1
|
Han Y, Zhou S, Xiang H, Zhang Q, Zhang H, Zhang Y, Lin Q, Zhang Z, Zhang L, Huang S. A brain-targeting dl-3-n-butylphthalide prodrug to treat ischemic stroke. Eur J Med Chem 2025; 290:117474. [PMID: 40120494 DOI: 10.1016/j.ejmech.2025.117474] [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: 01/03/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Stroke is an acute cerebrovascular disease worldwide, featured by stubbornly high mortality and morbidity. 3-n-butylphthalide (NBP) is a first-line agent to treat ischemic stroke, but its unsatisfactorily intracephalic bioavailability is detrimental to the therapeutic efficacy. We have previously developed a series of prodrugs analogous to NBP, however, their conversion rate in the targeted sites cannot meet the clinical demand. Hence, it is imperative to ameliorate the bioavailability and cellular uptake efficiency of the prodrugs, which may be driven by potential energy consuming cationic transporters. Accordingly, we fabricated a novel prodrug named DB-10, which were used tertiary amine to possess active targeting capacity and connected through amide bonds. Moreover, it is noteworthy that DB-10 could rapidly convert into active original drug in the brain to treat acute injury caused by ischemia-reperfusion (I/R). The cytotoxicity of DB-10 was equivalent to that of NBP and would not cause obviously undesired damage in vivo. These encouraging results demonstrate that DB-10 could easily increase the accumulation in the brain site, and significantly improve the treatment effect for ischemic stroke, indicating this biosafety prodrug holds tremendous potential for clinical application prospects.
Collapse
Affiliation(s)
- Yikun Han
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Shuhao Zhou
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610000, China
| | - Honglin Xiang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Qiang Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Hanming Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Yicong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610000, China; Med-X Center for Materials, Sichuan University, Chengdu, 610000, China.
| | - Shiqi Huang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610000, China.
| |
Collapse
|
2
|
Chen Y, Zhang J, Xu H. Recent developments in cuproptosis of glioblastoma. Pathol Res Pract 2025; 269:155939. [PMID: 40164043 DOI: 10.1016/j.prp.2025.155939] [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: 12/18/2024] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Glioblastoma (GBM) is the most malignant tumor within the central nervous system, attributed to its high-grade malignancy, propensity for recurrence, refractoriness to conventional therapeutic modalities, and the suboptimal efficacy of current targeted therapies. Hence, there is an urgent need to identify more efficacious molecular targets for the therapeutic intervention of GBM. The regulated cell death (RCD) has specific signaling factors and signaling pathways. Hence, targeting RCD is considered to be one of the effective targeted therapies for GBM. At present, cuproptosis is a novel form of RCD, characterized by a distinct molecular mechanism that differentiates it from apoptosis, pyroptosis, necroptosis, and ferroptosis. It is characterized by its principal mechanisms, which include copper dependency, the accumulation of acylated proteins, and the reduction of Fe-S cluster-containing proteins. These processes collectively induce proteotoxic stress, culminating in cell death. In previous studies, copper-ionized formulations have demonstrated cytotoxic effects on gliomas. Thus, the key factors of cuproptosis may be able to serve as a new target for GBM treatment. This review delves into several pivotal aspects, including the discovery of cuproptosis, the impact of copper homeostasis on tumorigenesis, the role of cuproptosis in GBM, and its potential as a therapeutic target in molecular targeted therapy for GBM. Hence, this article could reveal novel strategies for GBM treatment.
Collapse
Affiliation(s)
- Yajia Chen
- Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China
| | - Jingxian Zhang
- Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China
| | - Hongwu Xu
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Wandao Rd, 78, Wanjiang Subdistrict, Dongguan, Guangdong Province 523059, China; Shantou University Medical College, No. 22 Xinlin Rd, Jinping District, Shantou, Guangdong 515031, China.
| |
Collapse
|
3
|
Yang J, des Rieux A, Malfanti A. Stimuli-Responsive Nanomedicines for the Treatment of Non-cancer Related Inflammatory Diseases. ACS NANO 2025. [PMID: 40249331 DOI: 10.1021/acsnano.5c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Nanomedicines offer a means to overcome the limitations associated with traditional drug dosage formulations by affording drug protection, enhanced drug bioavailability, and targeted drug delivery to affected sites. Inflamed tissues possess unique microenvironmental characteristics (including excessive reactive oxygen species, low pH levels, and hypoxia) that stimuli-responsive nanoparticles can employ as triggers to support on-demand delivery, enhanced accumulation, controlled release, and activation of anti-inflammatory drugs. Stimuli-responsive nanomedicines respond to physicochemical and pathological factors associated with diseased tissues to improve the specificity of drug delivery, overcome multidrug resistance, ensure accurate diagnosis and precision therapy, and control drug release to improve efficacy and safety. Current stimuli-responsive nanoparticles react to intracellular/microenvironmental stimuli such as pH, redox, hypoxia, or specific enzymes and exogenous stimuli such as temperature, magnetic fields, light, and ultrasound via bioresponsive moieties. This review summarizes the general strategies employed to produce stimuli-responsive nanoparticles tailored for inflammatory diseases and all recent advances, reports their applications in drug delivery, and illustrates the progress made toward clinical translation.
Collapse
Affiliation(s)
- Jingjing Yang
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Anne des Rieux
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
4
|
Wang X, Zhou Z, Zhang Y, Liu J, Qin T, Zhou W, Li Q, Wu X, Xue K, Cao H, Su Y, Zhao S, Lu C, Jiang T, Yin G, Chen J. Exosome-shuttled miR-5121 from A2 astrocytes promotes BSCB repair after traumatic SCI by activating autophagy in vascular endothelial cells. J Nanobiotechnology 2025; 23:291. [PMID: 40229869 PMCID: PMC11998472 DOI: 10.1186/s12951-025-03365-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disorder that significantly impacts patients' quality of life. Following SCI, the blood-spinal cord barrier (BSCB) is destroyed, leading to ischemia and hypoxia, which further exacerbates the imbalance in the spinal cord microenvironment. A2-type astrocytes, which arise under ischemic and hypoxic conditions, have been reported to promote SCI repair. However, the roles of exosomes derived from A2 astrocytes (A2-Exos) in SCI have not been explored. This study aims to investigate the role of A2-Exos in SCI repair, particularly in BSCB restoration, and to elucidate its potential mechanisms. GEO database analysis, western blotting, and immunofluorescence were used to detect A2 astrocyte polarization after SCI in mice. In vitro, A2 astrocytes were obtained through hypoxia induction, and A2-Exos were extracted via ultracentrifugation. An in vivo SCI model and a series of in vitro experiments demonstrated the reparative effects of A2-Exos on BSCB following SCI. Furthermore, miRNA sequencing analysis and rescue experiments confirmed the role of miRNAs in A2-Exos-mediated BSCB repair. Finally, luciferase assays and western blotting were performed to investigate the underlying mechanisms. The results showed that A2-Exos promote motor function recovery and BSCB repair in mice following SCI. In vitro, A2-Exos facilitated BSCB reconstruction and endothelial cell autophagy. miRNA sequencing identified miR-5121 as the most significantly enriched miRNA in A2-Exos, suggesting its involvement in BSCB repair and autophagy regulation. AKT2 was identified as a potential downstream target of miR-5121. Functional gain- and loss-of-function experiments further validated the miR-5121/AKT2 axis. Finally, we demonstrated that the AKT2/mTOR/p70S6K pathway may mediate the effects of miR-5121 in A2-Exos on BSCB repair.
Collapse
Affiliation(s)
- Xiaowei Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
- Department of Orthopedics, Maanshan People's Hospital, Maanshan, Anhui, 243000, China
| | - Zihan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Yu Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Jiayun Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Tao Qin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Wei Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Xincan Wu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Kaixiao Xue
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Heng Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Yunxin Su
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Chun Lu
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, China.
| | - Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
5
|
Song M, Ren J, Zhu Z, Yi Z, Wang C, Liang L, Tian J, Mao G, Mao G, Chen M. The STING Signaling: A Novel Target for Central Nervous System Diseases. Cell Mol Neurobiol 2025; 45:33. [PMID: 40195137 PMCID: PMC11977075 DOI: 10.1007/s10571-025-01550-4] [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: 01/11/2025] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
The canonical cyclic GMP-AMP (cGAMP) synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway has been widely recognized as a crucial mediator of inflammation in many diseases, including tumors, infections, and tissue damage. STING signaling can also be activated in a cGAS- or cGAMP-independent manner, although the specific mechanisms remain unclear. In-depth studies on the structural and molecular biology of the STING pathway have led to the development of therapeutic strategies involving STING modulators and their targeted delivery. These strategies may effectively penetrate the blood-brain barrier (BBB) and target STING signaling in multiple central nervous system (CNS) diseases in humans. In this review, we outline both canonical and non-canonical pathways of STING activation and describe the general mechanisms and associations between STING activity and CNS diseases. Finally, we discuss the prospects for the targeted delivery and clinical application of STING agonists and inhibitors, highlighting the STING signaling pathway as a novel therapeutic target in CNS diseases.
Collapse
Affiliation(s)
- Min Song
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Jianxun Ren
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Zhipeng Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
- Department of Neurosurgery, Shangrao People's Hospital, Shangrao, Jiangxi Province, China
| | - Zhaohui Yi
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Chengyun Wang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Lirong Liang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Jiahui Tian
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Guofu Mao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China
| | - Guohua Mao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China.
| | - Min Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, Jiangxi, China.
| |
Collapse
|
6
|
Jing Y, Zhao G, Xu Y, McGuire T, Hou G, Zhao J, Chen M, Lopez O, Xue Y, Xie XQ. GCN-BBB: Deep Learning Blood-Brain Barrier (BBB) Permeability PharmacoAnalytics with Graph Convolutional Neural (GCN) Network. AAPS J 2025; 27:73. [PMID: 40180695 DOI: 10.1208/s12248-025-01059-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
The Blood-Brain Barrier (BBB) is a selective barrier between the Central Nervous System (CNS) and the peripheral system, regulating the distribution of molecules. BBB permeability has been crucial in CNS-targeting drug development, such as glioblastoma-related drug discovery. In addition, more CNS diseases still present significant challenges, for instance, neurological disorders like Alzheimer's Disease (AD) and drug abuse. Conversely, cannabinoid drugs that do not cross the BBB are needed to avoid off-target CNS psychotropic effects. In vitro and in vivo experiments measuring BBB permeability are costly and low throughput. Computational pharmacoanalytics modeling, particularly using deep-learning Graph Neural Networks (GNNs), offers a promising alternative. GNNs excel at capturing intricate relationships in graph-based information, such as small molecular structures. In this study, we developed GNNs model for BBB permeability using the graph representation of drugs. The GNNs were compared with other algorithms using molecular fingerprints or physical-chemical descriptors. With a dataset of 1924 molecules, the best GNNs model, a convolutional graph neural network using a normalized Laplacian matrix (GCN_2), achieved a precision of 0.94, recall of 0.96, F1 score of 0.95, and MCC score of 0.77. This outperformed other machine learning algorithms with molecular fingerprints. The findings indicate that the graphic representation of small molecules combined with GNNs architecture is powerful in predicting BBB permeability with high accuracy and recall. The developed GNNs model can be utilized in the initial screening stage for new drug development.
Collapse
Affiliation(s)
- Yankang Jing
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
| | - Guangyi Zhao
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
| | - Yuanyuan Xu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
| | - Terence McGuire
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
| | - Ganqian Hou
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
| | - Jack Zhao
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
| | - Maozi Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America
| | - Oscar Lopez
- Department of Neurology, Psychiatry and Clinical & Translational Sciences, Alzheimer'S Disease Research Center, University of Pittsburgh, Pittsburgh, 15260, United States of America.
| | - Ying Xue
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America.
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America.
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America.
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology (PSP) Pharmacoanalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America.
- National Center of Excellence for Computational Drug Abuse Research University of Pittsburgh, Pittsburgh, PA, 15261, United States of America.
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, 15261, United States of America.
- Department of Computational Biology and Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, United States of America.
| |
Collapse
|
7
|
Romero-Ben E, Goswami U, Soto-Cruz J, Mansoori-Kermani A, Mishra D, Martin-Saldaña S, Muñoz-Ugartemendia J, Sosnik A, Calderón M, Beloqui A, Larrañaga A. Polymer-based nanocarriers to transport therapeutic biomacromolecules across the blood-brain barrier. Acta Biomater 2025; 196:17-49. [PMID: 40032217 DOI: 10.1016/j.actbio.2025.02.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Therapeutic biomacromolecules such as genetic material, antibodies, growth factors and enzymes represent a novel therapeutic alternative for neurological diseases and disorders. In comparison to traditional therapeutics, which are mainly based on small molecular weight drugs that address the symptoms of these disorders, therapeutic biomacromolecules can reduce undesired side effects and target specific pathological pathways, thus paving the way towards personalized medicine. However, these biomacromolecules undergo degradation/denaturation processes in the physiological environment and show poor capacity to cross the blood-brain barrier (BBB). Consequently, they rarely reach the central nervous system (CNS) in their active form. Herein, we critically overview several polymeric nanocarriers that can protect and deliver therapeutic biomacromolecules across the BBB. Polymeric nanocarriers are first categorized based on their architecture (biodegradable solid nanoparticles, nanogels, dendrimers, self-assembled nanoparticles) that ultimately determines their physico-chemical properties and function. The available polymeric formulations are then thoroughly analyzed, placing particular attention on those strategies that ensure the stability of the biomacromolecules during their encapsulation process and promote their passage across the BBB by controlling their physical (e.g., mechanical properties, size, surface charge) and chemical (e.g., surface functional groups, targeting motifs) properties. Accordingly, this review gives a unique perspective on polymeric nanocarriers for the delivery of therapeutic biomacromolecules across the BBB, representing a concise, complete and easy-to-follow guide, which will be of high interest for chemists, material scientists, pharmacologists, and biologists. Besides, it also provides a critical perspective about the limited clinical translation of these systems. STATEMENT OF SIGNIFICANCE: The increasing incidence of central nervous system disorders is a major health concern. The use of therapeutic biomacromolecules has been placed in the spotlight of many investigations. However, reaching therapeutic concentration levels of biomacromolecules in the central nervous system is restricted by the blood-brain barrier and, thus, this represents the main clinical challenge when developing efficient therapies. Herein, we provide a critical discussion about the use of polymeric nanocarriers to deliver therapeutic biomacromolecules into the central nervous system, highlighting potential future directions to overcome the current challenges.
Collapse
Affiliation(s)
- Elena Romero-Ben
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Upashi Goswami
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain
| | - Jackeline Soto-Cruz
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Amirreza Mansoori-Kermani
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain; Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy; Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo PIaggio 34, Pontedera 56025, Italy
| | - Dhiraj Mishra
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Sergio Martin-Saldaña
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Jone Muñoz-Ugartemendia
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Ana Beloqui
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain.
| |
Collapse
|
8
|
Zhao Q, Li Y, Sun Q, Wang R, Lu H, Zhang X, Gao L, Cai Q, Liu B, Deng G. Self-assembled genistein nanoparticles suppress the epithelial-mesenchymal transition in glioblastoma by targeting MMP9. Mater Today Bio 2025; 31:101606. [PMID: 40104644 PMCID: PMC11919400 DOI: 10.1016/j.mtbio.2025.101606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive primary malignant brain tumor in adults, known for its poor prognosis and resistance to conventional treatments. The blood-brain barrier (BBB) presents a significant challenge in delivering effective treatments. In this study, we developed a carrier-free, self-assembled nanosystem using genistein (GE), a naturally occurring isoflavone, to enhance therapeutic delivery across the BBB. GE nanoparticles (GE NPs) were synthesized via solvent emulsification evaporation, in uniform spherical particles (∼180 nm), stabilized by hydrogen bonding and π-π interactions. The GE NPs demonstrated optimal physicochemical properties, including stability, high BBB permeability, prolonged circulation time. In vitro studies revealed that GE NPs inhibited GBM cell proliferation, induced apoptosis and suppressed epithelial-mesenchymal transition (EMT) by promoting the degradation of MMP9. In vivo, GE NPs significantly reduced tumor growth and extended survival in an orthotopic GBM mouse model, outperforming temozolomide treatment. Mechanistic analysis indicated that GE NPs inhibited the degradation of the extracellular matrix by targeting the catalytic domain of MMP9, thereby effectively suppressing the EMT of GBM. This research highlights the potential of GE NPs as a novel therapeutic approach for GBM, addressing drug delivery challenges while improving anti-tumor efficacy. Further optimization for enhanced tumor retention and exploration of combination therapies may improve clinical outcomes (Graphical Abstract).
Collapse
Affiliation(s)
- Qingyu Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Yong Li
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Qian Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Ronggui Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Haoran Lu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Xinyi Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Lun Gao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Gang Deng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| |
Collapse
|
9
|
Kasapkara ÇS, Civelek Ürey B, Bilginer Gürbüz B, Küçükçongar Yavaş A, Keçeli AM, Öncül Ü, Gündüz M, Biberoğlu G, Çıtak Kurt AN, Gürkaş E, Kılıç E, Güleç Ceylan G, Özbek NY. Clinical and Radiological Profile of Nine Patients with Metachromatic Leukodystrophy. Mol Syndromol 2025; 16:138-151. [PMID: 40176833 PMCID: PMC11961098 DOI: 10.1159/000540925] [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: 06/01/2024] [Accepted: 08/13/2024] [Indexed: 04/05/2025] Open
Abstract
Introduction Metachromatic leukodystrophy (MLD) is a rare, demyelinating, autosomal recessive lysosomal storage disease caused by a deficiency in the arylsulfatase A enzyme (ASA), which is encoded by ARSA gene. A lack of ASA activity results in an accumulation of sulfatides in the myelin sheaths of both the central and peripheral nervous systems, leading to developmental and neurocognitive progressive deterioration that can be observed in all age groups. Methods We present a total of 9 patients with MLD with an average age of 61 months, whose clinical, laboratory and cranial magnetic resonance imaging findings were evaluated, and who underwent an ARSA gene molecular analysis. Results Of the 9 patients, 7 had the late infantile form of the condition, 2 had the juvenile form, and 3 were identified through family screening. The median age at diagnosis was 30 months (min 3-max 73 months), the mean ASA activity value was 2 nmol/h/mgprt and the median cranial MR imaging severity score was 10 (min 5-max 18). The grey and white matter volumes of all patients, evaluated using volBrain software, were within the normal range. At an average age of 48 months, the late-infantile MLD patients were unable to control any body part. Conclusions Hematopoietic stem cell transplantation (HSCT), a treatment option for both the juvenile and adult forms of MLD in asymptomatic or early symptomatic patients, was performed on two of the asymptomatic and early symptomatic patients, and post-HSCT ASA activity settled within the normal range and their developmental milestones stabilized. It is important to diagnose MLD in the asymptomatic period and newborn screening can support early diagnosis.
Collapse
Affiliation(s)
- Çiğdem Seher Kasapkara
- Department of Pediatric Metabolism, Ankara Bilkent City Hospital, Ankara, Turkey
- Department of Pediatric Metabolic Diseases, Department of Pediatrics, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Burcu Civelek Ürey
- Department of Pediatric Metabolism, Ankara Bilkent City Hospital, Ankara, Turkey
| | | | | | - Avni Merter Keçeli
- Department of Pediatric Radiology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Ümmühan Öncül
- Department of Pediatric Metabolism, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Mehmet Gündüz
- Department of Pediatric Metabolism, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Gürsel Biberoğlu
- Department of Pediatric Metabolism, Gazi University Hospital, Ankara, Turkey
| | | | - Esra Gürkaş
- Department of Pediatric Neurology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Esra Kılıç
- Department of Pediatric Genetics, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Gülay Güleç Ceylan
- Department of Medical Genetics, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Namık Yaşar Özbek
- Department of Pediatric Hematology, Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
10
|
Gao Y, Zhang M, Wang G, Lai W, Liao S, Chen Y, Ning Q, Tang S. Metabolic cross-talk between glioblastoma and glioblastoma-associated microglia/macrophages: From basic insights to therapeutic strategies. Crit Rev Oncol Hematol 2025; 208:104649. [PMID: 39922398 DOI: 10.1016/j.critrevonc.2025.104649] [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: 01/06/2025] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Glioblastoma (GBM), a highly malignant "cold" tumor of the central nervous system, is characterized by its ability to remodel the GBM immune microenvironment (GME), leading to significant resistance to immunotherapy. GBM-associated microglia/macrophages (GAMs) are essential components of the GME. Targeting GAMs has emerged as a promising strategy against GBM. However, their highly immunosuppressive nature contributes to GBM progression and drug resistance, significantly impeding anti-GBM immunotherapy. Accumulating evidence suggests that metabolic reprogramming accompanies GBM progression and GAM polarization, which are in turn driven by specific metabolic abnormalities and altered cellular signaling pathways. Importantly, metabolic crosstalk between GBM and GAMs further promotes tumor progression. Clarifying and disrupting this metabolic crosstalk is expected to enhance the antitumor phenotype of GAMs and inhibit GBM malignant progression. This review explores metabolism-based interregulation between GBM and GAMs and summarizes recent therapeutic strategies targeting this crosstalk, offering new insights into GBM immunotherapy.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Mengxia Zhang
- Department of Histology and Embryology, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Guihua Wang
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Weiwei Lai
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Shuxian Liao
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Yao Chen
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| | - Shengsong Tang
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
11
|
Yao TT, Chen L, Du Y, Jiang ZY, Cheng Y. MicroRNAs as Regulators, Biomarkers, and Therapeutic Targets in Autism Spectrum Disorder. Mol Neurobiol 2025; 62:5039-5056. [PMID: 39503812 DOI: 10.1007/s12035-024-04582-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/22/2024] [Indexed: 03/05/2025]
Abstract
The pathogenesis of autism spectrum disorder (ASD) is complex and is mainly influenced by genetic and environmental factors. Some research has indicated that environmental aspects may interplay with genetic aspects to enhance the risk, and microRNAs (miRNAs) are probably factors in explaining this link between heredity and the environment. MiRNAs are single-stranded noncoding RNAs that can regulate gene expression at the posttranscriptional level. Some research has indicated that miRNAs are closely linked to neurological diseases. Many aberrantly expressed miRNAs have been observed in autism, and these dysregulated miRNAs are expected to be potential biomarkers and provide new strategies for the treatment of this disease. This article reviews the research progress of miRNAs in autism, including their biosynthesis and function. It is found that some miRNAs show aberrant expression patterns in brain tissue and peripheral blood of autistic patients, which may serve as biomarkers of the disease. In addition, the article explores the novel role of exosomes as carriers of miRNAs with the ability to cross the blood-brain barrier and unique expression profiles, offering new possibilities for diagnostic and therapeutic interventions in ASD. The potential of miRNAs in exosomes as diagnostic markers for ASD is specifically highlighted, as well as the prospect of using engineered exosome-encapsulated miRNAs for targeted therapies.
Collapse
Affiliation(s)
- Tong-Tong Yao
- Center On Translational Neuroscience, Institute of National Security, Minzu University of China, 27th South Zhongguancun Avenue, Beijing, 100081, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Zhong-Yong Jiang
- Department of Medical Laboratory, Affiliated Cancer Hospital of Chengdu Medical College, Chengdu Seventh People's Hospital, Chengdu, China.
| | - Yong Cheng
- Center On Translational Neuroscience, Institute of National Security, Minzu University of China, 27th South Zhongguancun Avenue, Beijing, 100081, China.
- Center On Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
| |
Collapse
|
12
|
Luo Q, Yang J, Yang M, Wang Y, Liu Y, Liu J, Kalvakolanu DV, Cong X, Zhang J, Zhang L, Guo B, Duo Y. Utilization of nanotechnology to surmount the blood-brain barrier in disorders of the central nervous system. Mater Today Bio 2025; 31:101457. [PMID: 39896289 PMCID: PMC11786670 DOI: 10.1016/j.mtbio.2025.101457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/27/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Central nervous system (CNS) diseases are a major cause of disability and death worldwide. Due to the blood-brain barrier (BBB), drug delivery for CNS diseases is extremely challenging. Nano-delivery systems can overcome the limitations of BBB to deliver drugs to the CNS, improve the ability of drugs to target the brain and provide potential therapeutic methods for CNS diseases. At the same time, the choice of different drug delivery methods (bypassing BBB or crossing BBB) can further optimize the therapeutic effect of the nano-drug delivery system. This article reviews the different methods of nano-delivery systems to overcome the way BBB enters the brain. Different kinds of nanoparticles to overcome BBB were discussed in depth.
Collapse
Affiliation(s)
- Qian Luo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Jiaying Yang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Mei Yang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yingtong Wang
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yiran Liu
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Jixuan Liu
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Dhan V. Kalvakolanu
- Greenebaum NCI Comprehensive Cancer Center, Department of Microbiology and Immunology University of Maryland School Medicine, Baltimore, MD, USA
| | - Xianling Cong
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Jinnan Zhang
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Yanhong Duo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
13
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [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: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
14
|
Zhang X, Artz N, Steindler DA, Hingtgen S, Satterlee AB. Exosomes: Traversing the blood-brain barrier and their therapeutic potential in brain cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189300. [PMID: 40097050 DOI: 10.1016/j.bbcan.2025.189300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
The blood-brain barrier (BBB) presents a major challenge for the effective delivery of therapeutic agents to the brain tumor cells from the peripheral blood circulation, making the treatment of central nervous system (CNS)-related cancers more difficult and resistant to both standard treatments and emerging therapies. Exosomes, which serve as messengers for intercellular communication throughout the body, can naturally or be modified to penetrate the BBB. Recently, exosomes have been increasingly explored as an invasive or non-invasive approach for delivering therapeutic agents to the CNS. With their low immunogenicity, ease of modification, excellent cargo protection, and inherent ability to cross the BBB, exosomes hold great promise for revolutionizing targeted therapy for CNS-related diseases, including brain cancer. In this review, we highlight recent discoveries and insights into the mechanisms exosomes use to penetrate the BBB, the methods they employ to payload diverse therapeutics, and their roles in transporting therapeutic compounds for brain cancer and other neurological disorders.
Collapse
Affiliation(s)
- Xiaopei Zhang
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Nichole Artz
- Department of Pediatric Hematology/Oncology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Dennis A Steindler
- Steindler Consulting, Boston, MA, USA; Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Shawn Hingtgen
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew Benson Satterlee
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
15
|
Fu Y, Zhang J, Qin R, Ren Y, Zhou T, Han B, Liu B. Activating autophagy to eliminate toxic protein aggregates with small molecules in neurodegenerative diseases. Pharmacol Rev 2025; 77:100053. [PMID: 40187044 DOI: 10.1016/j.pharmr.2025.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 12/05/2024] [Indexed: 04/07/2025] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer disease, Parkinson disease, Huntington disease, amyotrophic lateral sclerosis, and frontotemporal dementia, are well known to pose formidable challenges for their treatment due to their intricate pathogenesis and substantial variability among patients, including differences in environmental exposures and genetic predispositions. One of the defining characteristics of NDs is widely reported to be the buildup of misfolded proteins. For example, Alzheimer disease is marked by amyloid beta and hyperphosphorylated Tau aggregates, whereas Parkinson disease exhibits α-synuclein aggregates. Amyotrophic lateral sclerosis and frontotemporal dementia exhibit TAR DNA-binding protein 43, superoxide dismutase 1, and fused-in sarcoma protein aggregates, and Huntington disease involves mutant huntingtin and polyglutamine aggregates. These misfolded proteins are the key biomarkers of NDs and also serve as potential therapeutic targets, as they can be addressed through autophagy, a process that removes excess cellular inclusions to maintain homeostasis. Various forms of autophagy, including macroautophagy, chaperone-mediated autophagy, and microautophagy, hold a promise in eliminating toxic proteins implicated in NDs. In this review, we focus on elucidating the regulatory connections between autophagy and toxic proteins in NDs, summarizing the cause of the aggregates, exploring their impact on autophagy mechanisms, and discussing how autophagy can regulate toxic protein aggregation. Moreover, we underscore the activation of autophagy as a potential therapeutic strategy across different NDs and small molecules capable of activating autophagy pathways, such as rapamycin targeting the mTOR pathway to clear α-synuclein and Sertraline targeting the AMPK/mTOR/RPS6KB1 pathway to clear Tau, to further illustrate their potential in NDs' therapeutic intervention. Together, these findings would provide new insights into current research trends and propose small-molecule drugs targeting autophagy as promising potential strategies for the future ND therapies. SIGNIFICANCE STATEMENT: This review provides an in-depth overview of the potential of activating autophagy to eliminate toxic protein aggregates in the treatment of neurodegenerative diseases. It also elucidates the fascinating interrelationships between toxic proteins and the process of autophagy of "chasing and escaping" phenomenon. Moreover, the review further discusses the progress utilizing small molecules to activate autophagy to improve the efficacy of therapies for neurodegenerative diseases by removing toxic protein aggregates.
Collapse
Affiliation(s)
- Yuqi Fu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueting Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Brain Science, Faculty of Medicine, Imperial College, London, UK
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Mun JH, Jang MJ, Kim WS, Kim SS, Lee B, Moon H, Oh SJ, Ryu CH, Park KS, Cho IH, Hong GS, Choi CW, Lee C, Kim MS. Enhanced Cognitive and Memory Functions via Gold Nanoparticle-Mediated Delivery of Afzelin through Synaptic Modulation Pathways in Alzheimer's Disease Mouse Models. ACS Chem Neurosci 2025; 16:826-843. [PMID: 39976589 DOI: 10.1021/acschemneuro.4c00766] [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: 03/06/2025] Open
Abstract
Gold nanoparticles (AuNPs) are valuable tools in pharmacological and biological research, offering unique properties for drug delivery in the treatment of neurodegenerative diseases. This study investigates the potential of gold nanoparticles loaded with afzelin, a natural chemical extracted from Ribes fasciculatum, to enhance its therapeutic effects and overcome the limitations of using natural compounds regarding low productivity. We hypothesized that the combined treatment of AuNPs with afzelin (AuNP-afzelin) would remarkably enforce neuroprotective effects compared with the single treatment of afzelin. Central administration of AuNP-afzelin (10 ng of afzelin) indicated improvements in cognition and memory-involved assessments of behavioral tests, comparing single treatments of afzelin (10 or 100 ng of afzelin) in scopolamine-induced AD mice. AuNP-afzelin also performed superior neuroprotective effects of rescuing mature neuronal cells and recovered cholinergic dysfunction compared to afzelin alone, according to further investigations of BDNF-pCREB-pAkt signaling, long-term potentiation, and doublecortin (DCX) expression in the hippocampus. This study highlights the potential of afzelin with gold nanoparticles as a promising therapeutic approach for mitigating cognitive impairments associated with neurodegenerative diseases and offers a new avenue for future research and drug development.
Collapse
Affiliation(s)
- Ju Hee Mun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- School of Electrical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Min Jun Jang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Won Seok Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Seong-Seop Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Bonggi Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - HyunSeon Moon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Cheol-Hui Ryu
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 136-791, Republic of Korea
| | - Kyung Su Park
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 136-791, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Gyu-Sang Hong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Chun Whan Choi
- Natural Product Research Team, Gyeonggi Biocenter, Gyeonggi-do Business and Science Accelerator, Suwon 16229, Republic of Korea
| | - Changhyuk Lee
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KIST-SKKU Brain Research Center, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Min Soo Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
17
|
Ngcobo NN. Influence of Ageing on the Pharmacodynamics and Pharmacokinetics of Chronically Administered Medicines in Geriatric Patients: A Review. Clin Pharmacokinet 2025; 64:335-367. [PMID: 39798015 PMCID: PMC11954733 DOI: 10.1007/s40262-024-01466-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2024] [Indexed: 01/13/2025]
Abstract
As people age, the efficiency of various regulatory processes that ensure proper communication between cells and organs tends to decline. This deterioration can lead to difficulties in maintaining homeostasis during physiological stress. This includes but is not limited to cognitive impairments, functional difficulties, and issues related to caregivers which contribute significantly to medication errors and non-adherence. These factors can lead to higher morbidity, extended hospital stays, reduced quality of life, and even mortality. The decrease in homeostatic capacity varies among individuals, contributing to the greater variability observed in geriatric populations. Significant pharmacokinetic and pharmacodynamic alterations accompany ageing. Pharmacokinetic changes include decreased renal and hepatic clearance and an increased volume of distribution for lipid-soluble drugs, which prolong their elimination half-life. Pharmacodynamic changes typically involve increased sensitivity to various drug classes, such as anticoagulants, antidiabetic and psychotropic medications. This review examines the primary age-related physiological changes in geriatrics and their impact on the pharmacokinetics and pharmacodynamics of medications.
Collapse
Affiliation(s)
- Nokwanda N Ngcobo
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
18
|
Choi HJ, Han M, Jung B, Huh H, Lee EH, Choi JR, Park J. Evaluation of blood-tumor barrier permeability and doxorubicin delivery in rat brain tumor models using additional focused ultrasound stimulation. Sci Rep 2025; 15:6592. [PMID: 39994241 PMCID: PMC11850853 DOI: 10.1038/s41598-025-88379-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Focused ultrasound (FUS) has emerged as a promising technique for temporarily disrupting the blood-brain barrier (BBB) and blood-tumor barrier (BTB) to enhance the delivery of therapeutic agents. Despite its potential, optimizing FUS to maximize drug delivery while minimizing adverse effects remains a significant challenge. In this study, we evaluated a novel FUS protocol that incorporates additional FUS stimulation without microbubbles (MBs) ("FUS protocol") prior to conventional BBB disruption with MBs ("BBBD protocol") in a rat brain tumor model (n = 35). This approach aimed to validate its effectiveness in enhancing BBB/BTB disruption and facilitating doxorubicin delivery. T1-weighted contrast-enhanced and dynamic contrast-enhanced (DCE) MRI demonstrated significant increases in signal intensity and permeability (Ktrans) in the tumor region under the "FUS + BBBD protocol", with 2.65-fold and 2.08-fold increases, respectively, compared to the non-sonicated contralateral region. These values were also elevated compared to the conventional "BBBD protocol" by 1.45-fold and 1.25-fold, respectively. Furthermore, doxorubicin delivery in the targeted region increased by 1.91-fold under the "FUS + BBBD protocol", compared to a 1.44-fold increase using the conventional "BBBD protocol". This novel FUS approach offers a promising, cost-effective strategy for enhancing drug delivery to brain tumors. While further studies are required to assess its applicability with different chemotherapeutics and tumor types, it holds significant potential for improving brain tumor treatment in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Hyo Jin Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Mun Han
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Byeongjin Jung
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
- BioHealth Convergence Center, Daegu Technopark, 46-17 Seongseogongdan-ro, Dalseo-gu, Daegu, 42716, Republic of Korea
| | - Hyungkyu Huh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Eun-Hee Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Jong-Ryul Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Juyoung Park
- College of IT Convergence, Department of Biomedical Device, Gachon University, 1342, Seongnam-daero, Sujeong-gu, 1342, Seongnam, 13120, Gyeonggi, Republic of Korea.
- Neumous Inc., 1526, Seongnam-daero, Sujeong-gu, Seongnam-si, 13113, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
19
|
Ren R, Zhang G, Ma J, Zheng Y, Zhao Y, Zhang Y, Zhao L. Nebulized seabuckthorn seed oil inhalation attenuates Alzheimer's disease progression in APP/PS1 mice. Sci Rep 2025; 15:6368. [PMID: 39984555 PMCID: PMC11845625 DOI: 10.1038/s41598-025-89747-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] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
Seabuckthorn (Hippophae rhamnoides L.) is known for its medicinal properties in treating various diseases, including neurological conditions. However, the therapeutic effect of inhaled seabuckthorn seed oil (SSO) on Alzheimer's disease (AD) remains not fully understood. This study explores the effects of nebulized inhalation of SSO in 9-month-old APP/PS1 mice over 21 days. The results showed that nebulized SSO improved memory and cognition. Using 7.0T MRI to monitor blood oxygenation level dependent (BOLD) signals revealed that SSO altered the Amplitude of Low Frequency Fluctuations (ALFF) and Regional Homogeneity (ReHo) signaling such as in the amygdala and substantia innominate, and hippocampus. Enzyme-linked immuno sorbent assay (ELISA) and pathological analyses indicated reduced neuroinflammation in plasma and brain, decreased neuronal necrosis, lower β-amyloid (Aβ) protein levels, reduced amyloid deposition, and increased tyrosine hydroxylase activity. Additionally, SSO promoted gut microbiota remodeling by increasing alpha diversity and boosting levels of probiotics such as Verrucomicrobia, Bifidobacterium, Prevotella, and Akkermansia, without adverse effects on lung tissue. Nebulized inhalation of SSO may slow AD progression by modulating inflammation and amyloid deposition. Nebulized inhalation offered a potential method for enhancing drug delivery across the blood-brain barrier with reduced systemic side effects.
Collapse
Affiliation(s)
- Ruichen Ren
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Gaorui Zhang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Junqing Ma
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yongze Zheng
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yuxuan Zhao
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yang Zhang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Lin Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China.
| |
Collapse
|
20
|
Mochnáčová E, Bhide K, Kucková K, Jozefiaková J, Maľarik T, Bhide M. Antimicrobial cyclic peptides effectively inhibit multiple forms of Borrelia and cross the blood-brain barrier model. Sci Rep 2025; 15:6147. [PMID: 39979461 PMCID: PMC11842550 DOI: 10.1038/s41598-025-90605-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/14/2025] [Indexed: 02/22/2025] Open
Abstract
Infection caused by neuroinvasive Borrelia often manifests long-term CNS disorders and is difficult to treat as most antibiotics fail to attain an effective concentration within the brain or cannot kill the persister forms of Borrelia (cysts and round bodies). Thus, this study focused on developing antimicrobial cyclic peptides (AMPs) from a combinatorial phage display library that target phosphatidylcholine of the borrelial cell membrane. Isolated cyclic peptides with anti-Borrelia properties were then fused with the CNS homing peptide developed in this study (designated as O-BBB) to facilitate AMP transport across the blood-brain barrier. Among all O-BBB fused AMPs, Bor-18 had half maximal effective concentration (EC50) 0.83 µM when tested against spirochetal Borrelia. Bor-16, Bor-18, and Bor-26 inhibited the cystic form with EC50 0.83 µM, while Bor-11 had EC50 0.41 µM. Within an hour, all four peptides caused a permeability breach in the borrelial cell membrane, causing depolarization of the membrane. Bor peptides did not inhibit eukaryotic cell metabolism or proliferation, nor did they cause erythrocyte lysis. Peptides were stable in serum, could cross the BBB in-vitro, and remained effective against Borrelia. Cyclic AMPs fused with a CNS homing moiety, the Bor peptides, deserve further investigation for their potential use in neuroborreliosis therapy.
Collapse
Affiliation(s)
- Evelína Mochnáčová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Katarína Kucková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Jana Jozefiaková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Tomáš Maľarik
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia.
- Institute of Neuroimmunology of Slovak Academy of Sciences, 84510, Bratislava, Slovakia.
| |
Collapse
|
21
|
Tandon R, Kumar S, Handa M, Srivastava N. Exosomes in glioma: mechanistic insights on biological, therapeutic, and diagnostic perspective. Ther Deliv 2025:1-12. [PMID: 39957239 DOI: 10.1080/20415990.2025.2466410] [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: 07/27/2024] [Accepted: 02/10/2025] [Indexed: 02/18/2025] Open
Abstract
Gliomas are prominent and frequent primary malignant brain tumors, with a generally poor prognosis. Current treatment involves radiation, surgery and chemotherapy. Exosomes are nanoscale extracellular vesicles released by cells that enable biological molecule movement and encourage intercellular communication in the tumor microenvironment. This contributes to glioma development, radiation resistance, and overcomes chemotherapy. Exosome functional and structural properties are essential for understanding cancer molecular mechanisms. They can also treat invasive tumors like glioblastomas and serve as diagnostic markers. Recent research depicted exosomes' prominent role in cancer cell maintenance, intercellular signaling, and microenvironment modification. Exosomes hold nucleic acids, proteins, lipids, mRNAs, lncRNAs, miRNAs, and immunological regulatory molecules depending on the origin of the cell. This paper reviews exosomes, their role in glioma etiology, and perspective diagnostic and therapeutic uses.
Collapse
Affiliation(s)
- Reetika Tandon
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| | - Samarth Kumar
- Formulation Research & Development-Non Orals, Sun Pharmaceuticals Industries Limited, Vadodara, India
| | - Mayank Handa
- Formulation Research & Development-Non Orals, Sun Pharmaceuticals Industries Limited, Vadodara, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| |
Collapse
|
22
|
Alaei M, Koushki K, Taebi K, Yousefi Taba M, Keshavarz Hedayati S, Keshavarz Shahbaz S. Metal nanoparticles in neuroinflammation: impact on microglial dynamics and CNS function. RSC Adv 2025; 15:5426-5451. [PMID: 39967886 PMCID: PMC11833603 DOI: 10.1039/d4ra07798a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Microglia, the primary immune cells of the central nervous system (CNS), are crucial in maintaining brain homeostasis and responding to pathological changes. While they play protective roles, their activation can lead to neuroinflammation and the progression of neurodegenerative diseases. Metal nanoparticles (NPs), due to their unique ability to cross the blood-brain barrier (BBB), have emerged as promising agents for drug delivery to the CNS. In this way, we aim to review the dual role of metal-containing NPs, gold (AuNPs), silver (AgNPs), iron oxide (IONPs), zinc oxide (ZnONPs), cobalt (CoNPs), titanium dioxide (TiO2NPs), and silica (SiO2NPs) in modulating microglial activity. Some NPs promote anti-inflammatory effects, while others exacerbate neuroinflammation. We examine how these NPs influence microglial activation, focusing on their potential therapeutic benefits and risks. A deeper understanding of NP-microglia interactions is crucial for developing safe and efficient treatments for neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Masood Alaei
- Student Research Committee, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Khadijeh Koushki
- Department of Neurosurgery, University of Texas Houston Health Science Center (UTHealth) Houston TX USA
| | - Kimia Taebi
- Student Research Committee, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Mahdieh Yousefi Taba
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | | | - Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences Qazvin 34197-59811 Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| |
Collapse
|
23
|
Jorgensen C, Linville RM, Galea I, Lambden E, Vögele M, Chen C, Troendle EP, Ruggiu F, Ulmschneider MB, Schiøtt B, Lorenz CD. Permeability Benchmarking: Guidelines for Comparing in Silico, in Vitro, and in Vivo Measurements. J Chem Inf Model 2025; 65:1067-1084. [PMID: 39823383 DOI: 10.1021/acs.jcim.4c01815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Permeability is a measure of the degree to which cells can transport molecules across biological barriers. Units of permeability are distance per unit time (typically cm/s), where accurate measurements are needed to define drug delivery in homeostasis and to model dysfunction occurring during disease. This perspective offers a set of community-led guidelines to benchmark permeability data across multidisciplinary approaches and different biological contexts. First, we lay out the analytical framework for three methodologies to calculate permeability: in silico assays using either transition-based counting or the inhomogeneous-solubility diffusion approaches, in vitro permeability assays using cells cultured in 2D or 3D geometries, and in vivo assays utilizing in situ brain perfusion or multiple time-point regression analysis. Then, we demonstrate a systematic benchmarking of in silico to both in vitro and in vivo, depicting the ways in which each benchmarking is sensitive to the choices of assay design. Finally, we outline seven recommendations for best practices in permeability benchmarking and underscore the significance of tailored permeability assays in driving advancements in drug delivery research and development. Our exploration encompasses a discussion of "generic" and tissue-specific biological barriers, including the blood-brain barrier (BBB), which is a major hurdle for the delivery of therapeutic agents into the brain. By addressing challenges in reconciling simulated data with experimental assays, we aim to provide insights essential for optimizing accuracy and reliability in permeability modeling.
Collapse
Affiliation(s)
- Christian Jorgensen
- School of Medicine, Pharmacy and Biomedical Sciences, Faculty of Science & Health, University of Portsmouth, Portsmouth PO1 2DT, Hampshire, U.K
- Dept. of Chemistry, Aarhus University, Langelandsgade, 140 8000 Aarhus C, Denmark
| | - Raleigh M Linville
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, U.K
| | - Edward Lambden
- Dept. of Chemistry, King's College London, London WC2R 2LS, U.K
| | - Martin Vögele
- Department of Computer Science, Stanford University, Stanford, California 94305, United States
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, United States
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, California 94305, United States
| | - Charles Chen
- Synthetic Biology Group, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Evan P Troendle
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, County Antrim, BT9 7BL, Northern Ireland, U.K
| | - Fiorella Ruggiu
- Kimia Therapeutics, 740 Heinz Avenue, Berkeley, California 94710, United States
| | | | - Birgit Schiøtt
- Dept. of Chemistry, Aarhus University, Langelandsgade, 140 8000 Aarhus C, Denmark
| | | |
Collapse
|
24
|
Tripathi M, Gharti L, Bansal A, Kaurav H, Sheth S. Intranasal Mucoadhesive In Situ Gel of Glibenclamide-Loaded Bilosomes for Enhanced Therapeutic Drug Delivery to the Brain. Pharmaceutics 2025; 17:193. [PMID: 40006560 PMCID: PMC11859129 DOI: 10.3390/pharmaceutics17020193] [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: 12/06/2024] [Revised: 01/18/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND The neuroprotective efficacy of glibenclamide (GLIB) has been demonstrated in multiple rodent models of ischemia, hemorrhagic stroke, traumatic brain damage, spinal cord injury, and metastatic brain tumors. Due to its poor solubility, GLIB has low oral bioavailability, limiting its transportation to the brain via the oral route. OBJECTIVES Here, we attempted to develop and optimize an intranasal mucoadhesive in situ gel of GLIB-loaded bilosomes using a 32 Box-Behnken design for brain drug delivery. METHODS To facilitate a longer residence time of the administered dose within the nasal cavity, the prepared bilosomes were loaded into a mucoadhesive in situ gel providing resistance to rapid mucociliary clearance. The amounts of sodium deoxycholate, the cholesterol/Span 40 mixture, and the molar ratio between the mixture's components were chosen as independent variables, while the entrapment efficiency and in vitro drug release were selected as dependent variables. RESULTS AND CONCLUSIONS The optimal formulation was analyzed for particle size and entrapment efficiency, which were found to be 270.6 nm and 68.39%, respectively. In vitro drug release from optimal formulation after 12 h was 87.29 ± 1.98% as compared to 52.01 ± 2.04% of plain in situ gel of drug. An in vivo brain drug delivery study performed on Swiss albino mice showed that the brain concentration of drug through intranasal administration from mucoadhesive in situ gel of GLIB-bilosomes after 12 h was 2.12 ± 0.16 µg/mL as compared to 0.68 ± 0.04 µg/mL from plain in situ gel of drug. Conclusively, the developed bilosomal formulation offers a favorable intranasal substitute with enhanced therapeutic drug delivery to the brain.
Collapse
Affiliation(s)
- Meenakshi Tripathi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India; (M.T.); (L.G.)
| | - Laxmi Gharti
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India; (M.T.); (L.G.)
| | - Amit Bansal
- Formulation Research and Development, Perrigo Company plc, Allegan, MI 49010, USA;
| | - Hemlata Kaurav
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India; (M.T.); (L.G.)
| | - Sandeep Sheth
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA
| |
Collapse
|
25
|
Feng K, Zhang JA, Shen WT, Leng T, Zhou Z, Yu Y, Gao W, Zhang L. Recent Development of Nanoparticle Platforms for Organophosphate Nerve Agent Detoxification. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:2124-2140. [PMID: 39862147 PMCID: PMC11803738 DOI: 10.1021/acs.langmuir.4c04637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Poisoning by organophosphate (OP) nerve agents remains a pressing global threat due to their extensive use in chemical warfare agents and pesticides, potentially causing high morbidity and mortality worldwide. This urgent need for effective countermeasures has driven considerable interest in innovative detoxification approaches. Among these, nanoparticle technology stands out for its multifunctional potential and wide-ranging applications. This review highlights recent advancements in nanoparticle platforms developed for OP detoxification, focusing on five main types: inorganic nanoparticles, lipid-based nanoparticles, polymer-based nanoparticles, metal-organic framework nanoparticles, and cellular nanoparticles. For each platform, we discuss representative examples that illustrate how structural and functional properties enhance their effectiveness as nanocarriers, nanocatalysts, or nanoscavengers, ultimately enabling safe and efficient OP detoxification. This review aims to stimulate further technological innovation in OP-detoxifying nanoparticles and encourage broader development of detoxification strategies.
Collapse
Affiliation(s)
- Kailin Feng
- Aiiso Yufeng Li Family Department
of Chemical and Nano Engineering, Shu and K. C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Jiayuan Alex Zhang
- Aiiso Yufeng Li Family Department
of Chemical and Nano Engineering, Shu and K. C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Wei-Ting Shen
- Aiiso Yufeng Li Family Department
of Chemical and Nano Engineering, Shu and K. C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Tianle Leng
- Aiiso Yufeng Li Family Department
of Chemical and Nano Engineering, Shu and K. C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Zhidong Zhou
- Aiiso Yufeng Li Family Department
of Chemical and Nano Engineering, Shu and K. C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Yiyan Yu
- Aiiso Yufeng Li Family Department
of Chemical and Nano Engineering, Shu and K. C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Weiwei Gao
- Aiiso Yufeng Li Family Department
of Chemical and Nano Engineering, Shu and K. C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Liangfang Zhang
- Aiiso Yufeng Li Family Department
of Chemical and Nano Engineering, Shu and K. C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| |
Collapse
|
26
|
Liu S, Li H, Xi S, Zhang Y, Sun T. Advancing CNS Therapeutics: Enhancing Neurological Disorders with Nanoparticle-Based Gene and Enzyme Replacement Therapies. Int J Nanomedicine 2025; 20:1443-1490. [PMID: 39925682 PMCID: PMC11806685 DOI: 10.2147/ijn.s457393] [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: 12/31/2023] [Accepted: 12/12/2024] [Indexed: 02/11/2025] Open
Abstract
Given the complexity of the central nervous system (CNS) and the diversity of neurological conditions, the increasing prevalence of neurological disorders poses a significant challenge to modern medicine. These disorders, ranging from neurodegenerative diseases to psychiatric conditions, not only impact individuals but also place a substantial burden on healthcare systems and society. A major obstacle in treating these conditions is the blood-brain barrier (BBB), which restricts the passage of therapeutic agents to the brain. Nanotechnology, particularly the use of nanoparticles (NPs), offers a promising solution to this challenge. NPs possess unique properties such as small size, large surface area, and modifiable surface characteristics, enabling them to cross the BBB and deliver drugs directly to the affected brain regions. This review focuses on the application of NPs in gene therapy and enzyme replacement therapy (ERT) for neurological disorders. Gene therapy involves altering or manipulating gene expression and can be enhanced by NPs designed to carry various genetic materials. Similarly, NPs can improve the efficacy of ERT for lysosomal storage disorders (LSDs) by facilitating enzyme delivery to the brain, overcoming issues like immunogenicity and instability. Taken together, this review explores the potential of NPs in revolutionizing treatment options for neurological disorders, highlighting their advantages and the future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Haisong Li
- Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Shiwen Xi
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- International Center of Future Science, Jilin University, Changchun, People’s Republic of China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
27
|
Gao S, Li X, Han B. Bacterial and bacterial derivatives-based drug delivery systems: a novel approach for treating central nervous system disorders. Expert Opin Drug Deliv 2025; 22:163-180. [PMID: 39688950 DOI: 10.1080/17425247.2024.2444364] [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: 12/06/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/19/2024]
Abstract
INTRODUCTION Bacteria and their derivatives show great potential as drug delivery systems due to their unique chemotaxis, biocompatibility, and targeting abilities. In CNS disease treatment, bacterial carriers can cross the blood-brain barrier (BBB) and deliver drugs precisely, overcoming limitations of traditional methods. Advances in genetic engineering, synthetic biology, and nanotechnology have transformed these systems into multifunctional platforms for personalized CNS treatment. AREAS COVERED This review examines the latest research on bacterial carriers for treating ischemic brain injury, neurodegenerative diseases, and gliomas. Bacteria efficiently cross the blood-brain barrier via active targeting, endocytosis, paracellular transport, and the nose-to-brain route for precise drug delivery. Various bacterial drug delivery systems, such as OMVs and bacterial ghosts, are explored for their design and application. Databases were searched in Google Scholar for the period up to December 2024. EXPERT OPINION Future developments in bacterial drug delivery will rely on AI-driven design and high-throughput engineering, enhancing treatment precision. Personalized medicine will further optimize bacterial carriers for individual patients, but challenges such as biosafety, immune rejection, and scalability must be addressed. As multimodal diagnostic and therapeutic strategies advance, bacterial carriers are expected to play a central role in CNS disease treatment, offering novel precision medicine solutions.
Collapse
Affiliation(s)
- Shizhu Gao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Xin Li
- Orthopedic Medical Center, 2nd hospital of Jilin University, Changchun, PR China
| | - Bing Han
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| |
Collapse
|
28
|
Domínguez-López I, López-Yerena A, Vallverdú-Queralt A, Pallàs M, Lamuela-Raventós RM, Pérez M. From the gut to the brain: the long journey of phenolic compounds with neurocognitive effects. Nutr Rev 2025; 83:e533-e546. [PMID: 38687609 PMCID: PMC11723161 DOI: 10.1093/nutrit/nuae034] [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: 05/02/2024] Open
Abstract
The human gut microbiota is a complex community of micro-organisms that play a crucial role in maintaining overall health. Recent research has shown that gut microbes also have a profound impact on brain function and cognition, leading to the concept of the gut-brain axis. One way in which the gut microbiota can influence the brain is through the bioconversion of polyphenols to other bioactive molecules. Phenolic compounds are a group of natural plant metabolites widely available in the human diet, which have anti-inflammatory and other positive effects on health. Recent studies have also suggested that some gut microbiota-derived phenolic metabolites may have neurocognitive effects, such as improving memory and cognitive function. The specific mechanisms involved are still being studied, but it is believed that phenolic metabolites may modulate neurotransmitter signaling, reduce inflammation, and enhance neural plasticity. Therefore, to exert a protective effect on neurocognition, dietary polyphenols or their metabolites must reach the brain, or act indirectly by producing an increase in bioactive molecules such as neurotransmitters. Once ingested, phenolic compounds are subjected to various processes (eg, metabolization by gut microbiota, absorption, distribution) before they cross the blood-brain barrier, perhaps the most challenging stage of their trajectory. Understanding the role of phenolic compounds in the gut-brain axis has important implications for the development of new therapeutic strategies for neurological and psychiatric disorders. By targeting the gut microbiota and its production of phenolic metabolites, it may be possible to improve brain function and prevent cognitive decline. In this article, the current state of knowledge on the endogenous generation of phenolic metabolites by the gut microbiota and how these compounds can reach the brain and exert neurocognitive effects was reviewed.
Collapse
Affiliation(s)
- Inés Domínguez-López
- Polyphenol Research Group, Department of Nutrition, Food Science, and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Anallely López-Yerena
- Polyphenol Research Group, Department of Nutrition, Food Science, and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, Barcelona, Spain
| | - Anna Vallverdú-Queralt
- Polyphenol Research Group, Department of Nutrition, Food Science, and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Mercè Pallàs
- Pharmacology and Toxicology Section and Institute of Neuroscience, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Rosa M Lamuela-Raventós
- Polyphenol Research Group, Department of Nutrition, Food Science, and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Maria Pérez
- Polyphenol Research Group, Department of Nutrition, Food Science, and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Zhang Q, Wang J, Chen Z, Qin H, Zhang Q, Tian B, Li X. Transcytosis: an effective mechanism to enhance nanoparticle extravasation and infiltration through biological barriers. Biomed Mater 2025; 20:022003. [PMID: 39788078 DOI: 10.1088/1748-605x/ada85e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/09/2025] [Indexed: 01/12/2025]
Abstract
Nanoparticles (NPs)1have been explored as drugs carriers for treating tumors and central nervous system (CNS)2diseases and for oral administration. However, they lack satisfactory clinical efficacy due to poor extravasation and infiltration through biological barriers to target tissues. Most clinical antitumor NPs have been designed based on enhanced permeability and retention effects which are insufficient and heterogeneous in human tumors. The tight junctions33TJs: tight junctionsof the blood-brain barrier44BBB: blood-brain barrierand the small intestinal epithelium severely impede NPs from being transported into the CNS and blood circulation, respectively. By contrast, transcytosis enables NPs to bypass these physiological barriers and enhances their infiltration into target tissues by active transport. Here, we systematically review the mechanisms and putative application of NP transcytosis for targeting tumor and CNS tissues, explore oral NP administration, and propose future research directions in the field of NP transcytosis.
Collapse
Affiliation(s)
- Qianyi Zhang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Jiamian Wang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200120, People's Republic of China
| | - Zhiyang Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Hao Qin
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Qichen Zhang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Bo Tian
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | - Xilei Li
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
30
|
Li Y, Liu R, Zhao Z. Targeting Brain Drug Delivery with Macromolecules Through Receptor-Mediated Transcytosis. Pharmaceutics 2025; 17:109. [PMID: 39861756 PMCID: PMC11769103 DOI: 10.3390/pharmaceutics17010109] [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: 12/13/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Brain diseases pose significant treatment challenges due to the restrictive nature of the blood-brain barrier (BBB). Recent advances in targeting macromolecules offer promising avenues for overcoming these obstacles through receptor-mediated transcytosis (RMT). We summarize the current progress in targeting brain drug delivery with macromolecules for brain diseases. This exploration details the transport mechanisms across the BBB, focusing on RMT and its use of natural ligands for drug delivery. Furthermore, the review examines macromolecular ligands such as antibodies, peptides, and aptamers that leverage RMT for effective BBB traversal. Advancements in macromolecules-based delivery systems for brain diseases are summarized, emphasizing their therapeutic potential and limitations. Finally, emerging RMT strategies, including viral vectors, exosomes, and boron neutron capture therapy, are discussed for their precision in brain-targeted treatments. This comprehensive overview underscores the potential of RMT-based approaches to revolutionize brain disease therapy.
Collapse
Affiliation(s)
- Yuanke Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, China
| | - Ruiying Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Zhen Zhao
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China
| |
Collapse
|
31
|
Tripathy NS, Sahoo L, Paikray S, Dilnawaz F. Emerging nanoplatforms towards microenvironment-responsive glioma therapy. Med Oncol 2025; 42:46. [PMID: 39812745 DOI: 10.1007/s12032-024-02596-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025]
Abstract
Gliomas are aggressive intracranial tumors of the central nervous system with a poor prognosis, high risk of recurrence, and low survival rates. Radiation, surgery, and chemotherapy are traditional cancer therapies. It is very challenging to accurately image and differentiate the malignancy grade of gliomas due to their heterogeneous and infiltrating nature and the obstruction of the blood-brain barrier. Imaging plays a crucial role in gliomas which significantly plays an important role in the accuracy of the diagnosis followed by any subsequent surgery or therapy. Other diagnostic methods (such as biopsies or surgery) are often very invasive. Preoperative imaging and intraoperative image-guided surgery perform the most significant safe resection. In recent years, the rapid growth of nanotechnology has opened up new avenues for glioma diagnosis and treatment. For better therapeutic efficacy, developing microenvironment-responsive nanoplatforms, including novel nanotherapeutic platforms of sonodynamic therapy, photodynamic therapy, and photothermal treatments, are employed for improved patient survival and better clinical control outcome. In this review recent advancement of multifunctional nanoplatforms leading toward treatment of glioma is discussed.
Collapse
Affiliation(s)
- Nigam Sekhar Tripathy
- School of Biotechnology, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Liza Sahoo
- School of Biotechnology, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Safal Paikray
- School of Biotechnology, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Fahima Dilnawaz
- School of Biotechnology, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India.
| |
Collapse
|
32
|
Sharma G, Wadhwa K, Kumar S, Singh G, Pahwa R. Revolutionizing Parkinson's treatment: Harnessing the potential of intranasal nanoemulsions for targeted therapy. Drug Deliv Transl Res 2025:10.1007/s13346-024-01770-z. [PMID: 39777646 DOI: 10.1007/s13346-024-01770-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2024] [Indexed: 01/11/2025]
Abstract
Parkinson's disease (PD) is the most prominent and highly prevalent chronic neuro-degenerative disease generally recognized by classical motor symptoms which are linked with genetic mutation, Lewy bodies, and subsequently selective loss of nigrostriatal dopaminergic neurons. The blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier protect the central nervous system against toxins and are the most significant barriers to effective brain drug delivery in managing Parkinsonism. In recent years, intranasal delivery has attracted remarkable attention for brain targeting as the drug can be administered to the brain directly from the nose employing the trigeminal and olfactory pathways. For brain targeting through nasal delivery, several advanced and promising formulation techniques have been investigated globally. Nanoemulsions are regarded as an innovative carrier approach for PD, where these provide targeted administration and enhanced bioavailability of neurotherapeutics. This manuscript provides deeper insight into the pathophysiology of PD, various drug delivery strategies to overcome BBB, and the potential role of nanoemulsions via the intranasal route. Various research findings on the intranasal administration of nanoemulsions and their pivotal applications in the treatment of PD have also been embarked. The potential role of phytoconstituents and surface-modified nanoemulsions for the effective treatment of PD has also been reflected along with current challenges and future perspectives in this avenue.
Collapse
Affiliation(s)
- Gulshan Sharma
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, Haryana, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| | - Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology (MIET), NH-58 Delhi-Roorkee Highway, Meerut, 250005, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Rakesh Pahwa
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, Haryana, India.
| |
Collapse
|
33
|
Mohammed PN, Hussen NH, Hasan AH, Salh HJH, Jamalis J, Ahmed S, Bhat AR, Kamal MA. A review on the role of nanoparticles for targeted brain drug delivery: synthesis, characterization, and applications. EXCLI JOURNAL 2025; 24:34-59. [PMID: 39967907 PMCID: PMC11830919 DOI: 10.17179/excli2024-7163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 12/09/2024] [Indexed: 02/20/2025]
Abstract
Unfortunately, nowadays, brain disorders, which include both neurological and mental disorders, are the main cause of years spent living with a disability worldwide. There are serious diseases with a high prevalence and a high mortality rate. However, the outmoded technical infrastructure makes their treatment difficult. The blood-brain barrier (BBB) serves as a protective mechanism for the central nervous system (CNS) and regulates its homeostatic processes. The brain is protected against injury and illness by an extremely complex system that precisely regulates the flow of ions, very few tiny molecules, and an even smaller number of macromolecules from the blood to the brain. Nevertheless, the BBB also considerably inhibits the delivery of medications to the brain, making it impossible to treat a variety of neurological diseases. Several strategies are now being studied to enhance the transport of drugs over the BBB. According to this research, nanoparticles are one of the most promising agents for brain disease treatment while many conventional drugs are also capable of crossing this barrier but there are amazing facts about nanoparticles in brain drug delivery. For example, 1. Precision Targeting: Through mechanisms such as receptor-mediated transport, ligand attachment, or the use of external stimuli (e.g., magnetic or thermal guidance), nanoparticles can deliver drugs specifically to diseased areas of the brain while minimizing exposure to healthy tissues. This targeted approach reduces side effects and enhances therapeutic outcomes. 2. Improved Drug Stability: Drugs can be encapsulated by nanoparticles, which keeps them stable and shields them from deterioration while being transported to the brain. 3. Therapeutic Payload: Nanoparticles possess a high surface-area-to-volume ratio, enabling them to encapsulate a substantial quantity of therapeutic agents relative to their size. This allows for enhanced drug delivery efficiency, maximizing therapeutic outcomes while potentially reducing the required dosage to achieve the desired effect. 4. Imaging Properties: Certain nanoparticles can also act as contrast agents for magnetic resonance imaging (MRI), allowing for the real-time visualization of drug distribution and administration in the brain. 5. Combination Therapy Possibility: Nanoparticles can be designed to co-deliver multiple medications or therapeutic agents, which could enhance synergistic effects. There have been in vivo studies where nanoparticles were successfully used for combination therapies, demonstrating potential for personalized treatments. One notable example is in cancer treatment, where nanoparticles have been designed to co-deliver multiple chemotherapeutic agents. In general, brain medication delivery by nanoparticles is a novel strategy that has the potential to revolutionize neurological disease therapy and enhance patient outcomes. The study furthermore includes a concise depiction of the structural and physiological characteristics of the BBB, and it also provides an overview of the nanoparticles that are most often used in medicine. A brief overview of the structural and physiochemical characteristics of the NPs, as well as the most popular nanoparticles used in medicine, is also included in the review.
Collapse
Affiliation(s)
- Payam Nawzad Mohammed
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Sulaimani 46001, Kurdistan Region-Iraq, Iraq
| | - Narmin Hamaamin Hussen
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Sulaimani 46001, Kurdistan Region-Iraq, Iraq
| | - Aso Hameed Hasan
- Department of Chemistry, College of Science, University of Garmian, Kalar 46021, Kurdistan Region-Iraq, Iraq
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia- 81310 Johor Bahru, Johor, Malaysia
| | - Hozan Jaza Hama Salh
- Department of Clinical Pharmacy, College of Pharmacy, University of Sulaimani, Sulaimani 46001, Kurdistan Region, Iraq
| | - Joazaizulfazli Jamalis
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia- 81310 Johor Bahru, Johor, Malaysia
| | - Sumeer Ahmed
- Post-Graduate and Research Department of Chemistry, The New College (Autonomous), University of Madras, Chennai - 600014, India
| | - Ajmal R. Bhat
- Department of Chemistry, RTM Nagpur University, Nagpur- 440033, India
| | - Mohammad Amjad Kamal
- Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Birulia, Savar, Dhaka -1216, Bangladesh
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
- Novel Global Community Educational Foundation, Australia
| |
Collapse
|
34
|
Nayak U, Halagali P, Panchal KN, Tippavajhala VK, Mudgal J, Radhakrishnan R, Manikkath J. Nanoparticles in CNS Therapeutics: Pioneering Drug Delivery Advancements. Curr Pharm Des 2025; 31:443-460. [PMID: 39318210 DOI: 10.2174/0113816128328722240828184410] [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: 05/06/2024] [Revised: 07/19/2024] [Accepted: 07/29/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION The incidence of Central Nervous System (CNS) disorders, including Parkinson's disease, Alzheimer's disease, stroke, and malignancies, has risen significantly in recent decades, contributing to millions of deaths annually. Efficacious treatment of these disorders requires medicines targeting the brain. The Blood-brain Barrier (BBB) poses a formidable challenge to effective drug delivery to the brain, hindering progress in CNS therapeutics. This review explores the latest developments in nanoparticulate carriers, highlighting their potential to overcome BBB limitations. OBJECTIVE This study aimed to evaluate and summarise the critical factors and pathways in the nanoparticle- based CNS targeted drug delivery. METHODS An extensive literature search was conducted, comprising the initial development of nanoparticle- based CNS-targeted drug delivery approaches to the latest advancements using various online search tools. RESULTS The properties of nanoparticles, such as type of nanoparticles, size, shape, surface charge, hydrophobicity, and surface functionalisation, along with properties of the BBB during normal and pathological conditions and their impact on the delivery of nanoparticles across the BBB, are identified and discussed here. CONCLUSION Important properties and pathways that determine the penetration of nanoparticles across the CNS are reviewed in this article, along with recent advances in the field.
Collapse
Affiliation(s)
- Usha Nayak
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Praveen Halagali
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Khushi N Panchal
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Vamshi Krishna Tippavajhala
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S102TA, UK
| | - Jyothsna Manikkath
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| |
Collapse
|
35
|
Maity S, Bhuyan T, Jewell C, Kawakita S, Sharma S, Nguyen HT, Najafabadi AH, Ermis M, Falcone N, Chen J, Mandal K, Khorsandi D, Yilgor C, Choroomi A, Torres E, Mecwan M, John JV, Akbari M, Wang Z, Moniz-Garcia D, Quiñones-Hinojosa A, Jucaud V, Dokmeci MR, Khademhosseini A. Recent Developments in Glioblastoma-On-A-Chip for Advanced Drug Screening Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405511. [PMID: 39535474 PMCID: PMC11719323 DOI: 10.1002/smll.202405511] [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: 07/03/2024] [Revised: 09/08/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is an aggressive form of cancer, comprising ≈80% of malignant brain tumors. However, there are no effective treatments for GBM due to its heterogeneity and the presence of the blood-brain barrier (BBB), which restricts the delivery of therapeutics to the brain. Despite in vitro models contributing to the understanding of GBM, conventional 2D models oversimplify the complex tumor microenvironment. Organ-on-a-chip (OoC) models have emerged as promising platforms that recapitulate human tissue physiology, enabling disease modeling, drug screening, and personalized medicine. There is a sudden increase in GBM-on-a-chip models that can significantly advance the knowledge of GBM etiology and revolutionize drug development by reducing animal testing and enhancing translation to the clinic. In this review, an overview of GBM-on-a-chip models and their applications is reported for drug screening and discussed current challenges and potential future directions for GBM-on-a-chip models.
Collapse
Affiliation(s)
- Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Department of Orthopedic Surgery, Duke University School of
Medicine, Duke University, Durham, NC 27705
| | - Tamanna Bhuyan
- Department of Applied Biology, School of Biological
Sciences, University of Science & Technology Meghalaya, Meghalaya, 793101,
India
| | - Christopher Jewell
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Saurabh Sharma
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Center of Excellence in Biomaterials and Tissue
Engineering, Middle East Technical University, Ankara, Turkey
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Junjie Chen
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Emily Torres
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Johnson V. John
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Mohsen Akbari
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Laboratoryfor Innovations in Micro Engineering (LiME),
Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2,
Canada
- Biotechnology Center, Silesian University of Technology,
Akademicka 2A, 44-100 Gliwice, Poland
| | - Zhaohui Wang
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| |
Collapse
|
36
|
Deshmukh V, Pathan NS, Haldar N, Nalawade S, Narwade M, Gajbhiye KR, Gajbhiye V. Exploring intranasal drug delivery via nanocarriers: A promising glioblastoma therapy. Colloids Surf B Biointerfaces 2025; 245:114285. [PMID: 39366109 DOI: 10.1016/j.colsurfb.2024.114285] [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: 08/12/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
Glioblastoma is one of the most recurring types of glioma, having the highest mortality rate among all other gliomas. Traditionally, the standard course of treatment for glioblastoma involved maximum surgical resection, followed by chemotherapy and radiation therapy. Nanocarriers have recently focused on enhancing the chemotherapeutic administration to the brain to satisfy unmet therapeutic requirements for treating brain-related disorders. Due to the significant drawbacks and high recurrence rates of gliomas, intranasal administration of nanocarrier systems presents several advantages. These include low toxicity, non-invasiveness, and the ability to cross the blood-brain barrier. By customizing their size, encasing them with mucoadhesive agents, or undergoing surface modification that encourages movement over the nose's mucosa, we can exceptionally engineer nanocarriers for intranasal administration. Olfactory and trigeminal nerves absorb drugs administered nasally and transport them to the brain, serving as the primary delivery mechanism for nose-to-brain administration. This review sums up the latest developments in chemotherapeutic nanocarriers, such as metallic nanoparticles, polymeric nanoparticles, nanogels, nano vesicular carriers, genetic material-based nanocarriers, and polymeric micelles. These nanocarriers have demonstrated efficient drug delivery from the nose to the brain, effectively overcoming mucociliary clearance. However, challenges persist, such as limitations in targeted chemotherapy and restricted drug loading capacity for intranasal administration. Additionally, the review addresses regulatory considerations and prospects for these innovative drug delivery systems.
Collapse
Affiliation(s)
- Vishawambhar Deshmukh
- Poona College of Pharmacy, Bharati Vidyapeeth Deemed to be University, Erandwane, Pune 411038, India
| | - Nida Sayed Pathan
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India
| | - Niladri Haldar
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India
| | - Shubhangi Nalawade
- Poona College of Pharmacy, Bharati Vidyapeeth Deemed to be University, Erandwane, Pune 411038, India
| | - Mahavir Narwade
- Poona College of Pharmacy, Bharati Vidyapeeth Deemed to be University, Erandwane, Pune 411038, India
| | - Kavita R Gajbhiye
- Poona College of Pharmacy, Bharati Vidyapeeth Deemed to be University, Erandwane, Pune 411038, India.
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| |
Collapse
|
37
|
Daskoulidou N, Carpanini SM, Zelek WM, Morgan BP. Involvement of Complement in Alzheimer's Disease: From Genetics Through Pathology to Therapeutic Strategies. Curr Top Behav Neurosci 2025; 69:3-24. [PMID: 39455500 DOI: 10.1007/7854_2024_524] [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: 10/28/2024]
Abstract
Complement is a critical component of innate immunity, evolved to defend against pathogens and clear toxic debris ranging from dead and dying cells to immune complexes. These roles make complement a key player in homeostasis; however, complement has a dark side. When the rigid control mechanisms fail, complement becomes dysregulated, acting as a driver of inflammation and resultant pathology in numerous diseases. Roles of complement in Alzheimer's disease (AD) and other dementias have emerged in recent years, supported by genetic, biomarker and pathological evidence and animal model studies. Numerous questions remain regarding the precise roles of complement in the brain in health and disease, including where and when complement is expressed, how it contributes to immune defence and garbage disposal in the healthy brain, and exactly how complement contributes to pathology in dementias. In this brief review, we will summarise current knowledge on complement roles in brain, present the evidence implicating complement in AD and explore whether complement represents an attractive therapeutic target for AD.
Collapse
Affiliation(s)
| | - Sarah M Carpanini
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK
| | - Wioleta M Zelek
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK
| | - B Paul Morgan
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK.
| |
Collapse
|
38
|
Firuzpour F, Saleki K, Aram C, Rezaei N. Nanocarriers in glioblastoma treatment: a neuroimmunological perspective. Rev Neurosci 2024:revneuro-2024-0097. [PMID: 39733347 DOI: 10.1515/revneuro-2024-0097] [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: 07/19/2024] [Accepted: 12/08/2024] [Indexed: 12/31/2024]
Abstract
Glioblastoma multiforme (GBM) is the most fatal brain tumor with a poor prognosis with current treatments, mainly because of intrinsic resistance processes. GBM is also referred to as grade 4 astrocytoma, that makes up about 15.4 % of brain cancers globally as well as 60-75 % of astrocytoma. The most prevalent therapeutic choices for GBM comprise surgery in combination with radiotherapy and chemotherapy, providing patients with an average survival of 6-14 months. Nanocarriers provide various benefits such as enhanced drug solubility, biocompatibility, targeted activity, as well as minimized side effects. In addition, GBM treatment comes with several challenges such as the presence of the blood-brain barrier (BBB), blood-brain tumor barrier (BBTB), overexpressed efflux pumps, infiltration, invasion, drug resistance, as well as immune escape due to tumor microenvironment (TME) and cancer stem cells (CSC). Recent research has focused on nanocarriers due to their ability to self-assemble, improve bioavailability, provide controlled release, and penetrate the BBB. These nano-based components could potentially enhance drug accumulation in brain tumor tissues and reduce systemic toxicity, making them a compelling solution for GBM therapy. This review captures the complexities associated with multi-functional nano drug delivery systems (NDDS) in crossing the blood-brain barrier (BBB) and targeting cancer cells. In addition, it presents a succinct overview of various types of targeted multi-functional nano drug delivery system (NDDS) which has exhibited promising value for improving drug delivery to the brain.
Collapse
Affiliation(s)
- Faezeh Firuzpour
- USERN Office, Babol University of Medical Sciences, 47176-41367, Babol, Iran
- Student Research Committee, Babol University of Medical Sciences, 47176-41367, Babol, Iran
| | - Kiarash Saleki
- USERN Office, Babol University of Medical Sciences, 47176-41367, Babol, Iran
- Student Research Committee, Babol University of Medical Sciences, 47176-41367, Babol, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Cena Aram
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, 15719-14911, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| |
Collapse
|
39
|
Shen B, Liu N, Dai Y. Exosomes derived from umbilical cord mesenchymal stem cells ameliorate ischemic brain injury in mice by regulating AAK1 via miR-664a-5p. Int J Neurosci 2024:1-15. [PMID: 39655875 DOI: 10.1080/00207454.2024.2441120] [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: 11/11/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVE To identify the molecular targets of mesenchymal stem cell (MSC)-derived exosomes in treating cerebral ischemia and elucidate their therapeutic mechanisms. METHODS We utilized a mouse model of middle cerebral artery occlusion and treated mice with umbilical cord mesenchymal stem cells derived exosomes. Proteomic analysis identified AAK1(AP2 associated kinase 1) as a key target protein. Functional studies confirmed that AAK1 modulates the NF-κB signaling pathway in ischemic stroke. MicroRNA profiling, bioinformatic prediction and cell experiments identified miR-664a-5p as the specific microRNA regulating AAK1 expression. Finally, we validated the therapeutic effects of umbilical cord mesenchymal stem cell-derived exosomes using engineered miR-664a-5p-deficient exosomes. RESULTS Our findings demonstrate that umbilical cord mesenchymal stem cells-derived exosomes exert neuroprotective effects in ischemic stroke by modulating the AAK1/NF-κB axis via miR-664a-5p. CONCLUSION This study provides novel insights into the therapeutic mechanism of mesenchymal stem cell-derived exosomes in ischemic stroke, highlighting their potential for developing exosome-based therapies.
Collapse
Affiliation(s)
- Baoxi Shen
- Medical School of Chinese PLA, Beijing, China
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Ning Liu
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yiwu Dai
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
40
|
Hey G, Mehkri I, Mehkri Y, Maqbool H, Tahirkheli M, Woodford S, Lucke-Wold B. Nanoparticle-Based Therapies for Management of Subarachnoid Hemorrhage, Neurotrauma, and Stroke. Biomedicines 2024; 13:16. [PMID: 39857600 PMCID: PMC11760890 DOI: 10.3390/biomedicines13010016] [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: 12/02/2024] [Revised: 12/21/2024] [Accepted: 12/22/2024] [Indexed: 01/27/2025] Open
Abstract
Neurotrauma, stroke, and subarachnoid hemorrhage (SAH) are symptomatically diverse and etiologically complex central nervous system pathologies. Despite numerous therapeutic modalities that are available to minimize neurologic damage and secondary injury, the prognosis can still be dismal and unpredictable. Nanoparticle (NP) technology allows for deliberate, modular, and minimally invasive drug delivery. This literature review encompasses pertinent information on the impact and versatility of nanoparticle therapeutics when treating neurotrauma, stroke, and SAH. Currently, notable treatments such as Perfluorooctyl-Bromide (PFOB), PLGA nanoparticles, and ischemic relief-based NPs are promising new techniques for the management of these complex pathologies.
Collapse
Affiliation(s)
- Grace Hey
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Ilyas Mehkri
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Yusuf Mehkri
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Hasan Maqbool
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Mubariz Tahirkheli
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Samuel Woodford
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.H.); (Y.M.)
| | - Brandon Lucke-Wold
- Lillian S. Wells Department of Neurosurgery, College of Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL 32610, USA
| |
Collapse
|
41
|
Wang Y, Che H, Qu L, Lu X, Dong M, Sun B, Guan H. The role of nanomaterials in revolutionizing ischemic stroke treatment: Current trends and future prospects. iScience 2024; 27:111373. [PMID: 39669428 PMCID: PMC11634991 DOI: 10.1016/j.isci.2024.111373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024] Open
Abstract
Ischemic stroke has a high disability rate, which leads to irreversible neuronal death. The efficacy of conventional stroke treatments, including thrombolytic and neuroprotective therapies, is constrained by a number of factors, including safety concerns and inefficient drug delivery. The advent of nanomaterials has created new avenues for stroke therapy, facilitating enhanced pharmacokinetic behavior of drugs, effective drug accumulation at the target site, augmented therapeutic efficacy, and concomitant reduction in side effects. Therefore, this paper pioneers a research approach that summarized the development trend and clinical value of nanomaterials in the field of ischemic stroke through bibliometric analysis. This review provides an overview of the pathophysiological mechanisms of stroke and examines the current research trends in the use of nanomaterials in stroke management. It encompasses a multitude of domains, including targeted drug delivery systems, biosensors for the sensitive detection of biomarkers, and neuroprotective nanotechnologies capable of traversing the blood-brain barrier. Moreover, we investigate the challenges that nanomaterials encounter in the clinical translation context, including those pertaining to biocompatibility and long-term safety. These results have provided the clinical value and limitations of nanomaterials in the diagnosis and treatment of ischemic stroke from double perspectives, thereby offering new avenues for the further development of innovative nanotherapeutic tools.
Collapse
Affiliation(s)
- Yong Wang
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Huiying Che
- Department of General Practice, Yanbian University Hospital, Yanji 133002, China
| | - Linzhuo Qu
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Xin Lu
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Mingzhen Dong
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Bo Sun
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Hongjian Guan
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| |
Collapse
|
42
|
Tiwari S, Chaturvedi S, Kaul A, Choudhary V, Barthélémy P, Mishra AK. Development of amphiphilic self-assembled nucleolipid as BBB targeting probe based on SPECT. DISCOVER NANO 2024; 19:210. [PMID: 39690348 DOI: 10.1186/s11671-024-04129-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 10/14/2024] [Indexed: 12/19/2024]
Abstract
Several approaches have been utilised to deliver therapeutic nanoparticles inside the brain but rendered by certain limitation such as active efflux, non-stability, toxicity of the nanocarrier, transport, physicochemical properties and many more. In this context use of biocompatible nano carriers is currently investigated. We herein present the hypothesis that the nucleoside-lipid based conjugates (nucleolipids) which are biocompatible in nature and have molecular recognition can be tuned for improved permeation across blood-brain barrier (BBB). In this work, a di-C15-palmitoyl-ketal nucleolipid nanoparticle bearing an acyclic chelator has been formulated, radiolabeled with 99mTc and evaluated for in vivo fate using SPECT imaging. The mean particle size of particles was 113 nm and found to be nontoxic as depticted through haemolytic assay (2.33% erythrocyte destruction) and 75 ± 0.3% HEK(Human Embryonic Kidney) cells survived at 72 h as depicted in SRB (Sulforhodamine B) toxicity assay. The encapsulation efficiency (68 ± 2.75%) and drug loading capacity (22 ± 1.8%.) was calculated for nanoparticles using Methotrexate as model anti-cancer drug. The mathematical models indicate fickian release with a release constant KH = 20.70. With 98 ± 0.75% radiolabelling efficiency and established in vitro stability, nanoparticles showed brain uptake in normal mice as 0.91 times in comparison to BBB compromised mice (1.6% ± 0.03 ID/g)indicating higher brain uptake with rapid clearance as depicted through blood kinetics.
Collapse
Affiliation(s)
- Swastika Tiwari
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, 110054, India.
- NOMATEN Center of Excellence, National Center for Nuclear Research, Ul. Andrzeja Soltana 7, 05-400, Otwock, Poland.
| | - Shubhra Chaturvedi
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, 110054, India.
| | - Ankur Kaul
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, 110054, India
| | - Vishakha Choudhary
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, 110054, India
| | - Philippe Barthélémy
- Université de Bordeaux, INSERM, U1212, CNRS UMR 5320, ARNA, ARN: Régulations Naturelle Et Artificielle, ChemBioPharm, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France
| | - A K Mishra
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, 110054, India.
| |
Collapse
|
43
|
Raza MA, Sharma MK, Nagori K, Jain P, Ghosh V, Gupta U, Ajazuddin. Recent trends on polycaprolactone as sustainable polymer-based drug delivery system in the treatment of cancer: Biomedical applications and nanomedicine. Int J Pharm 2024; 666:124734. [PMID: 39343332 DOI: 10.1016/j.ijpharm.2024.124734] [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: 06/15/2024] [Revised: 09/05/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
The unique properties-such as biocompatibility, biodegradability, bio-absorbability, low cost, easy fabrication, and high versatility-have made polycaprolactone (PCL) the center of attraction for researchers. The derived introduction in this manuscript gives a pretty detailed overview of PCL, so you can first brush up on it. Discussion on the various PCL-based derivatives involves, but is not limited to, poly(ε-caprolactone-co-lactide) (PCL-co-LA), PCL-g-PEG, PCL-g-PMMA, PCL-g-chitosan, PCL-b-PEO, and PCL-g-PU specific properties and their probable applications in biomedicine. This paper has considered examining the differences in the diverse disease subtypes and the therapeutic value of using PCL. Advanced strategies for PCL in delivery systems are also considered. In addition, this review discusses recently patented products to provide a snapshot of recent updates in this field. Furthermore, the text probes into recent advances in PCL-based DDS, for example, nanoparticles, liposomes, hydrogels, and microparticles, while giving special attention to comparing the esters in the delivery of bioactive compounds such as anticancer drugs. Finally, we review future perspectives on using PCL in biomedical applications and the hurdles of PCL-based drug delivery, including fine-tuning mechanical strength/degradation rate, biocompatibility, and long-term effects in living systems.
Collapse
Affiliation(s)
- Mohammad Adnan Raza
- Department of Pharmaceutics, Rungta College of Pharmaceutical Science and Research, Bhilai 490024, Chhattisgarh, India
| | - Mukesh Kumar Sharma
- Department of Pharmaceutics, Rungta College of Pharmaceutical Science and Research, Bhilai 490024, Chhattisgarh, India
| | - Kushagra Nagori
- Department of Pharmaceutics, Rungta College of Pharmaceutical Science and Research, Bhilai 490024, Chhattisgarh, India
| | - Parag Jain
- Department of Pharmaceutics, Rungta College of Pharmaceutical Science and Research, Bhilai 490024, Chhattisgarh, India
| | - Vijayalakshmi Ghosh
- Department of Biotechnology, GD Rungta College of Science & Technology, Bhilai 490024, Chhattisgarh, India
| | - Umesh Gupta
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer 305817, Rajasthan, India
| | - Ajazuddin
- Department of Pharmaceutics, Rungta College of Pharmaceutical Science and Research, Bhilai 490024, Chhattisgarh, India; Rungta College of Engineering and Technology, Bhilai 490024, Chhattisgarh, India.
| |
Collapse
|
44
|
Rafati N, Zarepour A, Bigham A, Khosravi A, Naderi-Manesh H, Iravani S, Zarrabi A. Nanosystems for targeted drug Delivery: Innovations and challenges in overcoming the Blood-Brain barrier for neurodegenerative disease and cancer therapy. Int J Pharm 2024; 666:124800. [PMID: 39374818 DOI: 10.1016/j.ijpharm.2024.124800] [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: 07/18/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
The evolution of sophisticated nanosystems has revolutionized biomedicine, notably in treating neurodegenerative diseases and cancer. These systems show potential in delivering medication precisely to affected tissues, improving treatment effectiveness while minimizing side effects. Nevertheless, a major hurdle in targeted drug delivery is breaching the blood-brain barrier (BBB), a selective shield separating the bloodstream from the brain and spinal cord. The tight junctions between endothelial cells in brain capillaries create a formidable physical barrier, alongside efflux transporters that expel harmful molecules. This presents a notable challenge for brain drug delivery. Nanosystems present distinct advantages in overcoming BBB challenges, offering enhanced drug efficacy, reduced side effects, improved stability, and controlled release. Despite their promise, challenges persist, such as the BBB's regional variability hindering uniform drug distribution. Efflux transporters can also limit therapeutic agent efficacy, while nanosystem toxicity necessitates rigorous safety evaluations. Understanding the long-term impact of nanomaterials on the brain remains crucial. Additionally, addressing nanosystem scalability, cost-effectiveness, and safety profiles is vital for widespread clinical implementation. This review delves into the advancements and obstacles of advanced nanosystems in targeted drug delivery for neurodegenerative diseases and cancer therapy, with a focus on overcoming the BBB.
Collapse
Affiliation(s)
- Nesa Rafati
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Ashkan Bigham
- Institute of Polymers, Composites, and Biomaterials, National Research Council (IPCB-CNR), Naples 80125, Italy; Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Turkiye
| | - Hossein Naderi-Manesh
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran; Departments of Biophysics, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran.
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
45
|
Chua AJ, Di Francesco V, D'Souza A, Amiji M, Bleier BS. Murine model of minimally invasive nasal depot (MIND) technique for central nervous system delivery of blood-brain barrier-impermeant therapeutics. Lab Anim (NY) 2024; 53:363-375. [PMID: 39548349 DOI: 10.1038/s41684-024-01460-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 10/14/2024] [Indexed: 11/17/2024]
Abstract
The blood-brain barrier (BBB) poses a substantial obstacle to the successful delivery of therapeutics to the central nervous system (CNS). The transnasal route has been extensively explored, but success rates have been modest due to challenges related to the precise anatomical placement of drugs, the small volumes that the olfactory cleft can accommodate and short drug residence times due to mucociliary clearance. Here, to address these issues, we have developed a surgical technique known as the minimally invasive nasal depot (MIND), which allows the accurate placement of depot drugs into the submucosal space of the olfactory epithelium of rats. This technique exploits the unique anatomy of the olfactory apparatus to enable transnasal delivery of drugs into the CNS, bypassing the BBB. In our rat model, a bony window is created in the animal snout to expose the submucosal space. Using the MIND technique, we have successfully delivered oligonucleotides to the CNS in Sprague-Dawley and Long-Evans rats, leading to an upregulation of brain-derived neurotrophic factor in the substantia nigra and hippocampus. In this Protocol, we describe the procedural steps for MIND. This procedure takes about 45 min and can be performed by researchers with basic surgical skills. We additionally describe modifications to perform MIND in mice, which are anatomically smaller. The MIND procedure represents a unique platform that can be used to overcome the limitations posed by the BBB. This technique can potentially expand the therapeutic toolkit in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Andy J Chua
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA.
- Department of Otorhinolaryngology - Head and Neck Surgery, Sengkang General Hospital, Singapore, Singapore.
| | - Valentina Di Francesco
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Anisha D'Souza
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA, USA
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, USA
| | - Benjamin S Bleier
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| |
Collapse
|
46
|
Nengroo MA, Ben-Sahra I. Brain-penetrating molecule might offer a route to treat glioblastoma tumours. Nature 2024; 636:307-308. [PMID: 39567800 DOI: 10.1038/d41586-024-03556-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
|
47
|
Gorji FS, Mahdavian SF, Khodashenas S, Kiasari ZR, Valadan R, Khalili S, Mahdavi MR. Exosomes with Engineered Brain Derived Neurotrophic Factor on Their Surfaces Can Proliferate Menstrual Blood Derived Mesenchymal Stem Cells: Targeted Delivery for a Protein Drug. Protein J 2024; 43:1070-1082. [PMID: 39397145 DOI: 10.1007/s10930-024-10234-9] [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] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
Despite the efficacy of brain derived neurotrophic factor (BDNF) in neuro-regenerative medicine, it can't pass the blood-brain barrier. Recently, exosomes have been harnessed for targeted delivery of therapeutics into brain. Given these facts, an engineered exosome capable of BDNF expression on the surface would be an amenable tool for drug delivery. The BDNF gene was cloned into a plex-lamp lentiviral vector and virus particles were packaged using the Torano method. HEK293T cells were transduced by the purified viruses to produce and purify recombinant exosomes displaying the fusion protein on their surfaces. Western blot, Zeta sizer, TEM, and ELISA methods were used for exosome characterization. The effect of engineered exosomes on menstrual blood-derived mesenchymal stem cells (Mens-MSCs) proliferation was evaluated by cell counting assay, MTT assay, and qPCR on the bcl2 and nestin genes. Approximately, 1.8 × 108 TdU/ml of the viral particles was purified from the transfected cells and transduced into the HEK293T. Western blot and ELISA methods confirmed the surface display of the LAMP-BDNF fusion. TEM graphs and Zeta sizer results confirmed the morphology and the size of purified exosomes. Treatment of Mens-MSCs with the targeted exosomes augmented the expression level of bcl2 and nestin genes, increased the cell proliferation, and elevated the cell number. Chimeric BDNF on the exosome surface could retain its biological activity and elevate the expression of bcl2 and nestin genes. Moreover, these exosomes are capable of elevating the Mens-MSCs proliferation.
Collapse
Affiliation(s)
| | - Seyedeh Farzaneh Mahdavian
- Medical Biotechnology, School of Advance Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shabanali Khodashenas
- Medical Biotechnology, School of Advance Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
- Thalassemia Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Zeinab Rezaee Kiasari
- Medical Biotechnology, School of Advance Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Valadan
- Department of Immunology, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Mohammad Reza Mahdavi
- Thalassemia Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Laboratory Sciences, Faculty of Allied Medical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
48
|
Brockway DF, Crowley NA. Emerging pharmacological targets for alcohol use disorder. Alcohol 2024; 121:103-114. [PMID: 39069210 PMCID: PMC11638729 DOI: 10.1016/j.alcohol.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/27/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Alcohol Use Disorder (AUD) remains a challenging condition with limited effective treatment options; however new technology in drug delivery and advancements in pharmacology have paved the way for discovery of novel therapeutic targets. This review explores emerging pharmacological targets that offer new options for the management of AUD, focusing on the potential of somatostatin (SST), vasoactive intestinal peptide (VIP), glucagon-like peptide-1 (GLP-1), nociceptin (NOP), and neuropeptide S (NPS). These targets have been selected based on recent advancements in preclinical and clinical research, which suggest their significant roles in modulating alcohol consumption and related behaviors. SST dampens cortical circuits, and targeting both the SST neurons and the SST peptide itself presents promise for treating AUD and various related comorbidities. VIP neurons are modulated by alcohol and targeting the VIP system presents an unexplored avenue for addressing alcohol exposure at various stages of development. GLP-1 interacts with the dopaminergic reward system and reduces alcohol intake. Nociceptin modulates mesolimbic circuitry and agonism and antagonism of nociceptin receptor offers a complex but promising approach to reducing alcohol consumption. NPS stands out for its anxiolytic-like effects, particularly relevant for the anxiety associated with AUD. This review aims to synthesize the current understanding of these targets, highlighting their potential in developing more effective and personalized AUD therapies, and underscores the importance of continued research in identifying and validating novel targets for treatment of AUD and comorbid conditions.
Collapse
Affiliation(s)
- Dakota F Brockway
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA; Penn State Neuroscience Institute, Penn State University, University Park, PA, 16802, USA.
| | - Nicole A Crowley
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA; Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA; Penn State Neuroscience Institute, Penn State University, University Park, PA, 16802, USA.
| |
Collapse
|
49
|
Mesut B, Al-Mohaya M, Gholap AD, Yeşilkaya E, Das U, Akhtar MS, Sah R, Khan S, Moin A, Faiyazuddin M. Demystifying the potential of lipid-based nanocarriers in targeting brain malignancies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9243-9279. [PMID: 38963550 DOI: 10.1007/s00210-024-03212-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/02/2024] [Indexed: 07/05/2024]
Abstract
Drug targeting for brain malignancies is restricted due to the presence of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB), which act as barriers between the blood and brain parenchyma. Certainly, the limited therapeutic options for brain malignancies have made notable progress with enhanced biological understanding and innovative approaches, such as targeted therapies and immunotherapies. These advancements significantly contribute to improving patient prognoses and represent a promising shift in the landscape of brain malignancy treatments. A more comprehensive understanding of the histology and pathogenesis of brain malignancies is urgently needed. Continued research focused on unraveling the intricacies of brain malignancy biology holds the key to developing innovative and tailored therapies that can improve patient outcomes. Lipid nanocarriers are highly effective drug delivery systems that significantly improve their solubility, bioavailability, and stability while also minimizing unwanted side effects. Surface-modified lipid nanocarriers (liposomes, niosomes, solid lipid nanoparticles, nanostructured lipid carriers, lipid nanocapsules, lipid-polymer hybrid nanocarriers, lipoproteins, and lipoplexes) are employed to improve BBB penetration and uptake through various mechanisms. This systematic review illuminates and covers various topics related to brain malignancies. It explores the different methods of drug delivery used in treating brain malignancies and delves into the benefits, limitations, and types of brain-targeted lipid-based nanocarriers. Additionally, this review discusses ongoing clinical trials and patents related to brain malignancy therapies and provides a glance into future perspectives for treating this condition.
Collapse
Affiliation(s)
- Burcu Mesut
- Pharmaceutical Technology Department, Faculty of Pharmacy, Istanbul University, Istanbul, 34216, Turkey
| | - Mazen Al-Mohaya
- Institute of Health Sciences, Istanbul University, Istanbul, 34216, Turkey
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, 401404, Maharashtra, India
| | - Eda Yeşilkaya
- Institute of Health Sciences, Istanbul University, Istanbul, 34216, Turkey
| | - Ushasi Das
- Pharmaceutical Technology Department, Jadavpur University, Kolkata, West Bengal, India
| | - Mohammad Shabib Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Ranjit Sah
- Department of Microbiology, Institute of Medicine, Tribhuvan University Teaching Hospital, Kathmandu, 44600, Nepal.
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, 411018, Maharashtra, India.
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, 411018, Maharashtra, India.
| | | | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Hail, 2440, Hail, Saudi Arabia
| | - Md Faiyazuddin
- School of Pharmacy, Al - Karim University, Katihar, 854106, Bihar, India.
- Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| |
Collapse
|
50
|
Eades W, Abdolmohammadpourbonab S, Dinh L, Yan B. Ionic liquids and their potential use in development and improvement of drug delivery systems: evidence of their tendency to promote drug accumulation in the brain. Pharm Dev Technol 2024; 29:1065-1074. [PMID: 39403783 DOI: 10.1080/10837450.2024.2417004] [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: 05/21/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Ionic liquids (ILs) are considered salt in liquid state, which is composed of organic cations and anions with low melting points (<100 °C). ILs have become a major scientific area with an extensive range of applications including chemistry, electrochemistry, and pharmaceutics. ILs have received great research interest in the pharmaceutical field as solvents, anti-solvents, co-solvents, and reagents in synthesis and formulation. While therapeutic ILs have been investigated for oral and trans-dermal drug delivery systems showing promising compatibility with a wide range of therapeutics, enhanced drug permeation through the skin, and cell membrane solvation to open channels to facilitate molecular passage, their potential to cross the challenging blood-brain barrier (BBB) remains an unanswered question. IL-based therapies could potentially be a game changer for improving drug delivery to cellular targets both at and across the BBB. In this review, we discuss (1) the tunable physicochemical properties of ILs; (2) the vast and various applications of ILs in the development and improvement of drug delivery systems; and (3) ILs as a potential approach for increasing drug accumulation in the brain tissue.
Collapse
Affiliation(s)
- William Eades
- Division of Pharmaceutical Sciences, University of Cincinnati, Cincinnati, OH, USA
| | | | - Linh Dinh
- Division of Pharmaceutical Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|