1
|
Pastre M, Occéan B, Boudousq V, Conejero I, Fabbro‐Peray P, Collombier L, Mallet L, Lopez‐Castroman J. Serotonergic underpinnings of obsessive-compulsive disorder: A systematic review and meta-analysis of neuroimaging findings. Psychiatry Clin Neurosci 2025; 79:48-59. [PMID: 39511769 PMCID: PMC11789457 DOI: 10.1111/pcn.13760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024]
Abstract
Obsessive-compulsive disorder (OCD) is a frequent and disabling condition, with many patients being treatment-resistant. Improved understanding of its neurobiology is vital for better therapies. Evidence is still conflicting regarding specific serotonergic-related dysfunctions in OCD. We systematically reviewed the literature to provide a quantitative assessment of the role of serotonin (5-HT) in patients with untreated OCD through imaging. We searched for neuroimaging studies investigating central 5-HT tonus in unmedicated patients with OCD, excluding studies comprising treated patients to prevent bias from antidepressant-induced changes in serotonergic tonus. We also conducted a meta-analysis using a homogeneous group of positron emission tomography and single photon emission computed tomography articles that compared 5-HT transporter (SERT) and 5-HT2A receptor (HT2AR) binding potential in different brain regions of patients with untreated OCD and healthy controls. The systematic review encompassed 18 articles, with 13 included in the subsequent meta-analysis. Risk of bias was assessed by a revised form of the Newcastle-Ottawa Scale. We provided standardized mean difference (SMD) values for SERT and 5-HT2AR binding potential measures across 15 different brain regions. Patients with OCD showed lower SERT binding potential in the brainstem (SMD = -1.13, 95% CI [-1.81 to -0.46]), midbrain (SMD = -0.54, 95% CI [-0.92 to -0.16]), and thalamus/hypothalamus regions (SMD = -0.58, 95% CI [-0.99 to -0.18]) with neglectable to moderate heterogeneity. By combining results from 2 decades of molecular imaging studies, we show that individuals with OCD exhibit lower SERT binding potential in specific brain regions, providing compelling evidence of a 5-HT system dysfunction. However, the exact mechanisms underlying this phenotype remain elusive. The limitations include heterogeneity across studies in populations, imaging techniques, and radiotracer usage.
Collapse
Affiliation(s)
| | - Bob‐Valéry Occéan
- Laboratoire de Biostatistique, Epidémiologie clinique, Santé Publique Innovation et Méthodologie (BESPIM)CHU NimesNimesFrance
| | - Vincent Boudousq
- Département de Médecine Nucléaire et Biophysique MédicaleCHU NimesNimesFrance
| | | | - Pascale Fabbro‐Peray
- Laboratoire de Biostatistique, Epidémiologie clinique, Santé Publique Innovation et Méthodologie (BESPIM)CHU NimesNimesFrance
| | - Laurent Collombier
- Département de Médecine Nucléaire et Biophysique MédicaleCHU NimesNimesFrance
| | - Luc Mallet
- Université Paris‐Est Créteil, DMU IMPACT, Département Médical‐Universitaire de Psychiatrie et d'AddictologieHôpitaux Universitaires Henri Mondor‐Albert Chenevier, Assistance Publique‐Hôpitaux de ParisCréteilFrance
- Institut du Cerveau‐Paris Brain Institute–ICMSorbonne Université, Inserm, CNRSParisFrance
- Department of Mental Health and Psychiatry, Global Health InstituteUniversity of GenevaGenevaSwitzerland
| | - Jorge Lopez‐Castroman
- Department of PsychiatryCHU NimesNimesFrance
- CIBERSAM, ISCIIIMadridSpain
- Department of Psychiatry, Radiology, Public Health, Nursing and MedicineUniversity of Santiago de CompostelaSantiago de CompostelaSpain
| |
Collapse
|
2
|
Meijlink B, van der Kooij HR, Wang Y, Li H, Huveneers S, Kooiman K. Ultrasound-activated microbubbles mediate F-actin disruptions and endothelial gap formation during sonoporation. J Control Release 2024; 376:1176-1189. [PMID: 39500409 DOI: 10.1016/j.jconrel.2024.10.066] [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: 10/07/2024] [Accepted: 10/29/2024] [Indexed: 11/14/2024]
Abstract
Locally opening up the endothelial barrier in a safe and controlled way is beneficial for drug delivery into the extravascular tissue. Although ultrasound-induced microbubble oscillations can affect the endothelial barrier integrity, the mechanism remains unknown. Here we uncover a new role for F-actin in microbubble-mediated endothelial gap formation. Unique simultaneous high-resolution confocal microscopy and ultra-high-speed camera imaging (10 million frames per second) reveal that single oscillating microbubbles (radius 1.3-3.8 μm; n = 48) induce sonoporation in all cells in which F-actin remodeling occurred. F-actin disruption only mainly resulted in tunnel formation (75 %), while F-actin stress fiber severing and recoil mainly resulted in cell-cell contact opening within 15 s upon treatment (54 %) and tunnel formation (15 %). F-actin stress fiber severing occurred when the fibers were within reach of the microbubble's maximum radius during oscillation, requiring normal forces of ≥230 nN. In the absence of F-actin stress fibers, oscillating microbubbles induced F-actin remodeling but no cell-cell contact opening. Together, these findings reveal a novel mechanism of microbubble-mediated transendothelial drug delivery, which associates with the underlying cytoskeletal F-actin organization.
Collapse
Affiliation(s)
- Bram Meijlink
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - H Rhodé van der Kooij
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Yuchen Wang
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Hongchen Li
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Stephan Huveneers
- Dept. Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Klazina Kooiman
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Han B, Liu Y, Zhou Q, Yu Y, Liu X, Guo Y, Zheng X, Zhou M, Yu H, Wang W. The advance of ultrasound-enabled diagnostics and therapeutics. J Control Release 2024; 375:1-19. [PMID: 39208935 DOI: 10.1016/j.jconrel.2024.08.039] [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: 04/30/2024] [Revised: 07/27/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Point-of-care ultrasound demonstrates significant potential in biomedical research due to its noninvasive, real-time visualization, cost-effectiveness, and other biological benefits. Ultrasound irradiation can precisely control the mechanical and physicochemical effects on pathogenic lesions, enabling real-time visualization, tunable tissue penetration depth, and therapeutic applications. This review summarizes recent advancements in ultrasound-enabled diagnostics and therapeutics, focusing on mechanochemical effects that can be directly integrated into biomedical applications. Additionally, the structure-functionality relationships of sonotheranostic nanoplatforms are systematically discussed, providing insights into the underlying biological effects. Finally, the limitations of current ultrasonic medicine are discussed, along with potential expansions to facilitate patient-centered translations.
Collapse
Affiliation(s)
- Biying Han
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yan Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Qianqian Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yuting Yu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xingxing Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yu Guo
- State Key Laboratory of Chemical Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Haijun Yu
- State Key Laboratory of Chemical Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
4
|
Lea-Banks H, Chauhan N, Hynynen K. Investigating the hypotensive effect of focused ultrasound neuromodulation and barbiturate-loaded nanodroplets in healthy and hypertensive rats. Brain Stimul 2024; 17:1317-1327. [PMID: 39643112 DOI: 10.1016/j.brs.2024.12.002] [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: 09/06/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Current strategies for reducing blood pressure (BP) are ineffective and unsafe for many patient populations, including drug-resistant hypertension and during pregnancy. Stimulating the periaqueductal grey (PAG) region has shown promise in treating drug-resistant hypertension in patients using deep brain stimulation. OBJECTIVE To develop a minimally invasive neuromodulation technique for the sustained treatment of hypertension. METHODS We have investigated BP reduction using focused ultrasound (FUS) (540 kHz) and anesthetic-loaded ultrasound-responsive nanodroplets to deliver pentobarbital to the PAG in normotensive (N = 27) and hypertensive (N = 20) male and female rats. BP, heart rate and plasma hormone content were collected before and after FUS exposure, and neuronal activity was mapped in the PAG using C-Fos and neuron subtype staining. Cavitation activity was monitored by detecting acoustic emissions from vaporizing nanodroplets, and neuromodulation was verified with immunohistochemistry. RESULTS Systolic and diastolic BP were reduced for 6 h following a single sonication of the PAG (-37/-28 mmHg systolic/diastolic), and the offline effect was extended to 4 days with consecutive sonications combined with systemically injected pentobarbital-loaded nanodroplets. In contrast, FUS applied to the frontal cortex had no effect on BP. Immunohistochemistry revealed stimulation of inhibitory neurons in the PAG region, indicating that the hypotensive effect was associated with a GABAergic pathway. The acoustic emissions from vaporizing droplets were found to correlate with neuron activity and change in BP, offering the potential for real-time treatment monitoring using ultrasound. CONCLUSIONS This work has implications for developing a new treatment for hypertension that has greater safety and broader applicability for vulnerable patient populations.
Collapse
Affiliation(s)
- Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Imaging, University of Toronto, Toronto, Canada.
| | - Neha Chauhan
- Physical Sciences Platform, Sunnybrook Research Institute, 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
|
5
|
Qiao L, Du X, Wang H, Wang Z, Gao S, Zhao CQ. Research Progress on the Strategies for Crossing the Blood-Brain Barrier. Mol Pharm 2024; 21:4786-4803. [PMID: 39231367 DOI: 10.1021/acs.molpharmaceut.4c00447] [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] [Indexed: 09/06/2024]
Abstract
Recently, the incidence of brain diseases, such as central nervous system degenerative diseases, brain tumors, and cerebrovascular diseases, has increased. However, the blood-brain barrier (BBB) limits the effective delivery of drugs to brain disease areas. Therefore, the mainstream direction of new drug development for these diseases is to engineer drugs that can better cross the BBB to exert their effects in the brain. This paper reviews the research progress and application of the main trans-BBB drug delivery strategies (receptor/transporter-mediated BBB crossing, focused ultrasound to open the BBB, adenosine agonist reversible opening of the BBB, aromatic resuscitation, transnasal administration, cell-mediated trans-BBB crossing, and viral vector system-mediated brain drug delivery). Meanwhile, the potential applications, advantages, and disadvantages of these strategies for crossing the BBB are analyzed. Finally, the future development prospects of strategies for crossing the BBB are also discussed. These strategies have potential value for treating brain diseases.
Collapse
Affiliation(s)
- Li Qiao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Xiuwei Du
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Hua Wang
- College of Intelligence and Information Engineering, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Zhiyi Wang
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Shijie Gao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Chun-Qin Zhao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| |
Collapse
|
6
|
Hong VM, Rade AD, Yan SM, Bhaskara A, Yousuf MS, Chen M, Martin SF, Liebl DJ, Price TJ, Kolber BJ. Loss of Sigma-2 Receptor/TMEM97 Is Associated with Neuropathic Injury-Induced Depression-Like Behaviors in Female Mice. eNeuro 2024; 11:ENEURO.0488-23.2024. [PMID: 38866499 PMCID: PMC11228697 DOI: 10.1523/eneuro.0488-23.2024] [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: 11/21/2023] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
Previous studies have shown that ligands that bind to sigma-2 receptor/TMEM97 (s2R/TMEM97), a transmembrane protein, have anxiolytic/antidepressant-like properties and relieve neuropathic pain-like effects in rodents. Despite medical interest in s2R/TMEM97, little affective and pain behavioral characterization has been done using transgenic mice, which limits the development of s2R/TMEM97 as a viable therapeutic target. Using wild-type (WT) and global Tmem97 knock-out (KO) mice, we sought to identify the contribution of Tmem97 in modulating affective and pain-like behaviors using a battery of affective and pain assays, including open field, light/dark preference, elevated plus maze, forced swim test, tail suspension test, and the mechanical sensitivity tests. Our results demonstrate that female Tmem97 KO mice show less anxiety-like and depressive-like behaviors in light/dark preference and tail suspension tests but not in an open field, elevated plus maze, and forced swim tests at baseline. We next performed spared nerve injury in WT and Tmem97 KO mice to assess the role of Tmem97 in neuropathic pain-induced anxiety and depression. WT mice, but not Tmem97 KO mice, developed a prolonged neuropathic pain-induced depressive-like phenotype when tested 10 weeks after nerve injury in females. Our results show that Tmem97 plays a role in modulating anxiety-like and depressive-like behaviors in naive animals with a significant change in the presence of nerve injury in female mice. Overall, these data demonstrate that Tmem97 could be a target to alleviate affective comorbidities of pain disorders.
Collapse
Affiliation(s)
- Veronica M Hong
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Avaneesh D Rade
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Shen M Yan
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Amulya Bhaskara
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Muhammad Saad Yousuf
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Min Chen
- Department of Mathematical Sciences, School of Natural Sciences and Mathematics, University of Texas at Dallas, Richardson, Texas 75080
| | - Stephen F Martin
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712
| | - Daniel J Liebl
- Department of Neurosurgery, University of Miami, Miller School of Medicine, Miami, Florida 33146
| | - Theodore J Price
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Benedict J Kolber
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| |
Collapse
|
7
|
Shi J, Tan C, Ge X, Qin Z, Xiong H. Recent advances in stimuli-responsive controlled release systems for neuromodulation. J Mater Chem B 2024; 12:5769-5786. [PMID: 38804184 DOI: 10.1039/d4tb00720d] [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: 05/29/2024]
Abstract
Neuromodulation aims to modulate the signaling activity of neurons or neural networks by the precise delivery of electrical stimuli or chemical agents and is crucial for understanding brain function and treating brain disorders. Conventional approaches, such as direct physical stimulation through electrical or acoustic methods, confront challenges stemming from their invasive nature, dependency on wired power sources, and unstable therapeutic outcomes. The emergence of stimulus-responsive delivery systems harbors the potential to revolutionize neuromodulation strategies through the precise and controlled release of neurochemicals in a specific brain region. This review comprehensively examines the biological barriers controlled release systems may encounter in vivo and the recent advances and applications of these systems in neuromodulation. We elucidate the intricate interplay between the molecular structure of delivery systems and response mechanisms to furnish insights for material selection and design. Additionally, the review contemplates the prospects and challenges associated with these systems in neuromodulation. The overarching objective is to propel the application of neuromodulation technology in analyzing brain functions, treating brain disorders, and providing insightful perspectives for exploiting new systems for biomedical applications.
Collapse
Affiliation(s)
- Jielin Shi
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Chao Tan
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Xiaoqian Ge
- Department of Mechanical Engineering, The University of Texas at Dallas Richardson, TX 75080, USA
| | - Zhenpeng Qin
- Department of Mechanical Engineering, The University of Texas at Dallas Richardson, TX 75080, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Hejian Xiong
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Bozuyuk U, Wrede P, Yildiz E, Sitti M. Roadmap for Clinical Translation of Mobile Microrobotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311462. [PMID: 38380776 DOI: 10.1002/adma.202311462] [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: 10/31/2023] [Revised: 01/24/2024] [Indexed: 02/22/2024]
Abstract
Medical microrobotics is an emerging field to revolutionize clinical applications in diagnostics and therapeutics of various diseases. On the other hand, the mobile microrobotics field has important obstacles to pass before clinical translation. This article focuses on these challenges and provides a roadmap of medical microrobots to enable their clinical use. From the concept of a "magic bullet" to the physicochemical interactions of microrobots in complex biological environments in medical applications, there are several translational steps to consider. Clinical translation of mobile microrobots is only possible with a close collaboration between clinical experts and microrobotics researchers to address the technical challenges in microfabrication, safety, and imaging. The clinical application potential can be materialized by designing microrobots that can solve the current main challenges, such as actuation limitations, material stability, and imaging constraints. The strengths and weaknesses of the current progress in the microrobotics field are discussed and a roadmap for their clinical applications in the near future is outlined.
Collapse
Affiliation(s)
- Ugur Bozuyuk
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Paul Wrede
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Erdost Yildiz
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- School of Medicine and College of Engineering, Koc University, Istanbul, 34450, Turkey
| |
Collapse
|
9
|
Zhu P, Simon I, Kokalari I, Kohane DS, Rwei AY. Miniaturized therapeutic systems for ultrasound-modulated drug delivery to the central and peripheral nervous system. Adv Drug Deliv Rev 2024; 208:115275. [PMID: 38442747 PMCID: PMC11031353 DOI: 10.1016/j.addr.2024.115275] [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: 12/12/2023] [Revised: 02/19/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Ultrasound is a promising technology to address challenges in drug delivery, including limited drug penetration across physiological barriers and ineffective targeting. Here we provide an overview of the significant advances made in recent years in overcoming technical and pharmacological barriers using ultrasound-assisted drug delivery to the central and peripheral nervous system. We commence by exploring the fundamental principles of ultrasound physics and its interaction with tissue. The mechanisms of ultrasonic-enhanced drug delivery are examined, as well as the relevant tissue barriers. We highlight drug transport through such tissue barriers utilizing insonation alone, in combination with ultrasound contrast agents (e.g., microbubbles), and through innovative particulate drug delivery systems. Furthermore, we review advances in systems and devices for providing therapeutic ultrasound, as their practicality and accessibility are crucial for clinical application.
Collapse
Affiliation(s)
- Pancheng Zhu
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands; State Key Laboratory of Mechanics and Control of Aerospace Structures, Nanjing University of Aeronautics & Astronautics, 210016, Nanjing, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Ignasi Simon
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands
| | - Ida Kokalari
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Alina Y Rwei
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands.
| |
Collapse
|
10
|
Pan X, Huang W, Nie G, Wang C, Wang H. Ultrasound-Sensitive Intelligent Nanosystems: A Promising Strategy for the Treatment of Neurological Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303180. [PMID: 37871967 DOI: 10.1002/adma.202303180] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/26/2023] [Indexed: 10/25/2023]
Abstract
Neurological diseases are a major global health challenge, affecting hundreds of millions of people worldwide. Ultrasound therapy plays an irreplaceable role in the treatment of neurological diseases due to its noninvasive, highly focused, and strong tissue penetration capabilities. However, the complexity of brain and nervous system and the safety risks associated with prolonged exposure to ultrasound therapy severely limit the applicability of ultrasound therapy. Ultrasound-sensitive intelligent nanosystems (USINs) are a novel therapeutic strategy for neurological diseases that bring greater spatiotemporal controllability and improve safety to overcome these challenges. This review provides a detailed overview of therapeutic strategies and clinical advances of ultrasound in neurological diseases, focusing on the potential of USINs-based ultrasound in the treatment of neurological diseases. Based on the physical and chemical effects induced by ultrasound, rational design of USINs is a prerequisite for improving the efficacy of ultrasound therapy. Recent developments of ultrasound-sensitive nanocarriers and nanoagents are systemically reviewed. Finally, the challenges and developing prospects of USINs are discussed in depth, with a view to providing useful insights and guidance for efficient ultrasound treatment of neurological diseases.
Collapse
Affiliation(s)
- Xueting Pan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Wenping Huang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Changyong Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
11
|
Perolina E, Meissner S, Raos B, Harland B, Thakur S, Svirskis D. Translating ultrasound-mediated drug delivery technologies for CNS applications. Adv Drug Deliv Rev 2024; 208:115274. [PMID: 38452815 DOI: 10.1016/j.addr.2024.115274] [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: 09/28/2023] [Revised: 02/18/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024]
Abstract
Ultrasound enhances drug delivery into the central nervous system (CNS) by opening barriers between the blood and CNS and by triggering release of drugs from carriers. A key challenge in translating setups from in vitro to in vivo settings is achieving equivalent acoustic energy delivery. Multiple devices have now been demonstrated to focus ultrasound to the brain, with concepts emerging to also target the spinal cord. Clinical trials to date have used ultrasound to facilitate the opening of the blood-brain barrier. While most have focused on feasibility and safety considerations, therapeutic benefits are beginning to emerge. To advance translation of these technologies for CNS applications, researchers should standardise exposure protocol and fine-tune ultrasound parameters. Computational modelling should be increasingly used as a core component to develop both in vitro and in vivo setups for delivering accurate and reproducible ultrasound to the CNS. This field holds promise for transformative advancements in the management and pharmacological treatment of complex and challenging CNS disorders.
Collapse
Affiliation(s)
- Ederlyn Perolina
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Svenja Meissner
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Brad Raos
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Bruce Harland
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Sachin Thakur
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand.
| |
Collapse
|
12
|
Del Campo Fonseca A, Ahmed D. Ultrasound robotics for precision therapy. Adv Drug Deliv Rev 2024; 205:115164. [PMID: 38145721 DOI: 10.1016/j.addr.2023.115164] [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: 09/30/2023] [Revised: 12/07/2023] [Accepted: 12/22/2023] [Indexed: 12/27/2023]
Abstract
In recent years, the application of microrobots in precision therapy has gained significant attention. The small size and maneuverability of these micromachines enable them to potentially access regions that are difficult to reach using traditional methods; thus, reducing off-target toxicities and maximizing treatment effectiveness. Specifically, acoustic actuation has emerged as a promising method to exert control. By harnessing the power of acoustic energy, these small machines potentially navigate the body, assemble at the desired sites, and deliver therapies with enhanced precision and effectiveness. Amidst the enthusiasm surrounding these miniature agents, their translation to clinical environments has proven difficult. The primary objectives of this review are threefold: firstly, to offer an overview of the fundamental acoustic principles employed in the field of microrobots; secondly, to assess their current applications in medical therapies, encompassing tissue targeting, drug delivery or even cell infiltration; and lastly, to delve into the continuous efforts aimed at integrating acoustic microrobots into in vivo applications.
Collapse
Affiliation(s)
- Alexia Del Campo Fonseca
- Department of Mechanical and Process Engineering, Acoustic Robotics Systems Lab, ETH Zurich, Säumerstrasse 4, 8803 Rüschlikon, Switzerland.
| | - Daniel Ahmed
- Department of Mechanical and Process Engineering, Acoustic Robotics Systems Lab, ETH Zurich, Säumerstrasse 4, 8803 Rüschlikon, Switzerland.
| |
Collapse
|
13
|
Tian M, Ma Z, Yang GZ. Micro/nanosystems for controllable drug delivery to the brain. Innovation (N Y) 2024; 5:100548. [PMID: 38161522 PMCID: PMC10757293 DOI: 10.1016/j.xinn.2023.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024] Open
Abstract
Drug delivery to the brain is crucial in the treatment for central nervous system disorders. While significant progress has been made in recent years, there are still major challenges in achieving controllable drug delivery to the brain. Unmet clinical needs arise from various factors, including controlled drug transport, handling large drug doses, methods for crossing biological barriers, the use of imaging guidance, and effective models for analyzing drug delivery. Recent advances in micro/nanosystems have shown promise in addressing some of these challenges. These include the utilization of microfluidic platforms to test and validate the drug delivery process in a controlled and biomimetic setting, the development of novel micro/nanocarriers for large drug loads across the blood-brain barrier, and the implementation of micro-intervention systems for delivering drugs through intraparenchymal or peripheral routes. In this article, we present a review of the latest developments in micro/nanosystems for controllable drug delivery to the brain. We also delve into the relevant diseases, biological barriers, and conventional methods. In addition, we discuss future prospects and the development of emerging robotic micro/nanosystems equipped with directed transportation, real-time image guidance, and closed-loop control.
Collapse
Affiliation(s)
- Mingzhen Tian
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang-Zhong Yang
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
14
|
Datta MS, Chen Y, Chauhan S, Zhang J, De La Cruz ED, Gong C, Tomer R. Whole-brain mapping reveals the divergent impact of ketamine on the dopamine system. Cell Rep 2023; 42:113491. [PMID: 38052211 PMCID: PMC10843582 DOI: 10.1016/j.celrep.2023.113491] [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: 11/07/2022] [Revised: 10/22/2023] [Accepted: 11/09/2023] [Indexed: 12/07/2023] Open
Abstract
Ketamine is a multifunctional drug with clinical applications as an anesthetic, pain management medication, and a fast-acting antidepressant. However, it is also recreationally abused for its dissociative effects. Recent studies in rodents are revealing the neuronal mechanisms mediating its actions, but the impact of prolonged exposure to ketamine on brain-wide networks remains less understood. Here, we develop a sub-cellular resolution whole-brain phenotyping approach and utilize it in male mice to show that repeated ketamine administration leads to a dose-dependent decrease in dopamine neurons in midbrain regions linked to behavioral states, alongside an increase in the hypothalamus. Additionally, diverse changes are observed in long-range innervations of the prefrontal cortex, striatum, and sensory areas. Furthermore, the data support a role for post-transcriptional regulation in enabling ketamine-induced neural plasticity. Through an unbiased, high-resolution whole-brain analysis, this study provides important insights into how chronic ketamine exposure reshapes brain-wide networks.
Collapse
Affiliation(s)
- Malika S Datta
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Yannan Chen
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Shradha Chauhan
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Jing Zhang
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | - Cheng Gong
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Raju Tomer
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
15
|
Wang M, Xu T, Li D, Wu Y, Zhang B, Zhang S. Enhanced and spatially controllable neuronal activity induced by transcranial focused ultrasound stimulation combined with phase-change nanodroplets. ULTRASONICS SONOCHEMISTRY 2023; 101:106686. [PMID: 37956511 PMCID: PMC10661601 DOI: 10.1016/j.ultsonch.2023.106686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/15/2023]
Abstract
Non-invasive ultrasound neuromodulation (USNM) is a powerful tool to explore neural circuits and treat neurological disorders. Due to the heterogeneity of the skull and regional variations in modulation and treatment objectives, it is necessary to develop an efficient and spatially controllable neuromodulation approach. Recently, transcranial focused ultrasound (tFUS) combined with external biomicro/nanomaterials for brain stimulation has garnered significant attention. This study focused on tFUS combined with perfluoropentane (PFP) nanodroplets (NDs) to improve the efficacy and spatial controllability of USNM. The developed two-stage variable pulse tFUS sequence that include the acoustic droplet vaporization (ADV) pulse for vaporizing PFP NDs into microbubbles (MBs) and the USNM sequence for inducing mechanical oscillations of the formed MBs to enhance neuronal activity. Further, adjusting the acoustic pressure of the ADV pulse generated the controllable vaporization regions, thereby achieving spatially controllable neuromodulation. The results showed that the mean densities of c-fos+ cells expression in the group of PFP NDs with ADV (109 ± 19 cells/mm2) were significantly higher compared to the group without ADV (37.34 ± 8.24 cells/mm2). The acoustic pressure of the ADV pulse with 1.98 MPa and 2.81 MPa in vitro generated the vaporization regions of 0.146 ± 0.032 cm2 and 0.349 ± 0.056 cm2, respectively. Under the same stimulation conditions, a larger vaporization region was also obtained with higher acoustic pressure in vivo, inducing a broader region of neuronal activation. Therefore, this study will serve as a valuable reference for developing the efficient and spatially controllable tFUS neuromodulation strategy.
Collapse
Affiliation(s)
- Mengke Wang
- The Key Laboratory of Biomedical Information Engineering of 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 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 Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yue Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Baochen Zhang
- The Key Laboratory of Biomedical Information Engineering of 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 Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
16
|
Wrede P, Aghakhani A, Bozuyuk U, Yildiz E, Sitti M. Acoustic Trapping and Manipulation of Hollow Microparticles under Fluid Flow Using a Single-Lens Focused Ultrasound Transducer. ACS APPLIED MATERIALS & INTERFACES 2023; 15. [PMID: 37917969 PMCID: PMC10658455 DOI: 10.1021/acsami.3c11656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
Microparticle manipulation and trapping play pivotal roles in biotechnology. To achieve effective manipulation within fluidic flow conditions and confined spaces, it is necessary to consider the physical properties of microparticles and the types of trapping forces applied. While acoustic waves have shown potential for manipulating microparticles, the existing setups involve complex actuation mechanisms and unstable microbubbles. Consequently, the need persists for an easily deployable acoustic actuation setup with stable microparticles. Here, we propose the use of hollow borosilicate microparticles possessing a rigid thin shell, which can be efficiently trapped and manipulated using a single-lens focused ultrasound (FUS) transducer under physiologically relevant flow conditions. These hollow microparticles offer stability and advantageous acoustic properties. They can be scaled up and mass-produced, making them suitable for systemic delivery. Our research demonstrates the successful trapping dynamics of FUS within circular tubings of varying diameters, validating the effectiveness of the method under realistic flow rates and ultrasound amplitudes. We also showcase the ability to remove hollow microparticles by steering the FUS transducer against the flow. Furthermore, we present potential biomedical applications, such as active cell tagging and navigation in bifurcated channels as well as ultrasound imaging in mouse cadaver liver tissue.
Collapse
Affiliation(s)
- Paul Wrede
- Physical
Intelligence Department, Max Planck Institute
for Intelligent Systems, 70569 Stuttgart, Germany
| | - Amirreza Aghakhani
- Physical
Intelligence Department, Max Planck Institute
for Intelligent Systems, 70569 Stuttgart, Germany
- Institute
of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569 Stuttgart, Germany
| | - Ugur Bozuyuk
- Physical
Intelligence Department, Max Planck Institute
for Intelligent Systems, 70569 Stuttgart, Germany
| | - Erdost Yildiz
- Physical
Intelligence Department, Max Planck Institute
for Intelligent Systems, 70569 Stuttgart, Germany
| | - Metin Sitti
- Physical
Intelligence Department, Max Planck Institute
for Intelligent Systems, 70569 Stuttgart, Germany
- Institute
for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
- School
of Medicine and School of Engineering, Koç
University, Istanbul, 34450, Turkey
| |
Collapse
|
17
|
Parks TV, Szuzupak D, Choi SH, Alikaya A, Mou Y, Silva AC, Schaeffer DJ. Noninvasive disruption of the blood-brain barrier in the marmoset monkey. Commun Biol 2023; 6:806. [PMID: 37532791 PMCID: PMC10397190 DOI: 10.1038/s42003-023-05185-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/26/2023] [Indexed: 08/04/2023] Open
Abstract
The common marmoset monkey (Callithrix jacchus) is a species of rising prominence in the neurosciences due to its small size, ease of handling, fast breeding, and its shared functional and structural brain characteristics with Old World primates. With increasing attention on modeling human brain diseases in marmosets, understanding how to deliver therapeutic or neurotropic agents to the marmoset brain noninvasively is of great preclinical importance. In other species, including humans, transcranial focused ultrasound (tFUS) aided by intravenously injected microbubbles has proven to be a transient, reliable, and safe method for disrupting the blood-brain barrier (BBB), allowing the focal passage of therapeutic agents that do not otherwise readily traverse the tight endothelial junctions of the BBB. The critical gap that we address here is to document parameters to disrupt the BBB reliably and safely in marmosets using tFUS. By integrating our marmoset brain atlases and the use of a marmoset-specific stereotactic targeting system, we conduct a series of systematic transcranial sonication experiments in nine marmosets. We demonstrate the effects of center frequency, acoustic pressure, burst period, and duration, establish a minimum microbubble dose, estimate microbubble clearance time, and estimate the duration that the BBB remains open to passage. Successful BBB disruption is reported in vivo with MRI-based contrast agents, as well as Evans blue staining assessed ex vivo. Histology (Hematoxylin and Eosin staining) and immunohistochemistry indicate that the BBB can be safely and reliably opened with the parameters derived from these experiments. The series of experiments presented here establish methods for safely, reproducibly, and focally perturbing the BBB using tFUS in the common marmoset monkey that can serve as a basis for noninvasive delivery of therapeutic or neurotropic agents.
Collapse
Affiliation(s)
- T Vincenza Parks
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Diego Szuzupak
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sang-Ho Choi
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aydin Alikaya
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yongshan Mou
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Afonso C Silva
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David J Schaeffer
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Wang K, Zhou W, Jin X, Shang X, Wu X, Wen L, Li S, Hong Y, Ke J, Xu Y, Yuan H, Hu F. Enhanced brain delivery of hypoxia-sensitive liposomes by hydroxyurea for rescue therapy of hyperacute ischemic stroke. NANOSCALE 2023. [PMID: 37377137 DOI: 10.1039/d3nr01071f] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Ischemic stroke is characterized by high morbidity, disability, and mortality. Unfortunately, the only FDA-approved pharmacological thrombolytic, alteplase, has a narrow therapeutic window of only 4.5 h. Other drugs like neuroprotective agents have not been clinically used because of their low efficacy. To improve the efficacy of neuroprotective agents and the effectiveness of rescue therapies for hyperacute ischemic stroke, we investigated and verified the variation trends of the blood-brain barrier (BBB) permeability and regional cerebral blood flow over 24 h in rats that had ischemic strokes. Hypoperfusion and the biphasic increase of BBB permeability are still the main limiting factors for lesion-specific drug distribution and drug brain penetration. Herein, the nitric oxide donor hydroxyurea (HYD) was reported to downregulate the expression of tight junction proteins and upregulate intracellular nitric oxide content in the brain microvascular endothelial cells subjected to oxygen-glucose deprivation, which was shown to facilitate the transport of liposomes across brain endothelial monolayer in an in vitro model. HYD also increased the BBB permeability and promoted microcirculation in the hyperacute phase of stroke. The neutrophil-like cell-membrane-fusogenic hypoxia-sensitive liposomes exhibited excellent performance in targeting the inflamed brain microvascular endothelial cells, enhancing cell association, and promoting rapid hypoxic-responsive release in the hypoxic microenvironment. Overall, the combined HYD and hypoxia-sensitive liposome dosing regimen effectively decreased the cerebral infarction volume and relieved neurological dysfunction in rats that had ischemic strokes; these therapies were involved in the anti-oxidative stress effect and the neurotrophic effect mediated by macrophage migration inhibitory factor.
Collapse
Affiliation(s)
- Kai Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Wentao Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Xiangyu Jin
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xuwei Shang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Xiaomei Wu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Lijuan Wen
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Sufen Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Yiling Hong
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Jia Ke
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Yichong Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
19
|
Honari A, Sirsi SR. The Evolution and Recent Trends in Acoustic Targeting of Encapsulated Drugs to Solid Tumors: Strategies beyond Sonoporation. Pharmaceutics 2023; 15:1705. [PMID: 37376152 DOI: 10.3390/pharmaceutics15061705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Despite recent advancements in ultrasound-mediated drug delivery and the remarkable success observed in pre-clinical studies, no delivery platform utilizing ultrasound contrast agents has yet received FDA approval. The sonoporation effect was a game-changing discovery with a promising future in clinical settings. Various clinical trials are underway to assess sonoporation's efficacy in treating solid tumors; however, there are disagreements on its applicability to the broader population due to long-term safety issues. In this review, we first discuss how acoustic targeting of drugs gained importance in cancer pharmaceutics. Then, we discuss ultrasound-targeting strategies that have been less explored yet hold a promising future. We aim to shed light on recent innovations in ultrasound-based drug delivery including newer designs of ultrasound-sensitive particles specifically tailored for pharmaceutical usage.
Collapse
Affiliation(s)
- Arvin Honari
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
20
|
Wu D, Chen Q, Chen X, Han F, Chen Z, Wang Y. The blood-brain barrier: structure, regulation, and drug delivery. Signal Transduct Target Ther 2023; 8:217. [PMID: 37231000 PMCID: PMC10212980 DOI: 10.1038/s41392-023-01481-w] [Citation(s) in RCA: 429] [Impact Index Per Article: 214.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Blood-brain barrier (BBB) is a natural protective membrane that prevents central nervous system (CNS) from toxins and pathogens in blood. However, the presence of BBB complicates the pharmacotherapy for CNS disorders as the most chemical drugs and biopharmaceuticals have been impeded to enter the brain. Insufficient drug delivery into the brain leads to low therapeutic efficacy as well as aggravated side effects due to the accumulation in other organs and tissues. Recent breakthrough in materials science and nanotechnology provides a library of advanced materials with customized structure and property serving as a powerful toolkit for targeted drug delivery. In-depth research in the field of anatomical and pathological study on brain and BBB further facilitates the development of brain-targeted strategies for enhanced BBB crossing. In this review, the physiological structure and different cells contributing to this barrier are summarized. Various emerging strategies for permeability regulation and BBB crossing including passive transcytosis, intranasal administration, ligands conjugation, membrane coating, stimuli-triggered BBB disruption, and other strategies to overcome BBB obstacle are highlighted. Versatile drug delivery systems ranging from organic, inorganic, and biologics-derived materials with their synthesis procedures and unique physio-chemical properties are summarized and analyzed. This review aims to provide an up-to-date and comprehensive guideline for researchers in diverse fields, offering perspectives on further development of brain-targeted drug delivery system.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| | - Qi Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| |
Collapse
|
21
|
Datta MS, Chen Y, Chauhan S, Zhang J, De La Cruz ED, Gong C, Tomer R. Whole-brain mapping reveals the divergent impact of ketamine on the dopamine system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536506. [PMID: 37090584 PMCID: PMC10120808 DOI: 10.1101/2023.04.12.536506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Ketamine is a multifunctional drug with clinical applications as an anesthetic, as a pain management medication and as a transformative fast-acting antidepressant. It is also abused as a recreational drug due to its dissociative property. Recent studies in rodents are revealing the neuronal mechanisms that mediate the complex actions of ketamine, however, its long-term impact due to prolonged exposure remains much less understood with profound scientific and clinical implications. Here, we develop and utilize a high-resolution whole-brain phenotyping approach to show that repeated ketamine administration leads to a dosage-dependent decrease of dopamine (DA) neurons in the behavior state-related midbrain regions and, conversely, an increase within the hypothalamus. Congruently, we show divergently altered innervations of prefrontal cortex, striatum, and sensory areas. Further, we present supporting data for the post-transcriptional regulation of ketamine-induced structural plasticity. Overall, through an unbiased whole-brain analysis, we reveal the divergent brain-wide impact of chronic ketamine exposure on the association and sensory pathways.
Collapse
Affiliation(s)
- Malika S. Datta
- Department of Biological Sciences, Columbia University
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University
| | - Yannan Chen
- Department of Biological Sciences, Columbia University
- Department of Biomedical Engineering, Columbia University
| | | | - Jing Zhang
- Department of Biological Sciences, Columbia University
| | | | - Cheng Gong
- Department of Biological Sciences, Columbia University
- Department of Biomedical Engineering, Columbia University
| | - Raju Tomer
- Department of Biological Sciences, Columbia University
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University
- Department of Biomedical Engineering, Columbia University
| |
Collapse
|
22
|
Rodrigues AF, Rebelo C, Reis T, Simões S, Bernardino L, Peça J, Ferreira L. Engineering optical tools for remotely controlled brain stimulation and regeneration. Biomater Sci 2023; 11:3034-3050. [PMID: 36947145 DOI: 10.1039/d2bm02059a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2023]
Abstract
Neurological disorders are one of the world's leading medical and societal challenges due to the lack of efficacy of the first line treatment. Although pharmacological and non-pharmacological interventions have been employed with the aim of regulating neuronal activity and survival, they have failed to avoid symptom relapse and disease progression in the vast majority of patients. In the last 5 years, advanced drug delivery systems delivering bioactive molecules and neuromodulation strategies have been developed to promote tissue regeneration and remodel neuronal circuitry. However, both approaches still have limited spatial and temporal precision over the desired target regions. While external stimuli such as electromagnetic fields and ultrasound have been employed in the clinic for non-invasive neuromodulation, they do not have the capability of offering single-cell spatial resolution as light stimulation. Herein, we review the latest progress in this area of study and discuss the prospects of using light-responsive nanomaterials to achieve on-demand delivery of drugs and neuromodulation, with the aim of achieving brain stimulation and regeneration.
Collapse
Affiliation(s)
- Artur Filipe Rodrigues
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Catarina Rebelo
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Tiago Reis
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Susana Simões
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Liliana Bernardino
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - João Peça
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Lino Ferreira
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| |
Collapse
|
23
|
Chu YC, Ho CY, Chuo Y, Wu HH, Wang JL. On minimal focal distance of a focused ultrasound probe for neuromodulation. JASA EXPRESS LETTERS 2023; 3:024002. [PMID: 36858995 DOI: 10.1121/10.0017106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Focal distance is a key parameter for a focused ultrasound probe, especially in mouse brain stimulation where targets are right below the skull. A closed-form solution for the minimal focal distance with a given transducer size was derived in this study to facilitate precise focal spot alignment with targets in the mouse brain. The spherical profile corresponding to the minimal focal distance does not produce accurate focusing. An iterative algorithm based on Snell's law was introduced for lens profile calculation. With a suitable step size, an accurate lens profile can be obtained for the minimal focal distance.
Collapse
Affiliation(s)
- Ya-Cherng Chu
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan , , , ,
| | - Chien-Ying Ho
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan , , , ,
| | - Yue Chuo
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan , , , ,
| | - Hao-Hsuan Wu
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan , , , ,
| | - Jaw-Lin Wang
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan , , , ,
| |
Collapse
|
24
|
Zhang T, Zheng Q, Xie C, Fan G, Wang Y, Wu Y, Fu Y, Huang J, Craig DQM, Cai X, Li X. Integration of Silica Nanorattles with Manganese-Doped In 2S 3/InOOH to Enable Ultrasound-Mediated Tumor Theranostics. ACS APPLIED MATERIALS & INTERFACES 2023; 15:4883-4894. [PMID: 36662514 DOI: 10.1021/acsami.2c18095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
As a result of their radiation-free nature and deep-penetration ability, tumor theranostics mediated by ultrasound have become increasingly recognized as a modality with high potential for translation into clinical cancer treatment. The effective integration of ultrasound imaging and sonodynamic therapy (SDT) into one nanoplatform remains an enormous challenge yet to be fully resolved. Here, a novel theranostic system, consisting of rattle-type SiO2 (r-SiO2) loaded with Mn-doped In2S3/InOOH (SMISO), was designed and synthesized to enable an improved ultrasound imaging-guided therapy. With Mn-doped In2S3/InOOH (MISO) and a heterojunction structure, this novel sonosensitizer facilitates the generation of reactive oxygen species (ROS) for SDT. By coupling interfaces between the shell and core in rattle-type SiO2, multiple reflections/scattering are generated, while MISO has high acoustic impedance. By integrating r-SiO2 and MISO, the SMISO composite nanoparticles (NPs) increase the acoustic reflection and provide enhanced contrast for ultrasound imaging. Through the effective accumulation in tumors, which was monitored by B-mode ultrasound imaging in vivo, SMISO composite NPs effectively inhibited tumor growth without adverse side effects under ultrasound irradiation treatment. This work therefore provides a new approach to integrate a novel gas-free ultrasound contrast agent and a semiconductor sonosensitizer for cancer theranostics.
Collapse
Affiliation(s)
- Tian Zhang
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, Zhejiang, P. R. China
| | - Qiang Zheng
- Key Laboratory of Endoscopic Technique Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 215123, P. R. China
| | - Congkun Xie
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, Zhejiang, P. R. China
| | - Gonglin Fan
- Key Laboratory of Endoscopic Technique Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 215123, P. R. China
| | - Yifan Wang
- Key Laboratory of Endoscopic Technique Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 215123, P. R. China
| | - Yongjun Wu
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, Zhejiang, P. R. China
| | - Yike Fu
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, Zhejiang, P. R. China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311200, P. R. China
| | - Jie Huang
- Department of Mechanical Engineering, University College London, London WC1E 7JE, U.K
| | - Duncan Q M Craig
- University College London School of Pharmacy, London WC1N 1AX, U.K
| | - Xiujun Cai
- Key Laboratory of Endoscopic Technique Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 215123, P. R. China
| | - Xiang Li
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, Zhejiang, P. R. China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311200, P. R. China
| |
Collapse
|
25
|
Deng Q, Mi J, Dong J, Chen Y, Chen L, He J, Zhou J. Superiorly Stable Three-Layer Air Microbubbles Generated by Versatile Ethanol-Water Exchange for Contrast-Enhanced Ultrasound Theranostics. ACS NANO 2023; 17:263-274. [PMID: 36354372 DOI: 10.1021/acsnano.2c07300] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Microbubbles have been widely used as ultrasound contrast agents in clinical diagnosis. Moreover, most current preparation methods for microbubbles are uncontrollable, and the as-obtained microbubbles are unstable in aqueous solution or under ultrasound. Here, we report a strategy to prepare superiorly stable microbubbles with three-layer structures by the ethanol-water exchange. This versatile method can also be applied to prepare different kinds of protein microbubbles with various sizes for advanced biomedical applications. To demonstrate this, the protein air microbubbles are created, which is stable in water for several days with intact structures and exhibits excellent contrast-enhanced ultrasound imaging. Moreover, the protein air microbubbles can also deliver a mass of drugs while maintaining their stable structures, making them a platform for ultrasound imaging-guided drug delivery. The versatile protein air microbubbles have great potential for the design and application of theranostic platforms.
Collapse
Affiliation(s)
- Qiurong Deng
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Jiaomei Mi
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Jianpei Dong
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Yin Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Lanxi Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Jinxu He
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou510006, China
| | - Jianhua Zhou
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou510006, China
| |
Collapse
|
26
|
Murphy KR, Farrell JS, Gomez JL, Stedman QG, Li N, Leung SA, Good CH, Qiu Z, Firouzi K, Butts Pauly K, Khuri-Yakub BPT, Michaelides M, Soltesz I, de Lecea L. A tool for monitoring cell type-specific focused ultrasound neuromodulation and control of chronic epilepsy. Proc Natl Acad Sci U S A 2022; 119:e2206828119. [PMID: 36343238 PMCID: PMC9674244 DOI: 10.1073/pnas.2206828119] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/16/2022] [Indexed: 11/09/2022] Open
Abstract
Focused ultrasound (FUS) is a powerful tool for noninvasive modulation of deep brain activity with promising therapeutic potential for refractory epilepsy; however, tools for examining FUS effects on specific cell types within the deep brain do not yet exist. Consequently, how cell types within heterogeneous networks can be modulated and whether parameters can be identified to bias these networks in the context of complex behaviors remains unknown. To address this, we developed a fiber Photometry Coupled focused Ultrasound System (PhoCUS) for simultaneously monitoring FUS effects on neural activity of subcortical genetically targeted cell types in freely behaving animals. We identified a parameter set that selectively increases activity of parvalbumin interneurons while suppressing excitatory neurons in the hippocampus. A net inhibitory effect localized to the hippocampus was further confirmed through whole brain metabolic imaging. Finally, these inhibitory selective parameters achieved significant spike suppression in the kainate model of chronic temporal lobe epilepsy, opening the door for future noninvasive therapies.
Collapse
Affiliation(s)
- Keith R. Murphy
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | | | - Juan L. Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse, Baltimore, MD 21224
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Quintin G. Stedman
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305
| | - Ningrui Li
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Steven A. Leung
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Cameron H. Good
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL 60601
| | - Zhihai Qiu
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Kamyar Firouzi
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305
| | - Kim Butts Pauly
- Department of Radiology, Stanford University, Stanford, CA 94305
| | | | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse, Baltimore, MD 21224
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA 94305
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| |
Collapse
|
27
|
Yang J, Li Y, Sun J, Zou H, Sun Y, Luo J, Xie Q, A R, Wang H, Li X, Wang K, Yang L, Ma T, Wu L, Sun X. An Osimertinib-Perfluorocarbon Nanoemulsion with Excellent Targeted Therapeutic Efficacy in Non-small Cell Lung Cancer: Achieving Intratracheal and Intravenous Administration. ACS NANO 2022; 16:12590-12605. [PMID: 35863049 DOI: 10.1021/acsnano.2c04159] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Low accumulation of anticancer drugs in tumors and serious systemic toxicity remain the main challenges to the clinical efficiency of pharmaceuticals. Pulmonary delivery of nanoscale-based drug delivery systems offered a strategy to increase antitumor activity with minimal adverse exposure. Herein, we report an osimertinib-loaded perfluoro-15-crown-5-ether (AZD9291-PFCE) nanoemulsion, through intratracheal and intravenous delivery, synergizes with 19F magnetic resonance imaging (19F MRI)-guided low-intensity focused ultrasound (LIFU) for lung cancer therapy. Pulmonary delivery of AZD9291-PFCE nanoemulsion in orthotopic lung carcinoma models achieves quick distribution of the nanoemulsion in lung tissues and tumors without short-term and long-term toxic effects. Furthermore, LIFU can trigger drug release from the AZD9291-PFCE nanoemulsion and specifically increases tumor vascular and tumor tissue permeability. 19F MRI was applied to quantify nanoemulsion accumulation in tumors in real time after LIFU irradiation. We validate the treatment effect of AZD9291-PFCE nanoemulsion in resected human lung cancer tissues, proving the translational potential to enhance clinical outcomes of lung cancer therapy. Thus, this work presents a promising pulmonary nanoemulsion delivery system of osimertinib (AZD9291) for targeted therapy of lung cancer without severe side effects.
Collapse
Affiliation(s)
- Jie Yang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Yingbo Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Jiemei Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Hongyan Zou
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Yige Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Jing Luo
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Qian Xie
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Rong A
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Hongbin Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Xiaona Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Kai Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Lili Yang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Teng Ma
- Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055 Shenzhen, Guangdong, China
| | - Lina Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| | - Xilin Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 150028 Harbin, Heilongjiang, China
- Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, 150028 Harbin, Heilongjiang, China
| |
Collapse
|
28
|
Fan CH, Ho YJ, Lin CW, Wu N, Chiang PH, Yeh CK. State-of-the-art of ultrasound-triggered drug delivery from ultrasound-responsive drug carriers. Expert Opin Drug Deliv 2022; 19:997-1009. [PMID: 35930441 DOI: 10.1080/17425247.2022.2110585] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The development of new tools to locally and non-invasively transferring therapeutic substances at the desired site in deep living tissue has been a long sought-after goal within the drug delivery field. Among the established methods, ultrasound (US) with US-responsive carriers holds great promise and demonstrates on-demand delivery of a variety of functional substances with spatial precision of several millimeters in deep-seated tissues in animal models and humans. These properties have motivated several explorations of US with US responsive carriers as a modality for neuromodulation and the treatment of various diseases, such as stroke and cancer. AREAS COVERED This article briefly discussed three specific mechanisms that enhance in vivo drug delivery via US with US-responsive carriers: 1) permeabilizing cellular membrane, 2) increasing the permeability of vessels, and 3) promoting cellular endocytotic uptake. Besides, a series of US-responsive drug carriers are discussed, with an emphasis on the relation between structural feature and therapeutic outcome. EXPERT OPINION This article summarized current development for each of US-responsive drug carrier, focusing on the routes of enhancing delivery and applications. The mechanisms of interaction between US-responsive carriers and US energy, such as cavitation, hyperthermia, and reactive oxygen species, as well as how these interactions can improve drug delivery into target cell/tissue. It can be expected that there are serval efforts to further identification of US-responsive particles, design of novel US waveform sequence, and survey of optimal combination between US parameters and US-responsive carriers for better controlling the spatiotemporal drug release profile, stability, and safety in vivo. The authors believe these will provide novel tools for precisely designing treatment strategies and significantly benefit the clinical management of several diseases.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ju Ho
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Nan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Hua Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
29
|
Huang WQ, Zhu YQ, You W, Chen J, Gao F, Nie X, Zhang Z, Chen G, Yu Y, Xia L, Hong CY, Wang LH, Hao ZY, You YZ. Tumor Microenvironment Triggered the In Situ Synthesis of an Excellent Sonosensitizer in Tumor for Sonodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:26469-26479. [PMID: 35670468 DOI: 10.1021/acsami.2c05369] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
An ultrasound-triggered sonodynamic therapy has shown great promise for cancer therapy. However, its clinical applications are very limited because the traditional sonosensitizers tend to suffer from very poor efficiency combined with low retention in cancer cells and low tumor selectivity. Therefore, sonosensitizers with higher effectivity, higher tumor cell retention, and higher tumor cell specificity are highly required. Herein, we constructed a Ti2C(OH)X nanosheet, which was a poor sonosensitizer but had a long circulation in the blood system. However, it was very interesting to find that the tumor microenvironment could in situ turn Ti2C(OH)X nanosheet into a novel and excellent sonosensitizer with a nanofiber structure in tumors, exhibiting excellent ability to generate reactive oxygen species (ROS) under ultrasound. Moreover, the nanofiber structure made it very difficult to get out of cancer cells, highly enhancing the retention of the sonosensitizer in the tumor, thereby enabling it to effectively and selectively kill cancer cells in vivo. Our findings demonstrate that the strategy of the tumor microenvironment triggering the in situ synthesis of an effective sonosensitizer in tumor provided a promising means to simultaneously increase the efficiency, sonosensitizer retention in cancer cells, and cancer selectivity, thereby effectively killing cancer cells but causing little damage to healthy tissues via the sonodynamic therapy.
Collapse
Affiliation(s)
- Wei-Qiang Huang
- The Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ya-Qi Zhu
- The Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Wei You
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jing Chen
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Fan Gao
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xuan Nie
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ze Zhang
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Guang Chen
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yue Yu
- The Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Lei Xia
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Chun-Yan Hong
- Hefei National Laboratory for Physical Science at Microscale, Hefei, Anhui 230026, China
| | - Long-Hai Wang
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zong-Yao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Ye-Zi You
- The Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
30
|
Luo J. TGF-β as a Key Modulator of Astrocyte Reactivity: Disease Relevance and Therapeutic Implications. Biomedicines 2022; 10:1206. [PMID: 35625943 PMCID: PMC9138510 DOI: 10.3390/biomedicines10051206] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are essential for normal brain development and functioning. They respond to brain injury and disease through a process referred to as reactive astrogliosis, where the reactivity is highly heterogenous and context-dependent. Reactive astrocytes are active contributors to brain pathology and can exert beneficial, detrimental, or mixed effects following brain insults. Transforming growth factor-β (TGF-β) has been identified as one of the key factors regulating astrocyte reactivity. The genetic and pharmacological manipulation of the TGF-β signaling pathway in animal models of central nervous system (CNS) injury and disease alters pathological and functional outcomes. This review aims to provide recent understanding regarding astrocyte reactivity and TGF-β signaling in brain injury, aging, and neurodegeneration. Further, it explores how TGF-β signaling modulates astrocyte reactivity and function in the context of CNS disease and injury.
Collapse
Affiliation(s)
- Jian Luo
- Palo Alto Veterans Institute for Research, VAPAHCS, Palo Alto, CA 94304, USA
| |
Collapse
|
31
|
Shamloo A, Boroumand A, Ebrahimi S, Kalantarnia F, Maleki S, Moradi H. Modeling of an Ultrasound System in Targeted Drug Delivery to Abdominal Aortic Aneurysm: A Patient-Specific in Silico Study Based on Ligand-Receptor Binding. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:967-974. [PMID: 34958631 DOI: 10.1109/tuffc.2021.3138868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Targeted drug delivery methods have shown a significant impact on enhancing drug delivery efficiency and reducing drug side effects. While various stimuli have been used to promote the drug delivery process, applying ultrasound (US) waves to control drug particles through the human body, noninvasively, has drawn the scientist's attention. However, microcarriers delivery reaches the aneurysmal artery by US waves that exert volumetric forces on blood, and drug carriers, which can therefore affect blood flow patterns and movement pathways of drug carriers, have not yet been studied. In this study, we developed a 3-D patient-specific model of abdominal aortic aneurysm (AAA) to evaluate the effect of US waves in enhancing the drug-containing microbubbles (MBs) adhered on the AAA lumen through ligand-receptor binding. Thus, a focused US (FUS) transducer with a resonance frequency of ~1.1 MHz was added to the geometry. Then, the surface density of MBs (SDM) adhered on the AAA lumen was calculated at peak acoustic pressure of ~1.1, ~2.2, and ~4.3 MPa. Results indicated that increasing the US pressure had a significant impact on improving the MBs adhered to the intended wall, whereby US waves with the maximum pressure of ~4.3 MPa could enhance ~1- [Formula: see text] MBs adhesion ~98% relative to not using the waves. While US waves have the advantage of more SDM adhered to the whole artery wall, they adversely affect the SDM adhered on the critical wall of the abdominal aorta. Furthermore, when the US strength goes up, a reduction occurs in the SDM adhered. This reduction is higher for smaller MBs, which is the mentioned MBs' size and US strength reduced SDM adhesion by about ~50% relative to systemic injection. Therefore, it can be concluded that drug delivery using the US field increases the SDM adhered to the whole AAA wall and decreases the SDM adhered to the critical wall of AAA.
Collapse
|
32
|
An On-Demand Drug Delivery System for Control of Epileptiform Seizures. Pharmaceutics 2022; 14:pharmaceutics14020468. [PMID: 35214199 PMCID: PMC8879600 DOI: 10.3390/pharmaceutics14020468] [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: 12/20/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 02/03/2023] Open
Abstract
Drug delivery systems have the potential to deliver high concentrations of drug to target areas on demand, while elsewhere and at other times encapsulating the drug, to limit unwanted actions. Here we show proof of concept in vivo and ex vivo tests of a novel drug delivery system based on hollow-gold nanoparticles tethered to liposomes (HGN-liposomes), which become transiently permeable when activated by optical or acoustic stimulation. We show that laser or ultrasound simulation of HGN-liposomes loaded with the GABAA receptor agonist, muscimol, triggers rapid and repeatable release in a sufficient concentration to inhibit neurons and suppress seizure activity. In particular, laser-stimulated release of muscimol from previously injected HGN-liposomes caused subsecond hyperpolarizations of the membrane potential of hippocampal pyramidal neurons, measured by whole cell intracellular recordings with patch electrodes. In hippocampal slices and hippocampal–entorhinal cortical wedges, seizure activity was immediately suppressed by muscimol release from HGN-liposomes triggered by laser or ultrasound pulses. After intravenous injection of HGN-liposomes in whole anesthetized rats, ultrasound stimulation applied to the brain through the dura attenuated the seizure activity induced by pentylenetetrazol. Ultrasound alone, or HGN-liposomes without ultrasound stimulation, had no effect. Intracerebrally-injected HGN-liposomes containing kainic acid retained their contents for at least one week, without damage to surrounding tissue. Thus, we demonstrate the feasibility of precise temporal control over exposure of neurons to the drug, potentially enabling therapeutic effects without continuous exposure. For future application, studies on the pharmacokinetics, pharmacodynamics, and toxicity of HGN-liposomes and their constituents, together with improved methods of targeting, are needed, to determine the utility and safety of the technology in humans.
Collapse
|
33
|
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: 4.3] [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
|
34
|
Jordan S, Zielinski M, Kortylewski M, Kuhn T, Bystritsky A. Noninvasive Delivery of Biologicals to the Brain. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2022; 20:64-70. [PMID: 35746928 PMCID: PMC9063603 DOI: 10.1176/appi.focus.20210028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the past, psychotherapy and neuropharmacological approaches have been the most common treatments for disordered thoughts, moods, and behaviors. One new path of brain therapeutics is in the deployment of noninvasive approaches designed to reprogram brain function at the cellular level. Treatment at the cellular level may be considered for a wide array of disorders, ranging from mood disorders to neurodegenerative disorders. Brain-targeted biological therapy may provide minimally invasive and accurate delivery of treatment. The present article discusses the hurdles and advances that characterize the pathway to this goal.
Collapse
|
35
|
Lo WC, Huang YL, Fan CH, Yeh CK. 3-D Ultrafast Ultrasound Imaging of Microbubbles Trapped Using an Acoustic Vortex. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:3507-3514. [PMID: 34228623 DOI: 10.1109/tuffc.2021.3095241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Increasing the local concentration of microbubbles (MBs) within the blood flow plays a crucial role in several medical applications, but there are few imaging modalities available for volumetric tracking of the aggregated MBs in real time. Here we describe a device integrating acoustic vortex tweezers (AVTs) and ultrasound plane-wave imaging (PWI) to achieve the goal of controlling the spatial distribution of MBs in blood vessels and simultaneously monitoring this process using the same probe. Experiments were conducted using a 5-MHz 2-D array ultrasound probe (with three cycles of excitation at an acoustic pressure of 2000 kPa) and 1.2- [Formula: see text]-diameter MBs at a flow rate of 20 mm/s. The AVT waveform was produced by modulating the repetition frequency of the transmitted pulse asymmetrically (4 and 8 kHz at the inflow and outflow ends, respectively). In order to simultaneously capture MBs and carry out imaging with the same probe, the asymmetric AVT pulse signal and the ultrasound-imaging pulse signal were arranged in a staggered series, and the imaging was carried out using plane-wave pulses at nine angles (-7° to 7°) in compounded PWI (volume rate: 200 Hz). Microscopy observations showed that freely suspended MBs could indeed be gathered by the asymmetric AVT in the flow field to form an MBs cluster with a spot size of about [Formula: see text], which could resist the flow to remain at a fixed location for about 22 s. After the asymmetric AVT signal and the ultrasound-imaging pulse signal were turned on for 1 s, the ultrasound 3-D image showed that the signal intensity of the MB clusters increased by 13.1 dB ± 2.9 dB in relation to the background area. These results show that the proposed strategy can be used to accumulate flowing MBs at a desired location and to simultaneously observe this phenomenon. This tool could be used in the future to improve the outcomes of MB-related treatments for various diseases.
Collapse
|
36
|
Zafar A, Hasan M, Tariq T, Dai Z. Enhancing Cancer Immunotherapeutic Efficacy with Sonotheranostic Strategies. Bioconjug Chem 2021; 33:1011-1034. [PMID: 34793138 DOI: 10.1021/acs.bioconjchem.1c00437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immunotherapy has revolutionized the modality for establishing a firm immune response and immunological memory. However, intrinsic limitations of conventional low responsive poor T cell infiltration and immune related adverse effects urge the coupling of cancer nanomedicines with immunotherapy for boosting antitumor response under ultrasound (US) sensitization to mimic dose-limiting toxicities for safe and effective therapy against advanced cancer. US is composed of high-frequency sound waves that mediate targeted spatiotemporal control over release and internalization of the drug. The unconventional US triggered immunogenic nanoengineered arena assists the limited immunogenic dose, limiting toxicities and efficacies. In this Review, we discuss current prospects of enhanced immunotherapy using nanomedicine under US. We highlight how nanotechnology designs and incorporates nanomedicines for the reprogramming of systematic immunity in the tumor microenvironment. We also emphasize the mechanical and biological potential of US, encompassing sonosensitizer activation for enhanced immunotherapeutic efficacies. Finally, the smartly converging combinational platform of US stimulated cancer nanomedicines for amending immunotherapy is summarized. This Review will widen scientists' ability to explore and understand the limiting factors for combating cancer in a precisely customized way.
Collapse
Affiliation(s)
- Ayesha Zafar
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Murtaza Hasan
- School of Chemistry and Chemical Engineering, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Tuba Tariq
- Department of Biochemistry and Biotechnology, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
37
|
Xia B, Sebesta C, Lee S, Nair V, Zhao X, Coffler S, Robinson JT, Szablowski JO. Biohybrid approaches to interface with the nervous system: the best of both worlds. Curr Opin Biotechnol 2021; 72:86-94. [PMID: 34735989 DOI: 10.1016/j.copbio.2021.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/27/2021] [Accepted: 10/13/2021] [Indexed: 10/20/2022]
Abstract
Synthetic materials and devices that interact with light, ultrasound, or magnetic fields can be used to modulate neural activity with high spatial and temporal precision; however, these approaches often lack the ability to target genetically defined cell types and signaling pathways. Genetically encoded proteins can be expressed to modify the host tissue and provide cellular and molecular specificity, but compared to synthetic materials, these proteins often interact weakly with externally applied energy sources. Synthetic materials can respond to optical, acoustic, and magnetic stimuli to focus, convert, and amplify forms of energy to ones that are more accessible to engineered cells and proteins. By combining the devices, synthetic materials, and genetically encoded proteins or cells, researchers can gain the ability to interface with the nervous system with improved spatiotemporal, cell-type and molecular precision. Here we review recent advances in these 'biohybrid' approaches that use optical, acoustic, and magnetic energy sources.
Collapse
Affiliation(s)
- Boao Xia
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77005, USA; Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, 77030, USA
| | - Charles Sebesta
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77005, USA; Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, 77030, USA
| | - Sangsin Lee
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77005, USA; Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, 77030, USA
| | - Vishnu Nair
- Department of Electrical and Computer Engineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77251, USA; Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, 77030, USA
| | - Xuan Zhao
- Department of Electrical and Computer Engineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77251, USA; Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, 77030, USA; Applied Physics Graduate Program, Smalley-Curl Institute, Rice University 77251, Houston, TX, USA
| | - Samantha Coffler
- Department of Electrical and Computer Engineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77251, USA; Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, 77030, USA; Systems, Synthetic, and Physical Biology PhD Program, Rice University, Houston, TX, 77251, USA
| | - Jacob T Robinson
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77005, USA; Department of Electrical and Computer Engineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77251, USA; Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, 77030, USA.
| | - Jerzy O Szablowski
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX, 77005, USA; Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, 77030, USA; Applied Physics Graduate Program, Smalley-Curl Institute, Rice University 77251, Houston, TX, USA; Systems, Synthetic, and Physical Biology PhD Program, Rice University, Houston, TX, 77251, USA.
| |
Collapse
|
38
|
Choukér A, Ngo-Anh TJ, Biesbroek R, Heldmaier G, Heppener M, Bereiter-Hahn J. European space agency's hibernation (torpor) strategy for deep space missions: Linking biology to engineering. Neurosci Biobehav Rev 2021; 131:618-626. [PMID: 34606822 DOI: 10.1016/j.neubiorev.2021.09.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/10/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022]
Abstract
Long-duration space missions to Mars will impose extreme stresses of physical and psychological nature on the crew, as well as significant logistical and technical challenges for life support and transportation. Main challenges include optimising overall mass and maintaining crew physical and mental health. These key scopes have been taken up as the baseline for a study by the European Space Agency (ESA) using its Concurrent Design Facility (CDF). It focussed on the biology of hibernation in reducing metabolism and hence stress, and its links to the infrastructure and life support. We concluded that torpor of crew members can reduce the payload with respect to oxygen, food and water but will require monitoring and artificial intelligence (AI) assisted monitoring of the crew. These studies additionally offer new potential applications for patient care on Earth. Keywords: Space flight, concurrent design facility, metabolic reduction.
Collapse
Affiliation(s)
- Alexander Choukér
- Laboratory of Translational Research "Stress and Immunity", Department of Anesthesiology, Hospital of the Ludwig-Maximilians-University, Marchioninistrasse 15, 81377, Munich, Germany
| | - Thu Jennifer Ngo-Anh
- Directorate of Human and Robotic Exploration Programmes, European Space Agency, P.O. Box 299, 2200 AG, Noordwijk, the Netherlands
| | - Robin Biesbroek
- Directorate of Technology, Engineering and Quality, European Space Agency, P.O. Box 299, 2200 AG, Noordwijk, the Netherlands
| | - Gerhard Heldmaier
- Animal Physiology, Department of Biology, Marburg University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Marc Heppener
- (c)/o Directorate of Human and Robotic Exploration Programmes, European Space Agency, P.O. Box 299, 2200 AG, Noordwijk, the Netherlands
| | - Jürgen Bereiter-Hahn
- Institute for Cell Biology and Neurosciences, Goethe University Frankfurt, Max-von-Lauestr. 19, D 6438, Frankfurt Am Main, Germany.
| |
Collapse
|
39
|
Zhang L, Sun H, Zhao J, Lee J, Ee Low L, Gong L, Chen Y, Wang N, Zhu C, Lin P, Liang Z, Wei M, Ling D, Li F. Dynamic nanoassemblies for imaging and therapy of neurological disorders. Adv Drug Deliv Rev 2021; 175:113832. [PMID: 34146626 DOI: 10.1016/j.addr.2021.113832] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/07/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
The past decades have witnessed an increased incidence of neurological disorders (NDs) such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, ischemic stroke, and epilepsy, which significantly lower patients' life quality and increase the economic and social burden. Recently, nanomedicines composed of imaging and/or therapeutic agents have been explored to diagnose and/or treat NDs due to their enhanced bioavailability, blood-brain barrier (BBB) permeability, and targeting capacity. Intriguingly, dynamic nanoassemblies self-assembled from functional nanoparticles to simultaneously interfere with multiple pathogenic substances and pathological changes, have been regarded as one of the foremost candidates to improve the diagnostic and therapeutic efficacy of NDs. To help readers better understand this emerging field, in this review, the pathogenic mechanism of different types of NDs is briefly introduced, then the functional nanoparticles used as building blocks in the construction of dynamic nanoassemblies for NDs theranostics are summarized. Furthermore, dynamic nanoassemblies that can actively cross the BBB to target brain lesions, sensitively and efficiently diagnose or treat NDs, and effectively promote neuroregeneration are highlighted. Finally, we conclude with our perspectives on the future development in this field.
Collapse
|
40
|
Franco-Urquijo CA, Navarro-Becerra JÁ, Ríos A, Escalante B. Release of vascular agonists from liposome-microbubble conjugate by ultrasound-mediated microbubble destruction: effect on vascular function. Drug Deliv Transl Res 2021; 12:1175-1186. [PMID: 33939122 DOI: 10.1007/s13346-021-00994-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 11/26/2022]
Abstract
The endothelium is a single cell layer of the vessel wall and a key regulator of blood flow in vascular beds. Local and systemic pathologies have been associated with alterations in endothelial function. However, targeting the endothelium with vasoconstrictor or vasodilator drugs is often accompanied by systemic effects. Here, we evaluated a liposome-microbubble delivery system as a vascular hydrophilic agonist carrier. Phenylephrine (Phe) or acetylcholine (Ach)-loaded liposomes were conjugated to microbubbles. The drug release was triggered by ultrasound (US), and the vascular response was assessed in rat aortic rings using an isolated organ chamber. Aortic rings incubated with Phe-liposome-microbubble conjugate, exposed to US showed a marked contractile response (0.79 ± 0.04 g) compared to empty liposomes conjugated to microbubbles, aortic rings exposed only to US, and Phe-liposome-microbubble conjugate without US exposure that elicited a minimal or no response. Expressed as %, contractile responses were 85.24 ± 4.31% and 12.62 ± 3.23% for Phe-Chol-liposome-microbubble conjugate and empty Chol-liposome-microbubble conjugate exposed to US, respectively. Addition of 1 × 10-5 M Ach to pre-contracted aortic rings decreased the contraction response from 1 to 0.21 g. The addition of Ach-liposome conjugate and exposure to US decreased the contraction response to 0.32 g. Additionally, the ED50 values for Phe and Ach released by US from liposome-microbubble conjugates were 3.6 × 10-8 M ± 2.8 × 10-9 M for Phe and 2.0 × 10-8 M ± 1.8 × 10-9 M. In conclusion, we evaluated a hybrid delivery system that consisted of loaded liposomes conjugated to microbubbles to deliver and release vascular agonists using UMMD.
Collapse
Affiliation(s)
- Carlos A Franco-Urquijo
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Vía del Conocimiento 201, PIIT, NL, Apodaca, Mexico
| | - J Ángel Navarro-Becerra
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Vía del Conocimiento 201, PIIT, NL, Apodaca, Mexico
- Department of Mechanical Engineering, University of Colorado, 1111 Engineering Drive, Boulder, CO, USA
| | - Amelia Ríos
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Vía del Conocimiento 201, PIIT, NL, Apodaca, Mexico.
| | - Bruno Escalante
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Vía del Conocimiento 201, PIIT, NL, Apodaca, Mexico
- Universidad de Monterrey, Av. Ignacio Morones Prieto 4500, San Pedro Garza García, NL, Mexico
| |
Collapse
|
41
|
Improving Release of Liposome-Encapsulated Drugs with Focused Ultrasound and Vaporizable Droplet-Liposome Nanoclusters. Pharmaceutics 2021; 13:pharmaceutics13050609. [PMID: 33922219 PMCID: PMC8145150 DOI: 10.3390/pharmaceutics13050609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/14/2021] [Accepted: 04/18/2021] [Indexed: 12/13/2022] Open
Abstract
Active targeted delivery of small molecule drugs is becoming increasingly important in personalized therapies, especially in cancer, brain disorders, and a wide variety of other diseases. However, effective means of spatial targeting and delivering high drug payloads in vivo are still lacking. Focused ultrasound combined with superheated phase-shift nanodroplets, which vaporize into microbubbles using heat and sound, are rapidly becoming a popular strategy for targeted drug delivery. Focused ultrasound can target deep tissue with excellent spatial precision and without using ionizing energy, thus can activate nanodroplets in circulation. One of the main limitations of this technology has been poor drug loading in the droplet core or the shell material. To address this need, we have developed a strategy to combine low-boiling point decafluorabutane and octafluoropropane (DFB and OFP) nanodroplets with drug-loaded liposomes, creating phase-changeable droplet-liposome clusters (PDLCs). We demonstrate a facile method of assembling submicron PDLCs with high drug-loading capacity on the droplet surface. Furthermore, we demonstrate that chemical tethering of liposomes in PDLCs enables a rapid release of their encapsulated cargo upon acoustic activation (>60% using OFP-based PDLCs). Rapid uncaging of small molecule drugs would make them immediately bioavailable in target tissue or promote better penetration in local tissue following intravascular release. PDLCs developed in this study can be used to deliver a wide variety of liposome-encapsulated therapeutics or imaging agents for multi-modal imaging applications. We also outline a strategy to deliver a surrogate encapsulated drug, fluorescein, to tumors in vivo using focused ultrasound energy and PDLCs.
Collapse
|
42
|
Industry News October 2020. Ther Deliv 2021; 12:111-117. [PMID: 33455454 DOI: 10.4155/tde-2020-0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|