1
|
Géraudie A, De Rossi P, Canney M, Carpentier A, Delatour B. Effects of blood-brain barrier opening using ultrasound on tauopathies: A systematic review. J Control Release 2025; 379:1029-1044. [PMID: 39875073 DOI: 10.1016/j.jconrel.2025.01.056] [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: 11/22/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
Blood-brain barrier opening with ultrasound can potentiate drug efficacy in the treatment of brain pathologies and also provides therapeutic effects on its own. It is an innovative tool to transiently, repeatedly and safely open the barrier, with studies showing beneficial effects in both preclinical models for Alzheimer's disease and recent clinical studies. The first preclinical and clinical work has mainly shown a decrease in amyloid burden in mice models and in patients. However, Alzheimer's disease pathology also encompasses tauopathy, which is closely related to cognitive decline, making it a crucial therapeutic target. The effects of blood-brain barrier opening with ultrasound have been rarely assessed on tau and are still unclear. METHODS This systematic review, conducted through searches using Pubmed, Embase, Web of Science and Cochrane Central databases, extracted results of 15 studies reporting effects of blood-brain barrier opening using ultrasound on tau proteins. RESULTS This review of the literature indicates that blood-brain barrier opening using ultrasound can decrease the extent of the tau pathology or potentialize the effect of a therapeutic drug. However, selected studies report paradoxically that blood-brain barrier opening can increase tau pathology burden and induce brain damage. DISCUSSION Apparent discrepancies between reports could originate from the variability in protocols or analytical methods that may impact the effects of blood-brain barrier opening with ultrasound on tauopathies, glial populations, tissue integrity and functional outcomes. CONCLUSION This calls for a better standardization effort combined with improved methodologies allowing between-studies comparisons, and for further understanding of the effects of blood-brain barrier opening on tau pathology as an essential prerequisite before translation to clinic.
Collapse
Affiliation(s)
- Amandine Géraudie
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France.
| | | | | | - Alexandre Carpentier
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France; Faculty of Medicine, Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France; Advanced Surgical Research Technology Lab, Sorbonne University, 75013 Paris, France
| | - Benoît Delatour
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France
| |
Collapse
|
2
|
Capilla-López MD, Deprada A, Andrade-Talavera Y, Martínez-Gallego I, Coatl-Cuaya H, Sotillo P, Rodríguez-Alvarez J, Rodríguez-Moreno A, Parra-Damas A, Saura CA. Synaptic vulnerability to amyloid-β and tau pathologies differentially disrupts emotional and memory neural circuits. Mol Psychiatry 2025:10.1038/s41380-025-02901-9. [PMID: 39885298 DOI: 10.1038/s41380-025-02901-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/22/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Alzheimer's disease (AD) is characterized by memory loss and neuropsychiatric symptoms associated with cerebral amyloid-β (Aβ) and tau pathologies, but whether and how these factors differentially disrupt neural circuits remains unclear. Here, we investigated the vulnerability of memory and emotional circuits to Aβ and tau pathologies in mice expressing mutant human amyloid precursor protein (APP), Tau or both APP/Tau in excitatory neurons. APP/Tau mice develop age- and sex-dependent Aβ and phosphorylated tau pathologies, the latter exacerbated at early stages, in vulnerable brain regions. Early memory deficits were associated with hippocampal tau pathology in Tau and APP/Tau mice, whereas anxiety and fear appeared linked to intracellular Aβ in the basolateral amygdala (BLA) of APP and APP/Tau mice. Transcriptome hippocampal profiling revealed gene changes affecting myelination and RNA processing in Tau mice, and inflammation and synaptic-related pathways in APP/Tau mice at 6 months. At 9 months, we detected common and region-specific changes in astrocytic, microglia and 63 AD-associated genes in the hippocampus and BLA of APP/Tau mice. Spatial learning deficits were associated with synaptic tau accumulation and synapse disruption in the hippocampus of Tau and APP/Tau mice, whereas emotional disturbances were linked to Aβ pathology but not synaptic tau in the BLA. Interestingly, Aβ and tau exhibited synergistic detrimental effects in long-term potentiation (LTP) in the hippocampus but they counteract with each other to mitigate LTP impairments in the amygdala. These findings indicate that Aβ and tau pathologies cause region-specific effects and synergize to induce synaptic dysfunction and immune responses, contributing to the differing vulnerability of memory and emotional neural circuits in AD.
Collapse
Affiliation(s)
- Maria Dolores Capilla-López
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Angel Deprada
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Irene Martínez-Gallego
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Sevilla, Spain
| | - Heriberto Coatl-Cuaya
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Sevilla, Spain
| | - Paula Sotillo
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José Rodríguez-Alvarez
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antonio Rodríguez-Moreno
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Sevilla, Spain
| | - Arnaldo Parra-Damas
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Carlos A Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
3
|
Șovrea AS, Boșca AB, Dronca E, Constantin AM, Crintea A, Suflețel R, Ștefan RA, Ștefan PA, Onofrei MM, Tschall C, Crivii CB. Non-Drug and Non-Invasive Therapeutic Options in Alzheimer's Disease. Biomedicines 2025; 13:84. [PMID: 39857667 PMCID: PMC11760896 DOI: 10.3390/biomedicines13010084] [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: 12/16/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Despite the massive efforts of modern medicine to stop the evolution of Alzheimer's disease (AD), it affects an increasing number of people, changing individual lives and imposing itself as a burden on families and the health systems. Considering that the vast majority of conventional drug therapies did not lead to the expected results, this review will discuss the newly developing therapies as an alternative in the effort to stop or slow AD. Focused Ultrasound (FUS) and its derived Transcranial Pulse Stimulation (TPS) are non-invasive therapeutic approaches. Singly or as an applied technique to change the permeability of the blood-brain-barrier (BBB), FUS and TPS have demonstrated the benefits of use in treating AD in animal and human studies. Adipose-derived stem Cells (ADSCs), gene therapy, and many other alternative methods (diet, sleep pattern, physical exercise, nanoparticle delivery) are also new potential treatments since multimodal approaches represent the modern trend in this disorder research therapies.
Collapse
Affiliation(s)
- Alina Simona Șovrea
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Adina Bianca Boșca
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Eleonora Dronca
- Molecular Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (E.D.); (A.C.)
| | - Anne-Marie Constantin
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Andreea Crintea
- Molecular Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (E.D.); (A.C.)
| | - Rada Suflețel
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Roxana Adelina Ștefan
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Paul Andrei Ștefan
- Radiology and Imaging Department, Emergency County Hospital Cluj, 400347 Cluj-Napoca, Romania;
| | - Mădălin Mihai Onofrei
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Christoph Tschall
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Carmen-Bianca Crivii
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| |
Collapse
|
4
|
Tang M, Guo JJ, Guo RX, Xu SJ, Lou Q, Hu QX, Li WY, Yu JB, Yao Q, Wang QW. Progress of research and application of non-pharmacologic intervention in Alzheimer's disease. J Alzheimers Dis 2024; 102:275-294. [PMID: 39573867 DOI: 10.1177/13872877241289396] [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: 11/26/2024]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by amyloid-β (Aβ) deposition and neurofibrillary tangles formed by high phosphorylation of tau protein. At present, drug therapy is the main strategy of AD treatment, but its effects are limited to delaying or alleviating AD. Recently, non-pharmacologic intervention has attracted more attention, and more studies have confirmed that non-pharmacologic intervention in AD can improve the patient's cognitive function and quality of life. This paper summarizes the current non-pharmacologic intervention in AD, to provide useful supplementary means for AD intervention.
Collapse
Affiliation(s)
- Min Tang
- Ningbo Rehabilitation Hospital, Ningbo, Zhejiang, China
| | - Jie-Jie Guo
- The First People's Hospital of Wenling, Taizhou, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Rong-Xia Guo
- School of Teacher Education, Ningbo University, Ningbo, Zhejiang, China
| | - Shu-Jun Xu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Qiong Lou
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qiao-Xia Hu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Wan-Yi Li
- Ningbo Rehabilitation Hospital, Ningbo, Zhejiang, China
| | - Jing-Bo Yu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qi Yao
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qin-Wen Wang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
5
|
Lu X, Sun W, Leng L, Yang Y, Gong S, Zou Q, Niu H, Wei C. Ultrasound Stimulation Modulates Microglia M1/M2 Polarization and Affects Hippocampal Proteomic Changes in a Mouse Model of Alzheimer's Disease. Immun Inflamm Dis 2024; 12:e70061. [PMID: 39588954 PMCID: PMC11590030 DOI: 10.1002/iid3.70061] [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: 06/13/2024] [Revised: 09/22/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND The effectiveness of ultrasound stimulation in treating Alzheimer's disease (AD) has been reported in previous studies, but the underlying mechanisms remain unclear. This study investigated the effects of ultrasound stimulation on the proportion and function of microglia of different phenotypes, as well as on the levels of inflammatory factors. Additionally, it revealed the alterations in proteomic molecules in the mouse hippocampus following ultrasound stimulation treatment, aiming to uncover potential new molecular mechanisms. METHODS Ultrasound stimulation was used to stimulate the hippocampus for 30 min per day for 5 days in the ultrasound stimulation-treated group. Amyloid plaque deposition was measured using immunofluorescence staining. M1 and M2 type microglia were labeled using immunofluorescent double staining, and the ratio was calculated. The levels of Aβ42, IL-10, and TNF-α were determined using ELISA kits. The quantitative proteomics method was employed to explore molecular changes in hippocampal proteins. RESULTS Ultrasound stimulation treatment reduced the average fluorescence intensity of amyloid plaques and the concentration of Aβ42. Compared to the AD group, ultrasound stimulation resulted in a 14% reduction in the proportion of M1 microglia and a 12% increase in the proportion of M2 microglia. The concentration of the anti-inflammatory factor IL-10 was significantly increased in the ultrasound stimulation-treated group. Proteomics analysis revealed 753 differentially expressed proteins between the ultrasound stimulation-treated and AD groups, with most being enriched in the oxidative phosphorylation pathway of mitochondria. Additionally, the activity of cytochrome c oxidase, involved in oxidative phosphorylation, was increased after ultrasound stimulation treatment. CONCLUSIONS Ultrasound stimulation affects microglial polarization, reduces amyloid plaque load, and enhances levels of anti-inflammatory factors in APP/PS1 mice. Proteomics analysis reveals molecular changes in hippocampal proteins after ultrasound stimulation treatment. The mechanism behind ultrasound stimulation-induced modulation of microglial polarization may be related to changes in mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Xinliang Lu
- Department of Neurology, Xuan Wu HospitalCapital Medical UniversityBeijingChina
- School of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Wenxian Sun
- Department of Neurology, Xuan Wu HospitalCapital Medical UniversityBeijingChina
| | - Li Leng
- School of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Yuting Yang
- Department of Neurology, Xuan Wu HospitalCapital Medical UniversityBeijingChina
- School of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Shuting Gong
- Department of Neurology, Xuan Wu HospitalCapital Medical UniversityBeijingChina
- School of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Qi Zou
- Department of Neurology, Xuan Wu HospitalCapital Medical UniversityBeijingChina
| | - Haijun Niu
- School of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Cuibai Wei
- Department of Neurology, Xuan Wu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
6
|
Mehta RI, Ranjan M, Haut MW, Carpenter JS, Rezai AR. Focused Ultrasound for Neurodegenerative Diseases. Magn Reson Imaging Clin N Am 2024; 32:681-698. [PMID: 39322357 DOI: 10.1016/j.mric.2024.03.001] [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: 09/27/2024]
Abstract
Neurodegenerative diseases are a leading cause of death and disability and pose a looming global public health crisis. Despite progress in understanding biological and molecular factors associated with these disorders and their progression, effective disease modifying treatments are presently limited. Focused ultrasound (FUS) is an emerging therapeutic strategy for Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In these contexts, applications of FUS include neuroablation, neuromodulation, and/or blood-brain barrier opening with and without facilitated intracerebral drug delivery. Here, the authors review preclinical evidence and current and emerging applications of FUS for neurodegenerative diseases and summarize future directions in the field.
Collapse
Affiliation(s)
- Rashi I Mehta
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University; Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University.
| | - Manish Ranjan
- Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University
| | - Marc W Haut
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University; Department of Behavioral Medicine and Psychiatry, Rockefeller Neuroscience Institute, West Virginia University; Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University
| | - Jeffrey S Carpenter
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University; Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University; Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University
| | - Ali R Rezai
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University; Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University
| |
Collapse
|
7
|
Zhang Y, Yu W, Zhang L, Li P. Application of engineered antibodies (scFvs and nanobodies) targeting pathological protein aggregates in Alzheimer's disease. Expert Opin Investig Drugs 2024; 33:1047-1062. [PMID: 39177331 DOI: 10.1080/13543784.2024.2396911] [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: 02/22/2024] [Revised: 07/07/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION The misfolding and aggregation of proteins are associated with various neurodegenerative diseases, such as Alzheimer's disease (AD). The small-molecule engineered antibodies, such as single-chain fragment variable (scFv) antibodies and nanobodies (Nbs), have gained attention in recent years due to their strong conformational specificity, ability to cross the blood-brain barrier (BBB), low immunogenicity, and enhanced proximity to active sites within aggregates. AREAS COVERED We have reviewed recent advances in therapies involving scFvs and Nbs that efficiently and specifically target pathological protein aggregates. Relevant publications were searched for in MEDLINE, GOOGLE SCHOLAR, Elsevier ScienceDirect and Wiley Online Library. EXPERT OPINION We reviewed the recent and specific targeting of pathological protein aggregates by scFvs and Nbs. These engineered antibodies can inhibit the aggregation or promote the disassembly of misfolded proteins by recognizing antigenic epitopes or through conformational specificity. Additionally, we discuss strategies for improving the effective application of engineered antibodies in treating AD. These technological strategies will lay the foundation for the clinical application of small-molecule antibody drugs in developing effective treatments for neurological diseases. Through rational application strategies, small-molecule engineered antibodies are expected to have significant potential in targeted therapy for neurological disorders.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wanpeng Yu
- Medical Collage, Qingdao University, Qingdao, China
| | - Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
8
|
McMahon D, Jones RM, Ramdoyal R, Zhuang JYX, Leavitt D, Hynynen K. Investigation of Sonication Parameters for Large-Volume Focused Ultrasound-Mediated Blood-Brain Barrier Permeability Enhancement Using a Clinical-Prototype Hemispherical Phased Array. Pharmaceutics 2024; 16:1289. [PMID: 39458618 PMCID: PMC11510584 DOI: 10.3390/pharmaceutics16101289] [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: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Focused ultrasound (FUS) and microbubble (MB) exposure is a promising technique for targeted drug delivery to the brain; however, refinement of protocols suitable for large-volume treatments in a clinical setting remains underexplored. Methods: Here, the impacts of various sonication parameters on blood-brain barrier (BBB) permeability enhancement and tissue damage were explored in rabbits using a clinical-prototype hemispherical phased array developed in-house, with real-time 3D MB cavitation imaging for exposure calibration. Initial experiments revealed that continuous manual agitation of MBs during infusion resulted in greater gadolinium (Gd) extravasation compared to gravity drip infusion. Subsequent experiments used low-dose MB infusion with continuous agitation and a low burst repetition frequency (0.2 Hz) to mimic conditions amenable to long-duration clinical treatments. Results: Key sonication parameters-target level (proportional to peak negative pressure), number of bursts, and burst length-significantly affected BBB permeability enhancement, with all parameters displaying a positive relationship with relative Gd contrast enhancement (p < 0.01). Even at high levels of BBB permeability enhancement, tissue damage was minimal, with low occurrences of hypointensities on T2*-weighted MRI. When accounting for relative Gd contrast enhancement, burst length had a significant impact on red blood cell extravasation detected in histological sections, with 1 ms bursts producing significantly greater levels compared to 10 ms bursts (p = 0.03), potentially due to the higher pressure levels required to generate equal levels of BBB permeability enhancement. Additionally, albumin and IgG extravasation correlated strongly with relative Gd contrast enhancement across sonication parameters, suggesting that protein extravasation can be predicted from non-invasive imaging. Conclusions: These findings contribute to the development of safer and more effective clinical protocols for FUS + MB exposure, potentially improving the efficacy of the approach.
Collapse
Affiliation(s)
- Dallan McMahon
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (R.M.J.); (K.H.)
| | - Ryan M. Jones
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (R.M.J.); (K.H.)
| | - Rohan Ramdoyal
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (R.M.J.); (K.H.)
| | - Joey Ying Xuan Zhuang
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (R.M.J.); (K.H.)
| | - Dallas Leavitt
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (R.M.J.); (K.H.)
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (R.M.J.); (K.H.)
- Department of Medical Biophysics, University of Toronto, Toronto, ON M4N 3M5, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
9
|
O'Reilly MA. Exploiting the mechanical effects of ultrasound for noninvasive therapy. Science 2024; 385:eadp7206. [PMID: 39265013 DOI: 10.1126/science.adp7206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/07/2024] [Indexed: 09/14/2024]
Abstract
Focused ultrasound is a platform technology capable of eliciting a wide range of biological responses with high spatial precision deep within the body. Although focused ultrasound is already in clinical use for focal thermal ablation of tissue, there has been a recent growth in development and translation of ultrasound-mediated nonthermal therapies. These approaches exploit the physical forces of ultrasound to produce a range of biological responses dependent on exposure conditions. This review discusses recent advances in four application areas that have seen particular growth and have immense clinical potential: brain drug delivery, neuromodulation, focal tissue destruction, and endogenous immune system activation. Owing to the maturation of transcranial ultrasound technology, the brain is a major target organ; however, clinical indications outside the brain are also discussed.
Collapse
Affiliation(s)
- Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Hang Z, Zhou L, Bian X, Liu G, Cui F, Du H, Wen Y. Potential application of aptamers combined with DNA nanoflowers in neurodegenerative diseases. Ageing Res Rev 2024; 100:102444. [PMID: 39084322 DOI: 10.1016/j.arr.2024.102444] [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: 07/09/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
The efficacy of neurotherapeutic drugs hinges on their ability to traverse the blood-brain barrier and access the brain, which is crucial for treating or alleviating neurodegenerative diseases (NDs). Given the absence of definitive cures for NDs, early diagnosis and intervention become paramount in impeding disease progression. However, conventional therapeutic drugs and existing diagnostic approaches must meet clinical demands. Consequently, there is a pressing need to advance drug delivery systems and early diagnostic methods tailored for NDs. Certain aptamers endowed with specific functionalities find widespread utility in the targeted therapy and diagnosis of NDs. DNA nanoflowers (DNFs), distinctive flower-shaped DNA nanomaterials, are intricately self-assembled through rolling ring amplification (RCA) of circular DNA templates. Notably, imbuing DNFs with diverse functionalities becomes seamlessly achievable by integrating aptamer sequences with specific functions into RCA templates, resulting in a novel nanomaterial, aptamer-bound DNFs (ADNFs) that amalgamates the advantageous features of both components. This article delves into the characteristics and applications of aptamers and DNFs, exploring the potential or application of ADNFs in drug-targeted delivery, direct treatment, early diagnosis, etc. The objective is to offer prospective ideas for the clinical treatment or diagnosis of NDs, thereby contributing to the ongoing efforts in this critical field.
Collapse
Affiliation(s)
- Zhongci Hang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China.
| | - Xiaochun Bian
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Guotao Liu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Fenghe Cui
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangdingdong Road, Zhifu District, Yantai, Shandong 264000, China.
| | - Hongwu Du
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China.
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China.
| |
Collapse
|
11
|
Wasielewska JM, Chaves JCS, Cabral-da-Silva MC, Pecoraro M, Viljoen SJ, Nguyen TH, Bella VL, Oikari LE, Ooi L, White AR. A patient-derived amyotrophic lateral sclerosis blood-brain barrier model for focused ultrasound-mediated anti-TDP-43 antibody delivery. Fluids Barriers CNS 2024; 21:65. [PMID: 39138578 PMCID: PMC11323367 DOI: 10.1186/s12987-024-00565-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disorder with minimally effective treatment options. An important hurdle in ALS drug development is the non-invasive therapeutic access to the motor cortex currently limited by the presence of the blood-brain barrier (BBB). Focused ultrasound and microbubble (FUS+ MB) treatment is an emerging technology that was successfully used in ALS patients to temporarily open the cortical BBB. However, FUS+ MB-mediated drug delivery across ALS patients' BBB has not yet been reported. Similarly, the effects of FUS+ MB on human ALS BBB cells remain unexplored. METHODS Here we established the first FUS+ MB-compatible, fully-human ALS patient-cell-derived BBB model based on induced brain endothelial-like cells (iBECs) to study anti-TDP-43 antibody delivery and FUS+ MB bioeffects in vitro. RESULTS Generated ALS iBECs recapitulated disease-specific hallmarks of BBB pathology, including reduced BBB integrity and permeability, and TDP-43 proteinopathy. The results also identified differences between sporadic ALS and familial (C9orf72 expansion carrying) ALS iBECs reflecting patient heterogeneity associated with disease subgroups. Studies in these models revealed successful ALS iBEC monolayer opening in vitro with no adverse cellular effects of FUS+ MB as reflected by lactate dehydrogenase (LDH) release viability assay and the lack of visible monolayer damage or morphology change in FUS+ MB treated cells. This was accompanied by the molecular bioeffects of FUS+ MB in ALS iBECs including changes in expression of tight and adherens junction markers, and drug transporter and inflammatory mediators, with sporadic and C9orf72 ALS iBECs generating transient specific responses. Additionally, we demonstrated an effective increase in the delivery of anti-TDP-43 antibody with FUS+ MB in C9orf72 (2.7-fold) and sporadic (1.9-fold) ALS iBECs providing the first proof-of-concept evidence that FUS+ MB can be used to enhance the permeability of large molecule therapeutics across the BBB in a human ALS in vitro model. CONCLUSIONS Together, this study describes the first characterisation of cellular and molecular responses of ALS iBECs to FUS+ MB and provides a fully-human platform for FUS+ MB-mediated drug delivery screening on an ALS BBB in vitro model.
Collapse
Affiliation(s)
- Joanna M Wasielewska
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, St. Lucia, QLD, Australia
| | - Juliana C S Chaves
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Mauricio Castro Cabral-da-Silva
- Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute (MCRI), Parkville, VIC, Australia
| | - Martina Pecoraro
- ALS Clinical Research Centre and Laboratory of Neurochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Stephani J Viljoen
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Science, University of Queensland, St. Lucia, QLD, Australia
| | - Tam Hong Nguyen
- Flow Cytometry and Imaging Facility, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Vincenzo La Bella
- ALS Clinical Research Centre and Laboratory of Neurochemistry, Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Lotta E Oikari
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lezanne Ooi
- Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, Australia
| | - Anthony R White
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia.
- School of Biomedical Science, University of Queensland, St. Lucia, QLD, Australia.
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
12
|
Leinenga G, To XV, Bodea LG, Yousef J, Richter-Stretton G, Palliyaguru T, Chicoteau A, Dagley L, Nasrallah F, Götz J. Scanning ultrasound-mediated memory and functional improvements do not require amyloid-β reduction. Mol Psychiatry 2024; 29:2408-2423. [PMID: 38499653 PMCID: PMC11412907 DOI: 10.1038/s41380-024-02509-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024]
Abstract
A prevalent view in treating age-dependent disorders including Alzheimer's disease (AD) is that the underlying amyloid plaque pathology must be targeted for cognitive improvements. In contrast, we report here that repeated scanning ultrasound (SUS) treatment at 1 MHz frequency can ameliorate memory deficits in the APP23 mouse model of AD without reducing amyloid-β (Aβ) burden. Different from previous studies that had shown Aβ clearance as a consequence of blood-brain barrier (BBB) opening, here, the BBB was not opened as no microbubbles were used. Quantitative SWATH proteomics and functional magnetic resonance imaging revealed that ultrasound induced long-lasting functional changes that correlate with the improvement in memory. Intriguingly, the treatment was more effective at a higher frequency (1 MHz) than at a frequency within the range currently explored in clinical trials in AD patients (286 kHz). Together, our data suggest frequency-dependent bio-effects of ultrasound and a dissociation of cognitive improvement and Aβ clearance, with important implications for the design of trials for AD therapies.
Collapse
Affiliation(s)
- Gerhard Leinenga
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Xuan Vinh To
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Liviu-Gabriel Bodea
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jumana Yousef
- Proteomics Facility, Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Gina Richter-Stretton
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Tishila Palliyaguru
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Antony Chicoteau
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Laura Dagley
- Proteomics Facility, Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Fatima Nasrallah
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
13
|
Lee JY, Lim MCX, Koh RY, Tsen MT, Chye SM. Blood-based therapies to combat neurodegenerative diseases. Metab Brain Dis 2024; 39:985-1004. [PMID: 38842660 DOI: 10.1007/s11011-024-01368-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Neurodegeneration, known as the progressive loss of neurons in terms of their structure and function, is the principal pathophysiological change found in the majority of brain-related disorders. Ageing has been considered the most well-established risk factor in most common neurodegenerative diseases, such as Parkinson's disease (PD) and Alzheimer's disease (AD). There is currently no effective treatment or cure for these diseases; the approved therapeutic options to date are only for palliative care. Ageing and neurodegenerative diseases are closely intertwined; reversing the aspects of brain ageing could theoretically mitigate age-related neurodegeneration. Ever since the regenerative properties of young blood on aged tissues came to light, substantial efforts have been focused on identifying and characterizing the circulating factors in the young and old systemic milieu that may attenuate or accentuate brain ageing and neurodegeneration. Later studies discovered the superiority of old plasma dilution in tissue rejuvenation, which is achieved through a molecular reset of the systemic proteome. These findings supported the use of therapeutic blood exchange for the treatment of degenerative diseases in older individuals. The first objective of this article is to explore the rejuvenating properties of blood-based therapies in the ageing brains and their therapeutic effects on AD. Then, we also look into the clinical applications, various limitations, and challenges associated with blood-based therapies for AD patients.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Mervyn Chen Xi Lim
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Min Tze Tsen
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
14
|
Duclos S, Choi SW, Andjelkovic AV, Chaudhary N, Camelo-Piragua S, Pandey A, Xu Z. Characterization of Blood-Brain Barrier Opening Induced by Transcranial Histotripsy in Murine Brains. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:639-646. [PMID: 38302370 PMCID: PMC11894761 DOI: 10.1016/j.ultrasmedbio.2023.12.014] [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: 10/18/2022] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 02/03/2024]
Abstract
OBJECTIVE Transcranial histotripsy has shown promise as a non-invasive neurosurgical tool, as it has the ability to treat a wide range of locations in the brain without overheating the skull. One important effect of histotripsy in the brain is the blood-brain barrier (BBB) opening (BBBO) at the ablation site, but there is a knowledge gap concerning the extent of histotripsy-induced BBBO. Here we describe induction of BBBO by transcranial histotripsy and use of magnetic resonance imaging (MRI) and histology to quantify changes in BBBO at the periphery of the histotripsy ablation zone over time in the healthy mouse brain. METHODS An eight-element, 1 MHz histotripsy transducer with a focal distance of 32.5 mm was used to treat the brains of 23 healthy female BL6 mice. T1-gadolinium (T1-Gd) MR images were acquired immediately following histotripsy treatment and during each of the subsequent 4 wk to quantify the size and intensity of BBB leakage. RESULTS The T1-Gd MRI results revealed that the hyperintense BBBO volume increased over the first week and subsided gradually over the following 3 wk. Histology revealed complete loss of tight junction proteins and blood vessels in the center of the ablation region immediately after histotripsy, partial recovery in the periphery of the ablation zone 1 wk following histotripsy and near-complete recovery of tight junction complex after 4 wk. CONCLUSION These results provide the first evidence of transcranial histotripsy-induced BBBO and repair at the periphery of the ablation zone.
Collapse
Affiliation(s)
- Sarah Duclos
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Sang Won Choi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Neeraj Chaudhary
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA; Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | | | - Aditya Pandey
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA; Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Zhen Xu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
15
|
Natera-Villalba E, Ruiz-Yanzi MA, Gasca-Salas C, Matarazzo M, Martínez-Fernández R. MR-guided focused ultrasound in movement disorders and beyond: Lessons learned and new frontiers. Parkinsonism Relat Disord 2024; 122:106040. [PMID: 38378311 DOI: 10.1016/j.parkreldis.2024.106040] [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: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
The development of MR-guided focused ultrasound (MRgFUS) has provided a new therapeutic tool for neuropsychiatric disorders. In contrast to previously available neurosurgical techniques, MRgFUS allows precise impact on deep brain structures without the need for incision and yields an immediate effect. In its high-intensity modality (MRgHIFU), it produces accurate therapeutic thermoablation in previously selected brain targets. Importantly, the production of the lesion is progressive and highly controlled in real-time by both neuroimaging and clinical means. MRgHIFU ablation is already an accepted and widely used treatment for medically-refractory Parkinson's disease and essential tremor. Notably, other neurological disorders and diverse brain targets, including bilateral treatments, are currently under examination. Conversely, the low-intensity modality (MRgLIFU) shows promising prospects in neuromodulation and transient blood-brain barrier opening (BBBO). In the former circumstance, MRgLIFU could serve as a powerful clinical and research tool for non-invasively modulating brain activity and function. BBBO, on the other hand, emerges as a potentially impactful method to influence disease pathogenesis and progression by increasing brain target engagement of putative therapeutic agents. While promising, these applications remain experimental. As a recently developed technology, MRgFUS is not without challenges and questions to be addressed. Further developments and broader experience are necessary to enhance MRgFUS capabilities in both research and clinical practice, as well as to define device constraints. This clinical mini-review aims to provide an overview of the main evidence of MRgFUS application and to highlight unmet needs and future potentialities of the technique.
Collapse
Affiliation(s)
- Elena Natera-Villalba
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain; PhD Medicine Program, Universidad Autónoma de Madrid, Madrid, Spain
| | - María-Agustina Ruiz-Yanzi
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain
| | - Carmen Gasca-Salas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain; University CEU-San Pablo, Madrid, Spain
| | - Michele Matarazzo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Raúl Martínez-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain; University CEU-San Pablo, Madrid, Spain.
| |
Collapse
|
16
|
Kofoed RH, Aubert I. Focused ultrasound gene delivery for the treatment of neurological disorders. Trends Mol Med 2024; 30:263-277. [PMID: 38216449 DOI: 10.1016/j.molmed.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024]
Abstract
The transformative potential of gene therapy has been demonstrated in humans. However, there is an unmet need for non-invasive targeted gene delivery and regulation in the treatment of brain disorders. Transcranial focused ultrasound (FUS) has gained tremendous momentum to address these challenges. FUS non-invasively modulates brain cells and their environment, and is a powerful tool to facilitate gene delivery across the blood-brain barrier (BBB) with millimeter precision and promptly regulate transgene expression. This review highlights technical aspects of FUS-mediated gene therapies for the central nervous system (CNS) and lessons learned from discoveries in other organs. Understanding the possibilities and remaining obstacles of FUS-mediated gene therapy will be necessary to harness remarkable technologies and create life-changing treatments for neurological disorders.
Collapse
Affiliation(s)
- Rikke Hahn Kofoed
- Department of Neurosurgery, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Center for Experimental Neuroscience (CENSE), Department of Neurosurgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.
| | - Isabelle Aubert
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
17
|
Liu N, Liang X, Chen Y, Xie L. Recent trends in treatment strategies for Alzheimer 's disease and the challenges: A topical advancement. Ageing Res Rev 2024; 94:102199. [PMID: 38232903 DOI: 10.1016/j.arr.2024.102199] [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: 11/14/2023] [Accepted: 01/12/2024] [Indexed: 01/19/2024]
Abstract
Alzheimer's Disease (AD) is an irreversible and progressive neurological disease that has affected at least 50 million people around the globe. Considering the severity of the disease and the continuous increase in the number of patients, the development of new effective drugs or intervention strategies for AD has become urgent. AD is caused by a combination of genetic, environmental, and lifestyle factors, but its exact cause has not yet been clarified. Given the current challenges being faced in the clinical treatment of AD, such as complex AD pathological network and insufficient early diagnosis, herein, we have focused on the three core pathological features of AD, including amyloid-β (Aβ) aggregation, tau phosphorylation and tangles, and activation of inflammatory factors. In this review, we have briefly underscored the primary evidence supporting each pathology and discuss AD pathological network among Aβ, tau, and inflammation. We have also comprehensively summarized the most instructive drugs and their treatment strategies against Aβ, tau, or neuroinflammation used in basic research and clinical trials. Finally, we have discussed and outlined the pros and cons of each pathological approach and looked forward to potential personalized diagnosis and treatment strategies that are beneficial to AD patients.
Collapse
Affiliation(s)
- Ni Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Xiaohan Liang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yu Chen
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Lihang Xie
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
18
|
Paranjape AN, D'Aiuto L, Zheng W, Chen X, Villanueva FS. A multicellular brain spheroid model for studying the mechanisms and bioeffects of ultrasound-enhanced drug penetration beyond the blood‒brain barrier. Sci Rep 2024; 14:1909. [PMID: 38253669 PMCID: PMC10803331 DOI: 10.1038/s41598-023-50203-3] [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: 08/16/2023] [Accepted: 12/16/2023] [Indexed: 01/24/2024] Open
Abstract
The blood‒brain barrier (BBB) acts as a hindrance to drug therapy reaching the brain. With an increasing incidence of neurovascular diseases and brain cancer metastases, there is a need for an ideal in vitro model to develop novel methodologies for enhancing drug delivery to the brain. Here, we established a multicellular human brain spheroid model that mimics the BBB both architecturally and functionally. Within the spheroids, endothelial cells and pericytes localized to the periphery, while neurons, astrocytes, and microglia were distributed throughout. Ultrasound-targeted microbubble cavitation (UTMC) is a novel noninvasive technology for enhancing endothelial drug permeability. We utilized our three-dimensional (3D) model to study the feasibility and mechanisms regulating UTMC-induced hyperpermeability. UTMC caused a significant increase in the penetration of 10 kDa Texas red dextran (TRD) into the spheroids, 100 µm beyond the BBB, without compromising cell viability. This hyperpermeability was dependent on UTMC-induced calcium (Ca2+) influx and endothelial nitric oxide synthase (eNOS) activation. Our 3D brain spheroid model, with its intact and functional BBB, offers a valuable platform for studying the bioeffects of UTMC, including effects occurring spatially distant from the endothelial barrier.
Collapse
Affiliation(s)
- Anurag N Paranjape
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leonardo D'Aiuto
- Department of Psychiatry, University of Pittsburgh School of Medicine Western Psychiatric Institute and Clinic, Pittsburgh, PA, USA
| | - Wenxiao Zheng
- Department of Psychiatry, University of Pittsburgh School of Medicine Western Psychiatric Institute and Clinic, Pittsburgh, PA, USA
- Department of Health and Human Development, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xucai Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Flordeliza S Villanueva
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
19
|
Li Y, Wu Y, Luo Q, Ye X, Chen J, Su Y, Zhao K, Li X, Lin J, Tong Z, Wang Q, Xu D. Neuropsychiatric Behavioral Assessments in Mice After Acute and Long-Term Treatments of Low-Intensity Pulsed Ultrasound. Am J Alzheimers Dis Other Demen 2024; 39:15333175231222695. [PMID: 38183177 PMCID: PMC10771054 DOI: 10.1177/15333175231222695] [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] [Indexed: 01/07/2024]
Abstract
Introduction: To evaluate whether both acute and chronic low-intensity pulsed ultrasound (LIPUS) affect brain functions of healthy male and female mice. Methods: Ultrasound (frequency: 1.5 MHz; pulse: 1.0 kHz; spatial average temporal average (SATA) intensity: 25 mW/cm2; and pulse duty cycle: 20%) was applied at mouse head in acute test for 20 minutes, and in chronic experiment for consecutive 10 days, respectively. Behaviors were then evaluated. Results: Both acute and chronic LIPUS at 25 mW/cm2 exposure did not affect the abilities of movements, mating, social interaction, and anxiety-like behaviors in the male and female mice. However, physical restraint caused struggle-like behaviors and short-time memory deficits in chronic LIPUS groups in the male mice. Conclusion: LIPUS at 25 mW/cm2 itself does not affect brain functions, while physical restraint for LIPUS therapy elicits struggle-like behaviors in the male mice. An unbound helmet targeted with ultrasound intensity at 25-50 mW/cm2 is proposed for clinical brain disease therapy.
Collapse
Affiliation(s)
- Ye Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Yiqing Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Qi Luo
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xuanjie Ye
- Department of Electrical and Computer Engineering, and Department of Biomedical Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Jie Chen
- Department of Electrical and Computer Engineering, and Department of Biomedical Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
- Academy for Engineering & Technology, Fudan University, Shanghai, China
| | - Yuanlin Su
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Ke Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xinmin Li
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jing Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Zhiqian Tong
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Qi Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Dongwu Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
20
|
Leinenga G, Padmanabhan P, Götz J. Improving Cognition Without Clearing Amyloid: Effects of Tau and Ultrasound Neuromodulation. J Alzheimers Dis 2024; 100:S211-S222. [PMID: 39058447 DOI: 10.3233/jad-240616] [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: 07/28/2024]
Abstract
Alzheimer's disease is characterized by progressive impairment of neuronal functions culminating in neuronal loss and dementia. A universal feature of dementia is protein aggregation, a process by which a monomer forms intermediate oligomeric assembly states and filaments that develop into end-stage hallmark lesions. In Alzheimer's disease, this is exemplified by extracellular amyloid-β (Aβ) plaques which have been placed upstream of tau, found in intracellular neurofibrillary tangles and dystrophic neurites. This implies causality that can be modeled as a linear activation cascade. When Aβ load is reduced, for example, in response to an anti-Aβ immunotherapy, cognitive functions improve in plaque-forming mice. They also deteriorate less in clinical trial cohorts although real-world clinical benefits remain to be demonstrated. Given the existence of aged humans with unimpaired cognition despite a high plaque load, the central role of Aβ has been challenged. A counter argument has been that clinical symptoms would eventually develop if these aged individuals were to live long enough. Alternatively, intrinsic mechanisms that protect the brain in the presence of pathology may exist. In fact, Aβ toxicity can be abolished by either reducing or manipulating tau (through which Aβ signals), at least in preclinical models. In addition to manipulating steps in this linear pathocascade model, mechanisms of restoring brain reserve can also counteract Aβ toxicity. Low-intensity ultrasound is a neuromodulatory modality that can improve cognitive functions in Aβ-depositing mice without the need for removing Aβ. Together, this highlights a dissociation of Aβ and cognition, with important implications for therapeutic interventions.
Collapse
Affiliation(s)
- Gerhard Leinenga
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
| | - Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
21
|
Ma X, Li T, Du L, Han T. Research and progress of focused ultrasound in the treatment of Alzheimer's disease. Front Neurol 2023; 14:1323386. [PMID: 38187144 PMCID: PMC10771294 DOI: 10.3389/fneur.2023.1323386] [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: 10/18/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Alzheimer's disease is one of the most common degenerative diseases of the central nervous system, with progressive cognitive and memory impairment and decreased ability of daily life as the cardinal symptoms, influencing the life quality of patients severely. There are currently approximately 46 million people living with Alzheimer's disease worldwide, and the number is expected to triple by 2050, which will pose a huge challenge for healthcare. At present, the Food and Drug Administration of the United States has approved five main drugs for the clinical treatment of Alzheimer's disease, which are cholinesterase inhibitors tacrine, galantamine, capalatine and donepezil, and N-methyl-d-aspartate receptor antagonist memantine, although these drugs have shown good efficacy in clinical trials, the actual clinical effect is less effective due to the existence of blood brain barrier. With the continuous development of ultrasound technology in recent years, focused ultrasound, as a non-invasive treatment technique, may target ultrasound energy to the deep brain for treatment without damaging the surrounding tissue. For the past few years, some studies could use focused ultrasound combined with microvesicles to induce blood brain barrier opening and targeted drug delivery to treat Alzheimer's disease, providing new opportunities for the treatment of Alzheimer's disease. This article reviews the application research and progress of focused ultrasound in the treatment of Alzheimer's disease, in order to provide new directions and ideas for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Xishun Ma
- Department of Ultrasound, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Tongxia Li
- Department of Tuberculosis, Qingdao Chest Hospital, Qingdao, China
| | - Lizhen Du
- Department of Ultrasound, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Tongliang Han
- Department of Ultrasound, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| |
Collapse
|
22
|
Congdon EE, Ji C, Tetlow AM, Jiang Y, Sigurdsson EM. Tau-targeting therapies for Alzheimer disease: current status and future directions. Nat Rev Neurol 2023; 19:715-736. [PMID: 37875627 PMCID: PMC10965012 DOI: 10.1038/s41582-023-00883-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Alzheimer disease (AD) is the most common cause of dementia in older individuals. AD is characterized pathologically by amyloid-β (Aβ) plaques and tau neurofibrillary tangles in the brain, with associated loss of synapses and neurons, which eventually results in dementia. Many of the early attempts to develop treatments for AD focused on Aβ, but a lack of efficacy of these treatments in terms of slowing disease progression led to a change of strategy towards targeting of tau pathology. Given that tau shows a stronger correlation with symptom severity than does Aβ, targeting of tau is more likely to be efficacious once cognitive decline begins. Anti-tau therapies initially focused on post-translational modifications, inhibition of tau aggregation and stabilization of microtubules. However, trials of many potential drugs were discontinued because of toxicity and/or lack of efficacy. Currently, the majority of tau-targeting agents in clinical trials are immunotherapies. In this Review, we provide an update on the results from the initial immunotherapy trials and an overview of new therapeutic candidates that are in clinical development, as well as considering future directions for tau-targeting therapies.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Changyi Ji
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Amber M Tetlow
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
23
|
Niazi SK. Non-Invasive Drug Delivery across the Blood-Brain Barrier: A Prospective Analysis. Pharmaceutics 2023; 15:2599. [PMID: 38004577 PMCID: PMC10674293 DOI: 10.3390/pharmaceutics15112599] [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: 10/12/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Non-invasive drug delivery across the blood-brain barrier (BBB) represents a significant advancement in treating neurological diseases. The BBB is a tightly packed layer of endothelial cells that shields the brain from harmful substances in the blood, allowing necessary nutrients to pass through. It is a highly selective barrier, which poses a challenge to delivering therapeutic agents into the brain. Several non-invasive procedures and devices have been developed or are currently being investigated to enhance drug delivery across the BBB. This paper presents a review and a prospective analysis of the art and science that address pharmacology, technology, delivery systems, regulatory approval, ethical concerns, and future possibilities.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
24
|
Hang Z, Zhou L, Xing C, Wen Y, Du H. The blood-brain barrier, a key bridge to treat neurodegenerative diseases. Ageing Res Rev 2023; 91:102070. [PMID: 37704051 DOI: 10.1016/j.arr.2023.102070] [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: 06/09/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
As a highly selective and semi-permeable barrier that separates the circulating blood from the brain and central nervous system (CNS), the blood-brain barrier (BBB) plays a critical role in the onset and treatment of neurodegenerative diseases (NDs). To delay or reverse the NDs progression, the dysfunction of BBB should be improved to protect the brain from harmful substances. Simultaneously, a highly efficient drug delivery across the BBB is indispensable. Here, we summarized several methods to improve BBB dysfunction in NDs, including knocking out risk geneAPOE4, regulating circadian rhythms, restoring the gut microenvironment, and activating the Wnt/β-catenin signaling pathway. Then we discussed the advances in BBB penetration techniques, such as transient BBB opening, carrier-mediated drug delivery, and nasal administration, which facilitates drug delivery across the BBB. Furthermore, various in vivo and in vitro BBB models and research methods related to NDs are reviewed. Based on the current research progress, the treatment of NDs in the long term should prioritize the integrity of the BBB. However, a treatment approach that combines precise control of transient BBB permeability and non-invasive targeted BBB drug delivery holds profound significance in improving treatment effectiveness, safety, and clinical feasibility during drug therapy. This review involves the cross application of biology, materials science, imaging, engineering and other disciplines in the field of BBB, aiming to provide multi-dimensional research directions and clinical ideas for the treating NDs.
Collapse
Affiliation(s)
- Zhongci Hang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Cencan Xing
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Yongqiang Wen
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China.
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China.
| |
Collapse
|
25
|
Géraudie A, Riche M, Lestra T, Trotier A, Dupuis L, Mathon B, Carpentier A, Delatour B. Effects of Low-Intensity Pulsed Ultrasound-Induced Blood-Brain Barrier Opening in P301S Mice Modeling Alzheimer's Disease Tauopathies. Int J Mol Sci 2023; 24:12411. [PMID: 37569786 PMCID: PMC10419069 DOI: 10.3390/ijms241512411] [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: 06/29/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia. No treatments have led to clinically meaningful impacts. A major obstacle for peripherally administered therapeutics targeting the central nervous system is related to the blood-brain barrier (BBB). Ultrasounds associated with microbubbles have been shown to transiently and safely open the BBB. In AD mouse models, the sole BBB opening with no adjunct drugs may be sufficient to reduce lesions and mitigate cognitive decline. However, these therapeutic effects are for now mainly assessed in preclinical mouse models of amyloidosis and remain less documented in tau lesions. The aim of the present study was therefore to evaluate the effects of repeated BBB opening using low-intensity pulsed ultrasounds (LIPU) in tau transgenic P301S mice with two main readouts: tau-positive lesions and microglial cells. Our results show that LIPU-induced BBB opening does not decrease tau pathology and may even potentiate the accumulation of pathological tau in selected brain regions. In addition, LIPU-BBB opening in P301S mice strongly reduced microglia densities in brain parenchyma, suggesting an anti-inflammatory action. These results provide a baseline for future studies using LIPU-BBB opening, such as adjunct drug therapies, in animal models and in AD patients.
Collapse
Affiliation(s)
- Amandine Géraudie
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France; (M.R.); (T.L.); (A.T.); (L.D.); (B.M.); (B.D.)
| | - Maximilien Riche
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France; (M.R.); (T.L.); (A.T.); (L.D.); (B.M.); (B.D.)
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France;
- Faculty of Medicine, Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France
- Advanced Surgical Research Technology Lab, Sorbonne University, 75013 Paris, France
| | - Thaïs Lestra
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France; (M.R.); (T.L.); (A.T.); (L.D.); (B.M.); (B.D.)
| | - Alexandre Trotier
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France; (M.R.); (T.L.); (A.T.); (L.D.); (B.M.); (B.D.)
| | - Léo Dupuis
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France; (M.R.); (T.L.); (A.T.); (L.D.); (B.M.); (B.D.)
- Laboratoire Des Maladies Neurodégénératives, Université Paris-Saclay, CEA, CNRS, 18 Route du Panorama, 92265 Fontenay-Aux-Roses, France
- Commissariat à l’Energie Atomique et aux Énergies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France
| | - Bertrand Mathon
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France; (M.R.); (T.L.); (A.T.); (L.D.); (B.M.); (B.D.)
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France;
- Faculty of Medicine, Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France
- Advanced Surgical Research Technology Lab, Sorbonne University, 75013 Paris, France
| | - Alexandre Carpentier
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France;
- Faculty of Medicine, Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France
- Advanced Surgical Research Technology Lab, Sorbonne University, 75013 Paris, France
| | - Benoît Delatour
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France; (M.R.); (T.L.); (A.T.); (L.D.); (B.M.); (B.D.)
| |
Collapse
|
26
|
Karakatsani ME, Ji R, Murillo MF, Kugelman T, Kwon N, Lao YH, Liu K, Pouliopoulos AN, Honig LS, Duff KE, Konofagou EE. Focused ultrasound mitigates pathology and improves spatial memory in Alzheimer's mice and patients. Theranostics 2023; 13:4102-4120. [PMID: 37554284 PMCID: PMC10405840 DOI: 10.7150/thno.79898] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/12/2023] [Indexed: 08/10/2023] Open
Abstract
Rationale: Bilateral sonication with focused ultrasound (FUS) in conjunction with microbubbles has been shown to separately reduce amyloid plaques and hyperphosphorylated tau protein in the hippocampal formation and the entorhinal cortex in different mouse models of Alzheimer's disease (AD) without any therapeutic agents. However, the two pathologies are expressed concurrently in human disease. Therefore, the objective of this study is to investigate the effects of repeated bilateral sonications in the presence of both pathologies. Methods: Herein, we investigate its functional and morphological outcomes on brains bearing both pathologies simultaneously. Eleven transgenic mice of the 3xTg-AD line (14 months old) expressing human amyloid beta and human tau and eleven age-matched wild-type littermates received four weekly bilateral sonications covering the hippocampus followed by working memory testing. Afterwards, immunohistochemistry and immunoassays (western blot and ELISA) were employed to assess any changes in amyloid beta and human tau. Furthermore, we present preliminary data from our clinical trial using a neuronavigation-guided FUS system for sonications in AD patients (NCT04118764). Results: Interestingly, both wild-type and transgenic animals that received FUS experienced improved working memory and spent significantly more time in the escape platform-quadrant, with wild-type animals spending 43.2% (sham: 37.7%) and transgenic animals spending 35.3% (sham: 31.0%) of the trial in the target quadrant. Furthermore, this behavioral amelioration in the transgenic animals correlated with a 58.3% decrease in the neuronal length affected by tau and a 27.2% reduction in total tau levels. Amyloid plaque population, volume and overall load were also reduced overall. Consistently, preliminary data from a clinical trial involving AD patients showed a 1.8% decrease of amyloid PET signal 3-weeks after treatment in the treated hemisphere compared to baseline. Conclusion: For the first time, it is shown that bilateral FUS-induced BBB opening significantly and simultaneously ameliorates both coexistent pathologies, which translated to improvements in spatial memory of transgenic animals with complex AD, the human mimicking phenotype. The level of cognitive improvement was significantly correlated with the volume of BBB opening. Non-transgenic animals were also shown to exhibit similar memory amelioration for the first time, indicating that BBB opening results into benefits in the neuronal function regardless of the existence of AD pathology. A potential mechanism of action for the reduction of the both pathologies investigated was the cholesterol metabolism, specifically the LRP1b receptor, which exhibited increased expression levels in transgenic mice following FUS-induced BBB opening. Initial clinical evidence supported that the beta amyloid reduction shown in rodents could be translatable to humans with significant amyloid reduction shown in the treated hemisphere.
Collapse
Affiliation(s)
| | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Maria F. Murillo
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Tara Kugelman
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Keyu Liu
- Department of Biomedical Engineering, Columbia University, New York, USA
| | | | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Karen E. Duff
- UK Dementia Research Institute, University College London, London, UK
| | - Elisa E. Konofagou
- Department of Radiology, Columbia University Irving Medical Center, New York, USA
| |
Collapse
|
27
|
Bao Y, Lu W. Targeting cerebral diseases with enhanced delivery of therapeutic proteins across the blood-brain barrier. Expert Opin Drug Deliv 2023; 20:1681-1698. [PMID: 36945117 DOI: 10.1080/17425247.2023.2193390] [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: 01/14/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Cerebral diseases have been threatening public physical and psychological health in the recent years. With the existence of the blood-brain barrier (BBB), it is particularly hard for therapeutic proteins like peptides, enzymes, antibodies, etc. to enter the central nervous system (CNS) and function in diagnosis and treatment in cerebral diseases. Fortunately, the past decade has witnessed some emerging strategies of delivering macromolecular therapeutic proteins across the BBB. AREAS COVERED Based on the structure, functions, and substances transport mechanisms, various enhanced delivery strategies of therapeutic proteins were reviewed, categorized by molecule-mediated delivery strategies, carrier-mediated delivery strategies, and other delivery strategies. EXPERT OPINION As for molecule-mediated delivery strategies, development of genetic engineering technology, optimization of protein expression and purification techniques, and mature of quality control systems all help to realize large-scale production of recombinant antibodies, making it possible to apply to the clinical practice. In terms of carrier-mediated delivery strategies and others, although nano-carriers/adeno-associated virus (AAV) are also promising candidates for delivering therapeutic proteins or genes across the BBB, some issues still remain to be further investigated, including safety concerns related to applied materials, large-scale production costs, quality control standards, combination therapies with auxiliary delivery strategies like focused ultrasound, etc.
Collapse
Affiliation(s)
- Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
- Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd. Lingang of Shanghai, China
| |
Collapse
|
28
|
Padmanabhan P, Götz J. Clinical relevance of animal models in aging-related dementia research. NATURE AGING 2023; 3:481-493. [PMID: 37202516 DOI: 10.1038/s43587-023-00402-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Alzheimer's disease (AD) and other, less prevalent dementias are complex age-related disorders that exhibit multiple etiologies. Over the past decades, animal models have provided pathomechanistic insight and evaluated countless therapeutics; however, their value is increasingly being questioned due to the long history of drug failures. In this Perspective, we dispute this criticism. First, the utility of the models is limited by their design, as neither the etiology of AD nor whether interventions should occur at a cellular or network level is fully understood. Second, we highlight unmet challenges shared between animals and humans, including impeded drug transport across the blood-brain barrier, limiting effective treatment development. Third, alternative human-derived models also suffer from the limitations mentioned above and can only act as complementary resources. Finally, age being the strongest AD risk factor should be better incorporated into the experimental design, with computational modeling expected to enhance the value of animal models.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
29
|
Johansen PM, Hansen PY, Mohamed AA, Girshfeld SJ, Feldmann M, Lucke-Wold B. Focused ultrasound for treatment of peripheral brain tumors. EXPLORATION OF DRUG SCIENCE 2023:107-125. [PMID: 37171968 PMCID: PMC10168685 DOI: 10.37349/eds.2023.00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/13/2023] [Indexed: 05/14/2023]
Abstract
Malignant brain tumors are the leading cause of cancer-related death in children and remain a significant cause of morbidity and mortality throughout all demographics. Central nervous system (CNS) tumors are classically treated with surgical resection and radiotherapy in addition to adjuvant chemotherapy. However, the therapeutic efficacy of chemotherapeutic agents is limited due to the blood-brain barrier (BBB). Magnetic resonance guided focused ultrasound (MRgFUS) is a new and promising intervention for CNS tumors, which has shown success in preclinical trials. High-intensity focused ultrasound (HIFU) has the capacity to serve as a direct therapeutic agent in the form of thermoablation and mechanical destruction of the tumor. Low-intensity focused ultrasound (LIFU) has been shown to disrupt the BBB and enhance the uptake of therapeutic agents in the brain and CNS. The authors present a review of MRgFUS in the treatment of CNS tumors. This treatment method has shown promising results in preclinical trials including minimal adverse effects, increased infiltration of the therapeutic agents into the CNS, decreased tumor progression, and improved survival rates.
Collapse
Affiliation(s)
| | - Payton Yerke Hansen
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Ali A. Mohamed
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Sarah J. Girshfeld
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Marc Feldmann
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
30
|
Blackmore DG, Razansky D, Götz J. Ultrasound as a versatile tool for short- and long-term improvement and monitoring of brain function. Neuron 2023; 111:1174-1190. [PMID: 36917978 DOI: 10.1016/j.neuron.2023.02.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/19/2023] [Accepted: 02/13/2023] [Indexed: 03/15/2023]
Abstract
Treating the brain with focused ultrasound (FUS) at low intensities elicits diverse responses in neurons, astroglia, and the extracellular matrix. In combination with intravenously injected microbubbles, FUS also opens the blood-brain barrier (BBB) and facilitates focal drug delivery. However, an incompletely understood cellular specificity and a wide parameter space currently limit the optimal application of FUS in preclinical and human studies. In this perspective, we discuss how different FUS modalities can be utilized to achieve short- and long-term improvements, thereby potentially treating brain disorders. We review the ongoing efforts to determine which parameters induce neuronal inhibition versus activation and how mechanoreceptors and signaling cascades are activated to induce long-term changes, including memory improvements. We suggest that optimal FUS treatments may require different FUS modalities and devices, depending on the targeted brain area or local pathology, and will be greatly enhanced by new techniques for monitoring FUS efficacy.
Collapse
Affiliation(s)
- Daniel G Blackmore
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Daniel Razansky
- Institute for Biomedical Engineering, Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, 8057 Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, 8093 Zurich, Switzerland
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
31
|
Meng Y, Goubran M, Rabin JS, McSweeney M, Ottoy J, Pople CB, Huang Y, Storace A, Ozzoude M, Bethune A, Lam B, Swardfager W, Heyn C, Abrahao A, Davidson B, Hamani C, Aubert I, Zetterberg H, Ashton NJ, Karikari TK, Blennow K, Black SE, Hynynen K, Lipsman N. Blood-brain barrier opening of the default mode network in Alzheimer's disease with magnetic resonance-guided focused ultrasound. Brain 2023; 146:865-872. [PMID: 36694943 PMCID: PMC10226733 DOI: 10.1093/brain/awac459] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 11/06/2022] [Indexed: 01/26/2023] Open
Abstract
The blood-brain barrier (BBB) protects the brain but is also an important obstacle for the effective delivery of therapeutics in Alzheimer's disease and other neurodegenerative disorders. Transcranial magnetic resonance-guided focused ultrasound (MRgFUS) has been shown to reversibly disrupt the BBB. However, treatment of diffuse regions across the brain along with the effect on Alzheimer's disease relevant pathology need to be better characterized. This study is an open-labelled single-arm trial (NCT04118764) to investigate the feasibility of modulating BBB permeability in the default mode network and the impact on cognition, amyloid and tau pathology as well as BBB integrity. Nine participants [mean age 70.2 ± 7.2 years, mean Mini-Mental State Examination (MMSE) 21.9] underwent three biweekly procedures with follow-up visits up to 6 months. The BBB permeability of the bilateral hippocampi, anterior cingulate cortex and precuneus was transiently increased without grade 3 or higher adverse events. Participants did not experience worsening trajectory of cognitive decline (ADAS-cog11, MMSE). Whole brain vertex-based analysis of the 18F-florbetaben PET imaging demonstrated clusters of modest SUVR reduction in the right parahippocampal and inferior temporal lobe. However, CSF and blood biomarkers did not demonstrate any amelioration of Alzheimer's disease pathology (P-tau181, amyloid-β42/40 ratio), nor did it show persistent BBB dysfunction (plasma PDGFRbeta and CSF-to-plasma albumin ratio). This study provides neuroimaging and fluid biomarker data to characterize the safety profile of MRgFUS BBB modulation in neurodegeneration as a potential strategy for enhanced therapeutic delivery.
Collapse
Affiliation(s)
- Ying Meng
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Maged Goubran
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Jennifer S Rabin
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON M5G 1V7, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Melissa McSweeney
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Julie Ottoy
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Christopher B Pople
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Yuexi Huang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Alexandra Storace
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Miracle Ozzoude
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Allison Bethune
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Benjamin Lam
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Walter Swardfager
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chinthaka Heyn
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Agessandro Abrahao
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Benjamin Davidson
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Clement Hamani
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Isabelle Aubert
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, 405 30 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London W1T 7NF, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, 405 30 Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London SE5 9RX, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London SE5 8AF, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, 405 30 Mölndal, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, 405 30 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
| | - Sandra E Black
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Kullervo Hynynen
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Nir Lipsman
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Hurvitz Brain Sciences Research Program, Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
32
|
Chang KW, Hong SW, Chang WS, Jung HH, Chang JW. Characteristics of Focused Ultrasound Mediated Blood-Brain Barrier Opening in Magnetic Resonance Images. J Korean Neurosurg Soc 2023; 66:172-182. [PMID: 36537034 PMCID: PMC10009247 DOI: 10.3340/jkns.2022.0236] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE The blood-brain barrier (BBB) is an obstacle for molecules to pass through from blood to the brain. Focused ultrasound is a new method which temporarily opens the BBB, which makes pharmaceutical delivery or removal of neurodegenerative proteins possible. This study was demonstrated to review our BBB opening procedure with magnetic resonance guided images and find specific patterns in the BBB opening. METHODS In this study, we reviewed the procedures and results of two clinical studies on BBB opening using focused ultrasound regarding its safety and clinical efficacy. Magnetic resonance images were also reviewed to discover any specific findings. RESULTS Two clinical trials showed clinical benefits. All clinical trials demonstrated safe BBB opening, with no specific side effects. Magnetic resonance imaging showed temporary T1 contrast enhancement in the sonication area, verifying the BBB opening. Several low-signal intensity spots were observed in the T2 susceptibility-weighted angiography images, which were also reversible and temporary. Although these spots can be considered as microbleeding, evidence suggests these are not ordinary microbleeding but an indicator for adequate BBB opening. CONCLUSION Magnetic resonance images proved safe and efficient BBB opening in humans, using focused ultrasound.
Collapse
Affiliation(s)
- Kyung Won Chang
- Brain Research Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Woo Hong
- Brain Research Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Won Seok Chang
- Brain Research Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Ho Jung
- Brain Research Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Woo Chang
- Brain Research Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
33
|
Advances in applications of head mounted devices (HMDs): Physical techniques for drug delivery and neuromodulation. J Control Release 2023; 354:810-820. [PMID: 36709924 DOI: 10.1016/j.jconrel.2023.01.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023]
Abstract
Head-mounted medical devices (HMDs) are disruptive inventions representing laboratories and clinical institutions worldwide are climbing the apexes of brain science. These complex devices are inextricably linked with a wide range knowledge containing the Physics, Imaging, Biomedical engineering, Biology and Pharmacology, particularly could be specifically designed for individuals, and finally exerting integrated bio-effect. The salient characteristics of them are non-invasive intervening in human brain's physiological structures, and alterating the biological process, such as thermal ablating the tumor, opening the BBB to deliver drugs and neuromodulating to enhance cognitive performance or manipulate prosthetic. The increasing demand and universally accepted of them have set off a dramatic upsurge in HMDs' studies, seminal applications of them span from clinical use to psychiatric disorders and neurological modulation. With subsequent pre-clinical studies and human trials emerging, the mechanisms of transcranial stimulation methods of them were widely studied, and could be basically came down to three notable approach: magnetic, electrical and ultrasonic stimulation. This review provides a comprehensive overviews of their stimulating mechanisms, and recent advances in clinic and military. We described the potential impact of HMDs on brain science, and current challenges to extensively adopt them as promising alternative treating tools.
Collapse
|
34
|
Sridharan B, Lim HG. Exosomes and ultrasound: The future of theranostic applications. Mater Today Bio 2023; 19:100556. [PMID: 36756211 PMCID: PMC9900624 DOI: 10.1016/j.mtbio.2023.100556] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Biomaterials and pertaining formulations have been very successful in various diagnostic and therapeutic applications because of its ability to overcome pharmacological limitations. Some of them have gained significant focus in the recent decade for their theranostic properties. Exosomes can be grouped as biomaterials, since they consist of various biological micro/macromolecules and possess all the properties of a stable biomaterial with size in nano range. Significant research has gone into isolation and exploitation of exosomes as potential theranostic agent. However, the limitations in terms of yield, efficacy, and target specificity are continuously being addressed. On the other hand, several nano/microformulations are responsive to physical or chemical alterations and were successfully stimulated by tweaking the physical characteristics of the surrounding environment they are in. Some of them are termed as photodynamic, sonodynamic or thermodynamic therapeutic systems. In this regard, ultrasound and acoustic systems were extensively studied for its ability towards altering the properties of the systems to which they were applied on. In this review, we have detailed about the diagnostic and therapeutic applications of exosomes and ultrasound separately, consisting of their conventional applications, drawbacks, and developments for addressing the challenges. The information were categorized into various sections that provide complete overview of the isolation strategies and theranostic applications of exosomes in various diseases. Then the ultrasound-based disease diagnosis and therapy were elaborated, with special interest towards the use of ultrasound in enhancing the efficacy of nanomedicines and nanodrug delivery systems, Finally, we discussed about the ability of ultrasound in enhancing the diagnostic and therapeutic properties of exosomes, which could be the future of theranostics.
Collapse
Affiliation(s)
| | - Hae Gyun Lim
- Corresponding author. Biomedical Ultrasound Lab, Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
35
|
Sousa JA, Bernardes C, Bernardo-Castro S, Lino M, Albino I, Ferreira L, Brás J, Guerreiro R, Tábuas-Pereira M, Baldeiras I, Santana I, Sargento-Freitas J. Reconsidering the role of blood-brain barrier in Alzheimer's disease: From delivery to target. Front Aging Neurosci 2023; 15:1102809. [PMID: 36875694 PMCID: PMC9978015 DOI: 10.3389/fnagi.2023.1102809] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
The existence of a selective blood-brain barrier (BBB) and neurovascular coupling are two unique central nervous system vasculature features that result in an intimate relationship between neurons, glia, and blood vessels. This leads to a significant pathophysiological overlap between neurodegenerative and cerebrovascular diseases. Alzheimer's disease (AD) is the most prevalent neurodegenerative disease whose pathogenesis is still to be unveiled but has mostly been explored under the light of the amyloid-cascade hypothesis. Either as a trigger, bystander, or consequence of neurodegeneration, vascular dysfunction is an early component of the pathological conundrum of AD. The anatomical and functional substrate of this neurovascular degeneration is the BBB, a dynamic and semi-permeable interface between blood and the central nervous system that has consistently been shown to be defective. Several molecular and genetic changes have been demonstrated to mediate vascular dysfunction and BBB disruption in AD. The isoform ε4 of Apolipoprotein E is at the same time the strongest genetic risk factor for AD and a known promoter of BBB dysfunction. Low-density lipoprotein receptor-related protein 1 (LRP-1), P-glycoprotein, and receptor for advanced glycation end products (RAGE) are examples of BBB transporters implicated in its pathogenesis due to their role in the trafficking of amyloid-β. This disease is currently devoid of strategies that change the natural course of this burdening illness. This unsuccess may partly be explained by our misunderstanding of the disease pathogenesis and our inability to develop drugs that are effectively delivered to the brain. BBB may represent a therapeutic opportunity as a target itself or as a therapeutic vehicle. In this review, we aim to explore the role of BBB in the pathogenesis of AD including the genetic background and detail how it can be targeted in future therapeutic research.
Collapse
Affiliation(s)
- João André Sousa
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Catarina Bernardes
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sara Bernardo-Castro
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miguel Lino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Inês Albino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José Brás
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Miguel Tábuas-Pereira
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - João Sargento-Freitas
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
36
|
Bok J, Ha J, Ahn BJ, Jang Y. Disease-Modifying Effects of Non-Invasive Electroceuticals on β-Amyloid Plaques and Tau Tangles for Alzheimer's Disease. Int J Mol Sci 2022; 24:ijms24010679. [PMID: 36614120 PMCID: PMC9821138 DOI: 10.3390/ijms24010679] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Electroceuticals refer to various forms of electronic neurostimulators used for therapy. Interdisciplinary advances in medical engineering and science have led to the development of the electroceutical approach, which involves therapeutic agents that specifically target neural circuits, to realize precision therapy for Alzheimer's disease (AD). To date, extensive studies have attempted to elucidate the disease-modifying effects of electroceuticals on areas in the brain of a patient with AD by the use of various physical stimuli, including electric, magnetic, and electromagnetic waves as well as ultrasound. Herein, we review non-invasive stimulatory systems and their effects on β-amyloid plaques and tau tangles, which are pathological molecular markers of AD. Therefore, this review will aid in better understanding the recent technological developments, applicable methods, and therapeutic effects of electronic stimulatory systems, including transcranial direct current stimulation, 40-Hz gamma oscillations, transcranial magnetic stimulation, electromagnetic field stimulation, infrared light stimulation and ionizing radiation therapy, and focused ultrasound for AD.
Collapse
Affiliation(s)
- Junsoo Bok
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
| | - Juchan Ha
- Department of Biomedical Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
| | - Bum Ju Ahn
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, Republic of Korea
| | - Yongwoo Jang
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, Republic of Korea
- Correspondence: ; Tel.: +82-2-2220-0655
| |
Collapse
|
37
|
Gorick CM, Breza VR, Nowak KM, Cheng VWT, Fisher DG, Debski AC, Hoch MR, Demir ZEF, Tran NM, Schwartz MR, Sheybani ND, Price RJ. Applications of focused ultrasound-mediated blood-brain barrier opening. Adv Drug Deliv Rev 2022; 191:114583. [PMID: 36272635 PMCID: PMC9712235 DOI: 10.1016/j.addr.2022.114583] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/01/2022] [Accepted: 10/14/2022] [Indexed: 01/25/2023]
Abstract
The blood brain barrier (BBB) plays a critically important role in the regulation of central nervous system (CNS) homeostasis, but also represents a major limitation to treatments of brain pathologies. In recent years, focused ultrasound (FUS) in conjunction with gas-filled microbubble contrast agents has emerged as a powerful tool for transiently and non-invasively disrupting the BBB in a targeted and image-guided manner, allowing for localized delivery of drugs, genes, or other therapeutic agents. Beyond the delivery of known therapeutics, FUS-mediated BBB opening also demonstrates the potential for use in neuromodulation and the stimulation of a range of cell- and tissue-level physiological responses that may prove beneficial in disease contexts. Clinical trials investigating the safety and efficacy of FUS-mediated BBB opening are well underway, and offer promising non-surgical approaches to treatment of devastating pathologies. This article reviews a range of pre-clinical and clinical studies demonstrating the tremendous potential of FUS to fundamentally change the paradigm of treatment for CNS diseases.
Collapse
Affiliation(s)
- Catherine M Gorick
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Victoria R Breza
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Katherine M Nowak
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Vinton W T Cheng
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Delaney G Fisher
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Anna C Debski
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Matthew R Hoch
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Zehra E F Demir
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Nghi M Tran
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Mark R Schwartz
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Natasha D Sheybani
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
38
|
Safarzadeh Kozani P, Naseri A, Mirarefin SMJ, Salem F, Nikbakht M, Evazi Bakhshi S, Safarzadeh Kozani P. Nanobody-based CAR-T cells for cancer immunotherapy. Biomark Res 2022; 10:24. [DOI: https:/doi.org/10.1186/s40364-022-00371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/29/2022] [Indexed: 09/15/2023] Open
Abstract
AbstractChimeric antigen receptor T-cell (CAR-T) therapy is the result of combining genetic engineering-based cancer immunotherapy with adoptive cell therapy (ACT). CAR-T therapy has been successful in treating various types of hematological cancers. CARs are receptors made of an extracellular domain, a membrane-spanning domain, and an intracellular domain. The extracellular domain of CARs harbors an antigen-targeting domain responsible for recognizing and binding cell surface-expressed target antigens. Conventionally, the single-chain fragment variable (scFv) of a monoclonal antibody (mAb) is used as the antigen-targeting domain of CARs. However, of late, researchers have exploited nanobodies for this aim based on numerous rationales including the small size of nanobodies, their stability, specificity, and high affinity, and their easy and feasible development process. Many findings have confirmed that nanobody-based CAR-Ts can be as functional as scFv-based CAR-Ts in preclinical and clinical settings. In this review, we discuss the advantages and disadvantages of scFvs and nanobodies in regards to their application as the targeting domain of CARs. Ultimately, we discuss various CAR target antigens which have been targeted using nanobody-based CAR-T cells for the treatment of different types of malignancies.
Collapse
|
39
|
Modern Advances in CARs Therapy and Creating a New Approach to Future Treatment. Int J Mol Sci 2022; 23:ijms232315006. [PMID: 36499331 PMCID: PMC9739283 DOI: 10.3390/ijms232315006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Genetically engineered T and NK cells expressing a chimeric antigen receptor (CAR) are promising cytotoxic cells for the treatment of hematological malignancies and solid tumors. Despite the successful therapies using CAR-T cells, they have some disadvantages, such as cytokine release syndrome (CRS), neurotoxicity, or graft-versus-host-disease (GVHD). CAR-NK cells have lack or minimal cytokine release syndrome and neurotoxicity, but also multiple mechanisms of cytotoxic activity. NK cells are suitable for developing an "off the shelf" therapeutic product that causes little or no graft versus host disease (GvHD), but they are more sensitive to apoptosis and have low levels of gene expression compared to CAR-T cells. To avoid these adverse effects, further developments need to be considered to enhance the effectiveness of adoptive cellular immunotherapy. A promising approach to enhance the effectiveness of adoptive cellular immunotherapy is overcoming terminal differentiation or senescence and exhaustion of T cells. In this case, EVs derived from immune cells in combination therapy with drugs may be considered in the treatment of cancer patients, especially effector T and NK cells-derived exosomes with the cytotoxic activity of their original cells.
Collapse
|
40
|
Mehkri Y, Woodford S, Pierre K, Dagra A, Hernandez J, Reza Hosseini Siyanaki M, Azab M, Lucke-Wold B. Focused Delivery of Chemotherapy to Augment Surgical Management of Brain Tumors. Curr Oncol 2022; 29:8846-8861. [PMID: 36421349 PMCID: PMC9689062 DOI: 10.3390/curroncol29110696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Chemotherapy as an adjuvant therapy that has largely failed to significantly improve outcomes for aggressive brain tumors; some reasons include a weak blood brain barrier penetration and tumor heterogeneity. Recently, there has been interest in designing effective ways to deliver chemotherapy to the tumor. In this review, we discuss the mechanisms of focused chemotherapies that are currently under investigation. Nanoparticle delivery demonstrates both a superior permeability and retention. However, thus far, it has not demonstrated a therapeutic efficacy for brain tumors. Convection-enhanced delivery is an invasive, yet versatile method, which appears to have the greatest potential. Other vehicles, such as angiopep-2 decorated gold nanoparticles, polyamidoamine dendrimers, and lipid nanostructures have demonstrated efficacy through sustained release of focused chemotherapy and have either improved cell death or survival in humans or animal models. Finally, focused ultrasound is a safe and effective way to disrupt the blood brain barrier and augment other delivery methods. Clinical trials are currently underway to study the safety and efficacy of these methods in combination with standard of care.
Collapse
Affiliation(s)
- Yusuf Mehkri
- Department of Neurosurgery, College of Medicine, University of Florida, 1505 SW Archer Rd, Gainesville, FL 32608, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Kofoed RH, Dibia CL, Noseworthy K, Xhima K, Vacaresse N, Hynynen K, Aubert I. Efficacy of gene delivery to the brain using AAV and ultrasound depends on serotypes and brain areas. J Control Release 2022; 351:667-680. [DOI: 10.1016/j.jconrel.2022.09.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 02/01/2023]
|
42
|
Opportunities and challenges in delivering biologics for Alzheimer's disease by low-intensity ultrasound. Adv Drug Deliv Rev 2022; 189:114517. [PMID: 36030018 DOI: 10.1016/j.addr.2022.114517] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/24/2023]
Abstract
Low-intensity ultrasound combined with intravenously injected microbubbles (US+MB) is a novel treatment modality for brain disorders, including Alzheimer's disease (AD), safely and transiently allowing therapeutic agents to overcome the blood-brain barrier (BBB) that constitutes a major barrier for therapeutic agents. Here, we first provide an update on immunotherapies in AD and how US+MB has been applied to AD mouse models and in clinical trials, considering the ultrasound and microbubble parameter space. In the second half of the review, we compare different in vitro BBB models and discuss strategies for combining US+MB with BBB modulators (targeting molecules such as claudin-5), and highlight the insight provided by super-resolution microscopy. Finally, we conclude with a short discussion on how in vitro findings can inform the design of animal studies, and how the insight gained may aid treatment optimization in the clinical ultrasound space.
Collapse
|
43
|
Bajracharya R, Cruz E, Götz J, Nisbet RM. Ultrasound-mediated delivery of novel tau-specific monoclonal antibody enhances brain uptake but not therapeutic efficacy. J Control Release 2022; 349:634-648. [PMID: 35901857 DOI: 10.1016/j.jconrel.2022.07.026] [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] [Received: 05/10/2022] [Revised: 07/16/2022] [Accepted: 07/21/2022] [Indexed: 11/25/2022]
Abstract
Tau-specific immunotherapy is an attractive strategy for the treatment of Alzheimer's disease and other tauopathies. However, effectively targeting tau in the brain remains a considerable challenge due to the restrictive nature of the blood-brain barrier (BBB), which excludes an estimated >99% of peripherally administered antibodies. However, their transport across the BBB can be facilitated by a novel modality, low-intensity scanning ultrasound used in combination with intravenously injected microbubbles (SUS+MB). We have previously shown that SUS+MB-mediated delivery of a tau-specific antibody in a single-chain (scFv) format to tau transgenic mice enhanced brain and neuronal uptake and subsequently, reduced tau pathology and improved behavioural outcomes to a larger extent than either scFv or SUS+MB on its own. Here we generated a novel tau-specific monoclonal antibody, RNF5, and validated it in its IgG format in the presence or absence of SUS+MB by treating K369I tau transgenic K3 mice once weekly for 12 weeks. We found that both RNF5 and SUS+MB treatments on their own significantly reduced tau pathology. In the combination group (RNF5 + SUS+MB), however, despite increased antibody localization in the brain, there were no further reductions in tau pathology when compared to RNF5 treatment alone. Furthermore, following SUS+MB, RNF5 accumulated heavily within cells across the pyramidal cell layer of the hippocampus, that were negative for MAP2 and p-tau, suggesting that SUS+MB may not facilitate enhanced RNF5 engagement of intraneuronal tau. Overall, our new findings reveal the complexities of combining tau immunotherapy with SUS+MB and challenge the view that this is a straight-forward approach.
Collapse
Affiliation(s)
- Rinie Bajracharya
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Queensland 4072, Australia
| | - Esteban Cruz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Queensland 4072, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Queensland 4072, Australia.
| | - Rebecca M Nisbet
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Queensland 4072, Australia; Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3052, Australia.
| |
Collapse
|
44
|
Pineda-Pardo JA, Gasca-Salas C, Fernández-Rodríguez B, Rodríguez-Rojas R, Del Álamo M, Obeso I, Hernández-Fernández F, Trompeta C, Martínez-Fernández R, Matarazzo M, Mata-Marín D, Guida P, Duque A, Albillo D, Plaza de Las Heras I, Montero JI, Foffani G, Toltsis G, Rachmilevitch I, Blesa J, Obeso JA. Striatal Blood-Brain Barrier Opening in Parkinson's Disease Dementia: A Pilot Exploratory Study. Mov Disord 2022; 37:2057-2065. [PMID: 35765711 DOI: 10.1002/mds.29134] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/02/2022] [Accepted: 06/01/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) exhibits a high prevalence of dementia as disease severity and duration progress. Focused ultrasound (FUS) has been applied for transient blood-brain barrier (BBB) opening of cortical regions in neurodegenerative disorders. The striatum is a primary target for delivery of putative therapeutic agents in PD. OBJECTIVE Here, we report a prospective, single-arm, nonrandomized, proof-of-concept, phase I clinical trial (NCT03608553 amended) in PD with dementia to test the safety and feasibility of striatal BBB opening in PD patients. METHODS Seven PD patients with cognitive impairment were treated for BBB opening in the posterior putamen. This was performed in two sessions separated by 2 to 4 weeks, where the second session included bilateral putamina opening in 3 patients. Primary outcome measures included safety and feasibility of focal striatal BBB opening. Changes in motor and cognitive functions, magnetic resonance imaging (MRI), 18 F-fluorodopa (FDOPA), and β-amyloid PET (positron emission tomography) images were determined. RESULTS The procedure was feasible and well tolerated, with no serious adverse events. No neurologically relevant change in motor and cognitive (battery of neuropsychological tests) functions was recognized at follow-up. MRI revealed putamen BBB closing shortly after treatment (24 hours to 14 days) and ruled out hemorrhagic and ischemic lesions. There was a discrete but significant reduction in β-amyloid uptake in the targeted region and no change in FDOPA PET. CONCLUSIONS These initial results indicate that FUS-mediated striatal BBB opening is feasible and safe and therefore could become an effective tool to facilitate the delivery of putative neurorestorative molecules in PD. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- José A Pineda-Pardo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.,University CEU-San Pablo, Madrid, Spain
| | - Carmen Gasca-Salas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.,University CEU-San Pablo, Madrid, Spain
| | - Beatriz Fernández-Rodríguez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
| | - Rafael Rodríguez-Rojas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Marta Del Álamo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Ignacio Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Frida Hernández-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Clara Trompeta
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Raúl Martínez-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Michele Matarazzo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - David Mata-Marín
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Pasqualina Guida
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Alicia Duque
- Neuroradiology Unit, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - David Albillo
- Neuroradiology Unit, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | | | - Juan I Montero
- Intensive Care Unit, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Guglielmo Foffani
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.,Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | | | | | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - José A Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Fundación Hospitales de Madrid, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.,University CEU-San Pablo, Madrid, Spain
| |
Collapse
|
45
|
Balbi M, Blackmore DG, Padmanabhan P, Götz J. Ultrasound-Mediated Bioeffects in Senescent Mice and Alzheimer's Mouse Models. Brain Sci 2022; 12:775. [PMID: 35741660 PMCID: PMC9221310 DOI: 10.3390/brainsci12060775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/01/2023] Open
Abstract
Ultrasound is routinely used for a wide range of diagnostic imaging applications. However, given that ultrasound can operate over a wide range of parameters that can all be modulated, its applicability extends far beyond the bioimaging field. In fact, the modality has emerged as a hybrid technology that effectively assists drug delivery by transiently opening the blood-brain barrier (BBB) when combined with intravenously injected microbubbles, and facilitates neuromodulation. Studies in aged mice contributed to an insight into how low-intensity ultrasound brings about its neuromodulatory effects, including increased synaptic plasticity and improved cognitive functions, with a potential role for neurogenesis and the modulation of NMDA receptor-mediated neuronal signalling. This work is complemented by studies in mouse models of Alzheimer's disease (AD), a form of pathological ageing. Here, ultrasound was mainly employed as a BBB-opening tool that clears protein aggregates via microglial activation and neuronal autophagy, thereby restoring cognition. We discuss the currently available ultrasound approaches and how studies in senescent mice are relevant for AD and can accelerate the application of low-intensity ultrasound in the clinic.
Collapse
Affiliation(s)
- Matilde Balbi
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
| | - Daniel G. Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Pranesh Padmanabhan
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jürgen Götz
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
46
|
Safarzadeh Kozani P, Naseri A, Mirarefin SMJ, Salem F, Nikbakht M, Evazi Bakhshi S, Safarzadeh Kozani P. Nanobody-based CAR-T cells for cancer immunotherapy. Biomark Res 2022; 10:24. [PMID: 35468841 PMCID: PMC9036779 DOI: 10.1186/s40364-022-00371-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/29/2022] [Indexed: 12/23/2022] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is the result of combining genetic engineering-based cancer immunotherapy with adoptive cell therapy (ACT). CAR-T therapy has been successful in treating various types of hematological cancers. CARs are receptors made of an extracellular domain, a membrane-spanning domain, and an intracellular domain. The extracellular domain of CARs harbors an antigen-targeting domain responsible for recognizing and binding cell surface-expressed target antigens. Conventionally, the single-chain fragment variable (scFv) of a monoclonal antibody (mAb) is used as the antigen-targeting domain of CARs. However, of late, researchers have exploited nanobodies for this aim based on numerous rationales including the small size of nanobodies, their stability, specificity, and high affinity, and their easy and feasible development process. Many findings have confirmed that nanobody-based CAR-Ts can be as functional as scFv-based CAR-Ts in preclinical and clinical settings. In this review, we discuss the advantages and disadvantages of scFvs and nanobodies in regards to their application as the targeting domain of CARs. Ultimately, we discuss various CAR target antigens which have been targeted using nanobody-based CAR-T cells for the treatment of different types of malignancies.
Collapse
Affiliation(s)
- Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Abdolhossein Naseri
- School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | | | - Faeze Salem
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Nikbakht
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Evazi Bakhshi
- Department of Anatomical Sciences, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
47
|
Barbato G, Nisticò R, Triaca V. Exploiting Focused Ultrasound to Aid Intranasal Drug Delivery for Brain Therapy. Front Pharmacol 2022; 13:786475. [PMID: 35496270 PMCID: PMC9046653 DOI: 10.3389/fphar.2022.786475] [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: 09/30/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Novel effective therapeutic strategies are needed to treat brain neurodegenerative diseases and to improve the quality of life of patients affected by Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), Amyotrophic Lateral sclerosis (ALS) as well as other brain conditions. At present no effective treatment options are available; current therapeutics for neurodegenerative diseases (NDs) improve cognitive symptoms only transiently and in a minor number of patients. Further, most of the amyloid-based phase III clinical trials recently failed in AD, in spite of promising preclinical and phase I-II clinical trials, further pinpointing the need for a better knowledge of the early mechanisms of disease as well as of more effective routes of drug administration. In fact, beyond common pathological events and molecular substrates, each of these diseases preferentially affect defined subpopulations of neurons in specific neuronal circuits (selective neuronal vulnerability), leading to the typical age-related clinical profile. In this perspective, key to successful drug discovery is a robust and reproducible biological validation of potential new molecular targets together with a concomitant set up of protocols/tools for efficient and targeted brain delivery to a specific area of interest. Here we propose and discuss Focused UltraSound aided drug administration as a specific and novel technical approach to achieve optimal concentration of the drug at the target area of interest. We will focus on drug delivery to the brain through the nasal route coupled to FUS as a promising approach to achieve neuroprotection and rescue of cognitive decline in several NDs.
Collapse
Affiliation(s)
- Gaetano Barbato
- Inno-Sol Srl, Rome, Italy
- Department of Biology, School of Pharmacy, University of Tor Vergata, Rome, Italy
- *Correspondence: Gaetano Barbato, ; Robert Nisticò, ; Viviana Triaca,
| | - Robert Nisticò
- Department of Biology, School of Pharmacy, University of Tor Vergata, Rome, Italy
- Laboratory of Pharmacology of Synaptic Plasticity, Fondazione EBRI Rita Levi Montalcini, Rome, Italy
- *Correspondence: Gaetano Barbato, ; Robert Nisticò, ; Viviana Triaca,
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), International Campus A. Buzzati-Traverso, Rome, Italy
- *Correspondence: Gaetano Barbato, ; Robert Nisticò, ; Viviana Triaca,
| |
Collapse
|
48
|
Guo P, Zeng M, Wang S, Cao B, Liu M, Zhang Y, Jia J, Zhang Q, Zhang B, Wang R, Zheng X, Feng W. Eriodictyol and Homoeriodictyol Improve Memory Impairment in Aβ 25-35-Induced Mice by Inhibiting the NLRP3 Inflammasome. Molecules 2022; 27:2488. [PMID: 35458684 PMCID: PMC9025671 DOI: 10.3390/molecules27082488] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/01/2022] [Accepted: 04/09/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Alzheimer's disease (AD) is a neurodegenerative disorder, and it is now widely accepted that neuroinflammation plays a key role in its pathogenesis. Eriodictyol (Eri) and homoeriodictyol (Hom), dihydroflavonoids extracted from a variety of plants, have been confirmed to display a relationship with neuroprotection. (2) Methods: An AD mouse model was constructed by intracerebroventricular (ICV) injection of the Aβ25-35 peptide, and Eri and Hom were administered orally for 4 weeks. UPLC-MS/MS was used to determine whether Eri and Hom cross the blood-brain barrier to exert their therapeutic effects. Histological changes in the brain and levels of Aβ were evaluated, and Y-maze and new object recognition experiments were conducted to assess the effects of Eri and Hom on Aβ25-35-induced memory impairment in mice. The levels of oxidative stress and apoptosis in peripheral immune cells and progenitor cells in the hippocampal region were analyzed by flow cytometry and in vitro assays. Western blotting and enzyme-linked immunosorbent assays (ELISA) were used to measure the expression levels of NLRP3 inflammasome-related proteins and inflammatory factors in the brain. The effect of nigericin (an agonist of the NLRP3 inflammasome) on Eri and Hom intervention in LPS-induced N9 microglia was examined using a High Content Screening System. (3) Results: Eri and Hom reduced neuronal damage in mouse brain tissue, decreased Aβ levels in the brain, downregulated oxidative stress and apoptosis levels, and improved learning and memory capacity by crossing the blood-brain barrier to exert its effects. Moreover, Eri and Hom inhibited NLRP3 inflammasome activation and ameliorated immune cell disorder. Furthermore, the effect of Eri and Hom on LPS-induced N9 microglia disappeared after the addition of nigericin to agonize NLRP3 receptors. (4) Conclusions: Eri and Hom improved Aβ25-35-induced memory impairment in mice by inhibiting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Pengli Guo
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Shengchao Wang
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Bing Cao
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Meng Liu
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yuhan Zhang
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Jufang Jia
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Qinqin Zhang
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Ru Wang
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; (P.G.); (M.Z.); (S.W.); (B.C.); (M.L.); (Y.Z.); (J.J.); (Q.Z.); (B.Z.); (R.W.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| |
Collapse
|
49
|
Wasielewska JM, White AR. "Focused Ultrasound-mediated Drug Delivery in Humans - a Path Towards Translation in Neurodegenerative Diseases". Pharm Res 2022; 39:427-439. [PMID: 35257286 PMCID: PMC8986691 DOI: 10.1007/s11095-022-03185-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/31/2022] [Indexed: 11/04/2022]
Abstract
The blood-brain barrier (BBB) has a major protective function in preventing the entry of harmful molecules into the brain, but is simultaneously limiting the delivery of drugs, restricting their potential clinical application in neurodegenerative diseases. Recent preclinical evidence demonstrates that following application of focused ultrasound with microbubbles (FUS+MB), the BBB becomes reversibly accessible to compounds that normally are brain-impermeable, suggesting FUS+MB as a promising new platform for delivery of therapeutic agents into the central nervous system. As a step towards translation, small cohort clinical studies were performed demonstrating safe BBB opening in Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis (ALS) patients following FUS+MB, however improved drug delivery has not yet been achieved in human. Simultaneously, rapid progress in the human induced pluripotent stem cell (hiPSC) modeling technology allowed for development of novel Alzheimer's disease patient-derived BBB in vitro model that reacts to FUS+MB with BBB opening and can be used to answer fundamental questions of human BBB responses to FUS+MB in health and disease. This review summarizes key features of the BBB that contribute to limited drug delivery, recapitulates recent advances in the FUS+MB mediated human BBB opening in vivo and in vitro in the context of neurodegenerative disorders, and highlights potential strategies for fast-track translation of the FUS+MB to improve bioavailability of drugs to the human brain. With safe and effective application, this innovative FUS+MB technology may open new avenues for therapeutic interventions in neurodegenerative diseases leading to improved clinical outcomes for patients.
Collapse
Affiliation(s)
- Joanna M Wasielewska
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| | - Anthony R White
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
50
|
Zhu Q, Xu X, Chen B, Liao Y, Guan X, He Y, Cui H, Rong Y, Liu Z, Xu Y. Ultrasound targeted microbubble destruction assists dual delivery of beta-amyloid antibody and neural stem cells to restore neural function in transgenic mice of Alzheimer's disease. Med Phys 2022; 49:1357-1367. [PMID: 35092698 DOI: 10.1002/mp.15500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To explore the feasibility, efficacy and safety of ultrasound targeted microbubbles destruction (UTMD) assisted dual delivery of beta-amyloid (Aβ) antibody loaded by microbubbles (MBAβ ) and neural stem cells (NSCs) on Alzheimer's disease (AD). METHODS 27 APP/PS1 double transgenic mice (Tg mice) and 33 wild-type mice were used. Wild-type mice were insonated by diagnostic ultrasound with microbubbles (MB) for 5 min to observe the blood brain barrier (BBB) opening. The survival situation of engrafted NSCs crossing the opened BBB mediated by UTMD in AD mice was evaluated by in vivo imaging system. We further explored the combination therapy of UTMD mediated Aβ antibody and NSCs dual delivery. Tg mice in each group were exposed to diagnostic ultrasound for 5 min once a week for four times, with MB, MBAβ , and/or NSCs administration. Cognition and memory functions were explored by Morris water maze test, Aβ plaques deposition was evaluated by immunohistochemical, and brain-derived neurotrophic factor (BDNF) and synaptophysin (SYN) expression were detected by western blot and immunofluorescence. RESULTS BBB was opened mediated by diagnostic ultrasound with MB and the duration of opening was about 10 h. The transplanted NSCs survived in Tg mice for no more than 72 h. Compared with control group, the mice in combined delivery of NSCs and Aβ antibody by UTMD group improved memory function and spatial learning with shorter latency to find the platform, longer distance traveled and longer time spent in targeted quadrant, and more crossing times (P < 0.05). Besides, the combination delivery group promoted the clearance of Aβ plaques compared with control group both in hippocampus (P < 0.01) and cortex (P < 0.05). Moreover, the expression of BDNF in combination delivery group was significantly higher than that in control group and US mediated MB group (P < 0.05). No significant change of SYN was observed in each group. CONCLUSION UTMD assisted dual delivery of Aβ antibody and NSCs to AD mice brain could help to clear Aβ plaques, increase the expression of BDNF and restore the impaired neural function. This finding may offer potential insight into treatment of AD. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China
| | - Xiaoxun Xu
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China
| | - Beibei Chen
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China.,Postgraduate Training base of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yiyi Liao
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China
| | - Xue Guan
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China
| | - Ying He
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China
| | - Hai Cui
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China.,Department of Ultrasound, Armed Police Force Hospital, Sichuan, 614000, China
| | - Yani Rong
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital of Army Medical University, Chongqing, 400038, China
| |
Collapse
|