1
|
Chea M, Bouvier S, Gris JC. The hemostatic system in chronic brain diseases: A new challenging frontier? Thromb Res 2024; 243:109154. [PMID: 39305718 DOI: 10.1016/j.thromres.2024.109154] [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: 06/13/2024] [Revised: 08/19/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Neurological diseases (ND), including neurodegenerative diseases (NDD) and psychiatric disorders (PD), present a significant public health challenge, ranking third in Europe for disability and premature death, following cardiovascular diseases and cancers. In 2017, approximately 540 million cases of ND were reported among Europe's 925 million people, with strokes, dementia, and headaches being most prevalent. Nowadays, more and more evidence highlight the hemostasis critical role in cerebral homeostasis and vascular events. Indeed, hemostasis, thrombosis, and brain abnormalities contributing to ND form a complex and poorly understood equilibrium. Alterations in vascular biology, particularly involving the blood-brain barrier, are implicated in ND, especially dementia, and PD. While the roles of key coagulation players such as thrombin and fibrinogen are established, the roles of other hemostasis components are less clear. Moreover, the involvement of these elements in psychiatric disease pathogenesis is virtually unstudied, except in specific pathological models such as antiphospholipid syndrome. Advanced imaging techniques, primarily functional magnetic resonance imaging and its derivatives like diffusion tensor imaging, have been developed to study brain areas affected by ND and to improve our understanding of the pathophysiology of these diseases. This literature review aims to clarify the current understanding of the connections between hemostasis, thrombosis, and neurological diseases, as well as explore potential future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mathias Chea
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France.
| | - Sylvie Bouvier
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Jean-Christophe Gris
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France; I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
2
|
Acharya P, Shrestha S, Joshi P, Choi NY, Lekkala VKR, Kang SY, Ni G, Lee MY. Dynamic culture of cerebral organoids using a pillar/perfusion plate for the assessment of developmental neurotoxicity. Biofabrication 2024; 17:10.1088/1758-5090/ad867e. [PMID: 39444222 PMCID: PMC11542746 DOI: 10.1088/1758-5090/ad867e] [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: 08/15/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Despite the potential toxicity of commercial chemicals to the development of the nervous system (known as developmental neurotoxicity or DNT), conventionalin vitrocell models have primarily been employed for the assessment of acute neuronal toxicity. On the other hand, animal models used for the assessment of DNT are not physiologically relevant due to the heterogenic difference between humans and animals. In addition, animal models are low-throughput, time-consuming, expensive, and ethically questionable. Recently, human brain organoids have emerged as a promising alternative to assess the detrimental effects of chemicals on the developing brain. However, conventional organoid culture systems have several technical limitations including low throughput, lack of reproducibility, insufficient maturity of organoids, and the formation of the necrotic core due to limited diffusion of nutrients and oxygen. To address these issues and establish predictive DNT models, cerebral organoids were differentiated in a dynamic condition in a unique pillar/perfusion plate, which were exposed to test compounds to evaluate DNT potential. The pillar/perfusion plate facilitated uniform, dynamic culture of cerebral organoids with improved proliferation and maturity by rapid, bidirectional flow generated on a digital rocker. Day 9 cerebral organoids in the pillar/perfusion plate were exposed to ascorbic acid (DNT negative) and methylmercury (DNT positive) in a dynamic condition for 1 and 3 weeks, and changes in organoid morphology and neural gene expression were measured to determine DNT potential. As expected, ascorbic acid did not induce any changes in organoid morphology and neural gene expression. However, exposure of day 9 cerebral organoids to methylmercury resulted in significant changes in organoid morphology and neural gene expression. Interestingly, methylmercury did not induce adverse changes in cerebral organoids in a static condition, thus highlighting the importance of dynamic organoid culture in DNT assessment.
Collapse
Affiliation(s)
- Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | | | - Na Young Choi
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | | | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Gabriel Ni
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
- Bioprinting Laboratories Inc., Dallas, Texas
| |
Collapse
|
3
|
Liu Y, Liu Y, Shi P, Hu X, Fan X, Wu Y, Pan J, Bai Q, Li Q. Single-atom nanozyme liposome-integrated microneedles for in situ drug delivery and anti-inflammatory therapy in Parkinson's disease. J Nanobiotechnology 2024; 22:643. [PMID: 39427214 PMCID: PMC11490154 DOI: 10.1186/s12951-024-02924-4] [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: 12/15/2023] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
Treatment for Parkinson's disease (PD) has been impeded by inefficient treatment results and multiple membrane barriers during drug delivery. This study reports the design, synthesis, and application of microneedles (MNs) loaded with mitochondrion-targeted liposome encapsulated iron (Fe)-isolated single-atom nanozymes (Mito@Fe-ISAzyme, MFeI), called MFeI MNs, for in situ drug delivery into the brain parenchyma and efficient enrichment of drugs in lesion sites. In in vitro experiments, MFeI can scavenge reactive oxygen species (ROS) and protect the neurons via mitochondrial targeting, guaranteeing the subsequent treatment of PD. Using PD mouse models, we compared the intravenous injection of MFeI with the brain in situ administration of MFeI MNs (in situ MFeI MNs). Results showed that in situ MFeI MNs significantly improved the deep penetration of the drug into brain parenchyma, especially in the vital pathological sites such as the substantia nigra pars compacta and striatum. Importantly, ROS elimination and neuroinflammatory remission in the lesion site were observed, thereby efficiently alleviating the behavioral disorders and pathological symptoms of PD mice. Therefore, the MNs system for in situ single-atom nanozyme liposome delivery exhibits great potential in PD treatment.
Collapse
Affiliation(s)
- Ying Liu
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Henan, 450000, China
- Henan Key Laboratory of Chronic Disease Management, Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Henan, 450000, China
| | - Ye Liu
- Department of Anaesthesiology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Peimiao Shi
- The Affiliated Tengzhou Central People's Hospital of Xuzhou Medical University, Shandong, 277500, China
| | - Xiaopeng Hu
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Henan, 450000, China
| | - Xiaowan Fan
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Henan, 450000, China
| | - Yalong Wu
- Henan Key Laboratory of Chronic Disease Management, Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Henan, 450000, China
| | - Jiangpeng Pan
- Henan Key Laboratory of Chronic Disease Management, Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Henan, 450000, China
| | - Qian Bai
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Henan, 450000, China
| | - Qing Li
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Henan, 450000, China.
| |
Collapse
|
4
|
Kan X, Ma J, Ma J, Li D, Li F, Cao Y, Huang C, Li Y, Liu P. Dual-targeted TfRA4-DNA1-Ag@AuNPs: An innovative radiosensitizer for enhancing radiotherapy in glioblastoma multiforme. Colloids Surf B Biointerfaces 2024; 245:114328. [PMID: 39442410 DOI: 10.1016/j.colsurfb.2024.114328] [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: 07/29/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Radiation therapy (RT) is one of the most effective and widely used treatment methods for glioblastoma multiforme (GBM). However, its efficacy is often compromised by the inherent radioresistance of tumor cells, while the restrictive nature of the blood-brain barrier (BBB) specifically impedes the delivery of radiosensitizer. Thus, we constructed and characterized polyethylene glycol (PEG)-functionalized silver-gold core-shell nanoparticles (PSGNPs) targeting both BBB (TfRA4) and GBM (DNA1) (TDSGNPs). Afterwards, studies conducted both in vitro and in vivo were employed to assess the BBB penetration capabilities, abilities of GBM targeting and radiosensitization effect. Transmission electron microscope images of PSGNPs showed a core-shell structure, and the results of ultraviolet-visible absorption spectroscopy and dynamic light scattering displayed that TDSGNPs were successfully constructed with excellent dispersion properties. TDSGNPs could be specifically taken up by U87MG cells and the uptake peaked at 24 h. TDSGNPs combined with RT obviously increased the apoptosis proportion of the cells. It was shown by the in vitro and in vivo investigations that TDSGNPs could target U87MG cells after crossing the BBB, and further study revealed that TDSGNPs showed an uptake peak in the tumor sites after 3 h intravenous injection. The radiosensitization of TDSGNPs was better than that of the nanoparticles modified with single aptamers and the median survival of tumor-bearing mice was greatly extended. This study demonstrated that TDSGNPs could penetrate BBB to target GBM, functioning as a promising radiosensitizer for the targeted therapy of GBM.
Collapse
Affiliation(s)
- Xuechun Kan
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Jing Ma
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Jun Ma
- Radiotherapy Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, PR China
| | - Dongdong Li
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Fan Li
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Yuyu Cao
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Cheng Huang
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Yan Li
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Peidang Liu
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China; Jiangsu Key Laboratory for Biomaterials & Devices, Southeast University, Nanjing, Jiangsu 210009, PR China.
| |
Collapse
|
5
|
Wang P, Jin L, Zhang M, Wu Y, Duan Z, Guo Y, Wang C, Guo Y, Chen W, Liao Z, Wang Y, Lai R, Lee LP, Qin J. Blood-brain barrier injury and neuroinflammation induced by SARS-CoV-2 in a lung-brain microphysiological system. Nat Biomed Eng 2024; 8:1053-1068. [PMID: 37349391 DOI: 10.1038/s41551-023-01054-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/11/2023] [Indexed: 06/24/2023]
Abstract
In some patients, COVID-19 can trigger neurological symptoms with unclear pathogenesis. Here we describe a microphysiological system integrating alveolus and blood-brain barrier (BBB) tissue chips that recapitulates neuropathogenesis associated with infection by SARS-CoV-2. Direct exposure of the BBB chip to SARS-CoV-2 caused mild changes to the BBB, and infusion of medium from the infected alveolus chip led to more severe injuries on the BBB chip, including endothelial dysfunction, pericyte detachment and neuroinflammation. Transcriptomic analyses indicated downregulated expression of the actin cytoskeleton in brain endothelium and upregulated expression of inflammatory genes in glial cells. We also observed early cerebral microvascular damage following lung infection with a low viral load in the brains of transgenic mice expressing human angiotensin-converting enzyme 2. Our findings suggest that systemic inflammation is probably contributing to neuropathogenesis following SARS-CoV-2 infection, and that direct viral neural invasion might not be a prerequisite for this neuropathogenesis. Lung-brain microphysiological systems should aid the further understanding of the systemic effects and neurological complications of viral infection.
Collapse
Affiliation(s)
- Peng Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Science and Technology of China, Hefei, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China
| | - Lin Jin
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Min Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yunsong Wu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zilei Duan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yaqiong Guo
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Chaoming Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yingqi Guo
- Core Technology Facility of Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wenwen Chen
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Zhiyi Liao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yaqing Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| | - Luke P Lee
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA.
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Korea.
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
- University of Science and Technology of China, Hefei, China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
6
|
Huang Y, Liu T, Huang Q, Wang Y. From Organ-on-a-Chip to Human-on-a-Chip: A Review of Research Progress and Latest Applications. ACS Sens 2024; 9:3466-3488. [PMID: 38991227 DOI: 10.1021/acssensors.4c00004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Organ-on-a-Chip (OOC) technology, which emulates the physiological environment and functionality of human organs on a microfluidic chip, is undergoing significant technological advancements. Despite its rapid evolution, this technology is also facing notable challenges, such as the lack of vascularization, the development of multiorgan-on-a-chip systems, and the replication of the human body on a single chip. The progress of microfluidic technology has played a crucial role in steering OOC toward mimicking the human microenvironment, including vascularization, microenvironment replication, and the development of multiorgan microphysiological systems. Additionally, advancements in detection, analysis, and organoid imaging technologies have enhanced the functionality and efficiency of Organs-on-Chips (OOCs). In particular, the integration of artificial intelligence has revolutionized organoid imaging, significantly enhancing high-throughput drug screening. Consequently, this review covers the research progress of OOC toward Human-on-a-chip, the integration of sensors in OOCs, and the latest applications of organoid imaging technologies in the biomedical field.
Collapse
Affiliation(s)
- Yisha Huang
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China
| | - Tong Liu
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Huang
- School of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
7
|
Jain S, Voulgaris D, Thongkorn S, Hesen R, Hägg A, Moslem M, Falk A, Herland A. On-Chip Neural Induction Boosts Neural Stem Cell Commitment: Toward a Pipeline for iPSC-Based Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401859. [PMID: 38655836 PMCID: PMC11220685 DOI: 10.1002/advs.202401859] [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: 02/21/2024] [Indexed: 04/26/2024]
Abstract
The clinical translation of induced pluripotent stem cells (iPSCs) holds great potential for personalized therapeutics. However, one of the main obstacles is that the current workflow to generate iPSCs is expensive, time-consuming, and requires standardization. A simplified and cost-effective microfluidic approach is presented for reprogramming fibroblasts into iPSCs and their subsequent differentiation into neural stem cells (NSCs). This method exploits microphysiological technology, providing a 100-fold reduction in reagents for reprogramming and a ninefold reduction in number of input cells. The iPSCs generated from microfluidic reprogramming of fibroblasts show upregulation of pluripotency markers and downregulation of fibroblast markers, on par with those reprogrammed in standard well-conditions. The NSCs differentiated in microfluidic chips show upregulation of neuroectodermal markers (ZIC1, PAX6, SOX1), highlighting their propensity for nervous system development. Cells obtained on conventional well plates and microfluidic chips are compared for reprogramming and neural induction by bulk RNA sequencing. Pathway enrichment analysis of NSCs from chip showed neural stem cell development enrichment and boosted commitment to neural stem cell lineage in initial phases of neural induction, attributed to a confined environment in a microfluidic chip. This method provides a cost-effective pipeline to reprogram and differentiate iPSCs for therapeutics compliant with current good manufacturing practices.
Collapse
Affiliation(s)
- Saumey Jain
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyMalvinas väg 10Stockholm100 44Sweden
- Division of NanobiotechnologyScience for Life LaboratoryKTH Royal Institute of TechnologyTomtebodavägen 23aSolna171 65Sweden
| | - Dimitrios Voulgaris
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyMalvinas väg 10Stockholm100 44Sweden
- Division of NanobiotechnologyScience for Life LaboratoryKTH Royal Institute of TechnologyTomtebodavägen 23aSolna171 65Sweden
- AIMESCenter for Integrated Medical and Engineering ScienceDepartment of NeuroscienceKarolinska InstitutetSolna171 65Sweden
| | - Surangrat Thongkorn
- Division of NanobiotechnologyScience for Life LaboratoryKTH Royal Institute of TechnologyTomtebodavägen 23aSolna171 65Sweden
- Chulalongkorn Autism Research and Innovation Center of Excellence (Chula ACE)Department of Clinical ChemistryFaculty of Allied Health SciencesChulalongkorn UniversityBangkok10330Thailand
| | - Rick Hesen
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyMalvinas väg 10Stockholm100 44Sweden
| | - Alice Hägg
- Neural Stem CellsDepartment of Experimental Medical ScienceLund Stem Cell CenterLund UniversityLund221 84Sweden
| | - Mohsen Moslem
- Department of NeuroscienceKarolinska InstitutetSolna171 65Sweden
| | - Anna Falk
- Neural Stem CellsDepartment of Experimental Medical ScienceLund Stem Cell CenterLund UniversityLund221 84Sweden
- Department of NeuroscienceKarolinska InstitutetSolna171 65Sweden
| | - Anna Herland
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyMalvinas väg 10Stockholm100 44Sweden
- Division of NanobiotechnologyScience for Life LaboratoryKTH Royal Institute of TechnologyTomtebodavägen 23aSolna171 65Sweden
- AIMESCenter for Integrated Medical and Engineering ScienceDepartment of NeuroscienceKarolinska InstitutetSolna171 65Sweden
- Department of NeuroscienceKarolinska InstitutetSolna171 65Sweden
| |
Collapse
|
8
|
Wan Y, Ding J, Jia Z, Hong Y, Tian G, Zheng S, Pan P, Wang J, Liang H. Current trends and research topics regarding organoids: A bibliometric analysis of global research from 2000 to 2023. Heliyon 2024; 10:e32965. [PMID: 39022082 PMCID: PMC11253259 DOI: 10.1016/j.heliyon.2024.e32965] [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: 08/16/2023] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 07/20/2024] Open
Abstract
The use of animal models for biological experiments is no longer sufficient for research related to human life and disease. The development of organ tissues has replaced animal models by mimicking the structure, function, development and homeostasis of natural organs. This provides more opportunities to study human diseases such as cancer, infectious diseases and genetic disorders. In this study, bibliometric methods were used to analyze organoid-related articles published over the last 20+ years to identify emerging trends and frontiers in organoid research. A total of 13,143 articles from 4125 institutions in 86 countries or regions were included in the analysis. The number of papers increased steadily over the 20-year period. The United States was the leading country in terms of number of papers and citations. Harvard Medical School had the highest number of papers published. Keyword analysis revealed research trends and focus areas such as organ tissues, stem cells, 3D culture and tissue engineering. In conclusion, this study used bibliometric and visualization methods to explore the field of organoid research and found that organ tissues are receiving increasing attention in areas such as cancer, drug discovery, personalized medicine, genetic disease modelling and gene repair, making them a current research hotspot and a future research trend.
Collapse
Affiliation(s)
- Yantong Wan
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jianan Ding
- School of Basic Medical Sciences, Southern Medical University Guangzhou, China
| | - Zixuan Jia
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yinghao Hong
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guijie Tian
- School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou, China
| | - Shuqian Zheng
- School of Basic Medical Sciences, Southern Medical University Guangzhou, China
| | - Pinfei Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jieyan Wang
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
| | - Hui Liang
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
| |
Collapse
|
9
|
Ugodnikov A, Persson H, Simmons CA. Bridging barriers: advances and challenges in modeling biological barriers and measuring barrier integrity in organ-on-chip systems. LAB ON A CHIP 2024; 24:3199-3225. [PMID: 38689569 DOI: 10.1039/d3lc01027a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Biological barriers such as the blood-brain barrier, skin, and intestinal mucosal barrier play key roles in homeostasis, disease physiology, and drug delivery - as such, it is important to create representative in vitro models to improve understanding of barrier biology and serve as tools for therapeutic development. Microfluidic cell culture and organ-on-a-chip (OOC) systems enable barrier modelling with greater physiological fidelity than conventional platforms by mimicking key environmental aspects such as fluid shear, accurate microscale dimensions, mechanical cues, extracellular matrix, and geometrically defined co-culture. As the prevalence of barrier-on-chip models increases, so does the importance of tools that can accurately assess barrier integrity and function without disturbing the carefully engineered microenvironment. In this review, we first provide a background on biological barriers and the physiological features that are emulated through in vitro barrier models. Then, we outline molecular permeability and electrical sensing barrier integrity assessment methods, and the related challenges specific to barrier-on-chip implementation. Finally, we discuss future directions in the field, as well important priorities to consider such as fabrication costs, standardization, and bridging gaps between disciplines and stakeholders.
Collapse
Affiliation(s)
- Alisa Ugodnikov
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Henrik Persson
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Craig A Simmons
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| |
Collapse
|
10
|
Roghani AK, Garcia RI, Roghani A, Reddy A, Khemka S, Reddy RP, Pattoor V, Jacob M, Reddy PH, Sehar U. Treating Alzheimer's disease using nanoparticle-mediated drug delivery strategies/systems. Ageing Res Rev 2024; 97:102291. [PMID: 38614367 DOI: 10.1016/j.arr.2024.102291] [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: 10/30/2023] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/15/2024]
Abstract
The administration of promising medications for the treatment of neurodegenerative disorders (NDDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) is significantly hampered by the blood-brain barrier (BBB). Nanotechnology has recently come to light as a viable strategy for overcoming this obstacle and improving drug delivery to the brain. With a focus on current developments and prospects, this review article examines the use of nanoparticles to overcome the BBB constraints to improve drug therapy for AD The potential for several nanoparticle-based approaches, such as those utilizing lipid-based, polymeric, and inorganic nanoparticles, to enhance drug transport across the BBB are highlighted. To shed insight on their involvement in aiding effective drug transport to the brain, methods of nanoparticle-mediated drug delivery, such as surface modifications, functionalization, and particular targeting ligands, are also investigated. The article also discusses the most recent findings on innovative medication formulations encapsulated within nanoparticles and the therapeutic effects they have shown in both preclinical and clinical testing. This sector has difficulties and restrictions, such as the need for increased safety, scalability, and translation to clinical applications. However, the major emphasis of this review aims to provide insight and contribute to the knowledge of how nanotechnology can potentially revolutionize the worldwide treatment of NDDs, particularly AD, to enhance clinical outcomes.
Collapse
Affiliation(s)
- Aryan Kia Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Frenship High School, Lubbock, TX 79382, USA.
| | - Ricardo Isaiah Garcia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ali Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Aananya Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Lubbock High School, Lubbock, TX 79401, USA.
| | - Sachi Khemka
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ruhananhad P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Lubbock High School, Lubbock, TX 79401, USA.
| | - Vasanthkumar Pattoor
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; University of South Florida, Tampa, FL 33620, USA.
| | - Michael Jacob
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Services, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
11
|
Kim NI, Chen J, Wang W, Kim JY, Kwon MK, Moradnia M, Pouladi S, Ryou JH. Skin-Attached Arrayed Piezoelectric Sensors for Continuous and Safe Monitoring of Oculomotor Movements. Adv Healthc Mater 2024; 13:e2303581. [PMID: 38386698 DOI: 10.1002/adhm.202303581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/08/2024] [Indexed: 02/24/2024]
Abstract
Abnormal oculomotor movements are known to be linked to various types of brain disorders, physical/mental shocks to the brain, and other neurological disorders, hence its monitoring can be developed into a simple but effective diagnostic tool. To overcome the limitations in the current eye-tracking system and electrooculography, a piezoelectric arrayed sensor system is developed using single-crystalline III-N thin-film transducers, which offers advantages of mechanical flexibility, biocompatibility, and high electromechanical conversion, for continuous monitoring of oculomotor movements by skin-attachable, safe, and highly sensitive sensors. The flexible piezoelectric eye movement sensor array (F-PEMSA), consisting of three transducers, is attached to the face temple area where it can be comfortably wearable and can detect the muscles' activity associated with the eye motions. Output voltages from upper, mid, and lower sensors (transducers) on different temple areas generate discernable patterns of output voltage signals with different combinations of positive/negative signs and their relative magnitudes for the various movements of eyeballs including 8 directional (lateral, vertical, and diagonal) and two rotational movements, which enable various types of saccade and pursuit tests. The F-PEMSA can be used in clinical studies on the brain-eye relationship to evaluate the functional integrity of multiple brain systems and cognitive processes.
Collapse
Affiliation(s)
- Nam-In Kim
- Department of Mechanical Engineering, University of Houston, Houston, TX, 77204-2004, USA
- Materials Science and Engineering Program, University of Houston, Houston, TX, 77204, USA
- Advanced Manufacturing Institute (AMI), University of Houston, Houston, TX, 77204, USA
- Texas Center for Superconductivity at UH (TcSUH), University of Houston, Houston, TX, 77204, USA
| | - Jie Chen
- Department of Mechanical Engineering, University of Houston, Houston, TX, 77204-2004, USA
- Materials Science and Engineering Program, University of Houston, Houston, TX, 77204, USA
- Advanced Manufacturing Institute (AMI), University of Houston, Houston, TX, 77204, USA
- Texas Center for Superconductivity at UH (TcSUH), University of Houston, Houston, TX, 77204, USA
| | - Weijie Wang
- Department of Mechanical Engineering, University of Houston, Houston, TX, 77204-2004, USA
- Advanced Manufacturing Institute (AMI), University of Houston, Houston, TX, 77204, USA
- Texas Center for Superconductivity at UH (TcSUH), University of Houston, Houston, TX, 77204, USA
| | - Ja-Yeon Kim
- Korea Photonics Technology Institute (KOPTI), Gwangju, 61007, Republic of Korea
| | - Min-Ki Kwon
- Department of Photonic Engineering, Chosun University, Gwangju, 61452, Republic of Korea
| | - Mina Moradnia
- Department of Mechanical Engineering, University of Houston, Houston, TX, 77204-2004, USA
- Advanced Manufacturing Institute (AMI), University of Houston, Houston, TX, 77204, USA
- Texas Center for Superconductivity at UH (TcSUH), University of Houston, Houston, TX, 77204, USA
| | - Sara Pouladi
- Department of Mechanical Engineering, University of Houston, Houston, TX, 77204-2004, USA
- Advanced Manufacturing Institute (AMI), University of Houston, Houston, TX, 77204, USA
- Texas Center for Superconductivity at UH (TcSUH), University of Houston, Houston, TX, 77204, USA
| | - Jae-Hyun Ryou
- Department of Mechanical Engineering, University of Houston, Houston, TX, 77204-2004, USA
- Materials Science and Engineering Program, University of Houston, Houston, TX, 77204, USA
- Advanced Manufacturing Institute (AMI), University of Houston, Houston, TX, 77204, USA
- Texas Center for Superconductivity at UH (TcSUH), University of Houston, Houston, TX, 77204, USA
- Department of Electrical & Computer Engineering, University of Houston, Houston, TX, 77204, USA
| |
Collapse
|
12
|
Liang Y, Yoon JY. Sensors for blood brain barrier on a chip. VITAMINS AND HORMONES 2024; 126:219-240. [PMID: 39029974 DOI: 10.1016/bs.vh.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
The blood-brain barrier (BBB) is a highly selective membrane that regulates the passage of substances between the bloodstream and the brain, thus safeguarding the central nervous system. This chapter provides an overview of current experimental models and detection methods utilized to study the BBB, along with the implementation of sensors and biosensors in BBB research. We discuss static and dynamic BBB models, highlighting their respective advantages and limitations. Additionally, we examine various detection methods employed in BBB research, including those specific to static and dynamic models. Furthermore, we explore the applications of physical sensors and biosensors in BBB models, focusing on their roles in monitoring barrier integrity and function. We also discuss recent advancements in sensor integration, such as robotic interrogators and integrated electrochemical and optical biosensors. Finally, we present a brief conclusion and future outlook, emphasizing the importance of continued innovation in BBB research to advance our understanding of neurological disorders and drug development.
Collapse
Affiliation(s)
- Yan Liang
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, United States
| | - Jeong-Yeol Yoon
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, United States; Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
13
|
Acharya P, Shrestha S, Joshi P, Choi NY, Lekkala VKR, Kang SY, Ni G, Lee MY. Dynamic culture of cerebral organoids using a pillar/perfusion plate for the assessment of developmental neurotoxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584506. [PMID: 38559002 PMCID: PMC10979904 DOI: 10.1101/2024.03.11.584506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite the potential toxicity of commercial chemicals to the development of the nervous system (known as developmental neurotoxicity or DNT), conventional in vitro cell models have primarily been employed for the assessment of acute neuronal toxicity. On the other hand, animal models used for the assessment of DNT are not physiologically relevant due to the heterogenic difference between humans and animals. In addition, animal models are low-throughput, time-consuming, expensive, and ethically questionable. Recently, human brain organoids have emerged as a promising alternative to assess the detrimental effects of chemicals on the developing brain. However, conventional organoid culture systems have several technical limitations including low throughput, lack of reproducibility, insufficient maturity of organoids, and the formation of the necrotic core due to limited diffusion of nutrients and oxygen. To address these issues and establish predictive DNT models, cerebral organoids were differentiated in a dynamic condition in a unique pillar/perfusion plate, which were exposed to test compounds to evaluate DNT potential. The pillar/perfusion plate facilitated uniform, dynamic culture of cerebral organoids with improved proliferation and maturity by rapid, bidirectional flow generated on a digital rocker. Day 9 cerebral organoids in the pillar/perfusion plate were exposed to ascorbic acid (DNT negative) and methylmercury (DNT positive) in a dynamic condition for 1 and 3 weeks, and changes in organoid morphology and neural gene expression were measured to determine DNT potential. As expected, ascorbic acid didn't induce any changes in organoid morphology and neural gene expression. However, exposure of day 9 cerebral organoids to methylmercury resulted in significant changes in organoid morphology and neural gene expression. Interestingly, methylmercury did not induce adverse changes in cerebral organoids in a static condition, thus highlighting the importance of dynamic organoid culture in DNT assessment.
Collapse
Affiliation(s)
- Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | | | - Na Young Choi
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | | | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Gabriel Ni
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
- Bioprinting Laboratories Inc., Dallas, Texas
| |
Collapse
|
14
|
Li M, Sun H, Hou Z, Hao S, Jin L, Wang B. Engineering the Physical Microenvironment into Neural Organoids for Neurogenesis and Neurodevelopment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306451. [PMID: 37771182 DOI: 10.1002/smll.202306451] [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: 07/29/2023] [Revised: 09/04/2023] [Indexed: 09/30/2023]
Abstract
Understanding the signals from the physical microenvironment is critical for deciphering the processes of neurogenesis and neurodevelopment. The discovery of how surrounding physical signals shape human developing neurons is hindered by the bottleneck of conventional cell culture and animal models. Notwithstanding neural organoids provide a promising platform for recapitulating human neurogenesis and neurodevelopment, building neuronal physical microenvironment that accurately mimics the native neurophysical features is largely ignored in current organoid technologies. Here, it is discussed how the physical microenvironment modulates critical events during the periods of neurogenesis and neurodevelopment, such as neural stem cell fates, neural tube closure, neuronal migration, axonal guidance, optic cup formation, and cortical folding. Although animal models are widely used to investigate the impacts of physical factors on neurodevelopment and neuropathy, the important roles of human stem cell-derived neural organoids in this field are particularly highlighted. Considering the great promise of human organoids, building neural organoid microenvironments with mechanical forces, electrophysiological microsystems, and light manipulation will help to fully understand the physical cues in neurodevelopmental processes. Neural organoids combined with cutting-edge techniques, such as advanced atomic force microscopes, microrobots, and structural color biomaterials might promote the development of neural organoid-based research and neuroscience.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Heng Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Zongkun Hou
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| |
Collapse
|
15
|
Tian M, Ma Z, Yang GZ. Micro/nanosystems for controllable drug delivery to the brain. Innovation (N Y) 2024; 5:100548. [PMID: 38161522 PMCID: PMC10757293 DOI: 10.1016/j.xinn.2023.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024] Open
Abstract
Drug delivery to the brain is crucial in the treatment for central nervous system disorders. While significant progress has been made in recent years, there are still major challenges in achieving controllable drug delivery to the brain. Unmet clinical needs arise from various factors, including controlled drug transport, handling large drug doses, methods for crossing biological barriers, the use of imaging guidance, and effective models for analyzing drug delivery. Recent advances in micro/nanosystems have shown promise in addressing some of these challenges. These include the utilization of microfluidic platforms to test and validate the drug delivery process in a controlled and biomimetic setting, the development of novel micro/nanocarriers for large drug loads across the blood-brain barrier, and the implementation of micro-intervention systems for delivering drugs through intraparenchymal or peripheral routes. In this article, we present a review of the latest developments in micro/nanosystems for controllable drug delivery to the brain. We also delve into the relevant diseases, biological barriers, and conventional methods. In addition, we discuss future prospects and the development of emerging robotic micro/nanosystems equipped with directed transportation, real-time image guidance, and closed-loop control.
Collapse
Affiliation(s)
- Mingzhen Tian
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang-Zhong Yang
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
16
|
Wang Y, Guo Z, Li J, Sui F, Dai W, Zhang W, Du H. Unraveling the differential perturbations of species-level functional profiling of gut microbiota among phases of methamphetamine-induced conditioned place preference. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110828. [PMID: 37459963 DOI: 10.1016/j.pnpbp.2023.110828] [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: 05/01/2023] [Revised: 06/27/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
The gut microbiome plays a significant role in methamphetamine addiction. Previous studies using short-read amplicon sequencing have described alterations in microbiota at the genus level and predicted function, in which taxonomic resolution is insufficient for accurate functional measurements. To address this limitation, we employed metagenome sequencing to intuitively associate species to functions of gut microbiota in methamphetamine-induced conditioned place preference. We observed differential perturbations of species-level functional profiling of the gut microbiota across phases of METH-induced CPP, with alterations in SCFA metabolism and bacterial motility at the acquisition phase and substance dependence-alcoholism pathway and amino acid metabolism at the extinction phase. Our findings suggest that reduced beneficial bacteria, i.e., Lactobacillus reuteri, contributed to the alteration of SCFA metabolism, while the increased abundance of Akkermansia muciniphila during the extinction phase may be associated with altered phenylalanine, tyrosine, and tryptophan metabolism and substance dependence pathway. Our study further supports the association between specific microbial taxa and METH-induced rewarding.
Collapse
Affiliation(s)
- Yinan Wang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China; School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zhonghao Guo
- School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jing Li
- University of Science and Technology of China, Anhui, China
| | - Fang Sui
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenkui Dai
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wenyong Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Hui Du
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
17
|
Choi JH, Choi HK, Lee KB. In Situ Detection of Neuroinflammation using Multi-cellular 3D Neurovascular Unit-on-a-Chip. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2304382. [PMID: 39308874 PMCID: PMC11412436 DOI: 10.1002/adfm.202304382] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 09/25/2024]
Abstract
The human neurovascular system is a complex network of blood vessels and brain cells that is essential to the proper functioning of the brain. In recent years, researchers have become increasingly interested in the role of this system in developing drugs to treat neuroinflammation. This process is believed to contribute to the development of several neurodegenerative diseases, including Alzheimer's and Parkinson's diseases. While much remains to be learned about the precise mechanisms by which the neurovascular system interacts with the brain and how it can be targeted for therapeutic purposes, this area of research holds great promise for the future of neurology and medicine. Currently, creating neurovascular models begins with animal models, followed by testing on humans in clinical trials. However, the high number of medication failures that pass through animal testing indicates that animal models do not always reflect the outcome of human clinical trials. To overcome the challenges of neurovascular systems and the issues with animal models, we have developed a one-of-a-kind in vitro neurovascular unit-on-a-chip to accurately replicate the in vivo human neurovascular microenvironment. This neuroinflammation-on-a-chip platform has the potential to enhance the current methods of drug development and testing to treat neurodegenerative diseases. By replicating the human neurovascular unit in vitro, a more accurate representation of human physiology can be achieved compared to animal models. The ability to detect pro-inflammatory cytokines in situ and monitor physiological changes, such as barrier function, in real-time can provide an invaluable tool for evaluating the efficacy and safety of drugs. Moreover, using nano-sized graphene oxide for in situ detection of inflammatory responses is an innovative approach that can advance the field of neuroinflammation research. Overall, our developed neuroinflammation-on-a-chip system has the potential to provide a more efficient and effective method for developing drugs for treating neurodegenerative diseases and other central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey,123 Bevier Road, Piscataway, NJ 08854, USA
- School of Chemical Engineering, Jeonbuk National University, Jeonju, 54896, Korea
| | - Hye Kyu Choi
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey,123 Bevier Road, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey,123 Bevier Road, Piscataway, NJ 08854, USA
| |
Collapse
|
18
|
Pereira I, Lopez-Martinez MJ, Samitier J. Advances in current in vitro models on neurodegenerative diseases. Front Bioeng Biotechnol 2023; 11:1260397. [PMID: 38026882 PMCID: PMC10658011 DOI: 10.3389/fbioe.2023.1260397] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.
Collapse
Affiliation(s)
- Inês Pereira
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria J. Lopez-Martinez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Geng X, Zhou ZA, Mi Y, Wang C, Wang M, Guo C, Qu C, Feng S, Kim I, Yu M, Ji H, Ren X. Glioma Single-Cell Biomechanical Analysis by Cyclic Conical Constricted Microfluidics. Anal Chem 2023; 95:15585-15594. [PMID: 37843131 DOI: 10.1021/acs.analchem.3c02434] [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: 10/17/2023]
Abstract
Determining the grade of glioma is a critical step in choosing patients' treatment plans in clinical practices. The pathological diagnosis of patient's glioma samples requires extensive staining and imaging procedures, which are expensive and time-consuming. Current advanced uniform-width-constriction-channel-based microfluidics have proven to be effective in distinguishing cancer cells from normal tissues, such as breast cancer, ovarian cancer, prostate cancer, etc. However, the uniform-width-constriction channels can result in low yields on glioma cells with irregular morphologies and high heterogeneity. In this research, we presented an innovative cyclic conical constricted (CCC) microfluidic device to better differentiate glioma cells from normal glial cells. Compared with the widely used uniform-width-constriction microchannels, the new CCC configuration forces single cells to deform gradually and obtains the biophysical attributes from each deformation. The human-derived glioma cell lines U-87 and U-251, as well as the human-derived normal glial astrocyte cell line HA-1800 were selected as the proof of concept. The results showed that CCC channels can effectively obtain the biomechanical characteristics of different 12-25 μm glial cell lines. The patient glioma samples with WHO grades II, III, and IV were tested by CCC channels and compared between Elastic Net (ENet) and Lasso analysis. The results demonstrated that CCC channels and the ENet can successfully select critical biomechanical parameters to differentiate the grades of single-glioma cells. This CCC device can be potentially further applied to the extensive family of brain tumors at the single-cell level.
Collapse
Affiliation(s)
- Xin Geng
- Department of Neurosurgery, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Zi-Ang Zhou
- Department of Microelectronics, Tianjin University, Tianjin 300072, China
| | - Yang Mi
- Department of Neurosurgery, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Chunhong Wang
- Department of Neurosurgery, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Meng Wang
- Department of Neurosurgery, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Chenjia Guo
- Department of Pathology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Chongxiao Qu
- Department of Pathology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Shilun Feng
- Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Inyoung Kim
- Department of Statistics, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Miao Yu
- Department of Research and Development, Stedical Scientific, Carlsbad, California 92010, United States
| | - Hongming Ji
- Department of Neurosurgery, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Xiang Ren
- Department of Neurosurgery, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030012, China
- Department of Microelectronics, Tianjin University, Tianjin 300072, China
| |
Collapse
|
20
|
Graybill PM, Jacobs EJ, Jana A, Agashe A, Nain AS, Davalos RV. Ultra-thin and ultra-porous nanofiber networks as a basement-membrane mimic. LAB ON A CHIP 2023; 23:4565-4578. [PMID: 37772328 PMCID: PMC10623910 DOI: 10.1039/d3lc00304c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Current basement membrane (BM) mimics used for modeling endothelial and epithelial barriers in vitro do not faithfully recapitulate key in vivo physiological properties such as BM thickness, porosity, stiffness, and fibrous composition. Here, we use networks of precisely arranged nanofibers to form ultra-thin (∼3 μm thick) and ultra-porous (∼90%) BM mimics for blood-brain barrier modeling. We show that these nanofiber networks enable close contact between endothelial monolayers and pericytes across the membrane, which are known to regulate barrier tightness. Cytoskeletal staining and transendothelial electrical resistance (TEER) measurements reveal barrier formation on nanofiber membranes integrated within microfluidic devices and transwell inserts. Further, significantly higher TEER values indicate a biological benefit for co-cultures formed on the ultra-thin nanofiber membranes. Our BM mimic overcomes critical technological challenges in forming co-cultures that are in proximity and facilitate cell-cell contact, while still being constrained to their respective sides. We anticipate that our nanofiber networks will find applications in drug discovery, cell migration, and barrier dysfunction studies.
Collapse
Affiliation(s)
- Philip M Graybill
- Bioelectromechanical Systems Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| | - Edward J Jacobs
- Bioelectromechanical Systems Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| | - Aniket Jana
- Spinneret-Based Tunable Engineering Parameters (STEP) Lab, Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, USA.
| | - Atharva Agashe
- Spinneret-Based Tunable Engineering Parameters (STEP) Lab, Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, USA.
| | - Amrinder S Nain
- Spinneret-Based Tunable Engineering Parameters (STEP) Lab, Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, USA.
| | - Rafael V Davalos
- Bioelectromechanical Systems Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
21
|
Perxés Perich M, Palma-Florez S, Solé C, Goberna-Ferrón S, Samitier J, Gómez-Romero P, Mir M, Lagunas A. Polyoxometalate-Decorated Gold Nanoparticles Inhibit β-Amyloid Aggregation and Cross the Blood-Brain Barrier in a µphysiological Model. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2697. [PMID: 37836338 PMCID: PMC10574493 DOI: 10.3390/nano13192697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023]
Abstract
Alzheimer's disease is characterized by a combination of several neuropathological hallmarks, such as extracellular aggregates of beta amyloid (Aβ). Numerous alternatives have been studied for inhibiting Aβ aggregation but, at this time, there are no effective treatments available. Here, we developed the tri-component nanohybrid system AuNPs@POM@PEG based on gold nanoparticles (AuNPs) covered with polyoxometalates (POMs) and polyethylene glycol (PEG). In this work, AuNPs@POM@PEG demonstrated the inhibition of the formation of amyloid fibrils, showing a 75% decrease in Aβ aggregation in vitro. As it is a potential candidate for the treatment of Alzheimer's disease, we evaluated the cytotoxicity of AuNPs@POM@PEG and its ability to cross the blood-brain barrier (BBB). We achieved a stable nanosystem that is non-cytotoxic below 2.5 nM to human neurovascular cells. The brain permeability of AuNPs@POM@PEG was analyzed in an in vitro microphysiological model of the BBB (BBB-on-a-chip), containing 3D human neurovascular cell co-cultures and microfluidics. The results show that AuNPs@POM@PEG was able to cross the brain endothelial barrier in the chip and demonstrated that POM does not affect the barrier integrity, giving the green light to further studies into this system as a nanotherapeutic.
Collapse
Affiliation(s)
- Marta Perxés Perich
- Catalan Institute of Nanoscience and Nanotechnology(ICN2) CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Materials Chemistry and Catalysis, Debye Institute for Nanomaterials Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Sujey Palma-Florez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Clara Solé
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Sara Goberna-Ferrón
- Catalan Institute of Nanoscience and Nanotechnology(ICN2) CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Instituto Universitario de Tecnología Química (CSIC-UPV), Universitat Politècnica de València, Avda. De los Naranjos s/n, 46022 Valencia, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Pedro Gómez-Romero
- Catalan Institute of Nanoscience and Nanotechnology(ICN2) CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Mònica Mir
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Anna Lagunas
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
| |
Collapse
|
22
|
Kawakita S, Li S, Nguyen HT, Maity S, Haghniaz R, Bahari J, Yu N, Mandal K, Bandaru P, Mou L, Ermis M, Khalil E, Khosravi S, Peirsman A, Nasiri R, Adachi A, Nakayama A, Bell R, Zhu Y, Jucaud V, Dokmeci MR, Khademhosseini A. Rapid integration of screen-printed electrodes into thermoplastic organ-on-a-chip devices for real-time monitoring of trans-endothelial electrical resistance. Biomed Microdevices 2023; 25:37. [PMID: 37740819 DOI: 10.1007/s10544-023-00669-9] [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: 07/24/2023] [Indexed: 09/25/2023]
Abstract
Trans-endothelial electrical resistance (TEER) is one of the most widely used indicators to quantify the barrier integrity of endothelial layers. Over the last decade, the integration of TEER sensors into organ-on-a-chip (OOC) platforms has gained increasing interest for its efficient and effective measurement of TEER in OOCs. To date, microfabricated electrodes or direct insertion of wires has been used to integrate TEER sensors into OOCs, with each method having advantages and disadvantages. In this study, we developed a TEER-SPE chip consisting of carbon-based screen-printed electrodes (SPEs) embedded in a poly(methyl methacrylate) (PMMA)-based multi-layered microfluidic device with a porous poly(ethylene terephthalate) membrane in-between. As proof of concept, we demonstrated the successful cultures of hCMEC/D3 cells and the formation of confluent monolayers in the TEER-SPE chip and obtained TEER measurements for 4 days. Additionally, the TEER-SPE chip could detect changes in the barrier integrity due to shear stress or an inflammatory cytokine (i.e., tumor necrosis factor-α). The novel approach enables a low-cost and facile fabrication of carbon-based SPEs on PMMA substrates and the subsequent assembly of PMMA layers for rapid prototyping. Being cost-effective and cleanroom-free, our method lowers the existing logistical and technical barriers presenting itself as another step forward to the broader adoption of OOCs with TEER measurement capability.
Collapse
Affiliation(s)
- Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Jamal Bahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Ning Yu
- Department of Chemical and Environmental Engineering, University of California-Riverside, Riverside, California, 92521, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Praveen Bandaru
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Lei Mou
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Enam Khalil
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, V6T1Z4, Canada
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Plastic, Aesthetic & Reconstructive Surgery and Laboratory of Experimental Cancer Research, Ghent University, 9000, Ghent, Belgium
| | - Rohollah Nasiri
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, 17165, Solna, Sweden
| | - Annie Adachi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Cardiovascular Research Institute, University of California, San Francisco, California, 94158, USA
| | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, 91125, USA
| | - Remy Bell
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, 29208, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| |
Collapse
|
23
|
Floryanzia SD, Nance E. Applications and Considerations for Microfluidic Systems To Model the Blood-Brain Barrier. ACS APPLIED BIO MATERIALS 2023; 6:3617-3632. [PMID: 37582179 DOI: 10.1021/acsabm.3c00364] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
In a myriad of developmental and degenerative brain diseases, characteristic pathological biomarkers are often associated with cerebral blood flow and vasculature. However, the relationship between vascular dysfunction and markers of brain disease is not well-defined. Additionally, it is difficult to deliver effective therapeutics to the brain due to the highly regulated blood-brain barrier (BBB) at the microvasculature interface of the brain. This Review first covers the need for modeling the BBB and the challenges of modeling the BBB. In vitro models of the BBB enable the study of the relationship between vascular dysfunction, BBB function, and disease progression and can serve as a platform to screen therapeutics. In particular, microfluidic-based in vitro BBB models are useful for studying brain vasculature as they support cell culture within the presence of continuous perfusion, which mirrors the in vivo flow and associated stress conditions in the brain. Early microfluidic models of the BBB created the most simplistic models possible that still displayed some functional aspects of the in vivo BBB. Therefore, this Review also discusses the emerging unique ways in which microfluidics in tandem with recent advancements in cell culture, biomaterials, and in vitro modeling can be used to develop more complex and physiologically relevant models of the BBB. Finally, we discuss the current and future state-of-the-art application of microfluidic BBB models for drug development and disease modeling, and the ongoing areas of needed innovation in this field.
Collapse
Affiliation(s)
- Sydney D Floryanzia
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
24
|
Kim J, Kim J, Jin Y, Cho SW. In situbiosensing technologies for an organ-on-a-chip. Biofabrication 2023; 15:042002. [PMID: 37587753 DOI: 10.1088/1758-5090/aceaae] [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: 12/19/2022] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Thein vitrosimulation of organs resolves the accuracy, ethical, and cost challenges accompanyingin vivoexperiments. Organoids and organs-on-chips have been developed to model thein vitro, real-time biological and physiological features of organs. Numerous studies have deployed these systems to assess thein vitro, real-time responses of an organ to external stimuli. Particularly, organs-on-chips can be most efficiently employed in pharmaceutical drug development to predict the responses of organs before approving such drugs. Furthermore, multi-organ-on-a-chip systems facilitate the close representations of thein vivoenvironment. In this review, we discuss the biosensing technology that facilitates thein situ, real-time measurements of organ responses as readouts on organ-on-a-chip systems, including multi-organ models. Notably, a human-on-a-chip system integrated with automated multi-sensing will be established by further advancing the development of chips, as well as their assessment techniques.
Collapse
Affiliation(s)
- Jinyoung Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Junghoon Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yoonhee Jin
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Institute for Basic Science (IBS), Center for Nanomedicine, Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
25
|
Brandl S, Reindl M. Blood-Brain Barrier Breakdown in Neuroinflammation: Current In Vitro Models. Int J Mol Sci 2023; 24:12699. [PMID: 37628879 PMCID: PMC10454051 DOI: 10.3390/ijms241612699] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The blood-brain barrier, which is formed by tightly interconnected microvascular endothelial cells, separates the brain from the peripheral circulation. Together with other central nervous system-resident cell types, including pericytes and astrocytes, the blood-brain barrier forms the neurovascular unit. Upon neuroinflammation, this barrier becomes leaky, allowing molecules and cells to enter the brain and to potentially harm the tissue of the central nervous system. Despite the significance of animal models in research, they may not always adequately reflect human pathophysiology. Therefore, human models are needed. This review will provide an overview of the blood-brain barrier in terms of both health and disease. It will describe all key elements of the in vitro models and will explore how different compositions can be utilized to effectively model a variety of neuroinflammatory conditions. Furthermore, it will explore the existing types of models that are used in basic research to study the respective pathologies thus far.
Collapse
Affiliation(s)
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
26
|
Cerneckis J, Bu G, Shi Y. Pushing the boundaries of brain organoids to study Alzheimer's disease. Trends Mol Med 2023; 29:659-672. [PMID: 37353408 PMCID: PMC10374393 DOI: 10.1016/j.molmed.2023.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 06/25/2023]
Abstract
Progression of Alzheimer's disease (AD) entails deterioration or aberrant function of multiple brain cell types, eventually leading to neurodegeneration and cognitive decline. Defining how complex cell-cell interactions become dysregulated in AD requires novel human cell-based in vitro platforms that could recapitulate the intricate cytoarchitecture and cell diversity of the human brain. Brain organoids (BOs) are 3D self-organizing tissues that partially resemble the human brain architecture and can recapitulate AD-relevant pathology. In this review, we highlight the versatile applications of different types of BOs to model AD pathogenesis, including amyloid-β and tau aggregation, neuroinflammation, myelin breakdown, vascular dysfunction, and other phenotypes, as well as to accelerate therapeutic development for AD.
Collapse
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Guojun Bu
- SciNeuro Pharmaceuticals, Rockville, MD 20850, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
27
|
Chavarria D, Abbaspour A, Celestino N, Shah N, Sankar S, Baker AB. A high throughput blood-brain barrier model incorporating shear stress with improved predictive power for drug discovery. BIOMICROFLUIDICS 2023; 17:044105. [PMID: 37614679 PMCID: PMC10444201 DOI: 10.1063/5.0150887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/01/2023] [Indexed: 08/25/2023]
Abstract
The blood-brain barrier is a key structure regulating the health of the brain and access of drugs and pathogens to neural tissue. Shear stress is a key regulator of the blood-brain barrier; however, the commonly used multi-well vitro models of the blood-brain barrier do not incorporate shear stress. In this work, we designed and validated a high-throughput system for simulating the blood-brain barrier that incorporates physiological flow and incorporates an optimized cellular model of the blood-brain barrier. This system can perform assays of blood-brain barrier function with shear stress, with 48 independent assays simultaneously. Using the high throughput assay, we conducted drug screening assays to explore the effects of compounds for opening or closing blood-brain barrier. Our studies revealed that assays with shear stress were more predictive and were able to identify compounds known to modify the blood-brain barrier function while static assays were not. Overall, we demonstrate an optimized, high throughput assay for simulating the blood-brain barrier that incorporates shear stress and is practical for use in drug screening and other high throughput studies of toxicology.
Collapse
Affiliation(s)
- Daniel Chavarria
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Ali Abbaspour
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Natalie Celestino
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Nehali Shah
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | | | - Aaron B. Baker
- Author to whom correspondence should be addressed:. Tel.:+512-232-7114
| |
Collapse
|
28
|
Li W, Zhou Z, Zhou X, Khoo BL, Gunawan R, Chin YR, Zhang L, Yi C, Guan X, Yang M. 3D Biomimetic Models to Reconstitute Tumor Microenvironment In Vitro: Spheroids, Organoids, and Tumor-on-a-Chip. Adv Healthc Mater 2023; 12:e2202609. [PMID: 36917657 PMCID: PMC11468819 DOI: 10.1002/adhm.202202609] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/22/2023] [Indexed: 03/16/2023]
Abstract
Decades of efforts in engineering in vitro cancer models have advanced drug discovery and the insight into cancer biology. However, the establishment of preclinical models that enable fully recapitulating the tumor microenvironment remains challenging owing to its intrinsic complexity. Recent progress in engineering techniques has allowed the development of a new generation of in vitro preclinical models that can recreate complex in vivo tumor microenvironments and accurately predict drug responses, including spheroids, organoids, and tumor-on-a-chip. These biomimetic 3D tumor models are of particular interest as they pave the way for better understanding of cancer biology and accelerating the development of new anticancer therapeutics with reducing animal use. Here, the recent advances in developing these in vitro platforms for cancer modeling and preclinical drug screening, focusing on incorporating hydrogels are reviewed to reconstitute physiologically relevant microenvironments. The combination of spheroids/organoids with microfluidic technologies is also highlighted to better mimic in vivo tumors and discuss the challenges and future directions in the clinical translation of such models for drug screening and personalized medicine.
Collapse
Affiliation(s)
- Wenxiu Li
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000China
- Department of Biomedical SciencesTung Biomedical Sciences CentreCity University of Hong KongHong KongSAR999077China
| | - Zhihang Zhou
- Department of Biomedical SciencesTung Biomedical Sciences CentreCity University of Hong KongHong KongSAR999077China
- Department of Gastroenterologythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000China
- Department of Biomedical SciencesTung Biomedical Sciences CentreCity University of Hong KongHong KongSAR999077China
| | - Bee Luan Khoo
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000China
- Department of Biomedical EngineeringCity University of Hong KongHong Kong999077China
| | - Renardi Gunawan
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000China
- Department of Biomedical SciencesTung Biomedical Sciences CentreCity University of Hong KongHong KongSAR999077China
| | - Y. Rebecca Chin
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000China
- Department of Biomedical SciencesTung Biomedical Sciences CentreCity University of Hong KongHong KongSAR999077China
| | - Liang Zhang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000China
- Department of Biomedical SciencesTung Biomedical Sciences CentreCity University of Hong KongHong KongSAR999077China
| | - Changqing Yi
- Guangdong Provincial Engineering and Technology Center of Advanced and Portable Medical DevicesSchool of Biomedical EngineeringSun Yat‐sen UniversityGuangzhou518107China
| | - Xinyuan Guan
- Department of Clinical OncologyState Key Laboratory for Liver ResearchThe University of Hong KongHong KongSAR999077China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000China
- Department of Biomedical SciencesTung Biomedical Sciences CentreCity University of Hong KongHong KongSAR999077China
| |
Collapse
|
29
|
Garcia L, Palma-Florez S, Espinosa V, Soleimani Rokni F, Lagunas A, Mir M, García-Celma MJ, Samitier J, Rodríguez-Abreu C, Grijalvo S. Ferulic acid-loaded polymeric nanoparticles prepared from nano-emulsion templates facilitate internalisation across the blood-brain barrier in model membranes. NANOSCALE 2023; 15:7929-7944. [PMID: 37067009 DOI: 10.1039/d2nr07256d] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
A hydroxycinnamic acid derivative, namely ferulic acid (FA) has been successfully encapsulated in polymeric nanoparticles (NPs) based on poly(lactic-co-glycolic acid) (PLGA). FA-loaded polymeric NPs were prepared from O/W nano-emulsion templates using the phase inversion composition (PIC) low-energy emulsification method. The obtained PLGA NPs exhibited high colloidal stability, good drug-loading capacity, and particle hydrodynamic diameters in the range of 74 to 117 nm, depending on the FA concentration used. In vitro drug release studies confirmed a diffusion-controlled mechanism through which the amount of released FA reached a plateau at 60% after 6 hours-incubation. Five kinetic models were used to fit the FA release data as a function of time. The Weibull distribution and Korsmeyer-Peppas equation models provided the best fit to our experimental data and suggested quasi-Fickian diffusion behaviour. Moderate dose-response antioxidant and radical scavenging activities of FA-loaded PLGA NPs were demonstrated using the DPPH˙ assay achieving inhibition activities close to 60 and 40%, respectively. Cell culture studies confirmed that FA-loaded NPs were not toxic according to the MTT colorimetric assay, were able to internalise efficiently SH-SY5Y neuronal cells and supressed the intracellular ROS-level induced by H2O2 leading to 52% and 24.7% of cellular viability at 0.082 and 0.041 mg mL-1, respectively. The permeability of the NPs through the blood brain barrier was tested with an in vitro organ-on-a-chip model to evaluate the ability of the FA-loaded PLGA and non-loaded PLGA NPs to penetrate to the brain. NPs were able to penetrate the barrier, but permeability decreased when FA was loaded. These results are promising for the use of loaded PLGA NPs for the management of neurological diseases.
Collapse
Affiliation(s)
- Luna Garcia
- IQAC, CSIC, Jordi Girona 18-26, E-08034-Barcelona, Spain.
| | - Sujey Palma-Florez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), E-08028-Barcelona, Spain
- CIBER-BBN, ISCIII, Spain.
| | | | | | - Anna Lagunas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), E-08028-Barcelona, Spain
- CIBER-BBN, ISCIII, Spain.
| | - Mònica Mir
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), E-08028-Barcelona, Spain
- Department of Electronics and Biomedical engineering, University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
- CIBER-BBN, ISCIII, Spain.
| | - María José García-Celma
- Department of Pharmacy, Pharmaceutical Technology, and Physical-chemistry, IN2UB, R+D Associated Unit to CSIC, Pharmaceutical Nanotechnology, University of Barcelona, Joan XXIII 27-31, E-08028-Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), E-08028-Barcelona, Spain
- CIBER-BBN, ISCIII, Spain.
| | | | | |
Collapse
|
30
|
Nazari H, Shrestha J, Naei VY, Bazaz SR, Sabbagh M, Thiery JP, Warkiani ME. Advances in TEER measurements of biological barriers in microphysiological systems. Biosens Bioelectron 2023; 234:115355. [PMID: 37159988 DOI: 10.1016/j.bios.2023.115355] [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: 08/11/2022] [Revised: 03/10/2023] [Accepted: 04/25/2023] [Indexed: 05/11/2023]
Abstract
Biological barriers are multicellular structures that precisely regulate the transport of ions, biomolecules, drugs, cells, and other organisms. Transendothelial/epithelial electrical resistance (TEER) is a label-free method for predicting the properties of biological barriers. Understanding the mechanisms that control TEER significantly enhances our knowledge of the physiopathology of different diseases and aids in the development of new drugs. Measuring TEER values within microphysiological systems called organ-on-a-chip devices that simulate the microenvironment, architecture, and physiology of biological barriers in the body provides valuable insight into the behavior of barriers in response to different drugs and pathogens. These integrated systems should increase the accuracy, reproducibility, sensitivity, resolution, high throughput, speed, cost-effectiveness, and reliable predictability of TEER measurements. Implementing advanced micro and nanoscale manufacturing techniques, surface modification methods, biomaterials, biosensors, electronics, and stem cell biology is necessary for integrating TEER measuring systems with organ-on-chip technology. This review focuses on the applications, advantages, and future perspectives of integrating organ-on-a-chip technology with TEER measurement methods for studying biological barriers. After briefly reviewing the role of TEER in the physiology and pathology of barriers, standard techniques for measuring TEER, including Ohm's law and impedance spectroscopy, and commercially available devices are described. Furthermore, advances in TEER measurement are discussed in multiple barrier-on-a-chip system models representing different organs. Finally, we outline future trends in implementing advanced technologies to design and fabricate nanostructured electrodes, complicated microfluidic chips, and membranes for more advanced and accurate TEER measurements.
Collapse
Affiliation(s)
- Hojjatollah Nazari
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | - Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | - Vahid Yaghoubi Naei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | - Milad Sabbagh
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia
| | | | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, 2007, New South Wales, Australia; Institute of Molecular Medicine, Sechenov University, 119991, Moscow, Russia.
| |
Collapse
|
31
|
Qiao R, Fu C, Forgham H, Javed I, Huang X, Zhu J, Whittaker AK, Davis TP. Magnetic Iron Oxide Nanoparticles for Brain Imaging and Drug Delivery. Adv Drug Deliv Rev 2023; 197:114822. [PMID: 37086918 DOI: 10.1016/j.addr.2023.114822] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/14/2023] [Accepted: 04/09/2023] [Indexed: 04/24/2023]
Abstract
Central nervous system (CNS) disorders affect as many as 1.5 billion people globally. The limited delivery of most imaging and therapeutic agents into the brain is a major challenge for treatment of CNS disorders. With the advent of nanotechnologies, controlled delivery of drugs with nanoparticles holds great promise in CNS disorders for overcoming the blood-brain barrier (BBB) and improving delivery efficacy. In recent years, magnetic iron oxide nanoparticles (MIONPs) have stood out as a promising theranostic nanoplatform for brain imaging and drug delivery as they possess unique physical properties and biodegradable characteristics. In this review, we summarize the recent advances in MIONP-based platforms as imaging and drug delivery agents for brain diseases. We firstly introduce the methods of synthesis and surface functionalization of MIONPs with emphasis on the inclusion of biocompatible polymers that allow for the addition of tailored physicochemical properties. We then discuss the recent advances in in vivo imaging and drug delivery applications using MIONPs. Finally, we present a perspective on the remaining challenges and possible future directions for MIONP-based brain delivery systems.
Collapse
Affiliation(s)
- Ruirui Qiao
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Changkui Fu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Helen Forgham
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ibrahim Javed
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xumin Huang
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jiayuan Zhu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrew K Whittaker
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Thomas P Davis
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
32
|
Biosensor integrated brain-on-a-chip platforms: Progress and prospects in clinical translation. Biosens Bioelectron 2023; 225:115100. [PMID: 36709589 DOI: 10.1016/j.bios.2023.115100] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/07/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
Because of the brain's complexity, developing effective treatments for neurological disorders is a formidable challenge. Research efforts to this end are advancing as in vitro systems have reached the point that they can imitate critical components of the brain's structure and function. Brain-on-a-chip (BoC) was first used for microfluidics-based systems with small synthetic tissues but has expanded recently to include in vitro simulation of the central nervous system (CNS). Defining the system's qualifying parameters may improve the BoC for the next generation of in vitro platforms. These parameters show how well a given platform solves the problems unique to in vitro CNS modeling (like recreating the brain's microenvironment and including essential parts like the blood-brain barrier (BBB)) and how much more value it offers than traditional cell culture systems. This review provides an overview of the practical concerns of creating and deploying BoC systems and elaborates on how these technologies might be used. Not only how advanced biosensing technologies could be integrated with BoC system but also how novel approaches will automate assays and improve point-of-care (PoC) diagnostics and accurate quantitative analyses are discussed. Key challenges providing opportunities for clinical translation of BoC in neurodegenerative disorders are also addressed.
Collapse
|
33
|
Pérez-López A, Isabel Torres-Suárez A, Martín-Sabroso C, Aparicio-Blanco J. An overview of in vitro 3D models of the blood-brain barrier as a tool to predict the in vivo permeability of nanomedicines. Adv Drug Deliv Rev 2023; 196:114816. [PMID: 37003488 DOI: 10.1016/j.addr.2023.114816] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
The blood-brain barrier (BBB) prevents efficient drug delivery to the central nervous system. As a result, brain diseases remain one of the greatest unmet medical needs. Understanding the tridimensional structure of the BBB helps gain insight into the pathology of the BBB and contributes to the development of novel therapies for brain diseases. Therefore, 3D models with an ever-growing sophisticated complexity are being developed to closely mimic the human neurovascular unit. Among these 3D models, hydrogel-, spheroid- and organoid-based static BBB models have been developed, and so have microfluidic-based BBB-on-a-chip models. The different 3D preclinical models of the BBB, both in health and disease, are here reviewed, from their development to their application for permeability testing of nanomedicines across the BBB, discussing the advantages and disadvantages of each model. The validation with data from in vivo preclinical data is also discussed in those cases where provided.
Collapse
Affiliation(s)
- Alexandre Pérez-López
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Institute of Industrial Pharmacy, Complutense University of Madrid, Madrid, Spain.
| | - Cristina Martín-Sabroso
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Institute of Industrial Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Juan Aparicio-Blanco
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain; Institute of Industrial Pharmacy, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
34
|
Fan Y, Xu C, Deng N, Gao Z, Jiang Z, Li X, Zhou Y, Pei H, Li L, Tang B. Understanding drug nanocarrier and blood-brain barrier interaction based on a microfluidic microphysiological model. LAB ON A CHIP 2023; 23:1935-1944. [PMID: 36891748 DOI: 10.1039/d2lc01077a] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
As many nanoparticles (NPs) have been exploited as drug carriers to overcome the resistance of the blood-brain barrier (BBB), reliable in vitro BBB models are urgently needed to help researchers to comprehensively understand drug nanocarrier-BBB interaction during penetration, which can prompt pre-clinical nanodrug exploitation. Herein, we developed a microfluidic microphysiological model, allowing the analysis of BBB homeostasis and NP penetration. We found that the BBB penetrability of gold nanoparticles (AuNPs) was size- and modification-dependent, which might be caused by a distinct transendocytosis pathway. Notably, transferrin-modified 13 nm AuNPs held the strongest BBB penetrability and induced the slightest BBB dysfunction, while bare 80 nm and 120 nm AuNPs showed opposite results. Moreover, further analysis of the protein corona showed that PEGylation reduced the protein absorption, and some proteins facilitated the BBB penetration of NPs. The developed microphysiological model provides a powerful tool for understanding the drug nanocarrier-BBB interaction, which is vital for exploiting high-efficiency and biocompatible nanodrugs.
Collapse
Affiliation(s)
- Yuanyuan Fan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Chang Xu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Ning Deng
- Shandong Institute for Product Quality Inspection, Jinan 250101, P. R. China
| | - Ze Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Zhongyao Jiang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Xiaoxiao Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Yingshun Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Haimeng Pei
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Lu Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
35
|
Gonzales-Aloy E, Ahmed-Cox A, Tsoli M, Ziegler DS, Kavallaris M. From cells to organoids: The evolution of blood-brain barrier technology for modelling drug delivery in brain cancer. Adv Drug Deliv Rev 2023; 196:114777. [PMID: 36931346 DOI: 10.1016/j.addr.2023.114777] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/13/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Brain cancer remains the deadliest cancer. The blood-brain barrier (BBB) is impenetrable to most drugs and is a complex 3D network of multiple cell types including endothelial cells, astrocytes, and pericytes. In brain cancers, the BBB becomes disrupted during tumor progression and forms the blood-brain tumor barrier (BBTB). To advance therapeutic development, there is a critical need for physiologically relevant BBB in vitro models. 3D cell systems are emerging as valuable preclinical models to accelerate discoveries for diseases. Given the versatility and capability of 3D cell models, their potential for modelling the BBB and BBTB is reviewed. Technological advances of BBB models and challenges of in vitro modelling the BBTB, and application of these models as tools for assessing therapeutics and nano drug delivery, are discussed. Quantitative, in vitro BBB models that are predictive of effective brain cancer therapies will be invaluable for accelerating advancing new treatments to the clinic.
Collapse
Affiliation(s)
- Estrella Gonzales-Aloy
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia
| | - Aria Ahmed-Cox
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; Katharina Gaus Light Microscopy Facility, Mark Wainright Analytical Center, UNSW Sydney, NSW, Australia
| | - Maria Tsoli
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia
| | - David S Ziegler
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; Kids Cancer Center, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; UNSW RNA Institute, UNSW Sydney, NSW, Australia.
| |
Collapse
|
36
|
Su SH, Song Y, Stephens A, Situ M, McCloskey MC, McGrath JL, Andjelkovic AV, Singer BH, Kurabayashi K. A tissue chip with integrated digital immunosensors: In situ brain endothelial barrier cytokine secretion monitoring. Biosens Bioelectron 2023; 224:115030. [PMID: 36603283 PMCID: PMC10401069 DOI: 10.1016/j.bios.2022.115030] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
Organ-on-a-chip platforms have potential to offer more cost-effective, ethical, and human-resembling models than animal models for disease study and drug discovery. Particularly, the Blood-Brain-Barrier-on-a-chip (BBB-oC) has emerged as a promising tool to investigate several neurological disorders since it promises to provide a model of the multifunctional tissue working as an important node to control pathogen entry, drug delivery and neuroinflammation. A comprehensive understanding of the multiple physiological functions of the tissue model requires biosensors detecting several tissue-secreted substances in a BBB-oC system. However, current sensor-integrated BBB-oC platforms are only available for tissue membrane integrity characterization based on permeability measurement. Protein secretory pathways are closely associated with the tissue's various diseased conditions. At present, no biosensor-integrated BBB-oC platform exists that permits in situ tissue protein secretion analysis over time, which prohibits researchers from fully understanding the time-evolving pathology of a tissue barrier. Herein, the authors present a platform named "Digital Tissue-BArrier-CytoKine-counting-on-a-chip (DigiTACK)," which integrates digital immunosensors into a tissue chip system and demonstrates on-chip multiplexed, ultrasensitive, longitudinal cytokine secretion profiling of cultured brain endothelial barrier tissues. The integrated digital sensors utilize a novel beadless microwell format to perform an ultrafast "digital fingerprinting" of the analytes while achieving a low limit of detection (LoD) around 100-500 fg/mL for mouse MCP1 (CCL2), IL-6 and KC (CXCL1). The DigiTACK platform is extensively applicable to profile temporal cytokine secretion of other barrier-related organ-on-a-chip systems and can provide new insight into the secretory dynamics of the BBB by sequentially controlled experiments.
Collapse
Affiliation(s)
- Shiuan-Haur Su
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yujing Song
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrew Stephens
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Muyu Situ
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Molly C McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Anuska V Andjelkovic
- Department of Pathology and Neurosurgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin H Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
37
|
Yin S, Lu R, Li Y, Sun D, Liu C, Liu B, Li J. A microfluidic device inspired by leaky tumor vessels for hematogenous metastasis mechanism research. Analyst 2023; 148:1570-1578. [PMID: 36892183 DOI: 10.1039/d2an02081e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Endothelial intercellular pores of tumor vessels generally lead to enhanced interstitial flow and may facilitate the migration of tumor cells. The permeability of tumor vessels causes a concentration gradient of growth factors (CGGF) from blood vessels to tumor tissues, which is opposite to the direction of interstitial flow. In this work, exogenous chemotaxis under the CGGF is demonstrated as a mechanism of hematogenous metastasis. A bionic microfluidic device inspired by endothelial intercellular pores of tumor vessels has been designed to study the mechanism. A porous membrane vertically integrated into the device using a novel compound mold is utilized to mimic the leaky vascular wall. The formation mechanism of the CGGF caused by endothelial intercellular pores is numerically analyzed and experimentally verified. The migration behavior of U-2OS cells is studied in the microfluidic device. The device is divided into three regions of interest (ROI): primary site, migration zone, and tumor vessel. The number of cells in the migration zone increases significantly under the CGGF, but decreases under no CGGF, indicating tumor cells may be guided to the vascellum by exogenous chemotaxis. Transendothelial migration is subsequently monitored, demonstrating the successful replication of the key steps in vitro in the metastatic cascade by the bionic microfluidic device.
Collapse
Affiliation(s)
- Shuqing Yin
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Ruoyu Lu
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Yang Li
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Dexian Sun
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Chong Liu
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China. .,Key Laboratory for Precision and Non-traditional Machining Technology of Ministry of Education, Dalian University of Technology, Dalian, China
| | - Bo Liu
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, China.
| | - Jingmin Li
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| |
Collapse
|
38
|
Kincses A, Vigh JP, Petrovszki D, Valkai S, Kocsis AE, Walter FR, Lin HY, Jan JS, Deli MA, Dér A. The Use of Sensors in Blood-Brain Barrier-on-a-Chip Devices: Current Practice and Future Directions. BIOSENSORS 2023; 13:bios13030357. [PMID: 36979569 PMCID: PMC10046513 DOI: 10.3390/bios13030357] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 06/01/2023]
Abstract
The application of lab-on-a-chip technologies in in vitro cell culturing swiftly resulted in improved models of human organs compared to static culture insert-based ones. These chip devices provide controlled cell culture environments to mimic physiological functions and properties. Models of the blood-brain barrier (BBB) especially profited from this advanced technological approach. The BBB represents the tightest endothelial barrier within the vasculature with high electric resistance and low passive permeability, providing a controlled interface between the circulation and the brain. The multi-cell type dynamic BBB-on-chip models are in demand in several fields as alternatives to expensive animal studies or static culture inserts methods. Their combination with integrated biosensors provides real-time and noninvasive monitoring of the integrity of the BBB and of the presence and concentration of agents contributing to the physiological and metabolic functions and pathologies. In this review, we describe built-in sensors to characterize BBB models via quasi-direct current and electrical impedance measurements, as well as the different types of biosensors for the detection of metabolites, drugs, or toxic agents. We also give an outlook on the future of the field, with potential combinations of existing methods and possible improvements of current techniques.
Collapse
Affiliation(s)
- András Kincses
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Judit P. Vigh
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
- Doctoral School of Biology, University of Szeged, H-6720 Szeged, Hungary
| | - Dániel Petrovszki
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, H-6720 Szeged, Hungary
| | - Sándor Valkai
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Anna E. Kocsis
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Fruzsina R. Walter
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - András Dér
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| |
Collapse
|
39
|
Monteduro AG, Rizzato S, Caragnano G, Trapani A, Giannelli G, Maruccio G. Organs-on-chips technologies – A guide from disease models to opportunities for drug development. Biosens Bioelectron 2023; 231:115271. [PMID: 37060819 DOI: 10.1016/j.bios.2023.115271] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 11/24/2022] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Current in-vitro 2D cultures and animal models present severe limitations in recapitulating human physiopathology with striking discrepancies in estimating drug efficacy and side effects when compared to human trials. For these reasons, microphysiological systems, organ-on-chip and multiorgans microdevices attracted considerable attention as novel tools for high-throughput and high-content research to achieve an improved understanding of diseases and to accelerate the drug development process towards more precise and eventually personalized standards. This review takes the form of a guide on this fast-growing field, providing useful introduction to major themes and indications for further readings. We start analyzing Organs-on-chips (OOC) technologies for testing the major drug administration routes: (1) oral/rectal route by intestine-on-a-chip, (2) inhalation by lung-on-a-chip, (3) transdermal by skin-on-a-chip and (4) intravenous through vascularization models, considering how drugs penetrate in the bloodstream and are conveyed to their targets. Then, we focus on OOC models for (other) specific organs and diseases: (1) neurodegenerative diseases with brain models and blood brain barriers, (2) tumor models including their vascularization, organoids/spheroids, engineering and screening of antitumor drugs, (3) liver/kidney on chips and multiorgan models for gastrointestinal diseases and metabolic assessment of drugs and (4) biomechanical systems recapitulating heart, muscles and bones structures and related diseases. Successively, we discuss technologies and materials for organ on chips, analyzing (1) microfluidic tools for organs-on-chips, (2) sensor integration for real-time monitoring, (3) materials and (4) cell lines for organs on chips. (Nano)delivery approaches for therapeutics and their on chip assessment are also described. Finally, we conclude with a critical discussion on current significance/relevance, trends, limitations, challenges and future prospects in terms of revolutionary impact on biomedical research, preclinical models and drug development.
Collapse
Affiliation(s)
- Anna Grazia Monteduro
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Silvia Rizzato
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Giusi Caragnano
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Adriana Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology IRCCS "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
| | - Giuseppe Maruccio
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy.
| |
Collapse
|
40
|
Bi W, Cai S, Lei T, Wang L. Implementation of blood-brain barrier on microfluidic chip: recent advance and future prospects. Ageing Res Rev 2023; 87:101921. [PMID: 37004842 DOI: 10.1016/j.arr.2023.101921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/02/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023]
Abstract
The complex structure of the blood-brain barrier (BBB) hinders its modeling and the treatment of brain diseases. The microfluidic technology promotes the development of BBB-on-a-chip platforms, which can be used to reproduce the complex brain microenvironment and physiological reactions. Compared with traditional transwell technology, microfluidic BBB-on-a-chip shows great technical advantages in terms of flexible control of fluid shear stress in the chip and fabrication efficiency of the chip system, which can be enhanced by the development of lithography and three-dimensional (3D) printing. It is convenient to accurately monitor the dynamic changes of biochemical parameters of individual cells in the model by integrating an automatic super-resolution imaging sensing platform. In addition, biomaterials, especially hydrogels and conductive polymers, solve the limitations of microfluidic BBB-on-a-chip by compounding onto microfluidic chip to provide a 3D space and special performance on the microfluidic chip. The microfluidic BBB-on-a-chip promotes the development of basic research, including cell migration, mechanism exploration of neurodegenerative diseases, drug barrier permeability, SARS-CoV-2 pathology. This study summarizes the recent advances, challenges and future prospects of microfluidic BBB-on-a-chip, which can help to promote the development of personalized medicine and drug discovery.
Collapse
|
41
|
Rouleau N, Murugan NJ, Kaplan DL. Functional bioengineered models of the central nervous system. NATURE REVIEWS BIOENGINEERING 2023; 1:252-270. [PMID: 37064657 PMCID: PMC9903289 DOI: 10.1038/s44222-023-00027-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 02/10/2023]
Abstract
The functional complexity of the central nervous system (CNS) is unparalleled in living organisms. Its nested cells, circuits and networks encode memories, move bodies and generate experiences. Neural tissues can be engineered to assemble model systems that recapitulate essential features of the CNS and to investigate neurodevelopment, delineate pathophysiology, improve regeneration and accelerate drug discovery. In this Review, we discuss essential structure-function relationships of the CNS and examine materials and design considerations, including composition, scale, complexity and maturation, of cell biology-based and engineering-based CNS models. We highlight region-specific CNS models that can emulate functions of the cerebral cortex, hippocampus, spinal cord, neural-X interfaces and other regions, and investigate a range of applications for CNS models, including fundamental and clinical research. We conclude with an outlook to future possibilities of CNS models, highlighting the engineering challenges that remain to be overcome.
Collapse
Affiliation(s)
- Nicolas Rouleau
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, Ontario Canada
- Department of Biomedical Engineering, Tufts University, Medford, MA USA
| | - Nirosha J. Murugan
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, Ontario Canada
- Department of Biomedical Engineering, Tufts University, Medford, MA USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA USA
| |
Collapse
|
42
|
Endothelial Dysfunction in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24032909. [PMID: 36769234 PMCID: PMC9918222 DOI: 10.3390/ijms24032909] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
The cerebral vascular system stringently regulates cerebral blood flow (CBF). The components of the blood-brain barrier (BBB) protect the brain from pathogenic infections and harmful substances, efflux waste, and exchange substances; however, diseases develop in cases of blood vessel injuries and BBB dysregulation. Vascular pathology is concurrent with the mechanisms underlying aging, Alzheimer's disease (AD), and vascular dementia (VaD), which suggests its involvement in these mechanisms. Therefore, in the present study, we reviewed the role of vascular dysfunction in aging and neurodegenerative diseases, particularly AD and VaD. During the development of the aforementioned diseases, changes occur in the cerebral blood vessel morphology and local cells, which, in turn, alter CBF, fluid dynamics, and vascular integrity. Chronic vascular inflammation and blood vessel dysregulation further exacerbate vascular dysfunction. Multitudinous pathogenic processes affect the cerebrovascular system, whose dysfunction causes cognitive impairment. Knowledge regarding the pathophysiology of vascular dysfunction in neurodegenerative diseases and the underlying molecular mechanisms may lead to the discovery of clinically relevant vascular biomarkers, which may facilitate vascular imaging for disease prevention and treatment.
Collapse
|
43
|
Liu N, Zhu Y, Yu K, Gu Z, Lv S, Chen Y, He C, Fu J, He Y. Functional Blood-Brain Barrier Model with Tight Connected Minitissue by Liquid Substrates Culture. Adv Healthc Mater 2023; 12:e2201984. [PMID: 36394091 DOI: 10.1002/adhm.202201984] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/02/2022] [Indexed: 11/19/2022]
Abstract
The functional blood-brain barrier (BBB) model can provide a reliable tool for better understanding BBB transport mechanisms and in vitro preclinical experimentation. However, recapitulating microenvironmental complexities and physiological functions in an accessible approach remains a major challenge. Here, a new BBB model with a high-cell spatial density and tightly connected biomimetic minitissue is presented. The minitissue, pivotal functional structure of the BBB model, is fabricated by a novel and easy-to-use liquid substrate culture (LSC) method, which allows cells to self-assemble and self-heal into macrosized, tightly connected membranous minitissue. The minitissue with uniform thickness can be easily harvested in their entirety with extracellular matrix. Attributed to the tightly connected minitissue formed by LSC, the fabricated BBB biomimetic model has 1 to 2 orders of magnitude higher transendothelial electric resistance than the commonly reported BBB model. It also better prevents the transmission of large molecular substances, recapitulating the functional features of BBB. Furthermore, the BBB biomimetic model provides feedback regarding BBB-destructive drugs, exhibits selective transmission, and shows efflux pump activity. Overall, this model can serve as an accessible tool for life science or clinical medical researchers to enhance the understanding of human BBB and expedite the development of new brain-permeable drugs.
Collapse
Affiliation(s)
- Nian Liu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuanbo Zhu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Kang Yu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zeming Gu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Shang Lv
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuewei Chen
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chaofan He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.,Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, 450002, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
44
|
Sha L, Yong X, Shao Z, Duan Y, Hong Q, Zhang J, Zhang Y, Chen L. Targeting adverse effects of antiseizure medication on offspring: current evidence and new strategies for safety. Expert Rev Neurother 2023; 23:141-156. [PMID: 36731825 DOI: 10.1080/14737175.2023.2176751] [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: 02/04/2023]
Abstract
INTRODUCTION For women with epilepsy of reproductive age, antiseizure medications (ASMs) are associated with an increased risk of offspring malformations. There are safety concerns for most anti-seizure medications in the perinatal period, and there is a clear need to identify safe medications. ASMs must transport through biological barriers to exert toxic effects on the fetus, and transporters play essential roles in trans-barrier drug transport. Therefore, it is vital to understand the distribution and properties of ASM-related transporters in biological barriers. AREAS COVERED This study reviews the structure, transporter distribution, and properties of the blood-brain, placental, and blood-milk barrier, and summarizes the existing evidence for the trans-barrier transport mechanism of ASMs and standard experimental models of biological barriers. EXPERT OPINION Ideal ASMs in the perinatal period should have the following characteristics: 1) Increased transport through the blood-brain barrier, and 2) Reduced transport of the placental and blood-milk barriers. Thus, only low-dose or almost no antiseizure medication could enter the fetus's body, which could decrease medication-induced fetal abnormalities. Based on the stimulated structure and molecular docking, we propose a development strategy for new ASMs targeting transporters of biological barriers to improve the perinatal treatment of female patients with epilepsy.
Collapse
Affiliation(s)
- Leihao Sha
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| | - Xihao Yong
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yifei Duan
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| | - Qiulei Hong
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| | - Yunwu Zhang
- The current form, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Lei Chen
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| |
Collapse
|
45
|
Experimental Models of In Vitro Blood-Brain Barrier for CNS Drug Delivery: An Evolutionary Perspective. Int J Mol Sci 2023; 24:ijms24032710. [PMID: 36769032 PMCID: PMC9916529 DOI: 10.3390/ijms24032710] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Central nervous system (CNS) disorders represent one of the leading causes of global health burden. Nonetheless, new therapies approved against these disorders are among the lowest compared to their counterparts. The absence of reliable and efficient in vitro blood-brain barrier (BBB) models resembling in vivo barrier properties stands out as a significant roadblock in developing successful therapy for CNS disorders. Therefore, advancement in the creation of robust and sensitive in vitro BBB models for drug screening might allow us to expedite neurological drug development. This review discusses the major in vitro BBB models developed as of now for exploring the barrier properties of the cerebral vasculature. Our main focus is describing existing in vitro models, including the 2D transwell models covering both single-layer and co-culture models, 3D organoid models, and microfluidic models with their construction, permeability measurement, applications, and limitations. Although microfluidic models are better at recapitulating the in vivo properties of BBB than other models, significant gaps still exist for their use in predicting the performance of neurotherapeutics. However, this comprehensive account of in vitro BBB models can be useful for researchers to create improved models in the future.
Collapse
|
46
|
Luo M, Li Y, Peng B, White J, Mäkilä E, Tong WY, Jonathan Choi CH, Day B, Voelcker NH. A Multifunctional Porous Silicon Nanocarrier for Glioblastoma Treatment. Mol Pharm 2023; 20:545-560. [PMID: 36484477 DOI: 10.1021/acs.molpharmaceut.2c00763] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clinical treatment of glioblastoma (GBM) remains a major challenge because of the blood-brain barrier, chemotherapeutic resistance, and aggressive tumor metastasis. The development of advanced nanoplatforms that can efficiently deliver drugs and gene therapies across the BBB to the brain tumors is urgently needed. The protein "downregulated in renal cell carcinoma" (DRR) is one of the key drivers of GBM invasion. Here, we engineered porous silicon nanoparticles (pSiNPs) with antisense oligonucleotide (AON) for DRR gene knockdown as a targeted gene and drug delivery platform for GBM treatment. These AON-modified pSiNPs (AON@pSiNPs) were selectively internalized by GBM and human cerebral microvascular endothelial cells (hCMEC/D3) cells expressing Class A scavenger receptors (SR-A). AON was released from AON@pSiNPs, knocked down DRR and inhibited GBM cell migration. Additionally, a penetration study in a microfluidic-based BBB model and a biodistribution study in a glioma mice model showed that AON@pSiNPs could specifically cross the BBB and enter the brain. We further demonstrated that AON@pSiNPs could carry a large payload of the chemotherapy drug temozolomide (TMZ, 1.3 mg of TMZ per mg of NPs) and induce a significant cytotoxicity in GBM cells. On the basis of these results, the nanocarrier and its multifunctional strategy provide a strong potential for clinical treatment of GBM and research for targeted drug and gene delivery.
Collapse
Affiliation(s)
- Meihua Luo
- Monash Institute of Pharmaceutics Science, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria3052, Australia.,Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St. Lucia, Queensland4072, Australia.,Cell and Molecular Biology Department, QIMR Berghofer Medical Research Institute, Sid Faithfull Brain Cancer Laboratory, Brisbane, Queensland4006, Australia
| | - Yuchen Li
- Cell and Molecular Biology Department, QIMR Berghofer Medical Research Institute, Sid Faithfull Brain Cancer Laboratory, Brisbane, Queensland4006, Australia
| | - Bo Peng
- Monash Institute of Pharmaceutics Science, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria3052, Australia.,Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical materials & Engineering, Northwestern Polytechnical University, Xi'an710072, China
| | - Jacinta White
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria3168, Australia
| | - Ermei Mäkilä
- Industrial Physics Laboratory, Department of Physics and Astronomy, University of Turku, Turku20014, Finland
| | - Wing Yin Tong
- Monash Institute of Pharmaceutics Science, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria3052, Australia
| | - Chung Hang Jonathan Choi
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Bryan Day
- Cell and Molecular Biology Department, QIMR Berghofer Medical Research Institute, Sid Faithfull Brain Cancer Laboratory, Brisbane, Queensland4006, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland4072, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland4059, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutics Science, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria3052, Australia.,Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria3168, Australia.,Materials Science and Engineering, Monash University, 14 Alliance Lane, Clayton, Victoria3800, Australia
| |
Collapse
|
47
|
Blood brain barrier-on-a-chip to model neurological diseases. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
48
|
Human mini-blood-brain barrier models for biomedical neuroscience research: a review. Biomater Res 2022; 26:82. [PMID: 36527159 PMCID: PMC9756735 DOI: 10.1186/s40824-022-00332-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
The human blood-brain barrier (BBB) is a unique multicellular structure that is in critical demand for fundamental neuroscience studies and therapeutic evaluation. Despite substantial achievements in creating in vitro human BBB platforms, challenges in generating specifics of physiopathological relevance are viewed as impediments to the establishment of in vitro models. In this review, we provide insight into the development and deployment of in vitro BBB models that allow investigation of the physiology and pathology of neurological therapeutic avenues. First, we highlight the critical components, including cell sources, biomaterial glue collections, and engineering techniques to reconstruct a miniaturized human BBB. Second, we describe recent breakthroughs in human mini-BBBs for investigating biological mechanisms in neurology. Finally, we discuss the application of human mini-BBBs to medical approaches. This review provides strategies for understanding neurological diseases, a validation model for drug discovery, and a potential approach for generating personalized medicine.
Collapse
|
49
|
Nimbkar S, Leena MM, Moses JA, Anandharamakrishnan C. Microfluidic assessment of nutritional biomarkers: Concepts, approaches and advances. Crit Rev Food Sci Nutr 2022; 64:5113-5127. [PMID: 36503314 DOI: 10.1080/10408398.2022.2150597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Among various approaches to understand the health status of an individual, nutritional biomarkers can provide valuable information, particularly in terms of deficiencies, if any, and their severity. Commonly, the approach revolves around molecular sciences, and the information gained can support prognosis, diagnosis, remediation, and impact assessment of therapies. Microfluidic platforms can offer benefits of low sample and reagent requirements, low cost, high precision, and lower detection limits, with simplicity in handling and the provision for complete automation and integration with information and communication technologies (ICTs). While several advances are being made, this work details the underlying concepts, with emphasis on different point-of-care devices for the analysis of macro and micronutrient biomarkers. In addition, the scope of using different wearable microfluidic sensors for real-time and noninvasive determination of biomarkers is detailed. While several challenges remain, a strong focus is given on recent advances, presenting the state-of-the-art of this field. With more such biomarkers being discovered and commercialization-driven research, trends indicate the wide prospects of this advancing field in supporting clinicians, food technologists, nutritionists, and others.
Collapse
Affiliation(s)
- Shubham Nimbkar
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology, Entrepreneurship and Management, Ministry of Food Processing Industries, Thanjavur, Tamil Nadu, India
| | - M Maria Leena
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology, Entrepreneurship and Management, Ministry of Food Processing Industries, Thanjavur, Tamil Nadu, India
| | - Jeyan Arthur Moses
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology, Entrepreneurship and Management, Ministry of Food Processing Industries, Thanjavur, Tamil Nadu, India
| | - Chinnaswamy Anandharamakrishnan
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology, Entrepreneurship and Management, Ministry of Food Processing Industries, Thanjavur, Tamil Nadu, India
| |
Collapse
|
50
|
Akide Ndunge OB, Kilian N, Salman MM. Cerebral Malaria and Neuronal Implications of Plasmodium Falciparum Infection: From Mechanisms to Advanced Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202944. [PMID: 36300890 PMCID: PMC9798991 DOI: 10.1002/advs.202202944] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/22/2022] [Indexed: 06/01/2023]
Abstract
Reorganization of host red blood cells by the malaria parasite Plasmodium falciparum enables their sequestration via attachment to the microvasculature. This artificially increases the dwelling time of the infected red blood cells within inner organs such as the brain, which can lead to cerebral malaria. Cerebral malaria is the deadliest complication patients infected with P. falciparum can experience and still remains a major public health concern despite effective antimalarial therapies. Here, the current understanding of the effect of P. falciparum cytoadherence and their secreted proteins on structural features of the human blood-brain barrier and their involvement in the pathogenesis of cerebral malaria are highlighted. Advanced 2D and 3D in vitro models are further assessed to study this devastating interaction between parasite and host. A better understanding of the molecular mechanisms leading to neuronal and cognitive deficits in cerebral malaria will be pivotal in devising new strategies to treat and prevent blood-brain barrier dysfunction and subsequent neurological damage in patients with cerebral malaria.
Collapse
Affiliation(s)
- Oscar Bate Akide Ndunge
- Department of Internal MedicineSection of Infectious DiseasesYale University School of Medicine300 Cedar StreetNew HavenCT06510USA
| | - Nicole Kilian
- Centre for Infectious Diseases, ParasitologyHeidelberg University HospitalIm Neuenheimer Feld 32469120HeidelbergGermany
| | - Mootaz M. Salman
- Department of PhysiologyAnatomy and GeneticsUniversity of OxfordOxfordOX1 3QUUK
- Kavli Institute for NanoScience DiscoveryUniversity of OxfordOxfordUK
- Oxford Parkinson's Disease CentreUniversity of OxfordOxfordUK
| |
Collapse
|