Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Camilo Acosta is active.

Publication


Featured researches published by Camilo Acosta.


Journal of Cerebral Blood Flow and Metabolism | 2015

Enhanced Delivery and Bioactivity of the Neurturin Neurotrophic Factor through Focused Ultrasound—Mediated Blood—Brain Barrier Opening in vivo:

Gesthimani Samiotaki; Camilo Acosta; Shutao Wang; Elisa E. Konofagou

The blood—brain barrier (BBB) constitutes a major obstacle in brain drug delivery. Focused ultrasound (FUS) in conjunction with microbubbles has been shown to open the BBB noninvasively, locally, and transiently to allow large molecules diffusion. Neurturin (NTN), a member of the glial-derived neurotrophic factor (GDNF) family, has been demonstrated to have neuroprotective and regenerative effects on dopaminergic neurons in vivo using invasive drug delivery methods. The brains ascending nigrostriatal pathway is severely damaged in Parkinsons disease (PD), and therefore the substantia nigra (SN) and striatal caudoputamen (CP) were selected as the target areas. The objective of the study was to investigate whether safe and efficient NTN delivery can be achieved through FUS-induced BBB opening via intravenous administration, and thus trigger the neuroregeneration cascade in the nigrostriatal pathway. After the optimization of FUS parameters and target locations in the murine brain, NTN bioavailability and downstream signaling were detected and characterized through immunostaining. FUS significantly enhanced the delivery of NTN compared with the direct injection technique, whereas triggering of the signaling cascade was detected downstream to the neuronal nuclei. These findings thus indicate the potential of the FUS method to mediate transport of proteins through the blood—brain barrier in a PD animal model.


PLOS ONE | 2014

A New Brain Drug Delivery Strategy: Focused Ultrasound-Enhanced Intranasal Drug Delivery

Hong Chen; Cherry C. Chen; Camilo Acosta; Shih-Ying Wu; Tao Sun; Elisa E. Konofagou

Central nervous system (CNS) diseases are difficult to treat because of the blood-brain barrier (BBB), which prevents most drugs from entering into the brain. Intranasal (IN) administration is a promising approach for drug delivery to the brain, bypassing the BBB; however, its application has been restricted to particularly potent substances and it does not offer localized delivery to specific brain sites. Focused ultrasound (FUS) in combination with microbubbles can deliver drugs to the brain at targeted locations. The present study proposed to combine these two different platform techniques (FUS+IN) for enhancing the delivery efficiency of intranasally administered drugs at a targeted location. After IN administration of 40 kDa fluorescently-labeled dextran as the model drug, FUS targeted at one region within the caudate putamen of mouse brains was applied in the presence of systemically administered microbubbles. To compare with the conventional FUS technique, in which intravenous (IV) drug injection is employed, FUS was also applied after IV injection of the same amount of dextran in another group of mice. Dextran delivery outcomes were evaluated using fluorescence imaging of brain slices. The results showed that FUS+IN enhanced drug delivery within the targeted region compared with that achieved by IN only. Despite the fact that the IN route has limited drug absorption across the nasal mucosa, the delivery efficiency of FUS+IN was not significantly different from that of FUS+IV. As a new drug delivery platform, the FUS+IN technique is potentially useful for treating CNS diseases.


Physics in Medicine and Biology | 2015

Acoustic cavitation-based monitoring of the reversibility and permeability of ultrasound-induced blood-brain barrier opening.

Tao Sun; Gesthimani Samiotaki; Shutao Wang; Camilo Acosta; Cherry C. Chen; Elisa E. Konofagou

Cavitation events seeded by microbubbles have been previously reported to be associated with MR- or fluorescent-contrast enhancement after focused ultrasound (FUS)-induced blood-brain barrier (BBB) opening. However, it is still unknown whether bubble activity can be correlated with the reversibility (the duration of opening and the likelihood of safe reinstatement) and the permeability of opened BBB, which is critical for the clinical translation of using passive cavitation detection to monitor, predict and control the opening. In this study, the dependence of acoustic cavitation on the BBB opening duration, permeability coefficient and histological damage occurrence were thus investigated. Transcranial pulsed FUS at 1.5 MHz in the presence of systemically circulating microbubbles was applied in the mouse hippocampi (n  =  60). The stable and inertial cavitation activities were monitored during sonication. Contrast-enhanced MRI was performed immediately after sonication and every 24 h up to 6 d thereafter, to assess BBB opening, brain tissue permeability and potential edema. Histological evaluations were used to assess the occurrence of neurovascular damages. It was found that stable cavitation was well correlated with: (1) the duration of the BBB opening (r(2)  =  0.77); (2) the permeability of the opened BBB (r(2)  =  0.82); (3) the likelihood of safe opening (P  <  0.05, safe opening compared to cases of damage; P  <  0.0001, no opening compared to safe opening). The inertial cavitation dose was correlated with the resulting BBB permeability (r(2)  =  0.72). Stable cavitation was found to be more reliable than inertial cavitation at assessing the BBB opening within the pressure range used in this study. This study demonstrates that the stable cavitation response during BBB opening holds promise for predicting and controlling the restoration and pharmacokinetics of FUS-opened BBB. The stable cavitation response therefore showed great promise in predicting the BBB opening duration, enabling thus control of opening according to the drug circulation time. In addition, avoiding adverse effects in the brain and assessing the pharmacokinetics of the compounds delivered can also be achieved by monitoring and controlling the stable cavitation emissions.


Physics in Medicine and Biology | 2015

Chirp- and random-based coded ultrasonic excitation for localized blood-brain barrier opening

Hermes Kamimura; Shougang Wang; S-Y Wu; Maria Eleni Karakatsani; Camilo Acosta; Antonio Adilton Oliveira Carneiro; Elisa E. Konofagou

Chirp- and random-based coded excitation methods have been proposed to reduce standing wave formation and improve focusing of transcranial ultrasound. However, no clear evidence has been shown to support the benefits of these ultrasonic excitation sequences in vivo. This study evaluates the chirp and periodic selection of random frequency (PSRF) coded-excitation methods for opening the blood-brain barrier (BBB) in mice. Three groups of mice (n  =  15) were injected with polydisperse microbubbles and sonicated in the caudate putamen using the chirp/PSRF coded (bandwidth: 1.5–1.9 MHz, peak negative pressure: 0.52 MPa, duration: 30 s) or standard ultrasound (frequency: 1.5 MHz, pressure: 0.52 MPa, burst duration: 20 ms, duration: 5 min) sequences. T1-weighted contrast-enhanced MRI scans were performed to quantitatively analyze focused ultrasound induced BBB opening. The mean opening volumes evaluated from the MRI were mm3, mm3and mm3 for the chirp, random and regular sonications, respectively. The mean cavitation levels were V.s, V.s and V.s for the chirp, random and regular sonications, respectively. The chirp and PSRF coded pulsing sequences improved the BBB opening localization by inducing lower cavitation levels and smaller opening volumes compared to results of the regular sonication technique. Larger bandwidths were associated with more focused targeting but were limited by the frequency response of the transducer, the skull attenuation and the microbubbles optimal frequency range. The coded methods could therefore facilitate highly localized drug delivery as well as benefit other transcranial ultrasound techniques that use higher pressure levels and higher precision to induce the necessary bioeffects in a brain region while avoiding damage to the surrounding healthy tissue.


Journal of Cerebral Blood Flow and Metabolism | 2017

Direct brain infusion can be enhanced with focused ultrasound and microbubbles.

Shutao Wang; Maria Eleni Karakatsani; Christine Fung; Tao Sun; Camilo Acosta; Elisa E. Konofagou

The delivery of most therapeutic agents is rendered ineffective for the treatment of brain diseases due to the presence of the blood–brain barrier (BBB). The goal of this study was to investigate the effect of pre-infusion focused ultrasound (FUS) and microbubbles on the distribution of direct brain infusion in vivo. A single-element FUS transducer was used in all sonications, which were carried out immediately prior to direct infusion procedures. Mice received direct infusion of either Gadolinium-labeled albumin (Gd-albumin, 74 kDa) or adeno-associated virus (AAV, ∼4 MDa). The volumes of Gd-albumin at 30 min were deemed comparable (P = 0.334) between the direct infusion (DI)-only group and the FUS + DI group. At 120 min, the FUS + DI group showed significantly higher contrast-enhanced volume (9.76 ± 0.74 mm3) than the DI-only group (7.14 ± 0.34 mm3). For mice infused with AAV, the total volume of transduction was estimated as GFP-positive regions and FUS + DI group demonstrated significantly higher (P = 0.017) transduction efficiency in vivo. In conclusion, enhanced bio-distribution of directly infused agents was observed when the targeted region was pre-conditioned with FUS and microbubbles. Focused ultrasound has the potential, as an adjuvant technique, to significantly enhance direct brain infusion and achieve the desired therapeutic outcomes.


Scientific Reports | 2016

Focused ultrasound-enhanced intranasal brain delivery of brain-derived neurotrophic factor

Hong Chen; Georgiana Yang; Hoheteberhan Getachew; Camilo Acosta; Carlos Sierra Sanchez; Elisa E. Konofagou

The objective of this study was to unveil the potential mechanism of focused ultrasound (FUS)-enhanced intranasal (IN) brain drug delivery and assess its feasibility in the delivery of therapeutic molecules. Delivery outcomes of fluorescently-labeled dextrans to mouse brains by IN administration either before or after FUS sonication were compared to evaluate whether FUS enhances IN delivery by active pumping or passive diffusion. Fluorescence imaging of brain slices found that IN administration followed by FUS sonication achieved significantly higher delivery than IN administration only, while pre-treatment by FUS sonication followed by IN administration was not significantly different from IN administration only. Brain-derived neurotrophic factor (BDNF), a promising neurotrophic factor for the treatment of many central nervous system diseases, was delivered by IN followed by FUS to demonstrate the feasibility of this technique and compared with the established FUS technique where drugs are injected intravenously. Immunohistochemistry staining of BDNF revealed that FUS-enhanced IN delivery achieved similar locally enhanced delivery as the established FUS technique. This study suggested that FUS enhances IN brain drug delivery by FUS-induced active pumping of the drug and demonstrated that FUS-enhanced IN delivery is a promising technique for noninvasive and localized delivery of therapeutic molecules to the brain.


Journal of Cerebral Blood Flow and Metabolism | 2017

Lipid microbubbles as a vehicle for targeted drug delivery using focused ultrasound-induced blood–brain barrier opening:

Carlos Sierra; Camilo Acosta; Cherry C. Chen; Shih-Ying Wu; Maria Eleni Karakatsani; Manuel Bernal; Elisa E. Konofagou

Focused ultrasound in conjunction with lipid microbubbles has fully demonstrated its ability to induce non-invasive, transient, and reversible blood–brain barrier opening. This study was aimed at testing the feasibility of our lipid-coated microbubbles as a vector for targeted drug delivery in the treatment of central nervous system diseases. These microbubbles were labeled with the fluorophore 5-dodecanoylaminfluorescein. Focused ultrasound targeted mouse brains in vivo in the presence of these microbubbles for trans-blood–brain barrier delivery of 5-dodecanoylaminfluorescein. This new approach, compared to previously studies of our group, where fluorescently labeled dextrans and microbubbles were co-administered, represents an appreciable improvement in safety outcome and targeted drug delivery. This novel technique allows the delivery of 5-dodecanoylaminfluorescein at the region of interest unlike the alternative of systemic exposure. 5-dodecanoylaminfluorescein delivery was assessed by ex vivo fluorescence imaging and by in vivo transcranial passive cavitation detection. Stable and inertial cavitation doses were quantified. The cavitation dose thresholds for estimating, a priori, successful targeted drug delivery were, for the first time, identified with inertial cavitation were concluded to be necessary for successful delivery. The findings presented herein indicate the feasibility and safety of the proposed microbubble-based targeted drug delivery and that, if successful, can be predicted by cavitation detection in vivo.


internaltional ultrasonics symposium | 2015

Pupil dilation and motor response elicitation by ultrasound neuromodulation

Hermes Kamimura; Shougang Wang; Hong Chen; Qi Wang; Christian Aurup; Camilo Acosta; Antonio Adilton Oliveira Carneiro; Elisa E. Konofagou

Focused ultrasound (FUS) neuromodulation has been previously proposed as a promising technique to drive neuronal activity. Here, we explored motor- and cognitive-related brain regions of mice by targeting specific brain structures using FUS neuromodulation in the mega-Hz range under a specific type of anesthesia. Contralateral motor responses were observed showing successful target specificity of the FUS neuromodulation achieved with 1.9 MHz. Higher acoustic pressures increased the success rate from 20% (at the threshold, 1.45 MPa) to 70% (1.79 MPa). The estimated latency measured by electromyography was 266 ± 37 ms. Pupil dilation was observed when neuromodulating regions in the brain covering the superior colliculus and other anxiety-related structures such as hippocampus and locus coeruleus. This study demonstrated the capability of FUS to modulate target specific regions in the brain including pupil dilation induced by FUS for the first time. Furthermore, evoked responses by cognitive regions demonstrated the capability of FUS to modulate deeper structures in the brain.


internaltional ultrasonics symposium | 2014

Enhancement of direct brain infusion with focused ultrasound and microbubbles

Shutao Wang; Tao Sun; Camilo Acosta; Maria Eleni Karakatsani; Oluyemi Olumolade; Gesthimani Samiotaki; Elisa E. Konofagou

Direct infusion to the brain is a frequently used technique in pre-clinical neuroscience research as well as several clinical applications. The relatively high intracranial pressure is one of the limiting factors that hinder efficient diffusion from the cannula tip. In this study, we utilized focused ultrasound (FUS) and microbubbles to condition the brain prior to performing the direct infusion. The acoustic parameters used for sonications were: 0.45 MPa peak rarefactional pressure, 6.7 ms pulse length, 5 Hz pulse repetition frequency, and a duration of 60 s. A 9.4 T magnetic resonance imaging (MRI) system was used to monitor the diffusion of an albumin-tagged MR contrast agent up to two hours. In addition, the diffusion of a commonly used gene therapy vector - adeno-associated virus (AAV) was evaluated via fluorescence imaging. Our results revealed that Pre-treatment with FUS and microbubbles significantly enhanced the total volume (P<;0.001) and volume increase (P<;0.05) of MR contrast agent in vivo.


internaltional ultrasonics symposium | 2017

Analysis of focused ultrasound with microbubbles induced BBB disruption on tight junction morphology

Tara Kugelman; Camilo Acosta; Shutao Wang; Marilena Karakatsani; Dritan Agalliu; Elisa E. Konofagou

Focused ultrasound (FUS) with systemically delivered microbubbles has been established to locally, reversibly and non-invasively increase the permeability of the blood brain barrier to facilitate targeted drug delivery to the brain. Despite FUS constituting a promising therapeutic technique, the cellular mechanism of FUS-mediated BBB opening has yet to be fully described. Immunoelectron microscopy of vessels has reported the removal of tight junction (TJ) proteins from the junctional cleft, yet no study has examined the structural integrity of the entire length of the TJ strand following FUS with microbubbles. Furthermore, permeability studies have identified differences in the rate of leakage between large and small vessels, but it remains to be studied if TJ strand morphology differs among vessel types. The aim of the current study is to investigate TJ strand morphology for structural abnormalities in different vessel types following FUS-induced BBB opening.

Collaboration


Dive into the Camilo Acosta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Chen

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge