1
|
Zhang Y, Wang J, Ghobadi SN, Zhou H, Huang A, Gerosa M, Hou Q, Keunen O, Golebiewska A, Habte FG, Grant GA, Paulmurugan R, Lee KS, Wintermark M. Molecular Identity Changes of Tumor-Associated Macrophages and Microglia After Magnetic Resonance Imaging-Guided Focused Ultrasound-Induced Blood-Brain Barrier Opening in a Mouse Glioblastoma Model. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1082-1090. [PMID: 36717283 PMCID: PMC10059983 DOI: 10.1016/j.ultrasmedbio.2022.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/11/2022] [Accepted: 12/10/2022] [Indexed: 05/11/2023]
Abstract
An orthotopically allografted mouse GL26 glioma model (Ccr2RFP/wt-Cx3cr1GFP/wt) was used to evaluate the effect of transient, focal opening of the blood-brain barrier (BBB) on the composition of tumor-associated macrophages and microglia (TAMs). BBB opening was induced by magnetic resonance imaging (MRI)-guided focused ultrasound (MRgFUS) combined with microbubbles. CX3CR1-GFP cells and CCR2-RFP cells in brain tumors were quantified in microscopic images. Tumors in animals treated with a single session of MRgFUS did not exhibit significant changes in cell numbers when compared with tumors in animals not receiving FUS. However, tumors that received two or three sessions of MRgFUS had significantly increased amounts of both CX3CR1-GFP and CCR2-RFP cells. The effect of MRgFUS on immune cell composition was also characterized and quantified using flow cytometry. Glioma implantation resulted in increased amounts of lymphocytes, monocytes and neutrophils in the brain parenchyma. Tumors administered MRgFUS exhibited increased numbers of monocytes and monocyte-derived TAMs. In addition, MRgFUS-treated tumors exhibited more CD80+ cells in monocytes and microglia. In summary, transient, focal opening of the BBB using MRgFUS combined with microbubbles can activate the homing and differentiation of monocytes and induce a shift toward a more pro-inflammatory status of the immune environment in glioblastoma.
Collapse
Affiliation(s)
- Yanrong Zhang
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA
| | - Jing Wang
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA; Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Sara Natasha Ghobadi
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA
| | - Haiyan Zhou
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA; Acupuncture and Tuina School/Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ai Huang
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Marco Gerosa
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA; Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Qingyi Hou
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA; Department of Nuclear Medicine, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Olivier Keunen
- In Vivo Imaging Facility, Luxembourg Institute of Health, Luxembourg
| | - Anna Golebiewska
- Department of Oncology, Luxembourg Institute of Health, Luxembourg
| | - Frezghi G Habte
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, CA, USA
| | - Gerald A Grant
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Canary Center for Cancer Early Detection, Department of Radiology, Stanford University, Stanford, CA, USA
| | - Kevin S Lee
- Departments of Neuroscience and Neurosurgery and Center for Brain, Immunology, and Glia, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Max Wintermark
- Department of Neuroradiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Mathon B, Navarro V, Lecas S, Roussel D, Charpier S, Carpentier A. Safety Profile of Low-Intensity Pulsed Ultrasound-Induced Blood-Brain Barrier Opening in Non-epileptic Mice and in a Mouse Model of Mesial Temporal Lobe Epilepsy. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1327-1336. [PMID: 36878831 DOI: 10.1016/j.ultrasmedbio.2023.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE It is unknown whether ultrasound-induced blood-brain barrier (BBB) disruption can promote epileptogenesis and how BBB integrity changes over time after sonication. METHODS To gain more insight into the safety profile of ultrasound (US)-induced BBB opening, we determined BBB permeability as well as histological modifications in C57BL/6 adult control mice and in the kainate (KA) model for mesial temporal lobe epilepsy in mice after sonication with low-intensity pulsed ultrasound (LIPU). Microglial and astroglial changes in ipsilateral hippocampus were examined at different time points following BBB disruption by respectively analyzing Iba1 and glial fibrillary acidic protein immunoreactivity. Using intracerebral EEG recordings, we further studied the possible electrophysiological repercussions of a repeated disrupted BBB for seizure generation in nine non-epileptic mice. RESULTS LIPU-induced BBB opening led to transient albumin extravasation and reversible mild astrogliosis, but not to microglial activation in the hippocampus of non-epileptic mice. In KA mice, the transient albumin extravasation into the hippocampus mediated by LIPU-induced BBB opening did not aggravate inflammatory processes and histologic changes that characterize the hippocampal sclerosis. Three LIPU-induced BBB opening did not induce epileptogenicity in non-epileptic mice implanted with depth EEG electrodes. CONCLUSION Our experiments in mice provide persuasive evidence of the safety of LIPU-induced BBB opening as a therapeutic modality for neurological diseases.
Collapse
Affiliation(s)
- Bertrand Mathon
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, Paris, France; Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France; Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, Paris, France; Advanced Surgical Research Technology Lab, Sorbonne University, Paris, France.
| | - Vincent Navarro
- Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France; Epileptology Unit, Department of Neurology, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, Paris, France
| | - Sarah Lecas
- Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France
| | - Delphine Roussel
- Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France
| | - Stéphane Charpier
- Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France
| | - Alexandre Carpentier
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, Paris, France; Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, Paris, France; Advanced Surgical Research Technology Lab, Sorbonne University, Paris, France
| |
Collapse
|
3
|
Lim Kee Chang W, Chan TG, Raguseo F, Mishra A, Chattenton D, de Rosales RTM, Long NJ, Morse SV. Rapid short-pulses of focused ultrasound and microbubbles deliver a range of agent sizes to the brain. Sci Rep 2023; 13:6963. [PMID: 37117169 PMCID: PMC10147927 DOI: 10.1038/s41598-023-33671-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023] Open
Abstract
Focused ultrasound and microbubbles can non-invasively and locally deliver therapeutics and imaging agents across the blood-brain barrier. Uniform treatment and minimal adverse bioeffects are critical to achieve reliable doses and enable safe routine use of this technique. Towards these aims, we have previously designed a rapid short-pulse ultrasound sequence and used it to deliver a 3 kDa model agent to mouse brains. We observed a homogeneous distribution in delivery and blood-brain barrier closing within 10 min. However, many therapeutics and imaging agents are larger than 3 kDa, such as antibody fragments and antisense oligonucleotides. Here, we evaluate the feasibility of using rapid short-pulses to deliver higher-molecular-weight model agents. 3, 10 and 70 kDa dextrans were successfully delivered to mouse brains, with decreasing doses and more heterogeneous distributions with increasing agent size. Minimal extravasation of endogenous albumin (66.5 kDa) was observed, while immunoglobulin (~ 150 kDa) and PEGylated liposomes (97.9 nm) were not detected. This study indicates that rapid short-pulses are versatile and, at an acoustic pressure of 0.35 MPa, can deliver therapeutics and imaging agents of sizes up to a hydrodynamic diameter between 8 nm (70 kDa dextran) and 11 nm (immunoglobulin). Increasing the acoustic pressure can extend the use of rapid short-pulses to deliver agents beyond this threshold, with little compromise on safety. This study demonstrates the potential for deliveries of higher-molecular-weight therapeutics and imaging agents using rapid short-pulses.
Collapse
Affiliation(s)
- William Lim Kee Chang
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Tiffany G Chan
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Federica Raguseo
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Aishwarya Mishra
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SW1 7EH, UK
| | - Dani Chattenton
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, London, SM2 5NG, UK
| | - Rafael T M de Rosales
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SW1 7EH, UK
| | - Nicholas J Long
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Sophie V Morse
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK.
| |
Collapse
|
4
|
Morphological and Functional Effects of Ultrasound on Blood-Brain Barrier Transitory Opening: An In Vitro Study on Rat Brain Endothelial Cells. Cells 2023; 12:cells12010192. [PMID: 36611987 PMCID: PMC9818236 DOI: 10.3390/cells12010192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 01/05/2023] Open
Abstract
With the recent advances in medicine, human life expectancy is increasing; however, the extra years of life are not necessarily spent in good health or free from disability, resulting in a significantly higher incidence of age-associated pathologies. Among these disorders, neurodegenerative diseases have a significant impact. To this end, the presence of the protective blood-brain barrier (BBB) represents a formidable obstacle to the delivery of therapeutics. Thus, this makes it imperative to define strategies to bypass the BBB in order to successfully target the brain with the appropriate drugs. It has been demonstrated that targeting the BBB by ultrasound (US) can transiently make this anatomical barrier permeable and in so doing, allow the delivery of therapeutics. Thus, our aim was to carry out an in depth in vitro molecular and morphological study on the effects of US treatment on the BBB. The rat brain endothelial (RBE4) cell line was challenged with exposure to 12 MHz diagnostic US treatment for 10, 20, and 30 min. Cell viability assays, Western blotting analysis on the endoplasmic reticulum (ER), and oxidative stress marker evaluation were then performed, along with cytological and immunofluorescence staining, in order to evaluate the effects of US on the intercellular spaces and tight junction distribution of the brain endothelial cells. We observed that the US treatment exerted no toxic effects on either RBE4 cell viability or the upregulation/dislocation of the ER and oxidative stress marker (GRP78 and cytochrome C, respectively). Further, we observed that the application of US induced an increase in the intercellular spaces, as shown by Papanicolaou staining, mainly due to the altered distribution of the tight junction protein zonula occludens-1 (ZO-1). This latter US-dependent effect was transient and disappeared 20 min after the removal of the stimulus. In conclusion, our results show that US induces a transient alteration of the BBB, without altering the intracellular signaling pathways such as the ER and oxidative stress that could potentially be toxic for endothelial cells. These results suggested that US treatment could represent a potential strategy for improving drug delivery to the brain.
Collapse
|
5
|
Presset A, Bodard S, Lefèvre A, Millet A, Oujagir E, Dupuy C, Iazourène T, Bouakaz A, Emond P, Escoffre JM, Nadal-Desbarats L. First Metabolomic Signature of Blood-Brain Barrier Opening Induced by Microbubble-Assisted Ultrasound. Front Mol Neurosci 2022; 15:888318. [PMID: 35795688 PMCID: PMC9251546 DOI: 10.3389/fnmol.2022.888318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Microbubble (MB)-assisted ultrasound (US) is a promising physical method to increase non-invasively, transiently, and precisely the permeability of the blood-brain barrier (BBB) to therapeutic molecules. Previous preclinical studies established the innocuity of this procedure using complementary analytical strategies including transcriptomics, histology, brain imaging, and behavioral tests. This cross-sectional study using rats aimed to investigate the metabolic processes following acoustically-mediated BBB opening in vivo using multimodal and multimatrices metabolomics approaches. After intravenous injection of MBs, the right striata were exposed to 1-MHz sinusoidal US waves at 0.6 MPa peak negative pressure with a burst length of 10 ms, for 30 s. Then, the striata, cerebrospinal fluid (CSF), blood serum, and urine were collected during sacrifice in three experimental groups at 3 h, 2 days, and 1 week after BBB opening (BBBO) and were compared to a control group where no US was applied. A well-established analytical workflow using nuclear magnetic resonance spectrometry and non-targeted and targeted high-performance liquid chromatography coupled to mass spectrometry were performed on biological tissues and fluids. In our experimental conditions, a reversible BBBO was observed in the striatum without physical damage or a change in rodent weight and behavior. Cerebral, peri-cerebral, and peripheral metabolomes displayed specific and sequential metabolic kinetics. The blood serum metabolome was more impacted in terms of the number of perturbated metabolisms than in the CSF, the striatum, and the urine. In addition, perturbations of arginine and arginine-related metabolisms were detected in all matrices after BBBO, suggesting activation of vasomotor processes and bioenergetic supply. The exploration of the tryptophan metabolism revealed a transient vascular inflammation and a perturbation of serotoninergic neurotransmission in the striatum. For the first time, we characterized the metabolic signature following the acoustically-mediated BBBO within the striatum and its surrounding biological compartments.
Collapse
Affiliation(s)
- Antoine Presset
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Sylvie Bodard
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Antoine Lefèvre
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
- Département Analyses Chimique et Métabolomique, PST Analyses des Systèmes Biologiques, Université de Tours, Tours, France
| | - Anaïs Millet
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Edward Oujagir
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Camille Dupuy
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Tarik Iazourène
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Ayache Bouakaz
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Patrick Emond
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
- Département Analyses Chimique et Métabolomique, PST Analyses des Systèmes Biologiques, Université de Tours, Tours, France
- CHRU Tours, Serv Med Nucl in Vitro, Tours, France
| | - Jean-Michel Escoffre
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
- Jean-Michel Escoffre,
| | - Lydie Nadal-Desbarats
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
- Département Analyses Chimique et Métabolomique, PST Analyses des Systèmes Biologiques, Université de Tours, Tours, France
- *Correspondence: Lydie Nadal-Desbarats,
| |
Collapse
|
6
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Shojai S, Haeri Rohani SA, Moosavi-Movahedi AA, Habibi-Rezaei M. Human serum albumin in neurodegeneration. Rev Neurosci 2022; 33:803-817. [PMID: 35363449 DOI: 10.1515/revneuro-2021-0165] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/02/2022] [Indexed: 11/15/2022]
Abstract
Serum albumin (SA) exists in relatively high concentrations, in close contact with most cells. However, in the adult brain, except for cerebrospinal fluid (CSF), SA concentration is relatively low. It is mainly produced in the liver to serve as the main protein of the blood plasma. In the plasma, it functions as a carrier, chaperon, antioxidant, source of amino acids, osmoregulator, etc. As a carrier, it facilitates the stable presence and transport of the hydrophobic and hydrophilic molecules, including free fatty acids, steroid hormones, medicines, and metal ions. As a chaperon, SA binds to and protects other proteins. As an antioxidant, thanks to a free sulfhydryl group (-SH), albumin is responsible for most antioxidant properties of plasma. These functions qualify SA as a major player in, and a mirror of, overall health status, aging, and neurodegeneration. The low concentration of SA is associated with cognitive deterioration in the elderly and negative prognosis in multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS). SA has been shown to be structurally modified in neurological conditions such as Alzheimer's disease (AD). During blood-brain barrier damage albumin enters the brain tissue and could trigger epilepsy and neurodegeneration. SA is able to bind to the precursor agent of the AD, amyloid-beta (Aβ), preventing its toxic effects in the periphery, and is being tested for treating this disease. SA therapy may also be effective in brain rejuvenation. In the current review, we will bring forward the prominent properties and roles of SA in neurodegeneration.
Collapse
Affiliation(s)
- Sajjad Shojai
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | | | | | - Mehran Habibi-Rezaei
- School of Biology, College of Science, University of Tehran, Tehran, Iran
- Nano-Biomedicine Center of Excellence, Nanoscience and Nanotechnology Research Center, University of Tehran, Tehran, Iran
| |
Collapse
|
8
|
Ischemia-Modified Albumin: Origins and Clinical Implications. DISEASE MARKERS 2021; 2021:9945424. [PMID: 34336009 PMCID: PMC8315882 DOI: 10.1155/2021/9945424] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/02/2021] [Accepted: 06/29/2021] [Indexed: 11/18/2022]
Abstract
Albumin is one of the most abundant proteins in the body of mammals: about 40% of its pool is located in the intravascular space and the remainder is found in the interstitial space. The content of this multifunctional protein in blood is about 60-65% of total plasma proteins. A decrease in its synthesis or changes of functional activity can destabilize oncotic blood pressure, cause a violation of transporting hormones, fatty acids, metals, and drugs. Albumin properties change under ischemic attacks associated with oxidative stress, production of reactive oxygen species, and acidosis. Under these conditions, ischemia-modified albumin (IMA) is generated that has a reduced metal-binding capacity, especially for transition metals, such as copper, nickel, and cobalt. The method of determining the cobalt-binding capability of HSA was initially proposed to evaluate IMA level and then licensed as an ACB test for routine clinical analysis for myocardial ischemia. Subsequent studies have shown the viability of the ACB test in diagnosing other diseases associated with the development of oxidative stress. This review examines recent data on IMA generation mechanisms, describes principles, advantages, and limitations of methods for evaluation of IMA levels, and provides detailed analysis of its use in diagnostic and monitoring therapeutic efficacy in different diseases.
Collapse
|
9
|
Lynch M, Heinen S, Markham-Coultes K, O'Reilly M, Van Slyke P, Dumont DJ, Hynynen K, Aubert I. Vasculotide restores the blood-brain barrier after focused ultrasound-induced permeability in a mouse model of Alzheimer's disease. Int J Med Sci 2021; 18:482-493. [PMID: 33390817 PMCID: PMC7757142 DOI: 10.7150/ijms.36775] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Focused ultrasound (FUS) is used to locally and transiently induce blood-brain barrier (BBB) permeability, allowing targeted drug delivery to the brain. The purpose of the current study is to evaluate the potential of Vasculotide to accelerate the recovery of the BBB following FUS disruption in the TgCRND8 mouse model of amyloidosis, characteristic of Alzheimer's disease (AD). Accelerating the restoration of the BBB post-FUS would represent an additional safety procedure, which could be beneficial for clinical applications. Methods: TgCRND8 mice and their non-transgenic littermates were treated with Vasculotide (250 ng, intraperitoneal) every 48 hours for 3 months. BBB permeability was induced using FUS, in presence of intravenously injected microbubbles, in TgCRND8 and non-transgenic mice, and confirmed at time 0 by MRI enhancement using the contrast agent gadolinium. BBB closure was assessed at 6, 12 and 20 hours by MRI. In a separate cohort of animals, BBB closure was assessed at 24-hours post-FUS using Evans blue injected intravenously and followed by histological evaluation. Results: Chronic Vasculotide administration significantly reduces the ultra-harmonic threshold required for FUS-induced BBB permeability in the TgCRND8 mice. In addition, Vasculotide treatment led to a faster restoration of the BBB following FUS in TgCRND8 mice. BBB closure after FUS is not significantly different between TgCRND8 and non-transgenic mice. BBB permeability was assessed by gadolinium up to 20-hours post-FUS, demonstrating 87% closure in Vasculotide treated TgCRND8 mice, as opposed to 52% in PBS treated TgCRND8 mice, 58% in PBS treated non-transgenic mice, and 74% in Vasculotide treated non-transgenic mice. In both TgCRND8 mice and non-transgenic littermates the BBB was impermeable to Evans blue dye at 24-hours post-FUS. Conclusion: Vasculotide reduces the pressure required for microbubble ultra-harmonic onset for FUS-induced BBB permeability and it accelerates BBB restoration in a mouse model of amyloidosis, suggesting its potential clinical utility to promote vascular health, plasticity and repair in AD.
Collapse
Affiliation(s)
- Madelaine Lynch
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave. Toronto, ON, Canada M4N 3M5
- Laboratory Medicine & Pathobiology, University of Toronto, 27 King's College Circle, Toronto, ON, Canada, M5S 1A1
| | - Stefan Heinen
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave. Toronto, ON, Canada M4N 3M5
| | - Kelly Markham-Coultes
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave. Toronto, ON, Canada M4N 3M5
| | - Meaghan O'Reilly
- Physical Sciences, Sunnybrook Research Institute, 2075 Bayview Ave. Toronto, ON, Canada M4N 3M5
- Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, Canada, M5G 1L7
| | - Paul Van Slyke
- Vasomune Therapeutics, 661 University Ave #465, Toronto, ON M5G 1M1
| | - Daniel J. Dumont
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave. Toronto, ON, Canada M4N 3M5
- Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, Canada, M5G 1L7
| | - Kullervo Hynynen
- Physical Sciences, Sunnybrook Research Institute, 2075 Bayview Ave. Toronto, ON, Canada M4N 3M5
- Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, Canada, M5G 1L7
| | - Isabelle Aubert
- Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave. Toronto, ON, Canada M4N 3M5
- Laboratory Medicine & Pathobiology, University of Toronto, 27 King's College Circle, Toronto, ON, Canada, M5S 1A1
| |
Collapse
|
10
|
Ye D, Luan J, Pang H, Yang Y, Nazeri A, Rubin JB, Chen H. Characterization of focused ultrasound-mediated brainstem delivery of intranasally administered agents. J Control Release 2020; 328:276-285. [PMID: 32871204 PMCID: PMC7749082 DOI: 10.1016/j.jconrel.2020.08.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
Focused ultrasound-mediated intranasal (FUSIN) delivery is a recently proposed technique that bypasses the blood-brain barrier to achieve noninvasive and localized brain drug delivery. The goal of this study was to characterize FUSIN drug delivery outcome in mice with regard to its dependency on several critical experimental factors, including the time interval between IN administration and FUS sonication (Tlag1), the FUS pressure, and the time for sacrificing the mice post-FUS (Tlag2). Wild-type mice were treated by FUSIN delivery of near-infrared fluorescent dye-labeled bovine serum albumin (800CW-BSA, used as a model agent). 800CW-BSA was intranasally administered to the mice in vivo, followed by intravenous injection of microbubbles and FUS sonication at the brainstem. Fluorescence imaging of ex vivo mouse brain slices was used to quantify the delivery outcomes of 800CW-BSA. Major organs, along with the nasal tissue and trigeminal nerve, were harvested to assess the biodistribution of 800CW-BSA. The delivery outcome of 800CW-BSA was the highest at the brainstem when Tlag1 was 0.5 h, which was on average 24.5-fold, 5.4-fold, and 21.6-fold higher than those of the IN only, Tlag1 = 1.5 h, and Tlag1 = 4.0 h, respectively. The FUSIN delivery outcome at the lowest pressure level, 0.43 MPa, was on average 1.8-fold and 3.7-fold higher than those at 0.56 MPa and 0.70 MPa, respectively. The mean concentration of 800CW-BSA in the brainstem after FUSIN delivery decreased from 0.5 h to 4.0 h post-FUS. The accumulation of 800CW-BSA was low in the heart, lung, spleen, kidneys, and liver, but high in the stomach and intestines. This study revealed the unique characteristics of FUSIN as a noninvasive, efficient, and localized brain drug delivery technique.
Collapse
Affiliation(s)
- Dezhuang Ye
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Jingyi Luan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Hannah Pang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Arash Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO., 63110, USA
| | - Joshua B Rubin
- Departments of Pediatrics and Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA; Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, 63108, USA..
| |
Collapse
|
11
|
Clinically approved IVIg delivered to the hippocampus with focused ultrasound promotes neurogenesis in a model of Alzheimer's disease. Proc Natl Acad Sci U S A 2020; 117:32691-32700. [PMID: 33288687 DOI: 10.1073/pnas.1908658117] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Preclinical and clinical data support the use of focused ultrasound (FUS), in the presence of intravenously injected microbubbles, to safely and transiently increase the permeability of the blood-brain barrier (BBB). FUS-induced BBB permeability has been shown to enhance the bioavailability of administered intravenous therapeutics to the brain. Ideal therapeutics candidates for this mode of delivery are those capable of inducing benefits peripherally following intravenous injection and in the brain at FUS-targeted areas. In Alzheimer's disease, intravenous immunoglobulin (IVIg), a fractionated human blood product containing polyclonal antibodies, act as immunomodulator peripherally and centrally, and it can reduce amyloid pathology in the brain. Using the TgCRND8 mouse model of amyloidosis, we tested whether FUS can improve the delivery of IVIg, administered intravenously (0.4 g/kg), to the hippocampus and reach an effective dose to reduce amyloid plaque pathology and promote neurogenesis. Our results show that FUS-induced BBB permeability is required to deliver a significant amount of IVIg (489 ng/mg) to the targeted hippocampus of TgCRN8 mice. Two IVIg-FUS treatments, administered at days 1 and 8, significantly increased hippocampal neurogenesis by 4-, 3-, and 1.5-fold in comparison to saline, IVIg alone, and FUS alone, respectively. Amyloid plaque pathology was significantly reduced in all treatment groups: IVIg alone, FUS alone, and IVIg-FUS. Putative factors promoting neurogenesis in response to IVIg-FUS include the down-regulation of the proinflammatory cytokine TNF-α in the hippocampus. In summary, FUS was required to deliver an effective dose of IVIg to promote hippocampal neurogenesis and modulate the inflammatory milieu.
Collapse
|
12
|
Rich MC, Sherwood J, Bartley AF, Whitsitt QA, Lee M, Willoughby WR, Dobrunz LE, Bao Y, Lubin FD, Bolding M. Focused ultrasound blood brain barrier opening mediated delivery of MRI-visible albumin nanoclusters to the rat brain for localized drug delivery with temporal control. J Control Release 2020; 324:172-180. [PMID: 32376461 DOI: 10.1016/j.jconrel.2020.04.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022]
Abstract
There is an ongoing need for noninvasive tools to manipulate brain activity with molecular, spatial and temporal specificity. Here we have investigated the use of MRI-visible, albumin-based nanoclusters for noninvasive, localized and temporally specific drug delivery to the rat brain. We demonstrated that IV injected nanoclusters could be deposited into target brain regions via focused ultrasound facilitated blood brain barrier opening. We showed that nanocluster location could be confirmed in vivo with MRI. Additionally, following confirmation of nanocluster delivery, release of the nanocluster payload into brain tissue can be triggered by a second focused ultrasound treatment performed without circulating microbubbles. Release of glutamate from nanoclusters in vivo caused enhanced c-Fos expression, indicating that the loading capacity of the nanoclusters is sufficient to induce neuronal activation. This novel technique for noninvasive stereotactic drug delivery to the brain with temporal specificity could provide a new way to study brain circuits in vivo preclinically with high relevance for clinical translation.
Collapse
Affiliation(s)
- Megan C Rich
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Jennifer Sherwood
- Department of Chemical and Biological Engineering, University of Alabama at Tuscaloosa, Tuscaloosa, AL 35487, USA
| | - Aundrea F Bartley
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Quentin A Whitsitt
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Magdelene Lee
- Department of Chemical and Biological Engineering, University of Alabama at Tuscaloosa, Tuscaloosa, AL 35487, USA
| | - W R Willoughby
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Lynn E Dobrunz
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Yuping Bao
- Department of Chemical and Biological Engineering, University of Alabama at Tuscaloosa, Tuscaloosa, AL 35487, USA.
| | - Farah D Lubin
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Mark Bolding
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
13
|
Hamasaki S, Mukuda T, Koyama Y, Nakane H, Kaidoh T. Constitutive accessibility of circulating proteins to hippocampal neurons in physiologically normal rats. Brain Behav 2020; 10:e01544. [PMID: 31985144 PMCID: PMC7066366 DOI: 10.1002/brb3.1544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/28/2019] [Accepted: 01/04/2020] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION Although the hippocampus (HIP) is thought impermeable to blood-borne proteins because of the integrity of the blood-brain barrier (BBB), it was recently suggested to be susceptible to hydrophilic hormones. The present study determined the accessibility of blood-borne signal molecules such as hormones to hippocampal neurons in physiologically normal rats. METHODS As a probe for accessibility, Evans blue dye (EB) that rapidly binds to albumin (Alb), which is impermeable to the BBB, was injected intravenously. To increase the vascular permeability of the BBB, a daily single administration of angiotensin II (Ang II) was applied intravenously for seven consecutive days. RESULTS Fifteen minutes after the injection of EB, histological observation revealed that a number of neurons had entrapped and accumulated EB into their cell bodies in the hippocampal dentate gyrus in all rats. Of these, relatively large oval neurons (>15 µm) in the hilus and molecular layer showed parvalbumin immunopositivity, indicating they are GABAergic interneurons. The population of EB-accumulating neurons (approximately 10 µm) were localized in the inner margin of the granule cell layer, suggesting they were granule cells. However, the number of EB-positive neurons did not change in rats treated with Ang II compared with vehicle injection. CONCLUSIONS These findings suggest an intriguing possibility that blood-derived proteins such as hormones have access to hippocampal neurons constitutively in the absence of stimuli that increase the vascular permeability of the BBB in a physiologically normal state.
Collapse
Affiliation(s)
- Sawako Hamasaki
- Department of Anatomy, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Takao Mukuda
- Department of Anatomy, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yuka Koyama
- Department of Anatomy, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hironobu Nakane
- Department of Anatomy, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Toshiyuki Kaidoh
- Department of Anatomy, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
14
|
McMahon D, Oakden W, Hynynen K. Investigating the effects of dexamethasone on blood-brain barrier permeability and inflammatory response following focused ultrasound and microbubble exposure. Am J Cancer Res 2020; 10:1604-1618. [PMID: 32042325 PMCID: PMC6993222 DOI: 10.7150/thno.40908] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
Rationale: Clinical trials are currently underway to test the safety and efficacy of delivering therapeutic agents across the blood-brain barrier (BBB) using focused ultrasound and microbubbles (FUS+MBs). While acoustic feedback control strategies have largely minimized the risk of overt tissue damage, transient induction of inflammatory processes have been observed following sonication in preclinical studies. The goal of this work was to explore the potential of post-sonication dexamethasone (DEX) administration as a means to mitigate treatment risk. Vascular permeability, inflammatory protein expression, blood vessel growth, and astrocyte activation were assessed. Methods: A single-element focused transducer (transmit frequency = 580 kHz) and DefinityTM microbubbles were used to increase BBB permeability unilaterally in the dorsal hippocampi of adult male rats. Sonicating pressure was calibrated based on ultraharmonic emissions. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was used to quantitatively assess BBB permeability at 15 min (baseline) and 2 hrs following sonication. DEX was administered following baseline imaging and at 24 hrs post-FUS+MB exposure. Expression of key inflammatory proteins were assessed at 2 days, and astrocyte activation and blood vessel growth were assessed at 10 days post-FUS+MB exposure. Results: Compared to saline-treated control animals, DEX administration expedited the restoration of BBB integrity at 2 hrs, and significantly limited the production of key inflammation-related proteins at 2 days, following sonication. Indications of FUS+MB-induced astrocyte activation and vascular growth were diminished at 10 days in DEX-treated animals, compared to controls. Conclusions: These results suggest that DEX provides a means of modulating the duration of BBB permeability enhancement and may reduce the risk of inflammation-induced tissue damage, increasing the safety profile of this drug-delivery strategy. This effect may be especially relevant in scenarios for which the goal of treatment is to restore or preserve neural function and multiple sonications are required.
Collapse
|
15
|
Veersema TJ, Neef A, Scheppingen J, Ferrier CH, Eijsden P, Gosselaar PH, Rijen PC, Spliet WG, Braun KP, Mühlebner A, Aronica E. Changes in vascular density in resected tissue of 97 patients with mild malformation of cortical development, focal cortical dysplasia or TSC‐related cortical tubers. Int J Dev Neurosci 2019; 79:96-104. [DOI: 10.1016/j.ijdevneu.2019.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 10/13/2019] [Accepted: 11/22/2019] [Indexed: 11/30/2022] Open
Affiliation(s)
- Tim J. Veersema
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Andrew Neef
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jackelien Scheppingen
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Cyrille H. Ferrier
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Pieter Eijsden
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Peter H. Gosselaar
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Peter C. Rijen
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Wim G.M. Spliet
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Kees P.J. Braun
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Angelika Mühlebner
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Eleonora Aronica
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN)The Netherlands
| |
Collapse
|
16
|
Sajesh BV, On NH, Omar R, Alrushaid S, Kopec BM, Wang WG, Sun HD, Lillico R, Lakowski TM, Siahaan TJ, Davies NM, Puno PT, Vanan MI, Miller DW. Validation of Cadherin HAV6 Peptide in the Transient Modulation of the Blood-Brain Barrier for the Treatment of Brain Tumors. Pharmaceutics 2019; 11:pharmaceutics11090481. [PMID: 31533285 PMCID: PMC6781504 DOI: 10.3390/pharmaceutics11090481] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
The blood-brain barrier (BBB) poses a major obstacle by preventing potential therapeutic agents from reaching their intended brain targets at sufficient concentrations. While transient disruption of the BBB has been used to enhance chemotherapeutic efficacy in treating brain tumors, limitations in terms of magnitude and duration of BBB disruption exist. In the present study, the preliminary safety and efficacy profile of HAV6, a peptide that binds to the external domains of cadherin, to transiently open the BBB and improve the delivery of a therapeutic agent, was evaluated in a murine brain tumor model. Transient opening of the BBB in response to HAV6 peptide administration was quantitatively characterized using both a gadolinium magnetic resonance imaging (MRI) contrast agent and adenanthin (Ade), the intended therapeutic agent. The effects of HAV6 peptide on BBB integrity and the efficacy of concurrent administration of HAV6 peptide and the small molecule inhibitor, Ade, in the growth and progression of an orthotopic medulloblastoma mouse model using human D425 tumor cells was examined. Systemic administration of HAV6 peptide caused transient, reversible disruption of BBB in mice. Increases in BBB permeability produced by HAV6 were rapid in onset and observed in all regions of the brain examined. Concurrent administration of HAV6 peptide with Ade, a BBB impermeable inhibitor of Peroxiredoxin-1, caused reduced tumor growth and increased survival in mice bearing medulloblastoma. The rapid onset and transient nature of the BBB modulation produced with the HAV6 peptide along with its uniform disruption and biocompatibility is well-suited for CNS drug delivery applications, especially in the treatment of brain tumors.
Collapse
Affiliation(s)
- Babu V Sajesh
- Research Institute in Oncology and Hematology, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Ngoc H On
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| | - Refaat Omar
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| | - Samaa Alrushaid
- College of Pharmacy Pharmaceutical Analysis Laboratory, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, Safat 13110, Kuwait
| | - Brian M Kopec
- Department of Pharmaceutical Chemistry, University of Kansas, Kansas, KS 66205, USA
| | - Wei-Guang Wang
- Kunming Institute of Botany, Kunming 650201, Yunnan, China
| | - Han-Dong Sun
- Kunming Institute of Botany, Kunming 650201, Yunnan, China
| | - Ryan Lillico
- College of Pharmacy Pharmaceutical Analysis Laboratory, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Ted M Lakowski
- College of Pharmacy Pharmaceutical Analysis Laboratory, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Teruna J Siahaan
- Department of Pharmaceutical Chemistry, University of Kansas, Kansas, KS 66205, USA
| | - Neal M Davies
- Pharmacy and Pharmaceutical Sciences, University of Alberta, Alberta, AB T6G 2R3, Canada
| | | | - Magimairajan Issai Vanan
- Research Institute in Oncology and Hematology, University of Manitoba, Winnipeg, MB R3E 0V9, Canada.
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - Donald W Miller
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada.
| |
Collapse
|
17
|
Focused Ultrasonography-Mediated Blood-Brain Barrier Disruption in the Enhancement of Delivery of Brain Tumor Therapies. World Neurosurg 2019; 131:65-75. [PMID: 31323404 DOI: 10.1016/j.wneu.2019.07.096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 01/06/2023]
Abstract
Glioblastoma is the most common intracranial malignancy in adults and carries a poor prognosis. Chemotherapeutic treatment figures prominently in the management of primary and recurrent disease. However, the blood-brain barrier presents a significant and formidable impediment to the entry of oncotherapeutic compounds to target tumor tissue. Several strategies have been developed to effect disruption of the blood-brain barrier and in turn enhance the efficacy of cytotoxic chemotherapy, as well as newly developed biologic agents. Focused ultrasonography is one such treatment modality, using acoustic cavitation of parenterally administered microbubbles to mechanically effect disruption of the vascular endothelium. We review and discuss the preclinical and clinical studies evaluating the biophysical basis for, and efficacy of, focused ultrasonography in the enhancement of oncotherapeutic agent delivery. Further, we provide some perspectives regarding future directions for the role of focused ultrasound in facilitating and improving the safe and effective delivery of oncotherapeutic agents in the treatment of glioblastoma.
Collapse
|
18
|
Morse SV, Pouliopoulos AN, Chan TG, Copping MJ, Lin J, Long NJ, Choi JJ. Rapid Short-pulse Ultrasound Delivers Drugs Uniformly across the Murine Blood-Brain Barrier with Negligible Disruption. Radiology 2019; 291:459-466. [PMID: 30912718 PMCID: PMC6493324 DOI: 10.1148/radiol.2019181625] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/27/2018] [Accepted: 02/01/2019] [Indexed: 11/11/2022]
Abstract
Background Previous work has demonstrated that drugs can be delivered across the blood-brain barrier by exposing circulating microbubbles to a sequence of long ultrasound pulses. Although this sequence has successfully delivered drugs to the brain, concerns remain regarding potentially harmful effects from disrupting the brain vasculature. Purpose To determine whether a low-energy, rapid, short-pulse ultrasound sequence can efficiently and safely deliver drugs to the murine brain. Materials and Methods Twenty-eight female wild-type mice underwent focused ultrasound treatment after injections of microbubbles and a labeled model drug, while three control mice were not treated (May-November 2017). The left hippocampus of 14 mice was exposed to low-energy short pulses (1 MHz; five cycles; peak negative pressure, 0.35 MPa) of ultrasound emitted at a rapid rate (1.25 kHz) in bursts (0.5 Hz), and another 14 mice were exposed to standard long pulses (10 msec, 0.5 Hz) containing 150 times more acoustic energy. Mice were humanely killed at 0 (n = 5), 10 (n = 3), or 20 minutes (n = 3) after ultrasound treatment. Hematoxylin-eosin (H-E) staining was performed on three mice. The delivered drug dose and distribution were quantified with the normalized optical density and coefficient of variation. Safety was assessed by H-E staining, the amount of albumin released, and the duration of permeability change in the blood-brain barrier. Statistical analysis was performed by using the Student t test. Results The rapid short-pulse sequence delivered drugs uniformly throughout the parenchyma. The acoustic energy emitted from the microbubbles also predicted the delivered dose (r = 0.97). Disruption in the blood-brain barrier lasted less than 10 minutes and 3.4-fold less albumin was released into the brain than with long pulses. No vascular or tissue damage from rapid short-pulse exposure was observable using H-E staining. Conclusion The rapid short-pulse ultrasound sequence is a minimally disruptive and efficient drug delivery method that could improve the treatment, diagnosis, and study of neurologic diseases. © RSNA, 2019 Online supplemental material is available for this article. See also the editorial by Klibanov and McDannold in this issue.
Collapse
Affiliation(s)
- Sophie V. Morse
- From the Departments of Bioengineering (S.V.M., A.N.P., M.J.C., J.L.,
J.J.C.) and Chemistry (T.G.C., N.J.L.), Imperial College London, Exhibition
Road, London SW7 2AZ, United Kingdom
| | - Antonios N. Pouliopoulos
- From the Departments of Bioengineering (S.V.M., A.N.P., M.J.C., J.L.,
J.J.C.) and Chemistry (T.G.C., N.J.L.), Imperial College London, Exhibition
Road, London SW7 2AZ, United Kingdom
| | - Tiffany G. Chan
- From the Departments of Bioengineering (S.V.M., A.N.P., M.J.C., J.L.,
J.J.C.) and Chemistry (T.G.C., N.J.L.), Imperial College London, Exhibition
Road, London SW7 2AZ, United Kingdom
| | - Matthew J. Copping
- From the Departments of Bioengineering (S.V.M., A.N.P., M.J.C., J.L.,
J.J.C.) and Chemistry (T.G.C., N.J.L.), Imperial College London, Exhibition
Road, London SW7 2AZ, United Kingdom
| | - Julien Lin
- From the Departments of Bioengineering (S.V.M., A.N.P., M.J.C., J.L.,
J.J.C.) and Chemistry (T.G.C., N.J.L.), Imperial College London, Exhibition
Road, London SW7 2AZ, United Kingdom
| | - Nicholas J. Long
- From the Departments of Bioengineering (S.V.M., A.N.P., M.J.C., J.L.,
J.J.C.) and Chemistry (T.G.C., N.J.L.), Imperial College London, Exhibition
Road, London SW7 2AZ, United Kingdom
| | - James J. Choi
- From the Departments of Bioengineering (S.V.M., A.N.P., M.J.C., J.L.,
J.J.C.) and Chemistry (T.G.C., N.J.L.), Imperial College London, Exhibition
Road, London SW7 2AZ, United Kingdom
| |
Collapse
|
19
|
Chen KT, Wei KC, Liu HL. Theranostic Strategy of Focused Ultrasound Induced Blood-Brain Barrier Opening for CNS Disease Treatment. Front Pharmacol 2019; 10:86. [PMID: 30792657 PMCID: PMC6374338 DOI: 10.3389/fphar.2019.00086] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 01/21/2019] [Indexed: 12/22/2022] Open
Abstract
Focused Ultrasound (FUS) in combination with gaseous microbubbles has emerged as a potential new means of effective drug delivery to the brain. Recent research has shown that, under burst-type energy exposure with the presence of microbubbles, this modality can transiently permeate the blood-brain barrier (BBB). The bioavailability of therapeutic agents is site-specifically augmented only in the zone where the FUS energy is targeted. The non-invasiveness of this approach makes FUS-induced BBB opening a novel and attractive means to perform localized CNS therapeutic agent delivery. Over the past decade, FUS-BBB opening has been preclinically confirmed to successfully enhance CNS penetration of therapeutic agents including chemotherapeutic agents, therapeutic peptides, monoclonal antibodies, and nanoparticles. Recently, a number of clinical human trials have begun to explore clinical utility. This review article, explores this technology through its physical mechanisms, summarizes the existing preclinical findings (including current medical device designs and technical approaches), and summarizes current ongoing clinical trials.
Collapse
Affiliation(s)
- Ko-Ting Chen
- Ph.D. Program in Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan.,Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hao-Li Liu
- Ph.D. Program in Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan.,Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Department of Electrical Engineering, Chang-Gung University, Taoyuan, Taiwan
| |
Collapse
|
20
|
D'Arrigo JS. Targeting Early Dementia: Using Lipid Cubic Phase Nanocarriers to Cross the Blood⁻Brain Barrier. Biomimetics (Basel) 2018; 3:E4. [PMID: 31105226 PMCID: PMC6352688 DOI: 10.3390/biomimetics3010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022] Open
Abstract
Over the past decades, a frequent co-morbidity of cerebrovascular pathology and Alzheimer's disease has been observed. Numerous published studies indicate that the preservation of a healthy cerebrovascular endothelium can be an important therapeutic target. By incorporating the appropriate drug(s) into biomimetic (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic, which targets certain cell surface scavenger receptors, mainly class B type I (i.e., SR-BI), and crosses the blood⁻brain barrier. This targeting allows for various cell types related to Alzheimer's to be simultaneously searched out for localized drug treatment in vivo.
Collapse
|
21
|
Nanotherapy for Alzheimer's disease and vascular dementia: Targeting senile endothelium. Adv Colloid Interface Sci 2018; 251:44-54. [PMID: 29274774 DOI: 10.1016/j.cis.2017.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022]
Abstract
Due to the complexity of Alzheimer's disease, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted lipid nanoemulsion) are available. Versatile small molecule drug(s) targeting multiple pathways of Alzheimer's disease pathogenesis are known. By incorporating such drug(s) into the targeted "lipid-coated microbubble" [LCM]/"nanoparticle-derived" [ND] (or LCM/ND) nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly class B type I), or SR-BI, making possible for various Alzheimer's-related cell types to be simultaneously searched out for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble [LCM] subpopulation (i.e., a stable LCM suspension); such film-stabilized microbubbles are well known to substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer's patient.
Collapse
|
22
|
Park JH, Park JA, Ahn JH, Kim YH, Kang IJ, Won MH, Lee CH. Transient cerebral ischemia induces albumin expression in microglia only in the CA1 region of the gerbil hippocampus. Mol Med Rep 2017; 16:661-665. [PMID: 28586018 PMCID: PMC5482121 DOI: 10.3892/mmr.2017.6671] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 02/09/2017] [Indexed: 11/30/2022] Open
Abstract
Albumin, the most abundant plasma protein, is known to exhibit a neuroprotective effect in animal models of focal and global cerebral ischemia. In the present study, the expression and immunoreactivity of albumin was examined in the hippocampus following 5 min of transient cerebral ischemia in gerbils. Albumin immunoreactivity was observed in microglia of the CA1 hippocampal region 2 days post-ischemic insult, and it was significantly increased at 4 days following ischemia-reperfusion. In addition, at 4 days post-ischemic insult, albumin-immunoreactive microglia were abundant in the stratum pyramidale of the CA1 region. The present results demonstrated that albumin was newly expressed post-injury in microglia in the CA1 region, suggesting ischemia-induced neuronal loss. Albumin expression may therefore be associated with ischemia-induced delayed neuronal death in the CA1 region following transient cerebral ischemia.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Chuncheon, Gangwon 24252, Republic of Korea
| | - Jin-A Park
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, South Chungcheong 31116, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Chuncheon, Gangwon 24252, Republic of Korea
| | - Yang Hee Kim
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, South Chungcheong 31116, Republic of Korea
| |
Collapse
|
23
|
Prager O, Friedman A, Nebenzahl YM. Role of neural barriers in the pathogenesis and outcome of Streptococcus pneumoniae meningitis. Exp Ther Med 2017; 13:799-809. [PMID: 28450902 PMCID: PMC5403536 DOI: 10.3892/etm.2017.4082] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/10/2016] [Indexed: 12/20/2022] Open
Abstract
Bacterial meningitis is an inflammatory disease of the meninges of the central nervous system (CNS). Streptococcus pneumoniae (S. pneumoniae), Neisseria meningitidis, and Haemophilus influenzae are the major bacterial pathogens causing meningitis with S. pneumoniae being responsible for two thirds of meningitis cases in the developed world. To reach the CNS following nasopharyngeal colonization and bacteraemia, the bacteria traverse from the circulation across the blood brain barrier (BBB) and choroid plexus. While the BBB has a protective role in healthy individuals by shielding the CNS from neurotoxic substances circulating in the blood and maintaining the homeostasis within the brain environment, dysfunction of the BBB is associated with the pathophysiology of numerous neurologic disorders, including bacterial meningitis. Inflammatory processes, including release of a broad range of cytokines and free radicals, further increase vascular permeability and contribute to the excessive neural damage observed. Injury to the cerebral microvasculature and loss of blood flow auto-regulation promote increased intracranial pressure and may lead to vascular occlusion. Other common complications commonly associated with meningitis include abnormal neuronal hyper-excitability (e.g., seizures) and loss of hearing. Despite the existence of antibiotic treatment and adjuvant therapy, the relatively high mortality rate and the severe outcomes among survivors of pneumococcal meningitis in developing and developed countries increase the urgency in the requirement of discovering novel biomarkers for the early diagnosis as well as novel treatment approaches. The present review aimed to explore the changes in the brain vascular barriers, which allow S. pneumoniae to invade the CNS, and describe the resultant brain injuries following bacterial meningitis.
Collapse
Affiliation(s)
- Ofer Prager
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of The Negev, Beer-Sheva 84101, Israel.,Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of The Negev, Beer-Sheva 84105, Israel
| | - Alon Friedman
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of The Negev, Beer-Sheva 84101, Israel.,Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of The Negev, Beer-Sheva 84105, Israel.,Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Yaffa Mizrachi Nebenzahl
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of The Negev, Beer-Sheva 84101, Israel
| |
Collapse
|
24
|
Lasič E, Galland F, Vardjan N, Šribar J, Križaj I, Leite MC, Zorec R, Stenovec M. Time-dependent uptake and trafficking of vesicles capturing extracellular S100B in cultured rat astrocytes. J Neurochem 2016; 139:309-323. [PMID: 27488079 DOI: 10.1111/jnc.13754] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/21/2016] [Accepted: 07/26/2016] [Indexed: 01/16/2023]
Abstract
Astrocytes, the most heterogeneous glial cells in the central nervous system, contribute to brain homeostasis, by regulating a myriad of functions, including the clearance of extracellular debris. When cells are damaged, cytoplasmic proteins may exit into the extracellular space. One such protein is S100B, which may exert toxic effects on neighboring cells unless it is removed from the extracellular space, but the mechanisms of this clearance are poorly understood. By using time-lapse confocal microscopy and fluorescently labeled S100B (S100B-Alexa488 ) and fluorescent dextran (Dextran546 ), a fluid phase uptake marker, we examined the uptake of fluorescently labeled S100B-Alexa488 from extracellular space and monitored trafficking of vesicles that internalized S100B-Alexa488 . Initially, S100B-Alexa488 and Dextran546 internalized with distinct rates into different endocytotic vesicles; S100B-Alexa488 internalized into smaller vesicles than Dextran546 . At a later stage, S100B-Alexa488 -positive vesicles substantially co-localized with Dextran546 -positive endolysosomes and with acidic LysoTracker-positive vesicles. Cell treatment with anti-receptor for advanced glycation end products (RAGE) antibody, which binds to RAGE, a 'scavenger receptor', partially inhibited uptake of S100B-Alexa488 , but not of Dextran546 . The dynamin inhibitor dynole 34-2 inhibited internalization of both fluorescent probes. Directional mobility of S100B-Alexa488 -positive vesicles increased over time and was inhibited by ATP stimulation, an agent that increases cytosolic free calcium concentration ([Ca2+ ]i ). We conclude that astrocytes exhibit RAGE- and dynamin-dependent vesicular mechanism to efficiently remove S100B from the extracellular space. If a similar process occurs in vivo, astroglia may mitigate the toxic effects of extracellular S100B by this process under pathophysiologic conditions. This study reveals the vesicular clearance mechanism of extracellular S100B in astrocytes. Initially, fluorescent S100B internalizes into smaller endocytotic vesicles than dextran molecules. At a later stage, both probes co-localize within endolysosomes. S100B internalization is both dynamin- and RAGE-dependent, whereas dextran internalization is dependent on dynamin. Vesicle internalization likely mitigates the toxic effects of extracellular S100B and other waste products.
Collapse
Affiliation(s)
- Eva Lasič
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Fabiana Galland
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Celica Biomedical, Ljubljana, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Marina Concli Leite
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia. .,Celica Biomedical, Ljubljana, Slovenia.
| | - Matjaž Stenovec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia. .,Celica Biomedical, Ljubljana, Slovenia.
| |
Collapse
|
25
|
Cho H, Lee HY, Han M, Choi JR, Ahn S, Lee T, Chang Y, Park J. Localized Down-regulation of P-glycoprotein by Focused Ultrasound and Microbubbles induced Blood-Brain Barrier Disruption in Rat Brain. Sci Rep 2016; 6:31201. [PMID: 27510760 PMCID: PMC4980618 DOI: 10.1038/srep31201] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/13/2016] [Indexed: 01/14/2023] Open
Abstract
Multi-drug resistant efflux transporters found in Blood-Brain Barrier (BBB) acts as a functional barrier, by pumping out most of the drugs into the blood. Previous studies showed focused ultrasound (FUS) induced microbubble oscillation can disrupt the BBB by loosening the tight junctions in the brain endothelial cells; however, no study was performed to investigate its impact on the functional barrier of the BBB. In this study, the BBB in rat brains were disrupted using the MRI guided FUS and microbubbles. The immunofluorescence study evaluated the expression of the P-glycoprotein (P-gp), the most dominant multi-drug resistant protein found in the BBB. Intensity of the P-gp expression at the BBB disruption (BBBD) regions was significantly reduced (63.2 ± 18.4%) compared to the control area. The magnitude of the BBBD and the level of the P-gp down-regulation were significantly correlated. Both the immunofluorescence and histologic analysis at the BBBD regions revealed no apparent damage in the brain endothelial cells. The results demonstrate that the FUS and microbubbles can induce a localized down-regulation of P-gp expression in rat brain. The study suggests a clinically translation of this method to treat neural diseases through targeted delivery of the wide ranges of brain disorder related drugs.
Collapse
Affiliation(s)
- HongSeok Cho
- Daegu-Gyeongbuk Medical Innovation Foundation, Medical Device Development Center, Daegu, 41061, South Korea
| | - Hwa-Youn Lee
- Daegu-Gyeongbuk Medical Innovation Foundation, Medical Device Development Center, Daegu, 41061, South Korea
| | - Mun Han
- Kyungpook National University, Department of Medical &Biological Engineering, Daegu, 41566, South Korea
| | - Jong-Ryul Choi
- Daegu-Gyeongbuk Medical Innovation Foundation, Medical Device Development Center, Daegu, 41061, South Korea
| | - Sanghyun Ahn
- Daegu-Gyeongbuk Medical Innovation Foundation, Laboratory Animal Center, Daegu, 41061, South Korea
| | - Taekwan Lee
- Daegu-Gyeongbuk Medical Innovation Foundation, Laboratory Animal Center, Daegu, 41061, South Korea
| | - Yongmin Chang
- Kyungpook National University, Department of Medical &Biological Engineering, Daegu, 41566, South Korea
| | - Juyoung Park
- Daegu-Gyeongbuk Medical Innovation Foundation, Medical Device Development Center, Daegu, 41061, South Korea
| |
Collapse
|
26
|
Burgess A, Shah K, Hough O, Hynynen K. Focused ultrasound-mediated drug delivery through the blood-brain barrier. Expert Rev Neurother 2016; 15:477-91. [PMID: 25936845 DOI: 10.1586/14737175.2015.1028369] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite recent advances in blood-brain barrier (BBB) research, it remains a significant hurdle for the pharmaceutical treatment of brain diseases. Focused ultrasound (FUS) is one method to transiently increase permeability of the BBB to promote drug delivery to specific brain regions. An introduction to the BBB and a brief overview of the methods, which can be used to circumvent the BBB to promote drug delivery, is provided. In particular, we discuss the advantages and limitations of FUS technology and the efficacy of FUS-mediated drug delivery in models of disease. MRI for targeting and evaluating FUS treatments, combined with administration of microbubbles, allows for transient, reproducible BBB opening. The integration of a real-time acoustic feedback controller has improved treatment safety. Successful clinical translation of FUS has the potential to transform the treatment of brain disease worldwide without requiring the development of new pharmaceutical agents.
Collapse
Affiliation(s)
- Alison Burgess
- Physical Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S665, Toronto, ON M4N 3M5, Canada
| | | | | | | |
Collapse
|
27
|
Burgess A, Hynynen K. Microbubble-Assisted Ultrasound for Drug Delivery in the Brain and Central Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 880:293-308. [PMID: 26486344 DOI: 10.1007/978-3-319-22536-4_16] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier is a serious impediment to the delivery of pharmaceutical treatments for brain diseases, including cancer, neurodegenerative and neuropsychatric diseases. Focused ultrasound, when combined with microbubbles, has emerged as an effective method to transiently and locally open the blood-brain barrier to promote drug delivery to the brain. Focused ultrasound has been used to successfully deliver a wide variety of therapeutic agents to pre-clinical disease models. The requirement for clinical translation of focused ultrasound technology is considered.
Collapse
Affiliation(s)
- Alison Burgess
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kullervo Hynynen
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada. .,Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Leinenga G, Götz J. Scanning ultrasound removes amyloid-β and restores memory in an Alzheimer's disease mouse model. Sci Transl Med 2015; 7:278ra33. [PMID: 25761889 DOI: 10.1126/scitranslmed.aaa2512] [Citation(s) in RCA: 345] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Amyloid-β (Aβ) peptide has been implicated in the pathogenesis of Alzheimer's disease (AD). We present a nonpharmacological approach for removing Aβ and restoring memory function in a mouse model of AD in which Aβ is deposited in the brain. We used repeated scanning ultrasound (SUS) treatments of the mouse brain to remove Aβ, without the need for any additional therapeutic agent such as anti-Aβ antibody. Spinning disk confocal microscopy and high-resolution three-dimensional reconstruction revealed extensive internalization of Aβ into the lysosomes of activated microglia in mouse brains subjected to SUS, with no concomitant increase observed in the number of microglia. Plaque burden was reduced in SUS-treated AD mice compared to sham-treated animals, and cleared plaques were observed in 75% of SUS-treated mice. Treated AD mice also displayed improved performance on three memory tasks: the Y-maze, the novel object recognition test, and the active place avoidance task. Our findings suggest that repeated SUS is useful for removing Aβ in the mouse brain without causing overt damage, and should be explored further as a noninvasive method with therapeutic potential in AD.
Collapse
Affiliation(s)
- Gerhard Leinenga
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia Campus, Brisbane, Queensland 4072, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia Campus, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
29
|
Facilitation of Drug Transport across the Blood-Brain Barrier with Ultrasound and Microbubbles. Pharmaceutics 2015; 7:275-93. [PMID: 26404357 PMCID: PMC4588200 DOI: 10.3390/pharmaceutics7030275] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/13/2015] [Accepted: 08/14/2015] [Indexed: 11/09/2022] Open
Abstract
Medical treatment options for central nervous system (CNS) diseases are limited due to the inability of most therapeutic agents to penetrate the blood–brain barrier (BBB). Although a variety of approaches have been investigated to open the BBB for facilitation of drug delivery, none has achieved clinical applicability. Mounting evidence suggests that ultrasound in combination with microbubbles might be useful for delivery of drugs to the brain through transient opening of the BBB. This technique offers a unique non-invasive avenue to deliver a wide range of drugs to the brain and promises to provide treatments for CNS disorders with the advantage of being able to target specific brain regions without unnecessary drug exposure. If this method could be applied for a range of different drugs, new CNS therapeutic strategies could emerge at an accelerated pace that is not currently possible in the field of drug discovery and development. This article reviews both the merits and potential risks of this new approach. It assesses methods used to verify disruption of the BBB with MRI and examines the results of studies aimed at elucidating the mechanisms of opening the BBB with ultrasound and microbubbles. Possible interactions of this novel delivery method with brain disease, as well as safety aspects of BBB disruption with ultrasound and microbubbles are addressed. Initial translational research for treatment of brain tumors and Alzheimer’s disease is presented.
Collapse
|
30
|
Abstract
INTRODUCTION The presence of the blood-brain barrier (BBB) is a significant impediment to the delivery of therapeutic agents to the brain for treatment of brain diseases. Focused ultrasound (FUS) has been developed as a noninvasive method for transiently increasing the permeability of the BBB to promote drug delivery to targeted regions of the brain. AREAS COVERED The present review briefly compares the methods used to promote drug delivery to the brain and describes the benefits and limitations of FUS technology. We summarize the experimental data which shows that FUS, combined with intravascular microbubbles, increases therapeutic agent delivery into the brain leading to significant reductions in pathology in preclinical models of disease. The potential for translation of this technology to the clinic is also discussed. EXPERT OPINION The introduction of magnetic resonance imaging guidance and intravascular administration of microbubbles to FUS treatments permits the consistent, transient and targeted opening of the BBB. The development of feedback systems and real-time monitoring techniques improve the safety of BBB opening. Successful clinical translation of FUS has the potential to revolutionize the treatment of brain disease resulting in effective, less-invasive treatments without the need for expensive drug development.
Collapse
Affiliation(s)
- Alison Burgess
- Sunnybrook Research Institute, Physical Sciences , 2075 Bayview Avenue, S665, Toronto, ON M4N 3M5 , Canada +1 416 480 5765 ;
| | | |
Collapse
|
31
|
Zhang YK, Liu JT, Peng ZW, Fan H, Yao AH, Cheng P, Liu L, Ju G, Kuang F. Different TLR4 expression and microglia/macrophage activation induced by hemorrhage in the rat spinal cord after compressive injury. J Neuroinflammation 2013; 10:112. [PMID: 24015844 PMCID: PMC3847110 DOI: 10.1186/1742-2094-10-112] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 08/30/2013] [Indexed: 02/01/2023] Open
Abstract
Background Hemorrhage is a direct consequence of traumatic injury to the central nervous system and may cause innate immune reactions including cerebral Toll-like receptor (TLR) 4 upregulation which usually leads to poor outcome in the traumatic brain injury. In spinal cord injury (SCI), however, how hemorrhage induces innate immune reaction in spinal parenchyma remains unknown. The present study aimed to see whether blood component and/or other factor(s) induce TLR4 and microglia/macrophages involved innate immune reactions in the rat spinal cord after traumatic injury. Methods Using the compressive SCI model of the rat, hemorrhage in the spinal cord was identified by hematoxylin-eosin staining. Microglia/macrophage activation, TLR4 expression, and cell apoptosis were investigated by immunohistochemistry. Nuclear factor (NF)-κB p50 level of the two segments of the cord was detected by western blotting assay. With carbon powder injection, blood origination of the hematoma was explored. The blood-spinal cord barrier (BSCB) states of the lesion site and the hematoma were compared with immunohistochemistry and tannic acid-ferric chloride staining. Results Histological observation found blood accumulated in the center of compression lesion site (epicenter) and in the hematoma approximately 1.5 cm away from the epicenter. TLR4 expression, microglia//macrophage activation, and subsequent apoptosis in the area of far-away hematoma were late and weak in comparison to that in epicenter. In addition, TLR4 positive microglia/macrophages appeared to be phagocytotic in the far-away hematoma more obviously than that in the epicenter. Injected carbon powder indicated that accumulated blood of the far-away hematoma originated from the bleeding of the lesion epicenter, and the BSCB around the hematoma was not compromised in the early phase. Accordingly, at 3 days post injury, NF-κB p50 was upregulated based on the similar levels of blood component hemoglobin, and cell apoptosis was obvious in the epicenter but not in the far-away hematoma. Conclusion These data suggest that besides blood component, BSCB compromise and the extent of tissue injury contribute more to TLR4 and microglia/macrophage responses to the spinal cord hemorrhage. Therefore, the innate immune environment is a necessary consideration for the SCI therapy targeting TLR4 and microglia/macrophages.
Collapse
Affiliation(s)
- Yu-Kai Zhang
- Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chihara M, Ikebuchi R, Otsuka S, Ichii O, Hashimoto Y, Suzuki A, Saga Y, Kon Y. Mice stage-specific claudin 3 expression regulates progression of meiosis in early stage spermatocytes. Biol Reprod 2013; 89:3. [PMID: 23677978 DOI: 10.1095/biolreprod.113.107847] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Claudin 3 is a protein component of the tight junction strands. Tight junctions between adjacent Sertoli cells form the blood-testis barrier (BTB). During spermatogenesis, seminiferous stage-specific expression of claudin 3 is believed to regulate the migration of preleptotene/leptotene spermatocytes across the BTB. Here, we determined the cell types expressing claudin 3 in adult mouse testis and investigated spermatogenesis after testis-specific in vivo knockdown of claudin 3. The results of in situ hybridization revealed that claudin 3 mRNA was predominantly expressed in germ cells near the basal lamina of seminiferous tubules at stages VI-IX. Furthermore, claudin 3 protein was localized not only to the BTB but also to the cell membrane of STRA8-expressing preleptotene/leptotene spermatocytes in the testis of adult ICR.Cg-Tg(Stra8-EGFP)1Ysa/YsaRbrc mice. Although claudin 3 knockdown did not affect BTB integrity, it did cause a partial delay in spermatocyte migration across the BTB. Moreover, claudin 3 knockdown resulted in a prolonged preleptotene phase during spermatogenesis. These data indicate that the seminiferous stage-specific expression and localization of claudin 3 during spermatogenesis regulate the progression of meiosis by promoting germ cell migration across the BTB.
Collapse
Affiliation(s)
- Masataka Chihara
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Burgess A, Hynynen K. Noninvasive and targeted drug delivery to the brain using focused ultrasound. ACS Chem Neurosci 2013; 4:519-26. [PMID: 23379618 PMCID: PMC3629738 DOI: 10.1021/cn300191b] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 01/21/2013] [Indexed: 01/27/2023] Open
Abstract
Brain diseases are notoriously difficult to treat due to the presence of the blood-brain barrier (BBB). Here, we review the development of focused ultrasound (FUS) as a noninvasive method for BBB disruption, aiding in drug delivery to the brain. FUS can be applied through the skull to a targeted region in the brain. When combined with microbubbles, FUS causes localized and reversible disruption of the BBB. The cellular mechanisms of BBB disruption are presented. Several therapeutic agents have been delivered to the brain resulting in significant improvements in pathology in models of glioblastoma and Alzheimer's disease. The requirements for clinical translation of FUS will be discussed.
Collapse
Affiliation(s)
- Alison Burgess
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Medical Biophysics and Institute
of Biomaterials & Biomedical Engineering (IBBME), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Abstract
BACKGROUND Fetal hypoxia contributes significantly to the pathogenesis of permanent perinatal brain injury. We hypothesized that hypoxia-induced cerebral angiogenesis and microvascular changes would occur in fetal sheep subjected to a severe hypoxic insult produced by umbilical cord occlusion (UCO) for 10 min. METHODS At 124-126 d of gestation, singleton fetal sheep underwent surgery for implantation of catheters and placement of an inflatable cuff around the umbilical cord. A 10-min UCO or sham UCO (n = 5) was induced at 130 d gestation. The fetal brain was collected at 24 h (n = 5) or 48 h (n = 4) after UCO for immunohistochemical analysis of vascular endothelial growth factor (VEGF), Ki67, and serum albumin. RESULTS By 48 h after UCO, the percentage of blood vessels expressing VEGF had increased in the subventricular zone, periventricular and subcortical white matter, corpus callosum, and cortex. Alterations in vascular permeability (albumin extravasation) were observed only in the periventricular and subcortical white matter and the subventricular zone following UCO. CONCLUSION The upregulation of VEGF expression and increased leakage of plasma protein in the fetal sheep brain show that the microvasculature in white matter is sensitive to hypoxia in the near-term brain.
Collapse
|
35
|
Focal Delivery of AAV2/1-transgenes Into the Rat Brain by Localized Ultrasound-induced BBB Opening. MOLECULAR THERAPY-NUCLEIC ACIDS 2013; 2:e73. [PMID: 23423361 PMCID: PMC3586801 DOI: 10.1038/mtna.2012.64] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Delivery of drugs and macromolecules to the central nervous system (CNS) is hindered by the blood–brain barrier (BBB). Several approaches have been used to overcome this hindrance to facilitate the treatment of various CNS diseases. We now present results showing that chimeric adeno-associated virus 2/1 (AAV2/1) particles containing the coding region for the LacZ gene are efficiently delivered into the rat brain upon intravenous (IV) administration after BBB opening by focused ultrasound in the presence of vascular acoustic resonators. We show that the transgene is correctly and efficiently expressed in cells located in the neighborhood of the insonated focus, especially in the vicinity of small vessels and capillaries. Histochemical LacZ staining allows the identification of large amounts of cells expressing the enzymatically active protein. Using double immunofluorescence (IF) with antibodies against tubulinIII and bacterial LacZ, we identified these cells to be mostly neurons. A small proportion of the transduced cells was recognized as glial cells, reacting positive in the IF with antibodies against astrocytic markers. These results demonstrate that our approach allows a very specific, localized, and efficient expression of intravenously administered transgenes in the brain of rats upon ultrasound-induced BBB opening.
Collapse
|
36
|
Lapilover EG, Lippman K, Salar S, Maslarova A, Dreier JP, Heinemann U, Friedman A. Peri-infarct blood-brain barrier dysfunction facilitates induction of spreading depolarization associated with epileptiform discharges. Neurobiol Dis 2012; 48:495-506. [PMID: 22782081 PMCID: PMC3588590 DOI: 10.1016/j.nbd.2012.06.024] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 06/12/2012] [Accepted: 06/27/2012] [Indexed: 11/17/2022] Open
Abstract
Recent studies showed that spreading depolarizations (SDs) occurs abundantly in patients following ischemic stroke and experimental evidence suggests that SDs recruit tissue at risk into necrosis. We hypothesized that BBB opening with consequent alterations of the extracellular electrolyte composition and extravasation of albumin facilitates generation of SDs since albumin mediates an astrocyte transcriptional response with consequent disturbance of potassium and glutamate homeostasis. Here we show extravasation of Evans blue-albumin complex into the hippocampus following cortical photothrombotic stroke in the neighboring neocortex. Using extracellular field potential recordings and exposure to serum electrolytes we observed spontaneous SDs in 80% of hippocampal slices obtained from rats 24 h after cortical photothrombosis. Hippocampal exposure to albumin for 24 h through intraventricular application together with serum electrolytes lowered the threshold for the induction of SDs in most slices irrespective of the pathway of stimulation. Exposing acute slices from naive animals to albumin led also to a reduced SD threshold. In albumin-exposed slices the onset of SDs was usually associated with larger stimulus-induced accumulation of extracellular potassium, and preceded by epileptiform activity, which was also observed during the recovery phase of SDs. Application of ifenprodil (3 μM), an NMDA-receptor type 2 B antagonist, blocked stimulus dependent epileptiform discharges and generation of SDs in slices from animals treated with albumin in-vivo. We suggest that BBB opening facilitates the induction of peri-infarct SDs through impaired homeostasis of K+.
Collapse
Affiliation(s)
- EG Lapilover
- Institute of Neurophysiology, Charité Universitätsmedizin, 13347 Berlin, Germany
| | - K. Lippman
- Institute of Neurophysiology, Charité Universitätsmedizin, 13347 Berlin, Germany
| | - S. Salar
- Institute of Neurophysiology, Charité Universitätsmedizin, 13347 Berlin, Germany
| | - A. Maslarova
- Institute of Neurophysiology, Charité Universitätsmedizin, 13347 Berlin, Germany
| | - JP Dreier
- Center for Stroke Research Berlin, Charité Universitätsmedizin, 10117 Berlin, Germany
- Department of Experimental Neurology, Charité Universitätsmedizin, 10117 Berlin, Germany
- Department of Neurology, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - U. Heinemann
- Institute of Neurophysiology, Charité Universitätsmedizin, 13347 Berlin, Germany
- Neurocure Research Center, Charité Universitätsmedizin, 13347 Berlin, Germany
| | - A. Friedman
- Institute of Neurophysiology, Charité Universitätsmedizin, 13347 Berlin, Germany
- Department of Physiology and Neurobiology, Faculty of Health Sciences and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| |
Collapse
|
37
|
Castle J, Butts M, Healey A, Kent K, Marino M, Feinstein SB. Ultrasound-mediated targeted drug delivery: recent success and remaining challenges. Am J Physiol Heart Circ Physiol 2012. [PMID: 23203969 DOI: 10.1152/ajpheart.00265.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The potential clinical value of developing a novel, nonviral, ultrasound-directed gene and drug delivery system is immense. Investigators soon will initiate clinical trials with the goal of treating a wide variety of maladies using noninvasive, ultrasound-based technology. The ongoing, scientific validation associated with promising preclinical success portents a novel range of therapeutics. The clinical utility and eventual clinical successes await vigorous testing. This review highlights the recent successes and challenges within the field of ultrasound-mediated drug delivery.
Collapse
Affiliation(s)
- Jason Castle
- General Electric Global Research, Niskayuna, New York, USA
| | | | | | | | | | | |
Collapse
|
38
|
Baburamani AA, Ek CJ, Walker DW, Castillo-Melendez M. Vulnerability of the developing brain to hypoxic-ischemic damage: contribution of the cerebral vasculature to injury and repair? Front Physiol 2012; 3:424. [PMID: 23162470 PMCID: PMC3493883 DOI: 10.3389/fphys.2012.00424] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/17/2012] [Indexed: 11/13/2022] Open
Abstract
As clinicians attempt to understand the underlying reasons for the vulnerability of different regions of the developing brain to injury, it is apparent that little is known as to how hypoxia-ischemia may affect the cerebrovasculature in the developing infant. Most of the research investigating the pathogenesis of perinatal brain injury following hypoxia-ischemia has focused on excitotoxicity, oxidative stress and an inflammatory response, with the response of the developing cerebrovasculature receiving less attention. This is surprising as the presentation of devastating and permanent injury such as germinal matrix-intraventricular haemorrhage (GM-IVH) and perinatal stroke are of vascular origin, and the origin of periventricular leukomalacia (PVL) may also arise from poor perfusion of the white matter. This highlights that cerebrovasculature injury following hypoxia could primarily be responsible for the injury seen in the brain of many infants diagnosed with hypoxic-ischemic encephalopathy (HIE). Interestingly the highly dynamic nature of the cerebral blood vessels in the fetus, and the fluctuations of cerebral blood flow and metabolic demand that occur following hypoxia suggest that the response of blood vessels could explain both regional protection and vulnerability in the developing brain. However, research into how blood vessels respond following hypoxia-ischemia have mostly been conducted in adult models of ischemia or stroke, further highlighting the need to investigate how the developing cerebrovasculature responds and the possible contribution to perinatal brain injury following hypoxia. This review discusses the current concepts on the pathogenesis of perinatal brain injury, the development of the fetal cerebrovasculature and the blood brain barrier (BBB), and key mediators involved with the response of cerebral blood vessels to hypoxia.
Collapse
Affiliation(s)
- Ana A Baburamani
- The Ritchie Centre, Monash Medical Centre, Monash Institute of Medical Research, Clayton Melbourne, VIC, Australia ; Sahlgrenska Academy, Gothenburg University Göteborg, Sweden
| | | | | | | |
Collapse
|
39
|
O'Reilly MA, Hynynen K. Ultrasound enhanced drug delivery to the brain and central nervous system. Int J Hyperthermia 2012; 28:386-96. [PMID: 22621739 DOI: 10.3109/02656736.2012.666709] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There is an increasing interest in the use of ultrasound to enhance drug delivery to the brain and central nervous system. Disorders of the brain and CNS historically have had poor response to drug therapy due to the presence of the blood-brain barrier (BBB). Techniques for circumventing the BBB are typically highly invasive or involve disrupting large portions of the BBB, exposing the brain to pathogens. Ultrasound can be non-invasively delivered to the brain through the intact skull. When combined with preformed microbubbles, ultrasound can safely induce transient, localised and reversible disruption of the BBB, allowing therapeutics to be delivered. Investigations to date have shown positive response to ultrasound BBB disruption combined with therapeutic agent delivery in rodent models of primary and metastatic brain cancer and Alzheimer's disease. Recent work in non-human primates has demonstrated that the technique is feasible for use in humans. This review examines the current status of drug delivery to the brain and CNS both by disruption of the BBB, and by ultrasound enhancement of drug delivery through the already compromised BBB. Cellular and physical mechanisms of disruption are discussed, as well as treatment technique, safety and monitoring.
Collapse
|
40
|
Neurovascular Unit: a Focus on Pericytes. Mol Neurobiol 2012; 45:327-47. [DOI: 10.1007/s12035-012-8244-2] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 02/01/2012] [Indexed: 10/28/2022]
|