1
|
O'Reilly MA. Exploiting the mechanical effects of ultrasound for noninvasive therapy. Science 2024; 385:eadp7206. [PMID: 39265013 DOI: 10.1126/science.adp7206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/07/2024] [Indexed: 09/14/2024]
Abstract
Focused ultrasound is a platform technology capable of eliciting a wide range of biological responses with high spatial precision deep within the body. Although focused ultrasound is already in clinical use for focal thermal ablation of tissue, there has been a recent growth in development and translation of ultrasound-mediated nonthermal therapies. These approaches exploit the physical forces of ultrasound to produce a range of biological responses dependent on exposure conditions. This review discusses recent advances in four application areas that have seen particular growth and have immense clinical potential: brain drug delivery, neuromodulation, focal tissue destruction, and endogenous immune system activation. Owing to the maturation of transcranial ultrasound technology, the brain is a major target organ; however, clinical indications outside the brain are also discussed.
Collapse
Affiliation(s)
- Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
López-Aguirre M, Castillo-Ortiz M, Viña-González A, Blesa J, Pineda-Pardo JA. The road ahead to successful BBB opening and drug-delivery with focused ultrasound. J Control Release 2024; 372:901-913. [PMID: 38971426 DOI: 10.1016/j.jconrel.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
This review delves into the innovative technology of Blood-Brain Barrier (BBB) opening with low-intensity focused ultrasound in combination with microbubbles (LIFU-MB), a promising therapeutic modality aimed at enhancing drug delivery to the central nervous system (CNS). The BBB's selective permeability, while crucial for neuroprotection, significantly hampers the efficacy of pharmacological treatments for CNS disorders. LIFU-MB emerges as a non-invasive and localized method to transiently increase BBB permeability, facilitating the delivery of therapeutic molecules. Here, we review the procedural stages of LIFU-MB interventions, including planning and preparation, sonication, evaluation, and delivery, highlighting the technological diversity and methodological challenges encountered in current clinical applications. With an emphasis on safety and efficacy, we discuss the crucial aspects of ultrasound delivery, microbubble administration, acoustic feedback monitoring and assessment of BBB permeability. Finally, we explore the critical choices for effective BBB opening with LIFU-MB, focusing on selecting therapeutic agents, optimizing delivery methods, and timing for delivery. Overcoming existing barriers to integrate this technology into clinical practice could potentially revolutionize CNS drug delivery and treatment paradigms in the near future.
Collapse
Affiliation(s)
- Miguel López-Aguirre
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain; PhD Program in Physics, Complutense University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Castillo-Ortiz
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain; PhD Program in Technologies for Health and Well-being, Polytechnic University of Valencia, Valencia, Spain; Molecular Imaging Technologies Research Institute (I3M), Polytechnic University of Valencia, Valencia, Spain
| | - Ariel Viña-González
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain; PhD Program in Biomedical Engineering, Polytechnic University of Madrid, Madrid, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain; Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Madrid, Spain
| | - José A Pineda-Pardo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain.
| |
Collapse
|
3
|
Martinez PJ, Song JJ, Garay FG, Song KH, Mufford T, Steiner J, DeSisto J, Ellens N, Serkova NJ, Green AL, Borden M. Comprehensive assessment of blood-brain barrier opening and sterile inflammatory response: unraveling the therapeutic window. Sci Rep 2024; 14:17036. [PMID: 39043894 PMCID: PMC11266505 DOI: 10.1038/s41598-024-67916-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
Microbubbles (MBs) combined with focused ultrasound (FUS) has emerged as a promising noninvasive technique to permeabilize the blood-brain barrier (BBB) for drug delivery into the brain. However, the safety and biological consequences of BBB opening (BBBO) remain incompletely understood. This study aims to investigate the effects of two parameters mediating BBBO: microbubble volume dose (MVD) and mechanical index (MI). High-resolution MRI-guided FUS was employed in mouse brains to assess BBBO by manipulating these two parameters. Afterward, the sterile inflammatory response (SIR) was studied 6 h post-FUS treatment. Results demonstrated that both MVD and MI significantly influenced the extent of BBBO, with higher MVD and MI leading to increased permeability. Moreover, RNA sequencing revealed upregulation of major inflammatory pathways and immune cell infiltration after BBBO, indicating the presence and extent of SIR. Gene set enrichment analysis identified 12 gene sets associated with inflammatory responses that were significantly upregulated at higher MVD or MI. A therapeutic window was established between therapeutically relevant BBBO and the onset of SIR, providing operating regimes to avoid damage from stimulation of the NFκB pathway via TNFɑ signaling to apoptosis. These results contribute to the optimization and standardization of BBB opening parameters for safe and effective drug delivery to the brain and further elucidate the underlying molecular mechanisms driving sterile inflammation.
Collapse
Affiliation(s)
- Payton J Martinez
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Jane J Song
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Francis G Garay
- Department of Radiology, Cancer Center Animal Imaging Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kang-Ho Song
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Toni Mufford
- Department of Radiology, Cancer Center Animal Imaging Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jenna Steiner
- Department of Radiology, Cancer Center Animal Imaging Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - John DeSisto
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Nicholas Ellens
- Alpheus Medical, Inc., 1266 Park Rd., Chanhassen, MN, 55317, USA
| | - Natalie J Serkova
- Department of Radiology, Cancer Center Animal Imaging Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Adam L Green
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Mark Borden
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, 80303, USA.
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA.
| |
Collapse
|
4
|
Rigollet S, Rome C, Ador T, Dumont E, Pichon C, Delalande A, Barbier EL, Stupar V. FUS-mediated BBB opening leads to transient perfusion decrease and inflammation without acute or chronic brain lesion. Theranostics 2024; 14:4147-4160. [PMID: 38994025 PMCID: PMC11234282 DOI: 10.7150/thno.96721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
Impact: The permeabilization of the BBB to deliver therapeutics with MR-guided FUS redefines therapeutic strategies as it improves patient outcomes. To ensure the best translation towards clinical treatment, the evaluation of hemodynamic modifications in the CNS is necessary to refine treatment parameters. Methods: MR-guided FUS was applied at 1.5 MHz with a 50 ms burst every 1 s to open the BBB. CBF, BVf and ADC parameters were monitored with MRI. Cavitation was monitored with a PCD during the FUS sequence and classified with the IUD index into three cavitation levels. We distinctly applied the FUS in the cortex or the striatum. After the BBB permeabilization, neuroinflammation markers were quantified longitudinally. Results: The BBB was successfully opened in all animals in this study and only one animal was classified as "hard" and excluded from the rest of the study. 30 min after FUS-induced BBB opening in the cortex, we measured a 54% drop in CBF and a 13% drop in BVf compared to the contralateral side. After permeabilization of the striatum, a 38% drop in CBF and a 15% drop in BVf were measured. CBF values rapidly returned to baseline, and 90 min after BBB opening, no significant differences were observed. We quantified the subsequent neuroinflammation, noting a significant increase in astrocytic recruitment at 2 days and microglial activation at 1 day after FUS. After 7 days, no more inflammation was visible in the brain. Conclusion: FUS-induced BBB opening transiently modifies hemodynamic parameters such as CBF and BVf, suggesting limited nutrients and oxygen supply to the CNS in the hour following the procedure.
Collapse
Affiliation(s)
- Sébastien Rigollet
- Image Guided Therapy, Pessac, France
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Claire Rome
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Thomas Ador
- Université d'Orléans, LI²RSO, Orléans, France
- ART ARNm, Inserm US55, Orléans, France
- Laboratory of Experimental and Molecular Immunology and Neuromodulation (INEM), UMR 7355 CNRS-University of Orleans, Orleans, France
| | | | - Chantal Pichon
- Université d'Orléans, LI²RSO, Orléans, France
- ART ARNm, Inserm US55, Orléans, France
- Institut Universitaire de France, Paris, France
| | - Anthony Delalande
- Université d'Orléans, LI²RSO, Orléans, France
- ART ARNm, Inserm US55, Orléans, France
| | - Emmanuel L. Barbier
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CNRS, IRMaGe, Grenoble, France
| | - Vasile Stupar
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CNRS, IRMaGe, Grenoble, France
| |
Collapse
|
5
|
Pellow C, Pichardo S, Pike GB. A systematic review of preclinical and clinical transcranial ultrasound neuromodulation and opportunities for functional connectomics. Brain Stimul 2024; 17:734-751. [PMID: 38880207 DOI: 10.1016/j.brs.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/21/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Low-intensity transcranial ultrasound has surged forward as a non-invasive and disruptive tool for neuromodulation with applications in basic neuroscience research and the treatment of neurological and psychiatric conditions. OBJECTIVE To provide a comprehensive overview and update of preclinical and clinical transcranial low intensity ultrasound for neuromodulation and emphasize the emerging role of functional brain mapping to guide, better understand, and predict responses. METHODS A systematic review was conducted by searching the Web of Science and Scopus databases for studies on transcranial ultrasound neuromodulation, both in humans and animals. RESULTS 187 relevant studies were identified and reviewed, including 116 preclinical and 71 clinical reports with subjects belonging to diverse cohorts. Milestones of ultrasound neuromodulation are described within an overview of the broader landscape. General neural readouts and outcome measures are discussed, potential confounds are noted, and the emerging use of functional magnetic resonance imaging is highlighted. CONCLUSION Ultrasound neuromodulation has emerged as a powerful tool to study and treat a range of conditions and its combination with various neural readouts has significantly advanced this platform. In particular, the use of functional magnetic resonance imaging has yielded exciting inferences into ultrasound neuromodulation and has the potential to advance our understanding of brain function, neuromodulatory mechanisms, and ultimately clinical outcomes. It is anticipated that these preclinical and clinical trials are the first of many; that transcranial low intensity focused ultrasound, particularly in combination with functional magnetic resonance imaging, has the potential to enhance treatment for a spectrum of neurological conditions.
Collapse
Affiliation(s)
- Carly Pellow
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada.
| | - Samuel Pichardo
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| | - G Bruce Pike
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
6
|
Caffaratti H, Slater B, Shaheen N, Rhone A, Calmus R, Kritikos M, Kumar S, Dlouhy B, Oya H, Griffiths T, Boes AD, Trapp N, Kaiser M, Sallet J, Banks MI, Howard MA, Zanaty M, Petkov CI. Neuromodulation with Ultrasound: Hypotheses on the Directionality of Effects and a Community Resource. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.14.24308829. [PMID: 38947047 PMCID: PMC11213082 DOI: 10.1101/2024.06.14.24308829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Low-intensity Transcranial Ultrasound Stimulation (TUS) is a promising non-invasive technique for deep-brain stimulation and focal neuromodulation. Research with animal models and computational modelling has raised the possibility that TUS can be biased towards enhancing or suppressing neural function. Here, we first conduct a systematic review of human TUS studies for perturbing neural function and alleviating brain disorders. We then collate a set of hypotheses on the directionality of TUS effects and conduct an initial meta-analysis on the human TUS study reported outcomes to date (n = 32 studies, 37 experiments). We find that parameters such as the duty cycle show some predictability regarding whether the targeted area's function is likely to be enhanced or suppressed. Given that human TUS sample sizes are exponentially increasing, we recognize that results can stabilize or change as further studies are reported. Therefore, we conclude by establishing an Iowa-Newcastle (inTUS) resource for the systematic reporting of TUS parameters and outcomes to support further hypothesis testing for greater precision in brain stimulation and neuromodulation with TUS.
Collapse
Affiliation(s)
- Hugo Caffaratti
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Ben Slater
- Biosciences Institute, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Nour Shaheen
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Ariane Rhone
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Ryan Calmus
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Michael Kritikos
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Sukhbinder Kumar
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Brian Dlouhy
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Hiroyuki Oya
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Tim Griffiths
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
- Biosciences Institute, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Aaron D Boes
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Nicholas Trapp
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Marcus Kaiser
- NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
- Rui Jin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jérôme Sallet
- Stem Cell and Brain Research Institute, INSERM U1208, University of Lyon, Lyon, France
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Matthew I Banks
- Department of Anesthesiology, University of Wisconsin at Madison, WI, USA
| | - Matthew A Howard
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Mario Zanaty
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Christopher I Petkov
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
- Biosciences Institute, Newcastle University Medical School, Newcastle upon Tyne, UK
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
7
|
Quinlan JA, Inglut CT, Srivastava P, Rahman I, Stabile J, Gaitan B, Arnau Del Valle C, Baumiller K, Gaur A, Chiou W, Karim B, Connolly N, Robey RW, Woodworth GF, Gottesman MM, Huang H. Carrier-Free, Amorphous Verteporfin Nanodrug for Enhanced Photodynamic Cancer Therapy and Brain Drug Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302872. [PMID: 38445882 PMCID: PMC11077681 DOI: 10.1002/advs.202302872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 02/02/2024] [Indexed: 03/07/2024]
Abstract
Glioblastoma (GBM) is hard to treat due to cellular invasion into functioning brain tissues, limited drug delivery, and evolved treatment resistance. Recurrence is nearly universal even after surgery, chemotherapy, and radiation. Photodynamic therapy (PDT) involves photosensitizer administration followed by light activation to generate reactive oxygen species at tumor sites, thereby killing cells or inducing biological changes. PDT can ablate unresectable GBM and sensitize tumors to chemotherapy. Verteporfin (VP) is a promising photosensitizer that relies on liposomal carriers for clinical use. While lipids increase VP's solubility, they also reduce intracellular photosensitizer accumulation. Here, a pure-drug nanoformulation of VP, termed "NanoVP", eliminating the need for lipids, excipients, or stabilizers is reported. NanoVP has a tunable size (65-150 nm) and 1500-fold higher photosensitizer loading capacity than liposomal VP. NanoVP shows a 2-fold increase in photosensitizer uptake and superior PDT efficacy in GBM cells compared to liposomal VP. In mouse models, NanoVP-PDT improved tumor control and extended animal survival, outperforming liposomal VP and 5-aminolevulinic acid (5-ALA). Moreover, low-dose NanoVP-PDT can safely open the blood-brain barrier, increasing drug accumulation in rat brains by 5.5-fold compared to 5-ALA. NanoVP is a new photosensitizer formulation that has the potential to facilitate PDT for the treatment of GBM.
Collapse
Affiliation(s)
- John A. Quinlan
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
- Laboratory of Cell BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Collin T. Inglut
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
- Laboratory of Cell BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Payal Srivastava
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Idrisa Rahman
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
- Laboratory of Cell BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Jillian Stabile
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Brandon Gaitan
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | | | - Kaylin Baumiller
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Anandita Gaur
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Wen‐An Chiou
- Advanced Imaging and Microscopy LaboratoryMaryland Nano CenterUniversity of MarylandCollege ParkMD20742USA
| | - Baktiar Karim
- Molecular Histopathology LaboratoryLeidos Biomedical Research, Inc.Frederick National Laboratory for Cancer ResearchFrederickMD21701USA
| | - Nina Connolly
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Robert W. Robey
- Laboratory of Cell BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Graeme F. Woodworth
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Department of NeurosurgeryUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Michael M. Gottesman
- Laboratory of Cell BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Huang‐Chiao Huang
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| |
Collapse
|
8
|
Mascotte-Cruz JU, Vera A, Leija L, Lopez-Salas FE, Gradzielski M, Koetz J, Gatica-García B, Rodríguez-Oviedo CP, Valenzuela-Arzeta IE, Escobedo L, Reyes-Corona D, Gutierrez-Castillo ME, Maldonado-Berny M, Espadas-Alvarez AJ, Orozco-Barrios CE, Martinez-Fong D. Focused ultrasound on the substantia nigra enables safe neurotensin-polyplex nanoparticle-mediated gene delivery to dopaminergic neurons intranasally and by blood circulation. DISCOVER NANO 2024; 19:60. [PMID: 38564106 PMCID: PMC10987469 DOI: 10.1186/s11671-024-04005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Abstract
Neurotensin-polyplex nanoparticles provide efficient gene transfection of nigral dopaminergic neurons when intracerebrally injected in preclinical trials of Parkinson's disease because they do not cross the blood-brain barrier (BBB). Therefore, this study aimed to open BBB with focused ultrasound (FUS) on the substantia nigra to attain systemic and intranasal transfections and evaluate its detrimental effect in rats. Systemically injected Evans Blue showed that a two-pulse FUS opened the nigral BBB. Accordingly, 35 μL of neurotensin-polyplex nanoparticles encompassing the green fluorescent protein plasmid (79.6 nm mean size and + 1.3 mV Zeta-potential) caused its expression in tyrosine hydroxylase(+) cells (dopaminergic neurons) of both substantiae nigrae upon delivery via internal carotid artery, retro-orbital venous sinus, or nasal mucosa 30 min after FUS. The intracarotid delivery yielded the highest transgene expression, followed by intranasal and venous administration. However, FUS caused neuroinflammation displayed by infiltrated lymphocytes (positive to cluster of differentiation 45), activated microglia (positive to ionized calcium-binding adaptor molecule 1), neurotoxic A1 astrocytes (positive to glial fibrillary acidic protein and complement component 3), and neurotrophic A2 astrocytes (positive to glial fibrillary acidic protein and S100 calcium-binding protein A10), that ended 15 days after FUS. Dopaminergic neurons and axonal projections decreased but recuperated basal values on day 15 after transfection, correlating with a decrease and recovery of locomotor behavior. In conclusion, FUS caused transient neuroinflammation and reversible neuronal affection but allowed systemic and intranasal transfection of dopaminergic neurons in both substantiae nigrae. Therefore, FUS could advance neurotensin-polyplex nanotechnology to clinical trials for Parkinson's disease.
Collapse
Affiliation(s)
- Juan U Mascotte-Cruz
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional No. 2508, San Pedro Zacatenco, 07360, Ciudad de México, México
| | - Arturo Vera
- Departamento de Ingeniería Eléctrica-Bioelectrónica, Centro de Investigación y de Estudios Avanzados, Ciudad de Mexico, México
| | - Lorenzo Leija
- Departamento de Ingeniería Eléctrica-Bioelectrónica, Centro de Investigación y de Estudios Avanzados, Ciudad de Mexico, México
| | - Francisco E Lopez-Salas
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México Instituto de Investigaciones Biomédicas, Ciudad de Mexico, México
| | - Michael Gradzielski
- Institut für Chemie, Stranski-Laboratorium für Physikalische und Theoretische Chemie, Technische Universität Berlin, Berlin, Germany
| | - Joachim Koetz
- Institut für Chemie , Universität Potsdam, Potsdam, Germany
| | - Bismark Gatica-García
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional No. 2508, San Pedro Zacatenco, 07360, Ciudad de México, México
- Nanoparticle Therapy Institute, Aguascalientes, México
| | | | - Irais E Valenzuela-Arzeta
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional No. 2508, San Pedro Zacatenco, 07360, Ciudad de México, México
| | - Lourdes Escobedo
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional No. 2508, San Pedro Zacatenco, 07360, Ciudad de México, México
| | | | - M E Gutierrez-Castillo
- Centro Interdisciplinario de Investigaciones y Estudios Sobre Medio Ambiente y Desarrollo, Departamento de Biociencias e Ingeniería, Instituto Politécnico Nacional, Ciudad de Mexico, México
| | - Minerva Maldonado-Berny
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional No. 2508, San Pedro Zacatenco, 07360, Ciudad de México, México
| | - Armando J Espadas-Alvarez
- Centro Interdisciplinario de Investigaciones y Estudios Sobre Medio Ambiente y Desarrollo, Departamento de Biociencias e Ingeniería, Instituto Politécnico Nacional, Ciudad de Mexico, México
| | - Carlos E Orozco-Barrios
- CONAHCYT - Unidad de Investigaciones Médicas en Enfermedades Neurológicas, Hospital de Especialidades "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México
| | - Daniel Martinez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional No. 2508, San Pedro Zacatenco, 07360, Ciudad de México, México.
- Nanoparticle Therapy Institute, Aguascalientes, México.
| |
Collapse
|
9
|
Durham PG, Butnariu A, Alghorazi R, Pinton G, Krishna V, Dayton PA. Current clinical investigations of focused ultrasound blood-brain barrier disruption: A review. Neurotherapeutics 2024; 21:e00352. [PMID: 38636309 PMCID: PMC11044032 DOI: 10.1016/j.neurot.2024.e00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/20/2024] Open
Abstract
The blood-brain barrier (BBB) presents a formidable challenge in delivering therapeutic agents to the central nervous system. Ultrasound-mediated BBB disruption has emerged as a promising non-invasive technique to enhance drug delivery to the brain. This manuscript reviews fundamental principles of ultrasound-based techniques and their mechanisms of action in temporarily permeabilizing the BBB. Clinical trials employing ultrasound for BBB disruption are discussed, summarizing diverse applications ranging from the treatment of neurodegenerative diseases to targeted drug delivery for brain tumors. The review also addresses safety considerations, outlining the current understanding of potential risks and mitigation strategies associated with ultrasound exposure, including real-time monitoring and assessment of treatment efficacy. Among the large number of studies, significant successes are highlighted thus providing perspective on the future direction of the field.
Collapse
Affiliation(s)
- Phillip G Durham
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, NC, USA
| | | | - Rizk Alghorazi
- School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Gianmarco Pinton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, NC, USA
| | - Vibhor Krishna
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, NC, USA; School of Medicine, University of North Carolina, Chapel Hill, NC, United States.
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, NC, USA.
| |
Collapse
|
10
|
Chamkouri H, Liu Q, Zhang Y, Chen C, Chen L. Brain photobiomodulation therapy on neurological and psychological diseases. JOURNAL OF BIOPHOTONICS 2024; 17:e202300145. [PMID: 37403428 DOI: 10.1002/jbio.202300145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/06/2023]
Abstract
Photobiomodulation (PBM) therapy is an innovative treatment for neurological and psychological conditions. Complex IV of the mitochondrial respiratory chain can be stimulated by red light, which increases ATP synthesis. In addition, the ion channels' light absorption causes the release of Ca2+, which activates transcription factors and changes gene expression. Neuronal metabolism is improved by brain PBM therapy, which also promotes synaptogenesis and neurogenesis as well as anti-inflammatory. Its depression-treating potential is attracting attention for other conditions, including Parkinson's disease and dementia. Giving enough dosage for optimum stimulation using the transcranial PBM technique is challenging because of the rapidly increasing attenuation of light transmission in tissue. Different strategies like intranasal and intracranial light delivery systems have been proposed to overcome this restriction. The most recent preclinical and clinical data on the effectiveness of brain PBM therapy are studied in this review article.
Collapse
Affiliation(s)
- Hossein Chamkouri
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, China
| | - Qi Liu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, China
| | - Yuqin Zhang
- Department of Neurology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Changchun Chen
- Department of Neurology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Lei Chen
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, China
- Intelligent manufacturing institute of HFUT, Hefei, China
| |
Collapse
|
11
|
Zhang T, Guo B, Zuo Z, Long X, Hu S, Li S, Su X, Wang Y, Liu C. Excitatory-inhibitory modulation of transcranial focus ultrasound stimulation on human motor cortex. CNS Neurosci Ther 2023; 29:3829-3841. [PMID: 37309308 PMCID: PMC10651987 DOI: 10.1111/cns.14303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/10/2023] [Accepted: 05/27/2023] [Indexed: 06/14/2023] Open
Abstract
AIMS Transcranial focus ultrasound stimulation (tFUS) is a promising non-invasive neuromodulation technology. This study aimed to evaluate the modulatory effects of tFUS on human motor cortex (M1) excitability and explore the mechanism of neurotransmitter-related intracortical circuitry and plasticity. METHODS Single pulse transcranial magnetic stimulation (TMS)-eliciting motor-evoked potentials (MEPs) were used to assessed M1 excitability in 10 subjects. Paired-pulse TMS was used to measure the effects of tFUS on GABA- and glutamate-related intracortical excitability and 1 H-MRS was used to assess the effects of repetitive tFUS on GABA and Glx (glutamine + glutamate) neurometabolic concentrations in the targeting region in nine subjects. RESULTS The etFUS significantly increased M1 excitability, decreased short interval intracortical inhibition (SICI) and long interval intracortical inhibition (LICI). The itFUS significantly suppressed M1 excitability, increased SICI, LICI, and decreased intracortical facilitation (ICF). Seven times of etFUS decreased the GABA concentration (6.32%), increased the Glx concentration (12.40%), and decreased the GABA/Glx ratio measured by MRS, while itFUS increased the GABA concentration (18.59%), decreased Glx concentration (0.35%), and significantly increased GABA/Glx ratio. CONCLUSION The findings support that tFUS with different parameters can exert excitatory and inhibitory neuromodulatory effects on the human motor cortex. We provide novel insights that tFUS change cortical excitability and plasticity by regulating excitatory-inhibition balance related to the GABAergic and glutamatergic receptor function and neurotransmitter metabolic level.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeuromodulationBeijingChina
| | - Bingqi Guo
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeuromodulationBeijingChina
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Beijing MR Center for Brain Research, Institute of BiophysicsChinese Academy of SciencesBeijingChina
- Hefei Comprehensive National Science CenterInstitute of Artificial IntelligenceHefeiChina
- Sino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijingChina
| | - Xiaojing Long
- Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Shimin Hu
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeuromodulationBeijingChina
| | - Siran Li
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Xin Su
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Yuping Wang
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeuromodulationBeijingChina
- Institute of Sleep and Consciousness Disorders, Center of Epilepsy, Beijing Institute for Brain DisordersCapital Medical UniversityBeijingChina
- Hebei Hospital of Xuanwu HospitalCapital Medical UniversityShijiazhuangChina
- Neuromedical Technology Innovation Center of Hebei ProvinceShijiazhuangChina
| | - Chunyan Liu
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeuromodulationBeijingChina
| |
Collapse
|
12
|
Martinez P, Song JJ, Garay FG, Song KH, Mufford T, Steiner J, DeSisto J, Ellens N, Serkova NJ, Green AL, Borden M. Comprehensive Assessment of Blood-Brain Barrier Opening and Sterile Inflammatory Response: Unraveling the Therapeutic Window. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563613. [PMID: 37961395 PMCID: PMC10634745 DOI: 10.1101/2023.10.23.563613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Microbubbles (MBs) combined with focused ultrasound (FUS) have emerged as a promising noninvasive technique to permeabilize the blood-brain barrier (BBB) for drug delivery to the brain. However, the safety and biological consequences of BBB opening remain incompletely understood. This study investigates the effects of varying microbubble volume doses (MVD) and ultrasound mechanical indices (MI) on BBB opening and the sterile inflammatory response (SIR) using high-resolution ultra-high field MRI-guided FUS in mouse brains. The results demonstrate that both MVD and MI significantly influence the extent of BBB opening, with higher doses and mechanical indices leading to increased permeability. Moreover, RNA sequencing reveals upregulated inflammatory pathways and immune cell infiltration after BBB opening, suggesting the presence and extent of SIR. Gene set enrichment analysis identifies 12 gene sets associated with inflammatory responses that are upregulated at higher doses of MVD or MI. A therapeutic window is established between significant BBB opening and the onset of SIR, providing operating regimes for avoiding each three classes of increasing damage from stimulation of the NFκB pathway via TNFL signaling to apoptosis. This study contributes to the optimization and standardization of BBB opening parameters for safe and effective drug delivery to the brain and sheds light on the underlying molecular mechanisms of the sterile inflammatory response. Significance Statement The significance of this study lies in its comprehensive investigation of microbubble-facilitated focused ultrasound for blood-brain barrier (BBB) opening. By systematically exploring various combinations of microbubble volume doses and ultrasound mechanical indices, the study reveals their direct impact on the extent of BBB permeability and the induction of sterile inflammatory response (SIR). The establishment of a therapeutic window between significant BBB opening and the onset of SIR provides critical insights for safe and targeted drug delivery to the brain. These findings advance our understanding of the biological consequences of BBB opening and contribute to optimizing parameters for clinical applications, thus minimizing potential health risks, and maximizing the therapeutic potential of this technique.
Collapse
|
13
|
Liu D, Munoz F, Sanatkhani S, Pouliopoulos AN, Konofagou EE, Grinband J, Ferrera VP. Alteration of functional connectivity in the cortex and major brain networks of non-human primates following focused ultrasound exposure in the dorsal striatum. Brain Stimul 2023; 16:1196-1204. [PMID: 37558125 PMCID: PMC10530553 DOI: 10.1016/j.brs.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Focused ultrasound (FUS) is a non-invasive neuromodulation technology that is being investigated for potential treatment of neurological and psychiatric disorders. FUS combined with microbubbles can temporarily open the intact blood-brain barrier (BBB) of animals and humans, and facilitate drug delivery. FUS exposure, either with or without microbubbles, has been demonstrated to alter the behavior of non-human primates (NHP), and previous studies have demonstrated the transient and long-term effects of FUS neuromodulation on functional connectivity using resting state functional MRI. The behavioral effects of FUS vary depending on whether or not it is applied in conjunction with microbubbles to open the BBB, but it is unknown whether opening the BBB affects functional connectivity differently than FUS alone. OBJECTIVE To compare the effects of applying FUS alone (FUS neuromodulation) and FUS with microbubbles (FUS-BBB opening) on changes of resting state functional connectivity in NHP. METHODS We applied 2 min FUS exposure without (neuromodulation) and with microbubbles (BBB opening) in the dorsal striatum of lightly anesthetized non-human primates, and acquired resting state functional MRI 40 min respectively after FUS exposure. The functional connectivity (FC) in the cortex and major brain networks between the two approaches were measured and compared. RESULTS When applying FUS exposure to the caudate nucleus of NHP, we found that both FUS neuromodulation can activate FC between caudate and insular cortex, while inhibiting the FC between caudate and motor cortex. FUS-BBB opening can activate FC between the caudate and medial prefrontal cortex, and within the frontotemporal network (FTN). We also found both FUS and FUS-BBB opening can significantly activate FC within the default mode network (DMN). CONCLUSION The results suggest applying FUS to a deep brain structure can alter functional connectivity in the DMN and FTN, and that FUS neuromodulation and FUS-mediated BBB opening can have different effects on patterns of functional connectivity.
Collapse
Affiliation(s)
- Dong Liu
- Department of Neuroscience, Columbia University, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, USA.
| | - Fabian Munoz
- Department of Neuroscience, Columbia University, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, USA
| | - Soroosh Sanatkhani
- Department of Neuroscience, Columbia University, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, USA
| | - Antonios N Pouliopoulos
- Department of Surgical & Interventional Engineering, School of Biomedical Engineering & Imaging Science, King's College London, UK
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, USA; Department of Radiology, Columbia University, USA
| | - Jack Grinband
- Department of Radiology, Columbia University, USA; Department of Psychiatry, Columbia University, USA
| | - Vincent P Ferrera
- Department of Neuroscience, Columbia University, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, USA; Department of Psychiatry, Columbia University, USA
| |
Collapse
|
14
|
Qiao R, Fu C, Forgham H, Javed I, Huang X, Zhu J, Whittaker AK, Davis TP. Magnetic Iron Oxide Nanoparticles for Brain Imaging and Drug Delivery. Adv Drug Deliv Rev 2023; 197:114822. [PMID: 37086918 DOI: 10.1016/j.addr.2023.114822] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/14/2023] [Accepted: 04/09/2023] [Indexed: 04/24/2023]
Abstract
Central nervous system (CNS) disorders affect as many as 1.5 billion people globally. The limited delivery of most imaging and therapeutic agents into the brain is a major challenge for treatment of CNS disorders. With the advent of nanotechnologies, controlled delivery of drugs with nanoparticles holds great promise in CNS disorders for overcoming the blood-brain barrier (BBB) and improving delivery efficacy. In recent years, magnetic iron oxide nanoparticles (MIONPs) have stood out as a promising theranostic nanoplatform for brain imaging and drug delivery as they possess unique physical properties and biodegradable characteristics. In this review, we summarize the recent advances in MIONP-based platforms as imaging and drug delivery agents for brain diseases. We firstly introduce the methods of synthesis and surface functionalization of MIONPs with emphasis on the inclusion of biocompatible polymers that allow for the addition of tailored physicochemical properties. We then discuss the recent advances in in vivo imaging and drug delivery applications using MIONPs. Finally, we present a perspective on the remaining challenges and possible future directions for MIONP-based brain delivery systems.
Collapse
Affiliation(s)
- Ruirui Qiao
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Changkui Fu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Helen Forgham
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ibrahim Javed
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xumin Huang
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jiayuan Zhu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrew K Whittaker
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Thomas P Davis
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
15
|
Wu JR, Hernandez Y, Miyasaki KF, Kwon EJ. Engineered nanomaterials that exploit blood-brain barrier dysfunction fordelivery to the brain. Adv Drug Deliv Rev 2023; 197:114820. [PMID: 37054953 DOI: 10.1016/j.addr.2023.114820] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023]
Abstract
The blood-brain barrier (BBB) is a highly regulated physical and functional boundarythat tightly controls the transport of materials between the blood and the brain. There is an increasing recognition that the BBB is dysfunctional in a wide range of neurological disorders; this dysfunction can be symptomatic of the disease but can also play a role in disease etiology. BBB dysfunction can be exploited for the delivery of therapeutic nanomaterials. Forexample, there can be a transient, physical disruption of the BBB in diseases such as brain injury and stroke, which allows temporary access of nanomaterials into the brain. Physicaldisruption of the BBB through external energy sources is now being clinically pursued toincrease therapeutic delivery into the brain. In other diseases, the BBB takes on new properties that can beleveraged by delivery carriers. For instance, neuroinflammation induces the expression ofreceptors on the BBB that can be targeted by ligand-modified nanomaterials and theendogenous homing of immune cells into the diseased brain can be hijacked for the delivery ofnanomaterials. Lastly, BBB transport pathways can be altered to increase nanomaterial transport. In this review, we will describe changes that can occur in the BBB in disease, and how these changes have been exploited by engineered nanomaterials forincreased transport into the brain.
Collapse
Affiliation(s)
- Jason R Wu
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Yazmin Hernandez
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Katelyn F Miyasaki
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Ester J Kwon
- Department of Bioengineering, University of California San Diego, La Jolla, CA; Sanford Consortium for Regenerative Medicine.
| |
Collapse
|
16
|
Pini L, Salvalaggio A, Wennberg AM, Dimakou A, Matteoli M, Corbetta M. The pollutome-connectome axis: a putative mechanism to explain pollution effects on neurodegeneration. Ageing Res Rev 2023; 86:101867. [PMID: 36720351 DOI: 10.1016/j.arr.2023.101867] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
The study of pollutant effects is extremely important to address the epochal challenges we are facing, where world populations are increasingly moving from rural to urban centers, revolutionizing our world into an urban world. These transformations will exacerbate pollution, thus highlighting the necessity to unravel its effect on human health. Epidemiological studies have reported that pollution increases the risk of neurological diseases, with growing evidence on the risk of neurodegenerative disorders. Air pollution and water pollutants are the main chemicals driving this risk. These chemicals can promote inflammation, acting in synergy with genotype vulnerability. However, the biological underpinnings of this association are unknown. In this review, we focus on the link between pollution and brain network connectivity at the macro-scale level. We provide an updated overview of epidemiological findings and studies investigating brain network changes associated with pollution exposure, and discuss the mechanistic insights of pollution-induced brain changes through neural networks. We explain, in detail, the pollutome-connectome axis that might provide the functional substrate for pollution-induced processes leading to cognitive impairment and neurodegeneration. We describe this model within the framework of two pollutants, air pollution, a widely recognized threat, and polyfluoroalkyl substances, a large class of synthetic chemicals which are currently emerging as new neurotoxic source.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy.
| | | | - Alexandra M Wennberg
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Dimakou
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy
| | - Michela Matteoli
- Neuro Center, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milano, Italy; CNR Institute of Neuroscience, Milano, Italy
| | - Maurizio Corbetta
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy
| |
Collapse
|
17
|
Won SM, Cai L, Gutruf P, Rogers JA. Wireless and battery-free technologies for neuroengineering. Nat Biomed Eng 2023; 7:405-423. [PMID: 33686282 PMCID: PMC8423863 DOI: 10.1038/s41551-021-00683-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022]
Abstract
Tethered and battery-powered devices that interface with neural tissues can restrict natural motions and prevent social interactions in animal models, thereby limiting the utility of these devices in behavioural neuroscience research. In this Review Article, we discuss recent progress in the development of miniaturized and ultralightweight devices as neuroengineering platforms that are wireless, battery-free and fully implantable, with capabilities that match or exceed those of wired or battery-powered alternatives. Such classes of advanced neural interfaces with optical, electrical or fluidic functionality can also combine recording and stimulation modalities for closed-loop applications in basic studies or in the practical treatment of abnormal physiological processes.
Collapse
Affiliation(s)
- Sang Min Won
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Le Cai
- Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ, USA
| | - Philipp Gutruf
- Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ, USA.
- Bio5 Institute and Neuroscience GIDP, University of Arizona, Tucson, AZ, USA.
- Department of Electrical and Computer Engineering, University of Arizona, Tucson, AZ, USA.
| | - John A Rogers
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, IL, USA.
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
- Department of Neurological Surgery, Northwestern University, Evanston, IL, USA.
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
18
|
Kim H, Kim J, Kim J, Oh S, Choi K, Yoon J. Magnetothermal-based non-invasive focused magnetic stimulation for functional recovery in chronic stroke treatment. Sci Rep 2023; 13:4988. [PMID: 36973390 PMCID: PMC10042827 DOI: 10.1038/s41598-023-31979-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Magnetic heat-based brain stimulation of specific lesions could promote the restoration of impaired motor function caused by chronic stroke. We delivered localized stimulation by nanoparticle-mediated heat generation within the targeted brain area via focused magnetic stimulation. The middle cerebral artery occlusion model was prepared, and functional recovery in the chronic-phase stroke rat model was demonstrated by the therapeutic application of focused magnetic stimulation. We observed a transient increase in blood-brain barrier permeability at the target site of < 4 mm and metabolic brain activation at the target lesion. After focused magnetic stimulation, the rotarod score increased by 390 ± 28% (p < 0.05) compared to the control group. Standardized uptake value in the focused magnetic stimulation group increased by 2063 ± 748% (p < 0.01) compared to the control group. Moreover, an increase by 24 ± 5% (p < 0.05) was observed in the sham group as well. Our results show that non-invasive focused magnetic stimulation can safely modulate BBB permeability and enhance neural activation for chronic-phase stroke treatment in the targeted deep brain area.
Collapse
Affiliation(s)
- Hohyeon Kim
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jihye Kim
- Department of Neurology, Chonnam National University Hospital and Medical School, 8 Hak-dong, Dong-gu, Gwangju, 501-757, South Korea
| | - Jahae Kim
- Department of Nuclear Medicines, Chonnam National University Hospital and Medical School, 8 Hak-dong, Dong-gu, Gwangju, 501-757, South Korea
| | - Seungjun Oh
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Kangho Choi
- Department of Neurology, Chonnam National University Hospital and Medical School, 8 Hak-dong, Dong-gu, Gwangju, 501-757, South Korea.
| | - Jungwon Yoon
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea.
| |
Collapse
|
19
|
Ren L, Zhai Z, Xiang Q, Zhuo K, Zhang S, Zhang Y, Jiao X, Tong S, Liu D, Sun J. Transcranial ultrasound stimulation modulates the interhemispheric balance of excitability in human motor cortex. J Neural Eng 2023; 20. [PMID: 36669203 DOI: 10.1088/1741-2552/acb50d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
Background. Low-intensity transcranial ultrasound stimulation (TUS) could induce both immediate and long-lasting neuromodulatory effects in human brains. Interhemispheric imbalance at prefrontal or motor cortices generally associates with various cognitive decline in aging and mental disorders. However, whether TUS could modulate the interhemispheric balance of excitability in human brain remains unknown.Objective. This study aims to explore whether repetitive TUS (rTUS) intervention can modulate the interhemispheric balance of excitability between bilateral motor cortex (M1) in healthy subjects.Approach. Motor evoked potentials (MEPs) at bilateral M1 were measured at 15 min and 0 min before a 15 min active or sham rTUS intervention on left M1 and at 0 min, 15 min and 30 min after the intervention, and the Chinese version of brief neurocognitive test battery (C-BCT) was conducted before and after the intervention respectively. Cortical excitability was quantified by MEPs, and the long-lasting changes of MEP amplitude was used as an index of plasticity.Results. In the active rTUS group (n= 20), the ipsilateral MEP amplitude increased significantly compared with baselines and lasted for up to 30 min after intervention, while the contralateral MEP amplitude decreased lasting for 15 min, yielding increased laterality between bilateral MEPs. Furthermore, rTUS intervention induced changes in some C-BCT scores, and the changes of scores correlated with the changes of MEP amplitudes induced by rTUS intervention. The sham rTUS group (n= 20) showed no significant changes in MEPs and C-BCT scores. In addition, no participants reported any adverse effects during and after the rTUS intervention, and no obvious temperature increase appeared in skull or brain tissues in simulation.Significance. rTUS intervention modulated the plasticity of ipsilateral M1 and the interhemispheric balance of M1 excitability in human brain, and improved cognitive performance, suggesting a considerable potential of rTUS in clinical interventions.
Collapse
Affiliation(s)
- Liyuan Ren
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200230, People's Republic of China
| | - Zhaolin Zhai
- First-episode Schizophrenia and Early Psychosis Program, Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China
| | - Qiong Xiang
- First-episode Schizophrenia and Early Psychosis Program, Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China
| | - Kaiming Zhuo
- First-episode Schizophrenia and Early Psychosis Program, Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China
| | - Suzhen Zhang
- First-episode Schizophrenia and Early Psychosis Program, Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China.,Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Yi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200230, People's Republic of China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China
| | - Xiong Jiao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200230, People's Republic of China
| | - Shanbao Tong
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200230, People's Republic of China.,Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai 200230, People's Republic of China
| | - Dengtang Liu
- First-episode Schizophrenia and Early Psychosis Program, Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China.,Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China.,Institute of Mental Health, Fudan University, Shanghai 200030, People's Republic of China
| | - Junfeng Sun
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200230, People's Republic of China
| |
Collapse
|
20
|
Gorick CM, Breza VR, Nowak KM, Cheng VWT, Fisher DG, Debski AC, Hoch MR, Demir ZEF, Tran NM, Schwartz MR, Sheybani ND, Price RJ. Applications of focused ultrasound-mediated blood-brain barrier opening. Adv Drug Deliv Rev 2022; 191:114583. [PMID: 36272635 PMCID: PMC9712235 DOI: 10.1016/j.addr.2022.114583] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/01/2022] [Accepted: 10/14/2022] [Indexed: 01/25/2023]
Abstract
The blood brain barrier (BBB) plays a critically important role in the regulation of central nervous system (CNS) homeostasis, but also represents a major limitation to treatments of brain pathologies. In recent years, focused ultrasound (FUS) in conjunction with gas-filled microbubble contrast agents has emerged as a powerful tool for transiently and non-invasively disrupting the BBB in a targeted and image-guided manner, allowing for localized delivery of drugs, genes, or other therapeutic agents. Beyond the delivery of known therapeutics, FUS-mediated BBB opening also demonstrates the potential for use in neuromodulation and the stimulation of a range of cell- and tissue-level physiological responses that may prove beneficial in disease contexts. Clinical trials investigating the safety and efficacy of FUS-mediated BBB opening are well underway, and offer promising non-surgical approaches to treatment of devastating pathologies. This article reviews a range of pre-clinical and clinical studies demonstrating the tremendous potential of FUS to fundamentally change the paradigm of treatment for CNS diseases.
Collapse
Affiliation(s)
- Catherine M Gorick
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Victoria R Breza
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Katherine M Nowak
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Vinton W T Cheng
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Delaney G Fisher
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Anna C Debski
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Matthew R Hoch
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Zehra E F Demir
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Nghi M Tran
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Mark R Schwartz
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Natasha D Sheybani
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
21
|
Collins MN, Mesce KA. A review of the bioeffects of low-intensity focused ultrasound and the benefits of a cellular approach. Front Physiol 2022; 13:1047324. [PMID: 36439246 PMCID: PMC9685663 DOI: 10.3389/fphys.2022.1047324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 10/25/2022] [Indexed: 10/28/2023] Open
Abstract
This review article highlights the historical developments and current state of knowledge of an important neuromodulation technology: low-intensity focused ultrasound. Because compelling studies have shown that focused ultrasound can modulate neuronal activity non-invasively, especially in deep brain structures with high spatial specificity, there has been a renewed interest in attempting to understand the specific bioeffects of focused ultrasound at the cellular level. Such information is needed to facilitate the safe and effective use of focused ultrasound to treat a number of brain and nervous system disorders in humans. Unfortunately, to date, there appears to be no singular biological mechanism to account for the actions of focused ultrasound, and it is becoming increasingly clear that different types of nerve cells will respond to focused ultrasound differentially based on the complement of their ion channels, other membrane biophysical properties, and arrangement of synaptic connections. Furthermore, neurons are apparently not equally susceptible to the mechanical, thermal and cavitation-related consequences of focused ultrasound application-to complicate matters further, many studies often use distinctly different focused ultrasound stimulus parameters to achieve a reliable response in neural activity. In this review, we consider the benefits of studying more experimentally tractable invertebrate preparations, with an emphasis on the medicinal leech, where neurons can be studied as unique individual cells and be synaptically isolated from the indirect effects of focused ultrasound stimulation on mechanosensitive afferents. In the leech, we have concluded that heat is the primary effector of focused ultrasound neuromodulation, especially on motoneurons in which we observed a focused ultrasound-mediated blockade of action potentials. We discuss that the mechanical bioeffects of focused ultrasound, which are frequently described in the literature, are less reliably achieved as compared to thermal ones, and that observations ascribed to mechanical responses may be confounded by activation of synaptically-coupled sensory structures or artifacts associated with electrode resonance. Ultimately, both the mechanical and thermal components of focused ultrasound have significant potential to contribute to the sculpting of specific neural outcomes. Because focused ultrasound can generate significant modulation at a temperature <5°C, which is believed to be safe for moderate durations, we support the idea that focused ultrasound should be considered as a thermal neuromodulation technology for clinical use, especially targeting neural pathways in the peripheral nervous system.
Collapse
Affiliation(s)
- Morgan N. Collins
- Graduate Program in Neuroscience, University of Minnesota, Saint Paul, MN, United States
| | - Karen A. Mesce
- Department of Entomology and Graduate Program in Neuroscience, University of Minnesota, Saint Paul, MN, United States
| |
Collapse
|
22
|
Chen H, Felix C, Folloni D, Verhagen L, Sallet J, Jerusalem A. Modelling transcranial ultrasound neuromodulation: an energy-based multiscale framework. Acta Biomater 2022; 151:317-332. [PMID: 35902037 DOI: 10.1016/j.actbio.2022.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/26/2022]
Abstract
Several animal and human studies have now established the potential of low intensity, low frequency transcranial ultrasound (TUS) for non-invasive neuromodulation. Paradoxically, the underlying mechanisms through which TUS neuromodulation operates are still unclear, and a consensus on the identification of optimal sonication parameters still remains elusive. One emerging hypothesis based on thermodynamical considerations attributes the acoustic-induced nerve activity alterations to the mechanical energy and/or entropy conversions occurring during TUS action. Here, we propose a multiscale modelling framework to examine the energy states of neuromodulation under TUS. First, macroscopic tissue-level acoustic simulations of the sonication of a whole monkey brain are conducted under different sonication protocols. For each one of them, mechanical loading conditions of the received waves in the anterior cingulate cortex region are recorded and exported into a microscopic cell-level 3D viscoelastic finite element model of neuronal axon embedded extracellular medium. Pulse-averaged elastically stored and viscously dissipated energy rate densities during axon deformation are finally computed under different sonication incident angles and are mapped against distinct combinations of sonication parameters of the TUS. The proposed multiscale framework allows for the analysis of vibrational patterns of the axons and its comparison against the spectrograms of stimulating ultrasound. The results are in agreement with literature data on neuromodulation, demonstrating the potential of this framework to identify optimised acoustic parameters in TUS neuromodulation. The proposed approach is finally discussed in the context of multiphysics energetic considerations, argued here to be a promising avenue towards a scalable framework for TUS in silico predictions. STATEMENT OF SIGNIFICANCE: Low-intensity transcranial ultrasound (TUS) is poised to become a leading neuromodulation technique for the treatment of neurological disorders. Paradoxically, how it operates at the cellular scale remains unknown, hampering progress in personalised treatment. To this end, models of the multiphysics of neurons able to upscale results to the organ scale are required. We propose here to achieve this by considering an axon submitted to an ultrasound wave extracted from a simulation at the organ scale. Doing so, information pertaining to both stored and dissipated axonal energies can be extracted for a given head/brain morphology. This two-scale multiphysics energetic approach is a promising scalable framework for in silico predictions in the context of personalised TUS treatment.
Collapse
Affiliation(s)
- Haoyu Chen
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Ciara Felix
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Davide Folloni
- Wellcome Centre for Integrative Neuroimaging (WIN), Department of Experimental Psychology, University of Oxford, Oxford, UK; Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lennart Verhagen
- Wellcome Centre for Integrative Neuroimaging (WIN), Department of Experimental Psychology, University of Oxford, Oxford, UK; Donders Institute, Radboud University, Nijmegen, Netherlands
| | - Jérôme Sallet
- Wellcome Centre for Integrative Neuroimaging (WIN), Department of Experimental Psychology, University of Oxford, Oxford, UK; Inserm, Stem Cell and Brain Research Institute, Université Lyon 1, Bron, France
| | - Antoine Jerusalem
- Department of Engineering Science, University of Oxford, Oxford, UK.
| |
Collapse
|
23
|
Dell'Italia J, Sanguinetti JL, Monti MM, Bystritsky A, Reggente N. Current State of Potential Mechanisms Supporting Low Intensity Focused Ultrasound for Neuromodulation. Front Hum Neurosci 2022; 16:872639. [PMID: 35547195 PMCID: PMC9081930 DOI: 10.3389/fnhum.2022.872639] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/28/2022] [Indexed: 01/07/2023] Open
Abstract
Low intensity focused ultrasound (LIFU) has been gaining traction as a non-invasive neuromodulation technology due to its superior spatial specificity relative to transcranial electrical/magnetic stimulation. Despite a growing literature of LIFU-induced behavioral modifications, the mechanisms of action supporting LIFU's parameter-dependent excitatory and suppressive effects are not fully understood. This review provides a comprehensive introduction to the underlying mechanics of both acoustic energy and neuronal membranes, defining the primary variables for a subsequent review of the field's proposed mechanisms supporting LIFU's neuromodulatory effects. An exhaustive review of the empirical literature was also conducted and studies were grouped based on the sonication parameters used and behavioral effects observed, with the goal of linking empirical findings to the proposed theoretical mechanisms and evaluating which model best fits the existing data. A neuronal intramembrane cavitation excitation model, which accounts for differential effects as a function of cell-type, emerged as a possible explanation for the range of excitatory effects found in the literature. The suppressive and other findings need additional theoretical mechanisms and these theoretical mechanisms need to have established relationships to sonication parameters.
Collapse
Affiliation(s)
- John Dell'Italia
- Institute for Advanced Consciousness Studies, Santa Monica, CA, United States
- *Correspondence: John Dell'Italia
| | - Joseph L. Sanguinetti
- Department of Psychology, University of Arizona, Tuscon, AZ, United States
- Department of Psychology, University of New Mexico, Albuquerque, NM, United States
| | - Martin M. Monti
- Institute for Advanced Consciousness Studies, Santa Monica, CA, United States
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Alexander Bystritsky
- Institute for Advanced Consciousness Studies, Santa Monica, CA, United States
- Tiny Blue Dot Foundation, Santa Monica, CA, United States
| | - Nicco Reggente
- Institute for Advanced Consciousness Studies, Santa Monica, CA, United States
- Tiny Blue Dot Foundation, Santa Monica, CA, United States
| |
Collapse
|
24
|
Lechpammer M, Rao R, Shah S, Mirheydari M, Bhattacharya D, Koehler A, Toukam DK, Haworth KJ, Pomeranz Krummel D, Sengupta S. Advances in Immunotherapy for the Treatment of Adult Glioblastoma: Overcoming Chemical and Physical Barriers. Cancers (Basel) 2022; 14:1627. [PMID: 35406398 PMCID: PMC8997081 DOI: 10.3390/cancers14071627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma, or glioblastoma multiforme (GBM, WHO Grade IV), is a highly aggressive adult glioma. Despite extensive efforts to improve treatment, the current standard-of-care (SOC) regimen, which consists of maximal resection, radiotherapy, and temozolomide (TMZ), achieves only a 12-15 month survival. The clinical improvements achieved through immunotherapy in several extracranial solid tumors, including non-small-cell lung cancer, melanoma, and non-Hodgkin lymphoma, inspired investigations to pursue various immunotherapeutic interventions in adult glioblastoma patients. Despite some encouraging reports from preclinical and early-stage clinical trials, none of the tested agents have been convincing in Phase III clinical trials. One, but not the only, factor that is accountable for the slow progress is the blood-brain barrier, which prevents most antitumor drugs from reaching the target in appreciable amounts. Herein, we review the current state of immunotherapy in glioblastoma and discuss the significant challenges that prevent advancement. We also provide thoughts on steps that may be taken to remediate these challenges, including the application of ultrasound technologies.
Collapse
Affiliation(s)
- Mirna Lechpammer
- Foundation Medicine, Inc., Cambridge, MA 02141, USA;
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Rohan Rao
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Sanjit Shah
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Mona Mirheydari
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (M.M.); (K.J.H.)
| | - Debanjan Bhattacharya
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Abigail Koehler
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Donatien Kamdem Toukam
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Kevin J. Haworth
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (M.M.); (K.J.H.)
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| |
Collapse
|
25
|
Liu X, Qiu F, Hou L, Wang X. Review of Noninvasive or Minimally Invasive Deep Brain Stimulation. Front Behav Neurosci 2022; 15:820017. [PMID: 35145384 PMCID: PMC8823253 DOI: 10.3389/fnbeh.2021.820017] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
Brain stimulation is a critical technique in neuroscience research and clinical application. Traditional transcranial brain stimulation techniques, such as transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and deep brain stimulation (DBS) have been widely investigated in neuroscience for decades. However, TMS and tDCS have poor spatial resolution and penetration depth, and DBS requires electrode implantation in deep brain structures. These disadvantages have limited the clinical applications of these techniques. Owing to developments in science and technology, substantial advances in noninvasive and precise deep stimulation have been achieved by neuromodulation studies. Second-generation brain stimulation techniques that mainly rely on acoustic, electronic, optical, and magnetic signals, such as focused ultrasound, temporal interference, near-infrared optogenetic, and nanomaterial-enabled magnetic stimulation, offer great prospects for neuromodulation. This review summarized the mechanisms, development, applications, and strengths of these techniques and the prospects and challenges in their development. We believe that these second-generation brain stimulation techniques pave the way for brain disorder therapy.
Collapse
Affiliation(s)
- Xiaodong Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Fang Qiu
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Lijuan Hou
- College of Physical Education and Sports, Beijing Normal University, Beijing, China
- *Correspondence: Lijuan Hou Xiaohui Wang
| | - Xiaohui Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- *Correspondence: Lijuan Hou Xiaohui Wang
| |
Collapse
|
26
|
Chen S, Nazeri A, Baek H, Ye D, Yang Y, Yuan J, Rubin JB, Chen H. A review of bioeffects induced by focused ultrasound combined with microbubbles on the neurovascular unit. J Cereb Blood Flow Metab 2022; 42:3-26. [PMID: 34551608 PMCID: PMC8721781 DOI: 10.1177/0271678x211046129] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 01/29/2023]
Abstract
Focused ultrasound combined with circulating microbubbles (FUS+MB) can transiently enhance blood-brain barrier (BBB) permeability at targeted brain locations. Its great promise in improving drug delivery to the brain is reflected by a rapidly growing number of clinical trials using FUS+MB to treat various brain diseases. As the clinical applications of FUS+MB continue to expand, it is critical to have a better understanding of the molecular and cellular effects induced by FUS+MB to enhance the efficacy of current treatment and enable the discovery of new therapeutic strategies. Existing studies primarily focus on FUS+MB-induced effects on brain endothelial cells, the major cellular component of BBB. However, bioeffects induced by FUS+MB expand beyond the BBB to cells surrounding blood vessels, including astrocytes, microglia, and neurons. Together these cell types comprise the neurovascular unit (NVU). In this review, we examine cell-type-specific bioeffects of FUS+MB on different NVU components, including enhanced permeability in endothelial cells, activation of astrocytes and microglia, as well as increased intraneuron protein metabolism and neuronal activity. Finally, we discuss knowledge gaps that must be addressed to further advance clinical applications of FUS+MB.
Collapse
Affiliation(s)
- Si Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Arash Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hongchae Baek
- Imaging Institute and Neurological Institute, Cleveland Clinic, Cleveland Clinic, Cleveland, OH, USA
| | - Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Jinyun Yuan
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
27
|
Transcranial ultrasound stimulation of the human motor cortex. iScience 2021; 24:103429. [PMID: 34901788 PMCID: PMC8637484 DOI: 10.1016/j.isci.2021.103429] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/04/2021] [Accepted: 11/09/2021] [Indexed: 12/22/2022] Open
Abstract
It has been 40 years since the report of long-term synaptic plasticity on the rodent brain. Transcranial ultrasound stimulation (TUS) shows advantages in spatial resolution and penetration depth when compared with electrical or magnetic stimulation. The repetitive TUS (rTUS) can induce cortical excitability alteration on animals, and persistent aftereffects were observed. However, the effects of rTUS on synaptic plasticity in humans remain unelucidated. In the current study, we applied a 15-min rTUS protocol to stimulate left primary motor cortex (l-M1) in 24 male healthy participants. The single-pulsed transcranial magnetic stimulation-evoked motor evoked potential and Stop-signal task was applied to measure the rTUS aftereffects. Here, we report that conditioning the human motor cortex using rTUS may produce long-lasting and statistically significant effects on motor cortex excitability as well as motor behavior, without harmful side effects observed. These findings suggest a considerable potential of rTUS in cortical plasticity modulation and clinical intervention for impulsivity-related disorders.
Collapse
|
28
|
Monteiro F, Sotiropoulos I, Carvalho Ó, Sousa N, Silva FS. Multi-mechanical waves against Alzheimer's disease pathology: a systematic review. Transl Neurodegener 2021; 10:36. [PMID: 34560902 PMCID: PMC8464104 DOI: 10.1186/s40035-021-00256-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/03/2021] [Indexed: 01/18/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia, affecting approximately 40 million people worldwide. The ineffectiveness of the available pharmacological treatments against AD has fostered researchers to focus on alternative strategies to overcome this challenge. Mechanical vibrations delivered in different stimulation modes have been associated with marked improvements in cognitive and physical performance in both demented and non-demented elderly. Some of the mechanical-based stimulation modalities in efforts are earlier whole-body vibration, transcranial ultrasound stimulation with microbubble injection, and more recently, auditory stimulation. However, there is a huge variety of treatment specifications, and in many cases, conflicting results are reported. In this review, a search on Scopus, PubMed, and Web of Science databases was performed, resulting in 37 papers . These studies suggest that mechanical vibrations delivered through different stimulation modes are effective in attenuating many parameters of AD pathology including functional connectivity and neuronal circuit integrity deficits in the brains of AD patients, as well as in subjects with cognitive decline and non-demented older adults. Despite the evolving preclinical and clinical evidence on these therapeutic modalities, their translation into clinical practice is not consolidated yet. Thus, this comprehensive and critical systematic review aims to address the most important gaps in the reviewed protocols and propose optimal regimens for future clinical application.
Collapse
Affiliation(s)
- Francisca Monteiro
- Center for Microelectromechanical Systems (CMEMS), University of Minho, Campus Azurém, 4800-058, Guimarães, Portugal. .,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Institute of Biosciences & Applications NCSR "Demokritos", Athens, Greece
| | - Óscar Carvalho
- Center for Microelectromechanical Systems (CMEMS), University of Minho, Campus Azurém, 4800-058, Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Filipe S Silva
- Center for Microelectromechanical Systems (CMEMS), University of Minho, Campus Azurém, 4800-058, Guimarães, Portugal
| |
Collapse
|
29
|
Ho YJ, Huang CC, Fan CH, Liu HL, Yeh CK. Ultrasonic technologies in imaging and drug delivery. Cell Mol Life Sci 2021; 78:6119-6141. [PMID: 34297166 PMCID: PMC11072106 DOI: 10.1007/s00018-021-03904-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Ultrasonic technologies show great promise for diagnostic imaging and drug delivery in theranostic applications. The development of functional and molecular ultrasound imaging is based on the technical breakthrough of high frame-rate ultrasound. The evolution of shear wave elastography, high-frequency ultrasound imaging, ultrasound contrast imaging, and super-resolution blood flow imaging are described in this review. Recently, the therapeutic potential of the interaction of ultrasound with microbubble cavitation or droplet vaporization has become recognized. Microbubbles and phase-change droplets not only provide effective contrast media, but also show great therapeutic potential. Interaction with ultrasound induces unique and distinguishable biophysical features in microbubbles and droplets that promote drug loading and delivery. In particular, this approach demonstrates potential for central nervous system applications. Here, we systemically review the technological developments of theranostic ultrasound including novel ultrasound imaging techniques, the synergetic use of ultrasound with microbubbles and droplets, and microbubble/droplet drug-loading strategies for anticancer applications and disease modulation. These advancements have transformed ultrasound from a purely diagnostic utility into a promising theranostic tool.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Chung Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
30
|
Tretbar SH, Fournelle M, Speicher D, Becker FJ, Anastasiadis P, Landgraf L, Roy U, Melzer A. A novel matrix-array-based MR-conditional ultrasound system for local hyperthermia of small animals. IEEE Trans Biomed Eng 2021; 69:758-770. [PMID: 34398748 DOI: 10.1109/tbme.2021.3104865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE The goal of this work was to develop a novel modular focused ultrasound hyperthermia (FUS-HT) system for preclinical applications with the following characteristics: MR-compatible, compact probe for integration into a PET/MR small animal scanner, 3D-beam steering capabilities, high resolution focusing for generation of spatially confined FUS-HT effects. METHODS For 3D-beam steering capabilities, a matrix array approach with 11 11 elements was chosen. For reaching the required level of integration, the array was mounted with a conductive backing directly on the interconnection PCB. The array is driven by a modified version of our 128 channel ultrasound research platform DiPhAS. The system was characterized using sound field measurements and validated using tissue-mimicking phantoms. Preliminary MR-compatibility tests were performed using a 7T Bruker MRI scanner. RESULTS Four 11 11 arrays between 0.5 and 2 MHz were developed and characterized with respect to sound field properties and HT generation. Focus sizes between 1 and 4 mm were reached depending on depth and frequency. We showed heating by 4C within 60 s in phantoms. The integration concept allows a probe thickness of less than 12 mm. CONCLUSION We demonstrated FUS-HT capabilities of our modular system based on matrix arrays and a 128 channel electronics system within a 3D-steering range of up to 30. The suitability for integration into a small animal MR could be demonstrated in basic MR-compatibility tests. SIGNIFICANCE The developed system presents a new generation of FUS-HT for preclinical and translational work providing safe, reversible, localized, and controlled HT.
Collapse
|
31
|
Han M, Seo H, Choi H, Lee EH, Park J. Localized Modification of Water Molecule Transport After Focused Ultrasound-Induced Blood-Brain Barrier Disruption in Rat Brain. Front Neurosci 2021; 15:685977. [PMID: 34393708 PMCID: PMC8358078 DOI: 10.3389/fnins.2021.685977] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022] Open
Abstract
Interstitial solutes can be removed by various overlapping clearance systems, including blood-brain barrier (BBB) transport and glymphatic clearance. Recently, focused ultrasound (FUS)-induced BBB disruption (BBBD) has been applied to visualize glymphatic transport. Despite evidence that FUS-BBBD might facilitate glymphatic transport, the nature of fluid movement within the sonication region is yet to be determined. In this study, we sought to determine whether FUS-BBBD may facilitate the local movement of water molecules. Two different FUS conditions (0.60-0.65 MPa and 0.75-0.80 MPa) were used to induce BBBD in the caudate-putamen and thalamus regions of healthy Sprague-Dawley rats. The water diffusion caused by FUS-BBBD was analyzed using the apparent diffusion coefficient (ADC), axial diffusivity, radial diffusivity (RD), and fractional anisotropy, obtained at 5 min, 24 and 48 h, as well as the water channel expression of aquaporin-4 (AQP-4) immunostaining at 48 h after FUS-induced BBBD. In addition, hematoxylin and eosin histopathology and Fluoro-Jade C (FJC) immunostaining were performed to analyze brain damage. The signal changes in ADC and RD in the sonication groups showed significant and transient reduction at 5 min, with subsequent increases at 24 and 48 h after FUS-induced BBBD. When we applied higher sonication conditions, the ADC and RD showed enhancement until 48 h, and became comparable to contralateral values at 72 h. AQP-4 expression was upregulated after FUS-induced BBBD in both sonication conditions at 48 h. The results of this study provide preliminary evidence on how mechanical forces from FUS alter water dynamics through diffusion tensor imaging (DTI) measures and AQP4 expression.
Collapse
Affiliation(s)
- Mun Han
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Hyeon Seo
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Hyojin Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Eun-Hee Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Juyoung Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| |
Collapse
|
32
|
Pouliopoulos AN, Kwon N, Jensen G, Meaney A, Niimi Y, Burgess MT, Ji R, McLuckie AJ, Munoz FA, Kamimura HAS, Teich AF, Ferrera VP, Konofagou EE. Safety evaluation of a clinical focused ultrasound system for neuronavigation guided blood-brain barrier opening in non-human primates. Sci Rep 2021; 11:15043. [PMID: 34294761 PMCID: PMC8298475 DOI: 10.1038/s41598-021-94188-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
An emerging approach with potential in improving the treatment of neurodegenerative diseases and brain tumors is the use of focused ultrasound (FUS) to bypass the blood-brain barrier (BBB) in a non-invasive and localized manner. A large body of pre-clinical work has paved the way for the gradual clinical implementation of FUS-induced BBB opening. Even though the safety profile of FUS treatments in rodents has been extensively studied, the histological and behavioral effects of clinically relevant BBB opening in large animals are relatively understudied. Here, we examine the histological and behavioral safety profile following localized BBB opening in non-human primates (NHPs), using a neuronavigation-guided clinical system prototype. We show that FUS treatment triggers a short-lived immune response within the targeted region without exacerbating the touch accuracy or reaction time in visual-motor cognitive tasks. Our experiments were designed using a multiple-case-study approach, in order to maximize the acquired data and support translation of the FUS system into human studies. Four NHPs underwent a single session of FUS-mediated BBB opening in the prefrontal cortex. Two NHPs were treated bilaterally at different pressures, sacrificed on day 2 and 18 post-FUS, respectively, and their brains were histologically processed. In separate experiments, two NHPs that were earlier trained in a behavioral task were exposed to FUS unilaterally, and their performance was tracked for at least 3 weeks after BBB opening. An increased microglia density around blood vessels was detected on day 2, but was resolved by day 18. We also detected signs of enhanced immature neuron presence within areas that underwent BBB opening, compared to regions with an intact BBB, confirming previous rodent studies. Logistic regression analysis showed that the NHP cognitive performance did not deteriorate following BBB opening. These preliminary results demonstrate that neuronavigation-guided FUS with a single-element transducer is a non-invasive method capable of reversibly opening the BBB, without substantial histological or behavioral impact in an animal model closely resembling humans. Future work should confirm the observations of this multiple-case-study work across animals, species and tasks.
Collapse
Affiliation(s)
- Antonios N. Pouliopoulos
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Nancy Kwon
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Greg Jensen
- grid.21729.3f0000000419368729Department of Neuroscience, Columbia University, New York City, NY 10032 USA
| | - Anna Meaney
- grid.21729.3f0000000419368729Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY 10027 USA
| | - Yusuke Niimi
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Mark T. Burgess
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Robin Ji
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Alicia J. McLuckie
- grid.21729.3f0000000419368729Institute of Comparative Medicine, Columbia University, New York City, NY 10032 USA
| | - Fabian A. Munoz
- grid.21729.3f0000000419368729Department of Neuroscience, Columbia University, New York City, NY 10032 USA ,grid.21729.3f0000000419368729Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY 10027 USA
| | - Hermes A. S. Kamimura
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Andrew F. Teich
- grid.21729.3f0000000419368729Department of Pathology and Cell Biology, Columbia University, New York City, NY 10032 USA
| | - Vincent P. Ferrera
- grid.21729.3f0000000419368729Department of Neuroscience, Columbia University, New York City, NY 10032 USA ,grid.21729.3f0000000419368729Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY 10027 USA ,grid.21729.3f0000000419368729Department of Psychiatry, Columbia University, New York City, NY
10032
USA
| | - Elisa E. Konofagou
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA ,grid.21729.3f0000000419368729Department of Radiology, Columbia University, New York City, NY 10032 USA
| |
Collapse
|
33
|
Chen KT, Wei KC, Liu HL. Focused Ultrasound Combined with Microbubbles in Central Nervous System Applications. Pharmaceutics 2021; 13:pharmaceutics13071084. [PMID: 34371774 PMCID: PMC8308978 DOI: 10.3390/pharmaceutics13071084] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/20/2022] Open
Abstract
The blood–brain barrier (BBB) protects the central nervous system (CNS) from invasive pathogens and maintains the homeostasis of the brain. Penetrating the BBB has been a major challenge in the delivery of therapeutic agents for treating CNS diseases. Through a physical acoustic cavitation effect, focused ultrasound (FUS) combined with microbubbles achieves the local detachment of tight junctions of capillary endothelial cells without inducing neuronal damage. The bioavailability of therapeutic agents is increased only in the area targeted by FUS energy. FUS with circulating microbubbles is currently the only method for inducing precise, transient, reversible, and noninvasive BBB opening (BBBO). Over the past decade, FUS-induced BBBO (FUS-BBBO) has been preclinically confirmed to not only enhance the penetration of therapeutic agents in the CNS, but also modulate focal immunity and neuronal activity. Several recent clinical human trials have demonstrated both the feasibility and potential advantages of using FUS-BBBO in diseased patients. The promising results support adding FUS-BBBO as a multimodal therapeutic strategy in modern CNS disease management. This review article explores this technology by describing its physical mechanisms and the preclinical findings, including biological effects, therapeutic concepts, and translational design of human medical devices, and summarizes completed and ongoing clinical trials.
Collapse
Affiliation(s)
- Ko-Ting Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan;
- Ph.D. Program in Biomedical Engineering, Chang Gung University, Guishan, Taoyuan 333, Taiwan
- Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan;
- Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, TuCheng, New Taipei 236, Taiwan
- School of Medicine, Chang Gung University, Guishan, Taoyuan 333, Taiwan
- Correspondence: (K.-C.W.); (H.-L.L.)
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Da’an, Taipei 106, Taiwan
- Department of Biomedical Engineering, National Taiwan University, Da’an, Taipei 106, Taiwan
- Correspondence: (K.-C.W.); (H.-L.L.)
| |
Collapse
|
34
|
Inglut CT, Gray KM, Vig S, Jung JW, Stabile J, Zhang Y, Stroka KM, Huang HC. Photodynamic Priming Modulates Endothelial Cell-Cell Junction Phenotype for Light-activated Remote Control of Drug Delivery. IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS : A PUBLICATION OF THE IEEE LASERS AND ELECTRO-OPTICS SOCIETY 2021; 27:7200311. [PMID: 33519171 PMCID: PMC7839980 DOI: 10.1109/jstqe.2020.3024014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The blood-brain barrier (BBB) remains a major obstacle for drug delivery to the central nervous system. In particular, the tight and adherens junctions that join the brain capillary endothelial cells limit the diffusion of various molecules from the bloodstream into the brain. Photodynamic priming (PDP) is a non-cytotoxic modality that involves light activation of photosensitizers to photochemically modulate nearby molecules without killing the cells. Here we investigate the effects of sub-lethal photochemistry on junction phenotype (i.e., continuous, punctate, or perpendicular), as well as the BBB permeability in a transwell model of human brain microvascular endothelial cells (HBMECs). We showed that PDP decreases the continuous junction architecture by ~20%, increases the perpendicular junction architecture by ~40%, and has minimal impact on cell morphology in HBMECs. Furthermore, transwell permeability assay revealed that PDP improves the HBMEC permeability to dextran or nanoliposomes by up to 30-fold for 6-9 days. These results suggest that PDP could safely reverse the mature brain endothelial junctions without killing the HBMECs. This study not only emphasizes the critical roles of PDP in the modulation junction phenotype, but also highlights the opportunity to further develop PDP-based combinations that opens the cerebrum endothelium for enhanced drug transporter across the BBB.
Collapse
Affiliation(s)
- Collin T Inglut
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Kelsey M Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Shruti Vig
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Jae W Jung
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Jillian Stabile
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Yuji Zhang
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
35
|
Hwang BY, Mampre D, Ahmed AK, Suk I, Anderson WS, Manbachi A, Theodore N. Ultrasound in Traumatic Spinal Cord Injury: A Wide-Open Field. Neurosurgery 2021; 89:372-382. [PMID: 34098572 DOI: 10.1093/neuros/nyab177] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/19/2021] [Indexed: 02/02/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a common and devastating condition. In the absence of effective validated therapies, there is an urgent need for novel methods to achieve injury stabilization, regeneration, and functional restoration in SCI patients. Ultrasound is a versatile platform technology that can provide a foundation for viable diagnostic and therapeutic interventions in SCI. In particular, real-time perfusion and inflammatory biomarker monitoring, focal pharmaceutical delivery, and neuromodulation are capabilities that can be harnessed to advance our knowledge of SCI pathophysiology and to develop novel management and treatment options. Our review suggests that studies that evaluate the benefits and risks of ultrasound in SCI are severely lacking and our understanding of the technology's potential impact remains poorly understood. Although the complex anatomy and physiology of the spine and the spinal cord remain significant challenges, continued technological advances will help the field overcome the current barriers and bring ultrasound to the forefront of SCI research and development.
Collapse
Affiliation(s)
- Brian Y Hwang
- Division of Functional Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David Mampre
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - A Karim Ahmed
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ian Suk
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William S Anderson
- Division of Functional Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Manbachi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Ho YJ, Chang HC, Lin CW, Fan CH, Lin YC, Wei KC, Yeh CK. Oscillatory behavior of microbubbles impacts efficacy of cellular drug delivery. J Control Release 2021; 333:316-327. [PMID: 33811982 DOI: 10.1016/j.jconrel.2021.03.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 01/16/2023]
Abstract
Drug-loaded microbubbles have been proven to be an effective strategy for non-invasive and local drug delivery when combined with ultrasound excitation for targeted drug release. Inertial cavitation is speculated to be a major mechanism for releasing drugs from drug-loaded microbubbles, but it results in lethal cellular pore damage that greatly limits its application. Thus, we investigated the cellular vesicle attachment and uptake to evaluate the efficiency of drug delivery by modulating the behaviors of targeted microbubble oscillation. The efficiency of vesicle attachment on the targeted cell membrane was 36.5 ± 15.9% and 3.8 ± 2.3% under stable and inertial cavitation, respectively. Further, stable cavitation enhanced cell permeability (26.8 ± 3.2%), maintained cell viability (90.8 ± 2.1%), and showed 7.9 ± 1.9-fold enhancement of in vivo vesicle release on tumor vessels. Therefore, our results reveal the ability to improve drug delivery via stable cavitation induced by targeted microbubbles. We propose that this strategy might be suitable for tissue repair or neuromodulation.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ho-Chun Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan; Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan.
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, New Taipei City, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
37
|
Lea-Banks H, Meng Y, Wu SK, Belhadjhamida R, Hamani C, Hynynen K. Ultrasound-sensitive nanodroplets achieve targeted neuromodulation. J Control Release 2021; 332:30-39. [PMID: 33600879 DOI: 10.1016/j.jconrel.2021.02.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/20/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022]
Abstract
Focused ultrasound (FUS) offers an attractive tool for non-invasive neuromodulation, addressing a clinical need to develop more minimally invasive approaches that are safer, more tolerable and versatile. In combination with a cavitation agent, the effects of ultrasound can be amplified and localized for therapy. Using c-Fos expression mapping, we show how ultrasound-sensitive nanodroplets can be used to induce either neurosuppression or neurostimulation, without disrupting the blood-brain barrier in rats. By repurposing a commercial ultrasound contrast agent, Definity, lipid-shell decafluorobutane-core nanodroplets of 212.5 ± 2.0 nm were fabricated and loaded with or without pentobarbital. FUS was delivered with an atlas-based targeting system at 1.66 MHz to the motor cortex of rats, using a feedback-controller to detect successful nanodroplet vaporization and drug release. Neuromodulation was quantified through changes in sensorimotor function and c-Fos expression. Following FUS-triggered delivery, sham nanodroplets induced a 22.6 ± 21% increase in local c-Fos expression, whereas pentobarbital-loaded nanodroplets induced a 21.7 ± 13% decrease (n = 6). Nanodroplets, combined with FUS, offer an adaptable tool for neuromodulation, through local delivery of small molecule anesthetics or targeted mechanical effects.
Collapse
Affiliation(s)
- Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| | - Ying Meng
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Sheng-Kai Wu
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Clement Hamani
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada; Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
38
|
Ozdas MS, Shah AS, Johnson PM, Patel N, Marks M, Yasar TB, Stalder U, Bigler L, von der Behrens W, Sirsi SR, Yanik MF. Non-invasive molecularly-specific millimeter-resolution manipulation of brain circuits by ultrasound-mediated aggregation and uncaging of drug carriers. Nat Commun 2020; 11:4929. [PMID: 33004789 PMCID: PMC7529901 DOI: 10.1038/s41467-020-18059-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 07/31/2020] [Indexed: 12/13/2022] Open
Abstract
Non-invasive, molecularly-specific, focal modulation of brain circuits with low off-target effects can lead to breakthroughs in treatments of brain disorders. We systemically inject engineered ultrasound-controllable drug carriers and subsequently apply a novel two-component Aggregation and Uncaging Focused Ultrasound Sequence (AU-FUS) at the desired targets inside the brain. The first sequence aggregates drug carriers with millimeter-precision by orders of magnitude. The second sequence uncages the carrier's cargo locally to achieve high target specificity without compromising the blood-brain barrier (BBB). Upon release from the carriers, drugs locally cross the intact BBB. We show circuit-specific manipulation of sensory signaling in motor cortex in rats by locally concentrating and releasing a GABAA receptor agonist from ultrasound-controlled carriers. Our approach uses orders of magnitude (1300x) less drug than is otherwise required by systemic injection and requires very low ultrasound pressures (20-fold below FDA safety limits for diagnostic imaging). We show that the BBB remains intact using passive cavitation detection (PCD), MRI-contrast agents and, importantly, also by sensitive fluorescent dye extravasation and immunohistochemistry.
Collapse
Affiliation(s)
- Mehmet S Ozdas
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Aagam S Shah
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland. .,Neuroscience Center, Zurich, Switzerland.
| | - Paul M Johnson
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Nisheet Patel
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland
| | - Markus Marks
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Tansel Baran Yasar
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Urs Stalder
- Department of Chemistry, UZH, Zurich, Switzerland
| | | | - Wolfger von der Behrens
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Shashank R Sirsi
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Department of Bioengineering, UT at Dallas, Richardson, USA
| | - Mehmet Fatih Yanik
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland. .,Neuroscience Center, Zurich, Switzerland.
| |
Collapse
|
39
|
Ventre DM, Cluff A, Gagnon C, Diaz Vera D, Koppes RA, Koppes AN. The effects of low intensity focused ultrasonic stimulation on dorsal root ganglion neurons and Schwann cells in vitro. J Neurosci Res 2020; 99:374-391. [PMID: 32743823 DOI: 10.1002/jnr.24700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 01/14/2023]
Abstract
Satisfactory treatment of peripheral nerve injury (PNI) faces difficulties owing to the intrinsic biological barriers in larger injuries and invasive surgical interventions. Injury gaps >3 cm have low chances of full motor and sensory recovery, and the unmet need for PNI repair techniques which increase the likelihood of functional recovery while limiting invasiveness motivate this work. Building upon prior work in ultrasound stimulation (US) of dorsal root ganglion (DRG) neurons, the effects of US on DRG neuron and Schwann cell (SC) cocultures were investigated to uncover the role of SCs in mediating the neuronal response to US in vitro. Acoustic intensity-dependent alteration in selected neuromorphometrics of DRG neurons in coculture with SCs was observed in total outgrowth, primary neurites, and length compared to previously reported DRG monoculture in a calcium-independent manner. SC viability and proliferation were not impacted by US. Conditioned medium studies suggest secreted factors from SCs subjected to US impact DRG neuron morphology. These findings advance the current understanding of mechanisms by which these cell types respond to US, which may lead to new noninvasive US therapies for treating PNI.
Collapse
Affiliation(s)
- Daniel M Ventre
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Avery Cluff
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | | | - David Diaz Vera
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Ryan A Koppes
- Department of Biology, Northeastern University, Boston, MA, USA
| | - Abigail N Koppes
- Department of Biology, Northeastern University, Boston, MA, USA.,Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
40
|
Therapeutic Potentials of Localized Blood-Brain Barrier Disruption by Noninvasive Transcranial Focused Ultrasound: A Technical Review. J Clin Neurophysiol 2020; 37:104-117. [PMID: 32142021 DOI: 10.1097/wnp.0000000000000488] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The demands for region-specific, noninvasive therapies for neurologic/psychiatric conditions are growing. The rise of transcranial focused ultrasound technology has witnessed temporary and reversible disruptions of the blood-brain barrier in the brain with exceptional control over the spatial precisions and depth, all in a noninvasive manner. Starting with small animal studies about a decade ago, the technique is now being explored in nonhuman primates and humans for the assessment of its efficacy and safety. The ability to transfer exogenous/endogenous therapeutic agents, cells, and biomolecules across the blood-brain barrier opens up new therapeutic avenues for various neurologic conditions, with a possibility to modulate the excitability of regional brain function. This review addresses the technical fundamentals, sonication parameters, experimental protocols, and monitoring techniques to examine the efficacy/safety in focused ultrasound-mediated blood-brain barrier disruption and discuss its potential translations to clinical use.
Collapse
|
41
|
Wang S, Meng W, Ren Z, Li B, Zhu T, Chen H, Wang Z, He B, Zhao D, Jiang H. Ultrasonic Neuromodulation and Sonogenetics: A New Era for Neural Modulation. Front Physiol 2020; 11:787. [PMID: 32765294 PMCID: PMC7378787 DOI: 10.3389/fphys.2020.00787] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/15/2020] [Indexed: 12/19/2022] Open
Abstract
Non-invasive ultrasonic neural modulation (UNM), a non-invasive technique with enhanced spatial focus compared to conventional electrical neural modulation, has attracted much attention in recent decades and might become the mainstream regimen for neurological disorders. However, as ultrasonic bioeffects and its adjustments are still unclear, it remains difficult to be extensively applied for therapeutic purpose, much less in the setting of human skull. Hence to comprehensively understand the way ultrasound exerts bioeffects, we explored UNM from a basic perspective by illustrating the parameter settings and the underlying mechanisms. In addition, although the spatial resolution and precision of UNM are considerable, UNM is relatively non-specific to tissue or cell type and shows very low specificity at the molecular level. Surprisingly, Ibsen et al. (2015) first proposed the concept of sonogenetics, which combined UNM and mechanosensitive (MS) channel protein. This emerging approach is a valuable improvement, as it may markedly increase the precision and spatial resolution of UNM. It seemed to be an inspiring tool with high accuracy and specificity, however, little information about sonogenetics is currently available. Thus, in order to provide an overview of sonogenetics and prompt the researches on UNM, we summarized the potential mechanisms from a molecular level.
Collapse
Affiliation(s)
- Songyun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weilun Meng
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Medical Department, Nanjing Medical University, Nanjing, China
| | - Zhongyuan Ren
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Medical Department, Soochow University Medical College, Suzhou, China
| | - Binxun Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tongjian Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hui Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dongdong Zhao
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
42
|
Todd N, Angolano C, Ferran C, Devor A, Borsook D, McDannold N. Secondary effects on brain physiology caused by focused ultrasound-mediated disruption of the blood-brain barrier. J Control Release 2020; 324:450-459. [PMID: 32470359 DOI: 10.1016/j.jconrel.2020.05.040] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/12/2022]
Abstract
Focused ultrasound (FUS) combined with microbubbles is a non-invasive method for targeted, reversible disruption of the blood-brain barrier (FUS-BBB opening). This approach holds great promise for improving delivery of therapeutics to the brain. In order to achieve this clinically important goal, the approach necessarily breaks a protective barrier, temporarily, which plays a fundamental role in maintaining a homeostatic environment in the brain. Preclinical and clinical research has identified a set of treatment parameters under which this can be performed safely, whereby the BBB is disrupted to the point of being permeable to normally non-penetrant agents without causing significant acute damage to endothelial or neuronal cells. Much of the early work in this field focused on engineering questions around how to achieve optimal delivery of therapeutics via BBB disruption. However, there is increasing interest in addressing biological questions related to whether and how various aspects of neurophysiology might be affected when this fundamental protective barrier is compromised by the specific mechanisms of FUS-BBB opening. Improving our understanding of these secondary effects is becoming vital now that FUS-BBB opening treatments have entered clinical trials. Such information would help to safely expand FUS-BBB opening protocols into a wider range of drug delivery applications and may even lead to new types of treatments. In this paper, we will critically review our current knowledge of the secondary effects caused by FUS-BBB opening on brain physiology, identify areas that remain understudied, and discuss how a better understanding of these processes can be used to safely advance FUS-BBB opening into a wider range of clinical applications.
Collapse
Affiliation(s)
- Nick Todd
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| | - Cleide Angolano
- Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Christiane Ferran
- Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Anna Devor
- Department of Biomedical Engineering, Boston University, Boston, MA, United States; Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - David Borsook
- Center for Pain and the Brain, Boston Children's Hospital, Boston, MA, United States; Department of Anesthesia, Perioperative, and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Nathan McDannold
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Kung Y, Huang HY, Liao WH, Huang APH, Hsiao MY, Wu CH, Liu HL, Inserra C, Chen WS. A Single High-Intensity Shock Wave Pulse With Microbubbles Opens the Blood-Brain Barrier in Rats. Front Bioeng Biotechnol 2020; 8:402. [PMID: 32478046 PMCID: PMC7232561 DOI: 10.3389/fbioe.2020.00402] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/09/2020] [Indexed: 12/22/2022] Open
Abstract
Focused extracorporeal shockwave (FSW), one kind of focused high-intensity pulsed ultrasound, has been shown to induce blood-brain barrier (BBB) opening in targeted brain areas in rat animal models with minimal detrimental effects below threshold intensity levels or iterations. In the current study, we found that the thresholds could be further reduced by the addition of microbubbles (ultrasound contrast agents or UCA; SonoVue). FSW with 2 × 106 MBs/kg of UCA (20% of clinical dosage) at an intensity level of 0.1 (peak positive pressure 5.4 MPa; peak negative pressure -4.2 MPa; energy flux density 0.03 mJ/mm2) resulting in a 100% BBB opening rate without detectable hemorrhage or apoptosis in the brain. Significantly reduced free radical production was found compared with 0.5 MHz focused ultrasound at a peak negative pressure of 0.44 MPa (1% duty cycle and 4 × 107 MBs/kg of UCA). FSW devices offer advantages of commercial availability and high safety, and thus may facilitate future research and applications of focal BBB opening for oncological and pharmacological purposes.
Collapse
Affiliation(s)
- Yi Kung
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsin-Yu Huang
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wei-Hao Liao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Abel P.-H. Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Yen Hsiao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chueh-Hung Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Claude Inserra
- INSERM, U1032, LabTAU, Universiteì Claude Bernard Lyon 1, Lyon, France
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
44
|
Cui Z, Li D, Xu S, Xu T, Wu S, Bouakaz A, Wan M, Zhang S. Effect of scattered pressures from oscillating microbubbles on neuronal activity in mouse brain under transcranial focused ultrasound stimulation. ULTRASONICS SONOCHEMISTRY 2020; 63:104935. [PMID: 31945558 DOI: 10.1016/j.ultsonch.2019.104935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 06/10/2023]
Abstract
Previous studies have indicated that the presence of microbubbles (MBs) during sonication has an impact on neuronal activity, while the underlying mechanisms remain to be revealed. In this study, a model for the scattered pressures produced by the pulsating lipid-encapsulated MBs in mouse brain was developed to numerically investigate the effect of MBs on neuronal activity during transcranial focused ultrasound stimulation. The additional summed scattered pressure (Psummed_scat) from the oscillating MBs was calculated from the model. The level of neuronal activity was experimentally verified using an immunofluorescence assay with antibodies against c-fos. The pressure difference (ΔP) between acoustic pressures at which the same level of neuronal activity is excited by ultrasound stimulation with and without MBs was obtained from the experiments. The results showed that Psummed_scat accounts for about half of the ΔP when the MBs experience a "compression-only" response. The Psummed_scat suddenly increased at a critical acoustic pressure, around which a rapid enhancement of ΔP obtained from experiment also occurred. This work suggested that the additional scattered pressures from pulsating MBs are probably a mechanism that affects neuronal activity under transcranial focused ultrasound stimulation.
Collapse
Affiliation(s)
- Zhiwei Cui
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Dapeng Li
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shanshan Xu
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tianqi Xu
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shan Wu
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | | | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Siyuan Zhang
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
45
|
Constans C, Ahnine H, Santin M, Lehericy S, Tanter M, Pouget P, Aubry JF. Non-invasive ultrasonic modulation of visual evoked response by GABA delivery through the blood brain barrier. J Control Release 2020; 318:223-231. [DOI: 10.1016/j.jconrel.2019.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/25/2019] [Accepted: 12/05/2019] [Indexed: 11/25/2022]
|
46
|
Helms HCC, Kristensen M, Saaby L, Fricker G, Brodin B. Drug Delivery Strategies to Overcome the Blood-Brain Barrier (BBB). Handb Exp Pharmacol 2020; 273:151-183. [PMID: 33367937 DOI: 10.1007/164_2020_403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The brain capillary endothelium serves both as an exchange site for gases and solutes between blood and brain and as a protective fence against neurotoxic compounds from the blood. While this "blood-brain barrier" (BBB) function protects the fragile environment in the brain, it also poses a tremendous challenge for the delivery of drug compounds to the brain parenchyma. Paracellular brain uptake of drug compounds is limited by the physical tightness of the endothelium, which is tightly sealed with junction complexes. Transcellular uptake of lipophilic drug compounds is limited by the activity of active efflux pumps in the luminal membrane. As a result, the majority of registered CNS drug compounds are small lipophilic compounds which are not efflux transporter substrates. Small molecule CNS drug development therefore focuses on identifying compounds with CNS target affinity and modifies these in order to optimize lipophilicity and decrease efflux pump interactions. Since efflux pump activity is limiting drug uptake, it has been investigated whether coadministration of drug compounds with efflux pump inhibitors could increase drug uptake. While the concept works to some extent, a lot of challenges have been encountered in terms of obtaining efficient inhibition while avoiding adverse effects.Some CNS drug compounds enter the brain via nutrient transport proteins, an example is the levodopa, a prodrug of Dopamine, which crosses the BBB via the large neutral amino acid transporter LAT1. While carrier-mediated transport of drug compounds may seem attractive, the development of drugs targeting transporters is very challenging, since the compounds should have a good fit to the binding site, while still maintaining their CNS target affinity.Receptor-mediated transport of drug compounds, especially biotherapeutics, conjugated to a receptor-binding ligand has shown some promise, although the amounts transported are rather low. This also holds true for drug-conjugation to cell-penetrating peptides. Due to the low uptake of biotherapeutics, barrier-breaching approaches such as mannitol injections and focused ultrasound have been employed with some success to patient groups with no other treatment options.
Collapse
Affiliation(s)
| | - Mie Kristensen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Saaby
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Bioneer-Farma, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Birger Brodin
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
47
|
Cui Z, Li D, Feng Y, Xu T, Wu S, Li Y, Bouakaz A, Wan M, Zhang S. Enhanced neuronal activity in mouse motor cortex with microbubbles' oscillations by transcranial focused ultrasound stimulation. ULTRASONICS SONOCHEMISTRY 2019; 59:104745. [PMID: 31473423 DOI: 10.1016/j.ultsonch.2019.104745] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/29/2019] [Accepted: 08/22/2019] [Indexed: 06/10/2023]
Abstract
Microbubbles (MBs) are known to serve as an amplifier of the mechanical effects of ultrasound, which combined with ultrasound are widely used in brain. The goal of this study is to investigate the effect of oscillating MBs on the neuronal activity in the central nervous system (CNS) of mammals. The motor cortex of mice brain was subjected to ultrasound stimulation with and without MBs, and evoked electromyogram signals were recorded. A c-fos immunofluorescence assay was performed to evaluate the neuronal activation in the region of ultrasound stimulation. BBB integrity during ultrasound stimulation with MBs was assessed in this study. Moreover, the safety of ultrasound stimulation with MBs was examined. Using ultrasound at 620 kHz, the injection of MBs significantly increased the success rate of motor response from 0.065 ± 0.06 to 0.28 ± 0.10 when stimulation was applied at 0.12 MPa and from 0.38 ± 0.09 to 0.77 ± 0.18 at 0.25 MPa (p < 0.001). The results of the c-fos immunofluorescence assay showed that the mean densities of c-fos+ cells were significantly increased from 15.67 ± 3.51 to 53.01 ± 9.54 at 0.12 MPa acoustic pressure. At 0.25 MPa, the mean density of c-fos + cells was 81 ± 10.97 without MBs and it significantly increased to 124.12 ± 25.71 with MBs (p < 0.05). Enhanced neuronal activities were observed with 0.12 MPa ultrasound stimulation with MBs, while the integrity of BBB was not compromised, but 0.25 MPa ultrasound stimulation with MBs resulted in BBB disruption. These findings reveal that the oscillations of MBs can enhance neuronal activity in the CNS of mammals, and may provide an insight into the application of MBs combined with ultrasound in brain.
Collapse
Affiliation(s)
- Zhiwei Cui
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Dapeng Li
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yang Feng
- Xijing Hospital, Traditional Chinese Medicine, Xi'an 710032, China
| | - Tianqi Xu
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shan Wu
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yibao Li
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an 710049, China
| | | | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Siyuan Zhang
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
48
|
Foffani G, Trigo‐Damas I, Pineda‐Pardo JA, Blesa J, Rodríguez‐Rojas R, Martínez‐Fernández R, Obeso JA. Focused ultrasound in Parkinson's disease: A twofold path toward disease modification. Mov Disord 2019; 34:1262-1273. [DOI: 10.1002/mds.27805] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/14/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022] Open
Affiliation(s)
- Guglielmo Foffani
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- Hospital Nacional de Parapléjicos Toledo Spain
| | - Inés Trigo‐Damas
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - José A. Pineda‐Pardo
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Javier Blesa
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Rafael Rodríguez‐Rojas
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Raul Martínez‐Fernández
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - José A. Obeso
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| |
Collapse
|
49
|
Kang J, Zhang HK, Kadam SD, Fedorko J, Valentine H, Malla AP, Yan P, Harraz MM, Kang JU, Rahmim A, Gjedde A, Loew LM, Wong DF, Boctor EM. Transcranial Recording of Electrophysiological Neural Activity in the Rodent Brain in vivo Using Functional Photoacoustic Imaging of Near-Infrared Voltage-Sensitive Dye. Front Neurosci 2019; 13:579. [PMID: 31447622 PMCID: PMC6696882 DOI: 10.3389/fnins.2019.00579] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/22/2019] [Indexed: 12/27/2022] Open
Abstract
Minimally-invasive monitoring of electrophysiological neural activities in real-time-that enables quantification of neural functions without a need for invasive craniotomy and the longer time constants of fMRI and PET-presents a very challenging yet significant task for neuroimaging. In this paper, we present in vivo functional PA (fPA) imaging of chemoconvulsant rat seizure model with intact scalp using a fluorescence quenching-based cyanine voltage-sensitive dye (VSD) characterized by a lipid vesicle model mimicking different levels of membrane potential variation. The framework also involves use of a near-infrared VSD delivered through the blood-brain barrier (BBB), opened by pharmacological modulation of adenosine receptor signaling. Our normalized time-frequency analysis presented in vivo VSD response in the seizure group significantly distinguishable from those of the control groups at sub-mm spatial resolution. Electroencephalogram (EEG) recording confirmed the changes of severity and frequency of brain activities, induced by chemoconvulsant seizures of the rat brain. The findings demonstrate that the near-infrared fPA VSD imaging is a promising tool for in vivo recording of brain activities through intact scalp, which would pave a way to its future translation in real time human brain imaging.
Collapse
Affiliation(s)
- Jeeun Kang
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Haichong K. Zhang
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Shilpa D. Kadam
- Department of Neurology, Hugo W. Moser Research Institute at Kennedy Krieger, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Julie Fedorko
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Heather Valentine
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Adarsha P. Malla
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Ping Yan
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, United States
| | - Maged M. Harraz
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Jin U. Kang
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Arman Rahmim
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Albert Gjedde
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, United States
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Leslie M. Loew
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, United States
| | - Dean F. Wong
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
- Department of Environmental Sciences and Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Emad M. Boctor
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| |
Collapse
|
50
|
Wang P, Zhang J, Yu J, Smith C, Feng W. Brain Modulatory Effects by Low-Intensity Transcranial Ultrasound Stimulation (TUS): A Systematic Review on Both Animal and Human Studies. Front Neurosci 2019; 13:696. [PMID: 31396029 PMCID: PMC6667677 DOI: 10.3389/fnins.2019.00696] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/19/2019] [Indexed: 01/09/2023] Open
Abstract
Background and objective: Low Intensity Transcranial Ultrasound Stimulation (TUS) is a new form of non-invasive brain modulation with promising data; however, systematic reviews on the brain modulatory effects of TUS on both animals and humans have not been well-conducted. We aimed to conduct a systematic review on the studies using the TUS to modulate the brain functions and associated behavioral changes in both animals and humans. Methods: A literature search for published studies in the past 10 years was conducted. Two authors independently reviewed the relevant articles. Data were extracted and qualitatively summarized. Quality of studies was assessed by the SYRCLE's risk of bias tool for preclinical studies or the PEDro scale for clinical studies. Results: A total of 24 animal studies (506 animals) and 11 human studies (213 subjects) were included. Findings based on most animal studies demonstrated the excitatory or suppressive modulatory effects of ultrasonic stimulations on motor cortex, somatosensory cortex, thalamus, prefrontal cortex, auditory, and visual areas. Brain modulatory effects also were found among healthy human subjects in seven studies and two clinical studies suggested TUS may result in potential benefits on patients with disorder of consciousness or chronic pain. The safety concerns of TUS seem to be minor based on the human studies. Conclusions: TUS appears to be a viable technique in modulating the brain functions; however, research on TUS is still in its early stages, especially in human studies. Parameters need to be optimized before launching systematic investigations in humans.
Collapse
Affiliation(s)
- Pu Wang
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiaqi Zhang
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jiadan Yu
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, China
| | - Colin Smith
- Department of Neurology, Medical University of South Carolina, Charleston, SC, United States
| | - Wuwei Feng
- Department of Neurology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|