enrichment of common EV-associated markers, downregulation of markers associated with cancer progression and resistance and modulation of inflammation-associated markers. When DCs were pulsed with GEVs, we noted that naïve GEVs suppressed IL-12p70 production by DCs in a GEV dose-dependent manner. In contrast, GEVs from cells exposed to FUS hyperthermia promoted a significant upregulation in IL-12p70 production by DCs, consistent with a pro-inflammatory stimulus. Conclusion: FUS hyperthermia triggers release of proteomically distinct GEVs that are capable of facilitating an important component of innate immune activation, lending both to a potential mechanism by which FUS interfaces with the tumor-immune landscape and to a role for GEV-associated biomarkers in monitoring response to FUS.


Asunto(s)
Vesículas Extracelulares/efectos de la radiación , Glioma/terapia , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Hipertermia Inducida/métodos , Animales , Línea Celular Tumoral , Microscopía por Crioelectrón , Células Dendríticas/inmunología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Glioma/inmunología , Glioma/patología , Humanos , Inmunidad Innata , Interleucina-12/inmunología , Interleucina-12/metabolismo , Ratones , Proteómica , Linfocitos T Colaboradores-Inductores/inmunología , Escape del Tumor/efectos de la radiación , Regulación hacia Arriba/efectos de la radiación
7.
Theranostics ; 10(19): 8821-8833, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32754281

RESUMEN

Background: Focused ultrasound (FUS) activation of microbubbles (MBs) for blood-brain (BBB) and blood-tumor barrier (BTB) opening permits targeted therapeutic delivery. While the effects of FUS+MBs mediated BBB opening have been investigated for normal brain tissue, no such studies exist for intracranial tumors. As this technology advances into clinical immunotherapy trials, it will be crucial to understand how FUS+MBs modulates the tumor immune microenvironment. Methods and Results: Bulk RNA sequencing revealed that FUS+MBs BTB/BBB opening (1 MHz, 0.5 MPa peak-negative pressure) of intracranial B16F1cOVA tumors increases the expression of genes related to proinflammatory cytokine and chemokine signaling, pattern recognition receptor signaling, and antigen processing and presentation. Flow cytometry revealed increased maturation (i.e. CD86) of dendritic cells (DCs) in the meninges and altered antigen loading of DCs in both the tumor and meninges. For DCs in tumor draining lymph nodes, FUS+MBs had no effect on maturation and elicited only a trend towards increased presentation of tumor-derived peptide by MHC. Neither tumor endothelial cell adhesion molecule expression nor homing of activated T cells was affected by FUS+MBs. Conclusion: FUS+MBs-mediated BTB/BBB opening elicits signatures of inflammation; however, the response is mild, transient, and unlikely to elicit a systemic response independent of administration of immune adjuvants.


Asunto(s)
Neoplasias Encefálicas/terapia , Células Dendríticas/metabolismo , Melanoma Experimental/terapia , Ovalbúmina/inmunología , Terapia por Ultrasonido/métodos , Animales , Barrera Hematoencefálica , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Regulación Neoplásica de la Expresión Génica , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Microburbujas , Análisis de Secuencia de ARN , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
ACS Appl Mater Interfaces ; 9(20): 17603-17612, 2017 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-28485954

RESUMEN

Mechanochromic luminescent (ML) materials, which show a change in emission due to an applied mechanical stimulus, are useful components in a variety of applications, including organic light-emitting diodes, force sensors, optical memory storage, and next-generation lighting materials. While there are many different ML active derivatives, few show room temperature self-erasing. Thin films of the methoxy substituted ß-diketone, gbmOMe, initially exhibited blue (428 nm) emission; however, green (478 nm) emission was observed after smearing. The mechanically generated smeared state recovered so rapidly that characterization of its emission was difficult at room temperature using traditional luminescence techniques. Thus, a new complementary metal oxide semiconductor camera imaging method was developed and used to calculate the decay time of the mechanically generated smeared state (i.e., smeared-state decay; τSM) for gbmOMe thin films. Additionally, this method was used to evaluate substrate and film thickness effects on ML recovery for glass and weighing paper films. The recovery behavior of gbmOMe was largely substrate-independent for the indicated matrixes; however, thickness effects were observed. Thus, film thickness may be the main factor in determining ML recovery behavior and must be accounted for when comparing the recovery dynamics of different ML materials. Moreover, when heated above the melting point (Tm = 119 °C), bulk gbmOMe powders assumed a metastable state that eventually crystallized after a few minutes at room temperature. However, melted thin films remained in an amorphous state indefinitely despite annealing at different temperatures (50-110 °C). The amorphous phase was identified as a supercooled liquid via changing the rate of cooling in differential scanning calorimetry thermograms.

9.
Anal Methods ; 8(15): 3109-3114, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27909462

RESUMEN

Lifetime-based oxygen imaging is useful in many biological applications but instrumentation can be stationary, expensive, and complex. Herein, we present a portable, cost effective, simple alternative with high spatiotemporal resolution that uses a complementary metal oxide silicon (CMOS) camera to measure oxygen sensitive lifetimes on the millisecond scale. We demonstrate its compatibility with difluoroboron ß-diketonate poly(lactic acid) (BF2bdkPLA) polymers which are nontoxic and exhibit long-lived oxygen sensitive phosphorescence. Spatially resolved lifetimes of four BF2bdkPLA variants are measured using nonlinear least squares (NLS) and rapid lifetime determination (RLD) both of which are shown to be accurate and precise. Real-time imaging in a dynamic environment is demonstrated by determining lifetime pixel-wise. The setup costs less than $5000, easily fits into a backpack, and can operate on battery power alone. This versatility combined with the inherent utility of lifetime measurements make this system a useful tool for a wide variety of oxygen sensing applications. This study serves as an important foundation for the development of dual mode real time lifetime plus ratiometric imaging with bright, long lifetime difluoroboron ß-diketonate probes.

10.
ACS Sens ; 1(11): 1366-1373, 2016 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-28042606

RESUMEN

Difluoroboron ß-diketonate poly(lactic acid) materials exhibit both fluorescence (F) and oxygen sensitive room-temperature phosphorescence (RTP). Introduction of halide heavy atoms (Br and I) is an effective strategy to control the oxygen sensitivity in these materials. A series of naphthyl-phenyl (nbm) dye derivatives with hydrogen, bromide and iodide substituents were prepared for comparison. As nanoparticles, the hydrogen derivative was hypersensitive to oxygen (0-0.3%), while the bromide analogue was suited for hypoxia detection (0-3% O2). The iodo derivative, BF2nbm(I)PLA, showed excellent F to RTP peak separation and an 0-100% oxygen sensitivity range unprecedented for metal-free RTP emitting materials. Due to the dual emission and unconventionally long RTP lifetimes of these O2 sensing materials, a portable, cost-effective camera was used to quantify oxygen levels via lifetime and red/green/blue (RGB) ratiometry. The hypersensitive H dye was well matched to lifetime detection, simultaneous lifetime and ratiometric imaging was possible for the bromide analogue, whereas the iodide material, with intense RTP emission and a shorter lifetime, was suited for RGB ratiometry. To demonstrate the prospects of this camera/material design combination for bioimaging, iodide boron dye-PLA nanoparticles were applied to a murine wound model to detect oxygen levels. Surprisingly, wound oxygen imaging was achieved without covering (i.e. without isolating from ambient conditions, air). Additionally, would healing was monitored via wound size reduction and associated oxygen recovery, from hypoxic to normoxic. These single-component materials provide a simple tunable platform for biological oxygen sensing that can be deployed to spatially resolve oxygen in a variety of environments.

11.
ACS Appl Mater Interfaces ; 7(42): 23633-43, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-26480236

RESUMEN

The dual-emissive properties of solid-state difluoroboron ß-diketonate-poly(lactic acid) (BF2bdkPLA) materials have been utilized for biological oxygen sensing. In this work, BF2dbm(X)PLA materials were synthesized, where X = H, F, Cl, Br, and I. The effects of changing the halide substituent and PLA polymer chain length on the optical properties in dilute CH2Cl2 solutions and solid-state polymer films were studied. These luminescent materials show fluorescence, phosphorescence, and lifetime tunability on the basis of molecular weight, as well as lifetime modulation via the halide substituent. Short BF2dbm(Br)PLA (6.0 kDa) and both short and long BF2dbm(I)PLA polymers (6.0 or 20.3 kDa) have fluorescence and intense phosphorescence ideal for ratiometric oxygen sensing. The lighter halide-dye polymers with hydrogen, fluorine, and chlorine substitution have longer phosphorescence lifetimes and can be utilized as ultrasensitive oxygen sensors. Photostability was also analyzed for the polymer films.


Asunto(s)
Técnicas Biosensibles , Compuestos de Boro/química , Oxígeno/aislamiento & purificación , Poliésteres/química , Fluorescencia , Hidrógeno/química , Oxígeno/química , Polímeros/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA