1
|
Eltanameli B, Piñeiro-Llanes J, Cristofoletti R. Recent advances in cell-based in vitro models for predicting drug permeability across brain, intestinal, and pulmonary barriers. Expert Opin Drug Metab Toxicol 2024; 20:439-458. [PMID: 38850058 DOI: 10.1080/17425255.2024.2366390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/06/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION Recent years have witnessed remarkable progress in the development of cell-based in vitro models aimed at predicting drug permeability, particularly focusing on replicating the barrier properties of the blood-brain barrier (BBB), intestinal epithelium, and lung epithelium. AREA COVERED This review provides an overview of 2D in vitro platforms, including monocultures and co-culture systems, highlighting their respective advantages and limitations. Additionally, it discusses tools and techniques utilized to overcome these limitations, paving the way for more accurate predictions of drug permeability. Furthermore, this review delves into emerging technologies, particularly microphysiological systems (MPS), encompassing static platforms such as organoids and dynamic platforms like microfluidic devices. Literature searches were performed using PubMed and Google Scholar. We focus on key terms such as in vitro permeability models, MPS, organoids, intestine, BBB, and lungs. EXPERT OPINION The potential of these MPS to mimic physiological conditions more closely offers promising avenues for drug permeability assessment. However, transitioning these advanced models from bench to industry requires rigorous validation against regulatory standards. Thus, there is a pressing need to validate MPS to industry and regulatory agency standards to exploit their potential in drug permeability prediction fully. This review underscores the importance of such validation processes to facilitate the translation of these innovative technologies into routine pharmaceutical practice.
Collapse
Affiliation(s)
- Bassma Eltanameli
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Janny Piñeiro-Llanes
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
2
|
Lee J, Lee H, Lee H, Shin M, Shin MG, Seo J, Lee EJ, Park SA, Park S. ANKS1A regulates LDL receptor-related protein 1 (LRP1)-mediated cerebrovascular clearance in brain endothelial cells. Nat Commun 2023; 14:8463. [PMID: 38123547 PMCID: PMC10733300 DOI: 10.1038/s41467-023-44319-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Brain endothelial LDL receptor-related protein 1 (LRP1) is involved in the clearance of Aβ peptides across the blood-brain barrier (BBB). Here we show that endothelial deficiency of ankyrin repeat and SAM domain containing 1 A (ANKS1A) reduces both the cell surface levels of LRP1 and the Aβ clearance across the BBB. Association of ANKS1A with the NPXY motifs of LRP1 facilitates the transport of LRP1 from the endoplasmic reticulum toward the cell surface. ANKS1A deficiency in an Alzheimer's disease mouse model results in exacerbated Aβ pathology followed by cognitive impairments. These deficits are reversible by gene therapy with brain endothelial-specific ANKS1A. In addition, human induced pluripotent stem cell-derived BBBs (iBBBs) were generated from endothelial cells lacking ANKS1A or carrying the rs6930932 variant. Those iBBBs exhibit both reduced cell surface LRP1 and impaired Aβ clearance. Thus, our findings demonstrate that ANKS1A regulates LRP1-mediated Aβ clearance across the BBB.
Collapse
Affiliation(s)
- Jiyeon Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Korea
| | - Haeryung Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Korea
| | - Hyein Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, 42988, Korea
| | - Miram Shin
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Korea
| | - Min-Gi Shin
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Jinsoo Seo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, 42988, Korea
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Sun Ah Park
- Lab for Neurodegenerative Dementia, Department of Anatomy, and Department of Neurology, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Soochul Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Korea.
| |
Collapse
|
3
|
Chang HW, Hsu MJ, Chien LN, Chi NF, Yu MC, Chen HC, Lin YF, Hu CJ. Role of the Autism Risk Gene Shank3 in the Development of Atherosclerosis: Insights from Big Data and Mechanistic Analyses. Cells 2023; 12:2546. [PMID: 37947623 PMCID: PMC10647789 DOI: 10.3390/cells12212546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Increased medical attention is needed as the prevalence of autism spectrum disorder (ASD) rises. Both cardiovascular disorder (CVD) and hyperlipidemia are closely associated with adult ASD. Shank3 plays a key genetic role in ASD. We hypothesized that Shank3 contributes to CVD development in young adults with ASD. In this study, we investigated whether Shank3 facilitates the development of atherosclerosis. Using Gene Set Enrichment Analysis software (Version No.: GSEA-4.0.3), we analyzed the data obtained from Shank3 knockout mice (Gene Expression Omnibus database), a human population-based study cohort (from Taiwan's National Health Insurance Research Database), and a Shank3 knockdown cellular model. Shank3 knockout upregulated the expression of genes of cholesterol homeostasis and fatty acid metabolism but downregulated the expression of genes associated with inflammatory responses. Individuals with autism had higher risks of hyperlipidemia (adjusted hazard ratio (aHR): 1.39; p < 0.001), major adverse cardiac events (aHR: 2.67; p < 0.001), and stroke (aHR: 3.55; p < 0.001) than age- and sex-matched individuals without autism did. Shank3 downregulation suppressed tumor necrosis factor-α-induced fatty acid synthase expression; vascular cell adhesion molecule 1 expression; and downstream signaling pathways involving p38, Jun N-terminal kinase, and nuclear factor-κB. Thus, Shank3 may influence the development of early-onset atherosclerosis and CVD in ASD. Furthermore, regulating Shank3 expression may reduce inflammation-related disorders, such as atherosclerosis, by inhibiting tumor necrosis factor-alpha-mediated inflammatory cascades.
Collapse
Affiliation(s)
- Hsiu-Wen Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Neurology, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan
| | - Ming-Jen Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan (H.-C.C.)
| | - Li-Nien Chien
- Institute of Health and Welfare Policy, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
| | - Nai-Fang Chi
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei 11267, Taiwan;
| | - Meng-Chieh Yu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan (H.-C.C.)
| | - Hsiu-Chen Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan (H.-C.C.)
| | - Yuan-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
4
|
Wang S, Bai L, Hu X, Yao S, Hao Z, Zhou J, Li X, Lu H, He J, Wang L, Li D. 3D Bioprinting of Neurovascular Tissue Modeling with Collagen-Based Low-Viscosity Composites. Adv Healthc Mater 2023; 12:e2300004. [PMID: 37264745 PMCID: PMC11469067 DOI: 10.1002/adhm.202300004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 05/27/2023] [Indexed: 06/03/2023]
Abstract
In vitro neurovascular unit (NVU) models are valuable for investigating brain functions and developing drugs. However, it remains challenging to recapitulate the native architectural features and ultra-soft extracellular matrix (ECM) properties of the natural NVU. Cell-laden bioprinting is promising to prepare complex living tissues, but hard to balance the fidelity and cell growth. This study proposes a novel two-stage methodology for biomanufacturing functional 3D neurovascular constructs in vitro with low modulus of ECM. At the shaping stage, a low-viscosity alginate/collagen is printed through an embedded approach; at the culturing stage, the alginate is removed through targeted lysing. The low-viscosity and rapid crosslinking properties provide a printing resolution of ≈10 µm, and the lysis processing can decrease the hydrogels' modulus to ≈1 kPa and adjust the porosity of the microstructure, providing cells with an environment closing to the brain ECM. A 3D hollow coaxial neurovascular model is fabricated, in which the endothelial cells has expressed tight junction proteins and shown selective permeability, and the astrocytes outside of the endothelial layer are found to spread out with branches and directly interact with endothelial cells. The present study offers a promising modeling method for better understanding the NVU function and screening neuro-drugs.
Collapse
Affiliation(s)
- Sen Wang
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Luge Bai
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Xiaoxuan Hu
- Institute of NeurobiologySchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'an710061China
- Key Laboratory of Ministry of Education for Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterXi'an710061China
| | - Siqi Yao
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Zhiyan Hao
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - JiaJia Zhou
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Xiao Li
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Haixia Lu
- Institute of NeurobiologySchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'an710061China
- Key Laboratory of Ministry of Education for Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterXi'an710061China
- Department of Human Anatomy & HistoembryologySchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'an710061China
| | - Jiankang He
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Ling Wang
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Dichen Li
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| |
Collapse
|
5
|
Deng K, Lu Y, Finnema SJ, Vangjeli K, Huang J, Huang L, Goodearl A. Application of In vitro transcytosis models to brain targeted biologics. PLoS One 2023; 18:e0289970. [PMID: 37611031 PMCID: PMC10446226 DOI: 10.1371/journal.pone.0289970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
The blood brain barrier (BBB) efficiently limits the penetration of biologics drugs from blood to brain. Establishment of an in vitro BBB model can facilitate screening of central nervous system (CNS) drug candidates and accelerate CNS drug development. Despite many established in vitro models, their application to biologics drug selection has been limited. Here, we report the evaluation of in vitro transcytosis of anti-human transferrin receptor (TfR) antibodies across human, cynomolgus and mouse species. We first evaluated human models including human cerebral microvascular endothelial cell line hCMEC/D3 and human colon epithelial cell line Caco-2 models. hCMEC/D3 model displayed low trans-epithelial electrical resistance (TEER), strong paracellular transport, and similar transcytosis of anti-TfR and control antibodies. In contrast, the Caco-2 model displayed high TEER value and low paracellular transport. Anti-hTfR antibodies demonstrated up to 70-fold better transcytosis compared to control IgG. Transcytosis of anti-hTfR.B1 antibody in Caco-2 model was dose-dependent and saturated at 3 μg/mL. Enhanced transcytosis of anti-hTfR.B1 was also observed in a monkey brain endothelial cell based (MBT) model. Importantly, anti-hTfR.B1 showed relatively high brain radioactivity concentration in a non-human primate positron emission tomography study indicating that the in vitro transcytosis from both Caco-2 and MBT models aligns with in vivo brain exposure. Typically, brain exposure of CNS targeted biologics is evaluated in mice. However, antibodies, such as the anti-human TfR antibodies, do not cross-react with the mouse target. Therefore, validation of a mouse in vitro transcytosis model is needed to better understand the in vitro in vivo correlation. Here, we performed transcytosis of anti-mouse TfR antibodies in mouse brain endothelial cell-based models, bEnd3 and the murine intestinal epithelial cell line mIEC. There is a good correlation between in vitro transcytosis of anti-mTfR antibodies and bispecifics in mIEC model and their mouse brain uptake. These data strengthen our confidence in the predictive power of the in vitro transcytosis models. Both mouse and human in vitro models will serve as important screening assays for brain targeted biologics selection in CNS drug development.
Collapse
Affiliation(s)
- Kangwen Deng
- AbbVie Bioresearch Center, Worcester, MA, United States of America
| | - Yifeng Lu
- AbbVie Bioresearch Center, Worcester, MA, United States of America
| | | | - Kostika Vangjeli
- AbbVie Bioresearch Center, Worcester, MA, United States of America
| | - Junwei Huang
- AbbVie Bioresearch Center, Worcester, MA, United States of America
| | - Lili Huang
- AbbVie Bioresearch Center, Worcester, MA, United States of America
| | - Andrew Goodearl
- AbbVie Bioresearch Center, Worcester, MA, United States of America
| |
Collapse
|
6
|
Singh S, Agrawal M, Vashist R, Patel RK, Sangave SD, Alexander A. Recent advancements on in vitro blood-brain barrier model: A reliable and efficient screening approach for preclinical and clinical investigation. Expert Opin Drug Deliv 2023; 20:1839-1857. [PMID: 38100459 DOI: 10.1080/17425247.2023.2295940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION The efficiency of brain therapeutics is greatly hindered by the blood-brain barrier (BBB). BBB's protective function, selective permeability, and dynamic functionality maintain the harmony between the brain and peripheral region. Thus, the design of any novel drug carrier system requires the complete study and investigation of BBB permeability, efflux transport, and the effect of associated cellular and non-vascular unit trafficking on BBB penetrability. The in vitro BBB models offer a most promising, and reliable mode of initial investigation of BBB permeability and associated factors as strong evidence for further preclinical and clinical investigation. AREA COVERED This review work covers the structure and functions of BBB components and different types of in vitro BBB models along with factors affecting BBB model development and model selection criteria. EXPERT OPINION In vivo models assume to reciprocate the physiological environment to the maximum extent. However, the interspecies variability, NVUs trafficking, dynamic behavior of BBB, etc., lead to non-reproducible results. The in vitro models are comparatively less complex, and flexible, as per the study design, could generate substantial evidence and help identify suitable in vivo animal model selection.
Collapse
Affiliation(s)
- Snigdha Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Mukta Agrawal
- School of Pharmacy and Technology Management, Narsee Monjee Institute of Management Studies, Mahbubnagar, India
| | - Rajat Vashist
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Rohit K Patel
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | | | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| |
Collapse
|
7
|
Gupta R, Chauhan A, Kaur T, Kuanr BK, Sharma D. Transmigration of magnetite nanoparticles across the blood-brain barrier in a rodent model: influence of external and alternating magnetic fields. NANOSCALE 2022; 14:17589-17606. [PMID: 36409463 DOI: 10.1039/d2nr02210a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Despite advances in neurology, drug delivery to the central nervous system is considered a challenge due to the presence of the blood brain barrier (BBB). In this study, the role of magnetic hyperthermia induced by exposure of magnetic nanoparticles (MNPs) to an alternating magnetic field (AMF) in synergy with an external magnetic field (EMF) was investigated to transiently increase the permeability of the MNPs across the BBB. A dual magnetic targeting approach was employed by first dragging the MNPs by an EMF for an intended enhanced cellular association with the brain endothelial cells and then activating the MNPs by an AMF for the temporary disruption of the tight junctions of BBB. The efficacy of the BBB permeability for the MNPs under the influence of dual magnetic targeting was evaluated in vitro using transwell models developed by co-culturing murine brain endothelial cells with astrocytes, as well as in vivo in mouse models. The in vitro results revealed that the exposure to AMF transiently opened the tight junctions at the BBB, which, after 3 h of treatment, were observed to recover back to their comparable control levels. A biodistribution analysis of nanoparticles confirmed targeted accumulation of MNPs in the brain following dual targeting. This dual targeting approach was observed to open the tight junctions, thus increasing the transport of MNPs into the brain with higher specificity as compared to using EMF targeting alone, suggesting that a dual magnetic targeting-induced transport of MNPs across the BBB is an effective measure for delivery of therapeutics.
Collapse
Affiliation(s)
- Ruby Gupta
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab-140306, India.
| | - Anjali Chauhan
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab-140306, India.
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi-110067, India
| | - Tashmeen Kaur
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab-140306, India.
| | - Bijoy K Kuanr
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi-110067, India
| | - Deepika Sharma
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab-140306, India.
| |
Collapse
|
8
|
Gagliardi S, Truffi M, Tinelli V, Garofalo M, Pandini C, Cotta Ramusino M, Perini G, Costa A, Negri S, Mazzucchelli S, Bonizzi A, Sitia L, Busacca M, Sevieri M, Mocchi M, Ricciardi A, Prosperi D, Corsi F, Cereda C, Morasso C. Bisdemethoxycurcumin (BDC)-Loaded H-Ferritin-Nanocages Mediate the Regulation of Inflammation in Alzheimer's Disease Patients. Int J Mol Sci 2022; 23:9237. [PMID: 36012501 PMCID: PMC9409287 DOI: 10.3390/ijms23169237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/01/2022] [Accepted: 08/06/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Bisdemethoxycurcumin (BDC) might be an inflammation inhibitor in Alzheimer's Disease (AD). However, BDC is almost insoluble in water, poorly absorbed by the organism, and degrades rapidly. We thus developed a new nanoformulation of BDC based on H-Ferritin nanocages (BDC-HFn). METHODS We tested the BDC-HFn solubility, stability, and ability to cross a blood-brain barrier (BBB) model. We tested the effect of BDC-HFn on AD and control (CTR) PBMCs to evaluate the transcriptomic profile by RNA-seq. RESULTS We developed a nanoformulation with a diameter of 12 nm to improve the solubility and stability. The comparison of the transcriptomics analyses between AD patients before and after BDC-HFn treatment showed a major number of DEG (2517). The pathway analysis showed that chemokines and macrophages activation differed between AD patients and controls after BDC-HFn treatment. BDC-HFn binds endothelial cells from the cerebral cortex and crosses through a BBB in vitro model. CONCLUSIONS Our data showed how BDC-Hfn could improve the stability of BDC. Significant differences in genes associated with inflammation between the same patients before and after BDC-Hfn treatment have been found. Inflammatory genes that are upregulated between AD and CTR after BDC-HFn treatment are converted and downregulated, suggesting a possible therapeutic approach.
Collapse
Affiliation(s)
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS Spa SB, 27100 Pavia, Italy
| | - Veronica Tinelli
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, 20126 Milano, Italy
| | | | | | | | | | - Alfredo Costa
- IRCCS Mondino Foundation, 27100 Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Sara Negri
- Istituti Clinici Scientifici Maugeri IRCCS Spa SB, 27100 Pavia, Italy
| | - Serena Mazzucchelli
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Via G. B. Grassi 74, 20157 Milano, Italy
| | - Arianna Bonizzi
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Via G. B. Grassi 74, 20157 Milano, Italy
| | - Leopoldo Sitia
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Via G. B. Grassi 74, 20157 Milano, Italy
| | | | - Marta Sevieri
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Via G. B. Grassi 74, 20157 Milano, Italy
| | - Michela Mocchi
- Istituti Clinici Scientifici Maugeri IRCCS Spa SB, 27100 Pavia, Italy
| | | | - Davide Prosperi
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, 20126 Milano, Italy
| | - Fabio Corsi
- Istituti Clinici Scientifici Maugeri IRCCS Spa SB, 27100 Pavia, Italy
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, 20126 Milano, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Via G. B. Grassi 74, 20157 Milano, Italy
| | | | - Carlo Morasso
- Istituti Clinici Scientifici Maugeri IRCCS Spa SB, 27100 Pavia, Italy
| |
Collapse
|
9
|
Dana P, Pimpha N, Chaipuang A, Thumrongsiri N, Tanyapanyachon P, Taweechaipaisankul A, Chonniyom W, Watcharadulyarat N, Sathornsumetee S, Saengkrit N. Inhibiting Metastasis and Improving Chemosensitivity via Chitosan-Coated Selenium Nanoparticles for Brain Cancer Therapy. NANOMATERIALS 2022; 12:nano12152606. [PMID: 35957037 PMCID: PMC9370598 DOI: 10.3390/nano12152606] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 02/05/2023]
Abstract
Selenium nanoparticles (SeNPs) were synthesized to overcome the limitations of selenium, such as its narrow safe range and low water solubility. SeNPs reduce the toxicity and improve the bioavailability of selenium. Chitosan-coated SeNPs (Cs-SeNPs) were developed to further stabilize SeNPs and to test their effects against glioma cells. The effects of Cs-SeNPs on cell growth were evaluated in monolayer and 3D-tumor spheroid culture. Cell migration and cell invasion were determined using a trans-well assay. The effect of Cs-SeNPs on chemotherapeutic drug 5-fluorouracil (5-FU) sensitivity of glioma cells was determined in tumor spheroids. An in vitro blood–brain barrier (BBB) model was established to test the permeability of Cs-SeNPs. SeNPs and Cs-SeNPs can reduce the cell viability of glioma cells in a dose-dependent manner. Compared with SeNPs, Cs-SeNPs more strongly inhibited 3D-tumor spheroid growth. Cs-SeNPs exhibited stronger effects in inhibiting cell migration and cell invasion than SeNPs. Improved 5-FU sensitivity was observed in Cs-SeNP-treated cells. Cellular uptake in glioma cells indicated a higher uptake rate of coumarin-6-labeled Cs-SeNPs than SeNPs. The capability of coumarin-6 associated Cs-SeNPs to pass through the BBB was confirmed. Taken together, Cs-SeNPs provide exceptional performance and are a potential alternative therapeutic strategy for future glioma treatment.
Collapse
Affiliation(s)
- Paweena Dana
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
| | - Nuttaporn Pimpha
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
| | - Angkana Chaipuang
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
| | - Nutthanit Thumrongsiri
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
| | - Prattana Tanyapanyachon
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
| | - Anukul Taweechaipaisankul
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
| | - Walailuk Chonniyom
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
| | - Natsorn Watcharadulyarat
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
| | - Sith Sathornsumetee
- Research Network NANOTEC-Mahidol University in Theranostic Nanomedicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand;
- Department of Medicine (Neurology), Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand
| | - Nattika Saengkrit
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (P.D.); (N.P.); (A.C.); (N.T.); (P.T.); (A.T.); (W.C.); (N.W.)
- Correspondence: ; Tel.: +66-2117-6558
| |
Collapse
|
10
|
Adams Y, Jensen AR. Cerebral malaria - modelling interactions at the blood-brain barrier in vitro. Dis Model Mech 2022; 15:275963. [PMID: 35815443 PMCID: PMC9302004 DOI: 10.1242/dmm.049410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The blood–brain barrier (BBB) is a continuous endothelial barrier that is supported by pericytes and astrocytes and regulates the passage of solutes between the bloodstream and the brain. This structure is called the neurovascular unit and serves to protect the brain from blood-borne disease-causing agents and other risk factors. In the past decade, great strides have been made to investigate the neurovascular unit for delivery of chemotherapeutics and for understanding how pathogens can circumvent the barrier, leading to severe and, at times, fatal complications. One such complication is cerebral malaria, in which Plasmodium falciparum-infected red blood cells disrupt the barrier function of the BBB, causing severe brain swelling. Multiple in vitro models of the BBB are available to investigate the mechanisms underlying the pathogenesis of cerebral malaria and other diseases. These range from single-cell monolayer cultures to multicellular BBB organoids and highly complex cerebral organoids. Here, we review the technologies available in malaria research to investigate the interaction between P. falciparum-infected red blood cells and the BBB, and discuss the advantages and disadvantages of each model. Summary: This Review discusses the available in vitro models to investigate the impact of adhesion of Plasmodium falciparum-infected red blood cells on the blood–brain barrier, a process associated with cerebral malaria.
Collapse
Affiliation(s)
- Yvonne Adams
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Anja Ramstedt Jensen
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
11
|
Sun J, Ou W, Han D, Paganini-Hill A, Fisher MJ, Sumbria RK. Comparative studies between the murine immortalized brain endothelial cell line (bEnd.3) and induced pluripotent stem cell-derived human brain endothelial cells for paracellular transport. PLoS One 2022; 17:e0268860. [PMID: 35613139 PMCID: PMC9132315 DOI: 10.1371/journal.pone.0268860] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/09/2022] [Indexed: 01/11/2023] Open
Abstract
Brain microvascular endothelial cells, forming the anatomical site of the blood-brain barrier (BBB), are widely used as in vitro complements to in vivo BBB studies. Among the immortalized cells used as in vitro BBB models, the murine-derived bEnd.3 cells offer culturing consistency and low cost and are well characterized for functional and transport assays, but result in low transendothelial electrical resistance (TEER). Human-induced pluripotent stem cells differentiated into brain microvascular endothelial cells (ihBMECs) have superior barrier properties, but the process of differentiation is time-consuming and can result in mixed endothelial-epithelial gene expression. Here we performed a side-by-side comparison of the ihBMECs and bEnd.3 cells for key paracellular diffusional transport characteristics. The TEER across the ihBMECs was 45- to 68-fold higher than the bEnd.3 monolayer. The ihBMECs had significantly lower tracer permeability than the bEnd.3 cells. Both, however, could discriminate between the paracellular permeabilities of two tracers: sodium fluorescein (MW: 376 Da) and fluorescein isothiocyanate (FITC)-dextran (MW: 70 kDa). FITC-dextran permeability was a strong inverse-correlate of TEER in the bEnd.3 cells, whereas sodium fluorescein permeability was a strong inverse-correlate of TEER in the ihBMECs. Both bEnd.3 cells and ihBMECs showed the typical cobblestone morphology with robust uptake of acetylated LDL and strong immuno-positivity for vWF. Both models showed strong claudin-5 expression, albeit with differences in expression location. We further confirmed the vascular endothelial- (CD31 and tube-like formation) and erythrophagocytic-phenotypes and the response to inflammatory stimuli of ihBMECs. Overall, both bEnd.3 cells and ihBMECs express key brain endothelial phenotypic markers, and despite differential TEER measurements, these in vitro models can discriminate between the passage of different molecular weight tracers. Our results highlight the need to corroborate TEER measurements with different molecular weight tracers and that the bEnd.3 cells may be suitable for large molecule transport studies despite their low TEER.
Collapse
Affiliation(s)
- Jiahong Sun
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States of America
| | - Weijun Ou
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States of America
| | - Derick Han
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States of America
| | - Annlia Paganini-Hill
- Department of Neurology, University of California, Irvine, Irvine, CA, United States of America
| | - Mark J. Fisher
- Department of Neurology, University of California, Irvine, Irvine, CA, United States of America
- Department of Pathology & Laboratory Medicine, University of California, Irvine, Irvine, CA, United States of America
| | - Rachita K. Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States of America
- Department of Neurology, University of California, Irvine, Irvine, CA, United States of America
- * E-mail:
| |
Collapse
|
12
|
Piantino M, Louis F, Shigemoto-Mogami Y, Kitamura K, Sato K, Yamaguchi T, Kawabata K, Yamamoto S, Iwasaki S, Hirabayashi H, Matsusaki M. Brain microvascular endothelial cells derived from human induced pluripotent stem cells as in vitro model for assessing blood-brain barrier transferrin receptor-mediated transcytosis. Mater Today Bio 2022; 14:100232. [PMID: 35308041 PMCID: PMC8927846 DOI: 10.1016/j.mtbio.2022.100232] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB), a selective barrier formed by brain microvascular endothelial cells (BMEC), represents a major challenge for the efficient accumulation of pharmaceutical drugs into the brain. The receptor-mediated transcytosis (RMT) has recently gained increasing interest for pharmaceutical industry as it shows a great potential to shuttle large-sized therapeutic cargos across the BBB. Confirming the presence of the RMT pathway by BMEC is therefore important for the screening of peptides or antibody libraries that bind RMT receptors. Herein, a comparative study was performed between a human cell line of BMEC (HBEC) and human induced pluripotent stem cells-derived BMEC-like cells (hiPS-BMEC). The significantly higher gene and protein expressions of transporters and tight junction proteins, excepting CD31 and VE-cadherin were exhibited by hiPS-BMEC than by HBEC, suggesting more biomimetic BBB features of hiPS-BMEC. The presence and functionality of transferrin receptor (TfR), known to use RMT pathway, were confirmed using hiPS-BMEC by competitive binding assays and confocal microscopy observations. Finally, cysteine-modified T7 and cysteine modified-Tfr-T12 peptides, previously reported to be ligands of TfR, were compared regarding their permeability using hiPS-BMEC. The hiPS-BMEC could be useful for the identification of therapeutics that can be transported across the BBB using RMT pathway.
Collapse
Affiliation(s)
- Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN INC.) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Yukari Shigemoto-Mogami
- Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences (NIHS), Kawasaki, Kanagawa, Japan
| | - Kimiko Kitamura
- Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences (NIHS), Kawasaki, Kanagawa, Japan
| | - Kaoru Sato
- Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences (NIHS), Kawasaki, Kanagawa, Japan
| | - Tomoko Yamaguchi
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Syunsuke Yamamoto
- Drug Metabolism & Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Shinji Iwasaki
- Drug Metabolism & Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Hideki Hirabayashi
- Drug Metabolism & Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- Joint Research Laboratory (TOPPAN INC.) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
13
|
Pu Y, Zhao L, Xi Y, Xia Y, Qian Y. The protective effects of Mirtazapine against lipopolysaccharide (LPS)-induced brain vascular hyperpermeability. Bioengineered 2022; 13:3680-3693. [PMID: 35081868 PMCID: PMC8973832 DOI: 10.1080/21655979.2021.2024962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sepsis is mainly characterized by severe inflammation triggered by infection, and sepsis-associated encephalopathy (SAE) is defined as brain damage caused by sepsis. Disruption of the blood-brain barrier (BBB) triggered by injured brain microvascular endothelial cells (BMECs) and damaged tight junction (TJ) structure is closely associated with the pathogenesis of SAE. The present research proposed to evaluate the potential therapeutic effects of Mirtazapine, a central presynaptic α2 receptor antagonist, on LPS-induced BBB disruption. The mice were administered with normal saline and 10 mg/kg Mirtazapine for 8 consecutive days, and from day 6, the experiment group of mice received LPS for 2 days to induce SAE. We found that the increased BBB permeability, elevated concentrations of inflammatory factors in brain tissues, and downregulated zonula occludens -1 (ZO-1) were observed in LPS-stimulated mice, all of which were reversed by 10 mg/kg Mirtazapine. In the in vitro assay, bEnd.3 brain endothelial cells were treated with 1 μM LPS in the absence or presence of Mirtazapine (25, 50 μM). We found that LPS-treated cells had significantly declined transendothelial electrical resistance (TEER), increased monolayer permeability, elevated production of inflammatory factors, and downregulated ZO-1. However, 25 and 50 μM Mirtazapine ameliorated all these LPS- induced aberrations. Mirtazapine also mitigated the decreased level of NF-E2-related factor 2 (Nrf2) in LPS-challenged endothelial cells. The protective effect of Mirtazapine on endothelial permeability against LPS was significantly abolished by the knockdown of Nrf2. Collectively, we concluded that Mirtazapine exerted protective effects on LPS-induced endothelial cells hyperpermeability by upregulating Nrf2.
Collapse
Affiliation(s)
- Yuehong Pu
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhao
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yao Xi
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yichun Xia
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Qian
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Pokharel S, Gliyazova NS, Dandepally SR, Williams AL, Ibeanu GC. Neuroprotective effects of an in vitro BBB permeable phenoxythiophene sulfonamide small molecule in glutamate-induced oxidative injury. Exp Ther Med 2022; 23:79. [PMID: 34938365 PMCID: PMC8688931 DOI: 10.3892/etm.2021.11002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen species (ROS) play a central role in oxidative stress-associated neuronal cell death during ischemia. Further investigation into the inhibition of excessive ROS generation post-stroke is urgently required for the treatment of ischemic stroke. In the present study, the neuroprotective properties of the blood-brain barrier (BBB) penetrant B355227 were investigated. B355227 is a chemical analogue of B355252, and the role of the phenoxythiophene sulfonamide compound B355227 was further investigated in a glutamate-induced oxidative injury model. An in vitro model of the BBB was established in the immortalized mouse brain capillary endothelial cell line, bEnd.3. Formation of barrier in Transwell inserts was confirmed using EVOM resistance meter and Caffeine, Imatinib and Axitinib were used to validate the efficacy of the model. The validated BBB assay in combination with high performance liquid chromatography were used to analyse and verify the permeability of B355227 through the barrier. The integrity of the cell junctions after the BBB assays were confirmed using immunofluorescence to visualize the expression of the barrier junction protein zonula occludens-1. Cell survival was measured with Resazurin, a redox indicator dye, in HT22, a hippocampal neuronal cell treated with 5 mM glutamate or co-treated with the B355227 recovered from the BBB permeability experiment. Changes in glutathione levels were detected using a glutathione detection kit, while analyses of ROS, calcium (Ca2+), and mitochondrial membrane potential (MMP) were accomplished with the fluorescent dyes 2',7'-dichlorofluorescein diacetate, Fura-2 AM and MitoTracker Red dyes, respectively. Immunoblotting was also performed to detect the expression and activation of Erk1/2, p-38, JNK, Bax and Bcl-2. The results of the present study demonstrated that B355227 crossed the BBB in vitro and protected HT22 from oxidative injury induced by glutamate exposure. Treatment of cells with B355227 blocked the glutamate-dependent depletion of intracellular glutathione and significantly reduced ROS production. Increased Ca2+ influx and subsequent collapse of the MMP was attenuated by B355227. Furthermore, the results of the present study demonstrated that B355227 protected against oxidative stress via the MAPK pathway, by increasing the activation of Erk1/2, JNK and P38, and restoring anti-apoptotic Bcl-2. Collectively, the results of the present study indicate that B355227 has potent antioxidant and neuroprotective attributes in glutamate-induced neuronal cell death. Further investigation into the role of B355227 in the modulation of glutamate-dependent oxidative stress is required.
Collapse
Affiliation(s)
- Smritee Pokharel
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Nailya S. Gliyazova
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Srinivasa R. Dandepally
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Alfred L. Williams
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
- Department of Pharmaceutical Science, North Carolina Central University, Durham, NC 27707, USA
| | - Gordon C. Ibeanu
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
- Department of Pharmaceutical Science, North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
15
|
Low-Level Endothelial TRAIL-Receptor Expression Obstructs the CNS-Delivery of Angiopep-2 Functionalised TRAIL-Receptor Agonists for the Treatment of Glioblastoma. Molecules 2021; 26:molecules26247582. [PMID: 34946664 PMCID: PMC8706683 DOI: 10.3390/molecules26247582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant and aggressive form of glioma and is associated with a poor survival rate. Latest generation Tumour Necrosis Factor Related Apoptosis-Inducing Ligand (TRAIL)-based therapeutics potently induce apoptosis in cancer cells, including GBM cells, by binding to death receptors. However, the blood-brain barrier (BBB) is a major obstacle for these biologics to enter the central nervous system (CNS). We therefore investigated if antibody-based fusion proteins that combine hexavalent TRAIL and angiopep-2 (ANG2) moieties can be developed, with ANG2 promoting receptor-mediated transcytosis (RMT) across the BBB. We demonstrate that these fusion proteins retain the potent apoptosis induction of hexavalent TRAIL-receptor agonists. Importantly, blood-brain barrier cells instead remained highly resistant to this fusion protein. Binding studies indicated that ANG2 is active in these constructs but that TRAIL-ANG2 fusion proteins bind preferentially to BBB endothelial cells via the TRAIL moiety. Consequently, transport studies indicated that TRAIL-ANG2 fusion proteins can, in principle, be shuttled across BBB endothelial cells, but that low TRAIL receptor expression on BBB endothelial cells interferes with efficient transport. Our work therefore demonstrates that TRAIL-ANG2 fusion proteins remain highly potent in inducing apoptosis, but that therapeutic avenues will require combinatorial strategies, such as TRAIL-R masking, to achieve effective CNS transport.
Collapse
|
16
|
Lynch MJ, Gobbo OL. Advances in Non-Animal Testing Approaches towards Accelerated Clinical Translation of Novel Nanotheranostic Therapeutics for Central Nervous System Disorders. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2632. [PMID: 34685073 PMCID: PMC8538557 DOI: 10.3390/nano11102632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/21/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022]
Abstract
Nanotheranostics constitute a novel drug delivery system approach to improving systemic, brain-targeted delivery of diagnostic imaging agents and pharmacological moieties in one rational carrier platform. While there have been notable successes in this field, currently, the clinical translation of such delivery systems for the treatment of neurological disorders has been limited by the inadequacy of correlating in vitro and in vivo data on blood-brain barrier (BBB) permeation and biocompatibility of nanomaterials. This review aims to identify the most contemporary non-invasive approaches for BBB crossing using nanotheranostics as a novel drug delivery strategy and current non-animal-based models for assessing the safety and efficiency of such formulations. This review will also address current and future directions of select in vitro models for reducing the cumbersome and laborious mandate for testing exclusively in animals. It is hoped these non-animal-based modelling approaches will facilitate researchers in optimising promising multifunctional nanocarriers with a view to accelerating clinical testing and authorisation applications. By rational design and appropriate selection of characterised and validated models, ranging from monolayer cell cultures to organ-on-chip microfluidics, promising nanotheranostic particles with modular and rational design can be screened in high-throughput models with robust predictive power. Thus, this article serves to highlight abbreviated research and development possibilities with clinical translational relevance for developing novel nanomaterial-based neuropharmaceuticals for therapy in CNS disorders. By generating predictive data for prospective nanomedicines using validated in vitro models for supporting clinical applications in lieu of requiring extensive use of in vivo animal models that have notable limitations, it is hoped that there will be a burgeoning in the nanotherapy of CNS disorders by virtue of accelerated lead identification through screening, optimisation through rational design for brain-targeted delivery across the BBB and clinical testing and approval using fewer animals. Additionally, by using models with tissue of human origin, reproducible therapeutically relevant nanomedicine delivery and individualised therapy can be realised.
Collapse
Affiliation(s)
- Mark J. Lynch
- School of Pharmacy and Pharmaceutical Sciences, Panoz Building, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Oliviero L. Gobbo
- School of Pharmacy and Pharmaceutical Sciences, Panoz Building, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
17
|
Vézina A, Manglani M, Morris D, Foster B, McCord M, Song H, Zhang M, Davis D, Zhang W, Bills J, Nagashima K, Shankarappa P, Kindrick J, Walbridge S, Peer CJ, Figg WD, Gilbert MR, McGavern DB, Muldoon LL, Jackson S. Adenosine A2A Receptor Activation Enhances Blood-Tumor Barrier Permeability in a Rodent Glioma Model. Mol Cancer Res 2021; 19:2081-2095. [PMID: 34521765 DOI: 10.1158/1541-7786.mcr-19-0995] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/16/2020] [Accepted: 09/07/2021] [Indexed: 11/16/2022]
Abstract
The blood-tumor barrier (BTB) limits the entry of effective chemotherapeutic agents into the brain for treatment of malignant tumors like glioblastoma. Poor drug entry across the BTB allows infiltrative glioma stem cells to evade therapy and develop treatment resistance. Regadenoson, an FDA-approved adenosine A2A receptor (A2AR) agonist, has been shown to increase drug delivery across the blood-brain barrier in non-tumor-bearing rodents without a defined mechanism of enhancing BTB permeability. Here, we characterize the time-dependent impact of regadenoson on brain endothelial cell interactions and paracellular transport, using mouse and rat brain endothelial cells and tumor models. In vitro, A2AR activation leads to disorganization of cytoskeletal actin filaments by 30 minutes, downregulation of junctional protein expression by 4 hours, and reestablishment of endothelial cell integrity by 8 hours. In rats bearing intracranial gliomas, regadenoson treatment results in increase of intratumoral temozolomide concentrations, yet no increased survival noted with combined temozolomide therapy. These findings demonstrate regadenoson's ability to induce brain endothelial structural changes among glioma to increase BTB permeability. The use of vasoactive mediators, like regadenoson, which transiently influences paracellular transport, should further be explored to evaluate their potential to enhance central nervous system treatment delivery to aggressive brain tumors. IMPLICATIONS: This study provides insight on the use of a vasoactive agent to increase exposure of the BTB to chemotherapy with intention to improve glioma treatment efficacy.
Collapse
Affiliation(s)
- Amélie Vézina
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland.,Electron Microscope Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Monica Manglani
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - DreeAnna Morris
- Department of Neurology, Oregon Health & Sciences University, Portland, Oregon
| | - Brandon Foster
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | | | - Hua Song
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Meili Zhang
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Dionne Davis
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Wei Zhang
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Jessica Bills
- Department of Neurology, Oregon Health & Sciences University, Portland, Oregon
| | - Kunio Nagashima
- Electron Microscope Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Priya Shankarappa
- Genitourinary Malignancies Branch, Molecular Pharmacology Section, NCI, NIH, Bethesda, Maryland
| | - Jessica Kindrick
- Genitourinary Malignancies Branch, Molecular Pharmacology Section, NCI, NIH, Bethesda, Maryland
| | - Stuart Walbridge
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Cody J Peer
- Genitourinary Malignancies Branch, Molecular Pharmacology Section, NCI, NIH, Bethesda, Maryland
| | - William D Figg
- Genitourinary Malignancies Branch, Molecular Pharmacology Section, NCI, NIH, Bethesda, Maryland
| | | | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Leslie L Muldoon
- Department of Neurology, Oregon Health & Sciences University, Portland, Oregon
| | - Sadhana Jackson
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland. .,Electron Microscope Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
18
|
Mendonça DA, Bakker M, Cruz-Oliveira C, Neves V, Jiménez MA, Defaus S, Cavaco M, Veiga AS, Cadima-Couto I, Castanho MARB, Andreu D, Todorovski T. Penetrating the Blood-Brain Barrier with New Peptide-Porphyrin Conjugates Having anti-HIV Activity. Bioconjug Chem 2021; 32:1067-1077. [PMID: 34033716 PMCID: PMC8485325 DOI: 10.1021/acs.bioconjchem.1c00123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
![]()
Passing
through the blood-brain barrier (BBB) to treat neurological
conditions is one of the main hurdles in modern medicine. Many drugs
with promising in vitro profiles become ineffective in vivo due to
BBB restrictive permeability. In particular, this includes drugs such
as antiviral porphyrins, with the ability to fight brain-resident
viruses causing diseases such as HIV-associated neurocognitive disorders
(HAND). In the last two decades, BBB shuttles, particularly peptide-based
ones, have shown promise in carrying various payloads across the BBB.
Thus, peptide–drug conjugates (PDCs) formed by covalent attachment
of a BBB peptide shuttle and an antiviral drug may become key therapeutic
tools in treating neurological disorders of viral origin. In this
study, we have used various approaches (guanidinium, phosphonium,
and carbodiimide-based couplings) for on-resin synthesis of new peptide–porphyrin
conjugates (PPCs) with BBB-crossing and potential antiviral activity.
After careful fine-tuning of the synthetic chemistry, DIC/oxyma has
emerged as a preferred method, by which 14 different PPCs have been
made and satisfactorily characterized. The PPCs are prepared by coupling
a porphyrin carboxyl group to an amino group (either N-terminal or a Lys side chain) of the peptide shuttle and show effective
in vitro BBB translocation ability, low cytotoxicity toward mouse
brain endothelial cells, and low hemolytic activity. Three of the
PPCs, MP-P5, P4-MP, and P4-L-MP, effectively inhibiting HIV infectivity
in vitro, stand out as most promising. Their efficacy against other
brain-targeting viruses (Dengue, Zika, and SARS-CoV-2) is currently
under evaluation, with preliminary results confirming that PPCs are
a promising strategy to treat viral brain infections.
Collapse
Affiliation(s)
- Diogo A Mendonça
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Mariët Bakker
- Avans University of Applied Sciences, 5223 DE Breda, Netherlands
| | - Christine Cruz-Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Vera Neves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Maria Angeles Jiménez
- Department of Biological Physical Chemistry, Institute of Physical Chemistry Rocasolano (IQFR-CSIC), 28006 Madrid, Spain
| | - Sira Defaus
- Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Marco Cavaco
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Ana Salomé Veiga
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Iris Cadima-Couto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - David Andreu
- Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Toni Todorovski
- Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| |
Collapse
|
19
|
Tonali N, Hericks L, Schröder DC, Kracker O, Krzemieniecki R, Kaffy J, Le Joncour V, Laakkonen P, Marion A, Ongeri S, Dodero VI, Sewald N. Peptidotriazolamers Inhibit Aβ(1-42) Oligomerization and Cross a Blood-Brain-Barrier Model. Chempluschem 2021; 86:840-851. [PMID: 33905181 DOI: 10.1002/cplu.202000814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/07/2021] [Indexed: 12/25/2022]
Abstract
In peptidotriazolamers every second peptide bond is replaced by a 1H-1,2,3-triazole. Such foldamers are expected to bridge the gap in molecular weight between small-molecule drugs and protein-based drugs. Amyloid β (Aβ) aggregates play an important role in Alzheimer's disease. We studied the impact of amide bond replacements by 1,4-disubstituted 1H-1,2,3-triazoles on the inhibitory activity of the aggregation "hot spots" K16 LVFF20 and G39 VVIA42 in Aβ(1-42). We found that peptidotriazolamers act as modulators of the Aβ(1-42) oligomerization. Some peptidotriazolamers are able to interfere with the formation of toxic early Aβ oligomers, depending on the position of the triazoles, which is also supported by computational studies. Preliminary in vitro results demonstrate that a highly active peptidotriazolamer is also able to cross the blood-brain-barrier.
Collapse
Affiliation(s)
- Nicolo Tonali
- Organic and Bioorganic Chemistry, Department of Chemistry Bielefeld University, PO Box, 100131, 33501, Bielefeld, Germany.,BioCIS, CNRS, Université Paris Saclay, 5 rue Jean-Baptiste Clément, 92296, Châtenay-Malabry, France
| | - Loreen Hericks
- Organic and Bioorganic Chemistry, Department of Chemistry Bielefeld University, PO Box, 100131, 33501, Bielefeld, Germany
| | - David C Schröder
- Organic and Bioorganic Chemistry, Department of Chemistry Bielefeld University, PO Box, 100131, 33501, Bielefeld, Germany
| | - Oliver Kracker
- Organic and Bioorganic Chemistry, Department of Chemistry Bielefeld University, PO Box, 100131, 33501, Bielefeld, Germany
| | - Radosław Krzemieniecki
- Organic and Bioorganic Chemistry, Department of Chemistry Bielefeld University, PO Box, 100131, 33501, Bielefeld, Germany
| | - Julia Kaffy
- BioCIS, CNRS, Université Paris Saclay, 5 rue Jean-Baptiste Clément, 92296, Châtenay-Malabry, France
| | - Vadim Le Joncour
- Research Programs Unit, Translational Cancer Medicine Research Program, University of Helsinki, 00014, Helsinki, Finland
| | - Pirjo Laakkonen
- Research Programs Unit, Translational Cancer Medicine Research Program, University of Helsinki, 00014, Helsinki, Finland
| | - Antoine Marion
- Department of Chemistry, Middle East Technical University, 06800, Ankara, Turkey
| | - Sandrine Ongeri
- BioCIS, CNRS, Université Paris Saclay, 5 rue Jean-Baptiste Clément, 92296, Châtenay-Malabry, France
| | - Veronica I Dodero
- Organic and Bioorganic Chemistry, Department of Chemistry Bielefeld University, PO Box, 100131, 33501, Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Department of Chemistry Bielefeld University, PO Box, 100131, 33501, Bielefeld, Germany
| |
Collapse
|
20
|
Desmarais F, Hervé V, Bergeron KF, Ravaut G, Perrotte M, Fyfe-Desmarais G, Rassart E, Ramassamy C, Mounier C. Cerebral Apolipoprotein D Exits the Brain and Accumulates in Peripheral Tissues. Int J Mol Sci 2021; 22:ijms22084118. [PMID: 33923459 PMCID: PMC8073497 DOI: 10.3390/ijms22084118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Apolipoprotein D (ApoD) is a secreted lipocalin associated with neuroprotection and lipid metabolism. In rodent, the bulk of its expression occurs in the central nervous system. Despite this, ApoD has profound effects in peripheral tissues, indicating that neural ApoD may reach peripheral organs. We endeavor to determine if cerebral ApoD can reach the circulation and accumulate in peripheral tissues. Three hours was necessary for over 40% of all the radiolabeled human ApoD (hApoD), injected bilaterally, to exit the central nervous system (CNS). Once in circulation, hApoD accumulates mostly in the kidneys/urine, liver, and muscles. Accumulation specificity of hApoD in these tissues was strongly correlated with the expression of lowly glycosylated basigin (BSG, CD147). hApoD was observed to pass through bEnd.3 blood brain barrier endothelial cells monolayers. However, cyclophilin A did not impact hApoD internalization rates in bEnd.3, indicating that ApoD exit from the brain is either independent of BSG or relies on additional cell types. Overall, our data showed that ApoD can quickly and efficiently exit the CNS and reach the liver and kidneys/urine, organs linked to the recycling and excretion of lipids and toxins. This indicated that cerebral overexpression during neurodegenerative episodes may serve to evacuate neurotoxic ApoD ligands from the CNS.
Collapse
Affiliation(s)
- Frederik Desmarais
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Vincent Hervé
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
| | - Karl F. Bergeron
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
| | - Gaétan Ravaut
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
| | - Morgane Perrotte
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
| | - Guillaume Fyfe-Desmarais
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Eric Rassart
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Charles Ramassamy
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
- Correspondence: (C.R.); (C.M.)
| | - Catherine Mounier
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Correspondence: (C.R.); (C.M.)
| |
Collapse
|
21
|
Prashanth A, Donaghy H, Stoner SP, Hudson AL, Wheeler HR, Diakos CI, Howell VM, Grau GE, McKelvey KJ. Are In Vitro Human Blood-Brain-Tumor-Barriers Suitable Replacements for In Vivo Models of Brain Permeability for Novel Therapeutics? Cancers (Basel) 2021; 13:955. [PMID: 33668807 PMCID: PMC7956470 DOI: 10.3390/cancers13050955] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND High grade gliomas (HGG) are incapacitating and prematurely fatal diseases. To overcome the poor prognosis, novel therapies must overcome the selective and restricted permeability of the blood-brain barrier (BBB). This study critically evaluated whether in vitro human normal BBB and tumor BBB (BBTB) are suitable alternatives to "gold standard" in vivo models to determine brain permeability. METHODS A systematic review utilizing the PRISMA guidelines used English and full-text articles from the past 5 years in the PubMed, Embase, Medline and Scopus databases. Experimental studies employing human cell lines were included. RESULTS Of 1335 articles, the search identified 24 articles for evaluation after duplicates were removed. Eight in vitro and five in vivo models were identified with the advantages and disadvantages compared within and between models, and against patient clinical data where available. The greatest in vitro barrier integrity and stability, comparable to in vivo and clinical permeability data, were achieved in the presence of all cell types of the neurovascular unit: endothelial cells, astrocytes/glioma cells, pericytes and neurons. CONCLUSIONS In vitro co-culture BBB models utilizing stem cell-derived or primary cells are a suitable proxy for brain permeability studies in order to reduce animal use in medical research.
Collapse
Affiliation(s)
- Archana Prashanth
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Heather Donaghy
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Shihani P. Stoner
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Amanda L. Hudson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Helen R. Wheeler
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
- Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Connie I. Diakos
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
- Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Viive M. Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Georges E. Grau
- Vascular Immunology, Department of Pathology, School of Pathology, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia;
| | - Kelly J. McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| |
Collapse
|
22
|
Taïlé J, Patché J, Veeren B, Gonthier MP. Hyperglycemic Condition Causes Pro-Inflammatory and Permeability Alterations Associated with Monocyte Recruitment and Deregulated NFκB/PPARγ Pathways on Cerebral Endothelial Cells: Evidence for Polyphenols Uptake and Protective Effect. Int J Mol Sci 2021; 22:ijms22031385. [PMID: 33573189 PMCID: PMC7866545 DOI: 10.3390/ijms22031385] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Hyperglycemia alters the function of cerebral endothelial cells from the blood-brain barrier, increasing the risk of cerebrovascular complications during diabetes. This study evaluated the protective effect of polyphenols on inflammatory and permeability markers on bEnd3 cerebral endothelial cells exposed to high glucose concentration. Results show that hyperglycemic condition increased nuclear factor kappa B (NFκB) activity, deregulated the expression of interleukin-1 beta (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), interleukin-10 (IL-10) and endothelial-leukocyte adhesion molecule (E-selectin) genes, raised MCP-1 secretion and elevated monocyte adhesion and transendothelial migration. High glucose decreased occludin, claudin-5, zona occludens-1 (ZO-1) and zona occludens-2 (ZO-2) tight junctions production and altered the endothelial permeability. Characterized polyphenolic extracts from the French medicinal plants Antirhea borbonica, Ayapana triplinervis, Dodonaea viscosa and Terminalia bentzoe, and their major polyphenols quercetin, caffeic, chlorogenic and gallic acids limited the pro-inflammatory and permeability alterations caused by high glucose. Peroxisome proliferator-activated receptor gamma (PPARγ) agonist also attenuated these damages while PPARγ antagonist aggravated them, suggesting PPARγ protective action. Interestingly, polyphenols improved PPARγ gene expression lowered by high glucose. Moreover, polyphenols were detected at the intracellular level or membrane-bound to cells, with evidence for breast cancer resistance protein (BCRP) efflux transporter role. Altogether, these findings emphasize the ability of polyphenols to protect cerebral endothelial cells in hyperglycemic condition and their relevance for pharmacological strategies aiming to limit cerebrovascular disorders in diabetes.
Collapse
|
23
|
Lin Q, Wang W, Yang L, Duan X. 4-Methoxybenzylalcohol protects brain microvascular endothelial cells against oxygen-glucose deprivation/reperfusion-induced injury via activation of the PI3K/AKT signaling pathway. Exp Ther Med 2021; 21:252. [PMID: 33613705 PMCID: PMC7856387 DOI: 10.3892/etm.2021.9684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/02/2020] [Indexed: 11/07/2022] Open
Abstract
Damage to the blood-brain barrier (BBB) during the process of cerebral ischemic injury is a key factor that affects the treatment of this condition. The present study aimed to assess the potential effects of 4-methoxybenzyl alcohol (4-MA) on brain microvascular endothelial cells (bEnd.3) against oxygen-glucose deprivation/reperfusion (OGD/Rep) using an in vitro model that mimics in vivo ischemia/reperfusion injury. In addition, the present study aimed to explore whether this underlying mechanism was associated with the inhibition of pro-inflammatory factors and the activation status of the PI3K/Akt signaling pathway. bEnd.3 cells were subjected to OGD/Rep-induced injury before being treated with 4-MA, following which cell viability, lactate dehydrogenase (LDH) release and levels of nitric oxidase (NO) were detected by colorimetry, pro-inflammatory factors including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6, were detected by ELISA. The expression levels of occluding and claudin-5were evaluated by immunofluorescence staining. The expression levels of AKT, phosphorylated (p)-Akt, endothelial nitric oxide synthase (eNOS) and p-eNOS were also measured by western blot analysis. After bEnd.3 cells were subjected to OGD/Rep-induced injury, cell viability and NO levels were significantly decreased, whilst LDH leakage and inflammatory factor (TNF-α, IL-1β and IL-6) levels were significantly increased. Treatment with 4-MA significantly ameliorated cell viability, LDH release and the levels of NO and pro-inflammatory factors TNF-α, IL-1β and IL-6 as a result of OGD/Rep. Furthermore, treatment with 4-MA upregulated the expression of occludin, claudin-5, Akt and eNOS, in addition to increasing eNOS and AKT phosphorylation in bEnd.3 cells. These results suggest that 4-MA can alleviate OGD/Rep-induced injury in bEnd.3 cells by inhibiting inflammation and by activating the PI3K/AKT signaling pathway as a possible mechanism. Therefore, 4-MA can serve as a potential candidate for treating OGD/Rep-induced injury.
Collapse
Affiliation(s)
- Qing Lin
- Department of Pharmacology, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Weili Wang
- Department of Pharmacology, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Liping Yang
- Yunnan Key Laboratory of Dai and Yi Medicine, University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicine, University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
24
|
Hwang JS, Cha EH, Park B, Ha E, Seo JH. PBN inhibits a detrimental effect of methamphetamine on brain endothelial cells by alleviating the generation of reactive oxygen species. Arch Pharm Res 2020; 43:1347-1355. [PMID: 33200316 DOI: 10.1007/s12272-020-01284-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/04/2020] [Indexed: 12/20/2022]
Abstract
Methamphetamine (METH) is a powerful psychostimulant that is causing serious health problems worldwide owing to imprudent abuses. Recent studies have suggested that METH has deleterious effects on the blood-brain barrier (BBB). A few studies have also been conducted on the mechanisms whereby METH-induced oxidative stress causes BBB dysfunction. We investigated whether N-tert-butyl-α-phenylnitrone (PBN) has protective effects on BBB function against METH exposure in primary human brain microvascular endothelial cells (HBMECs). We found that METH significantly increased reactive oxygen species (ROS) generation in HBMECs. Pretreatment with PBN decreased METH-induced ROS production. With regard to BBB functional integrity, METH exposure elevated the paracellular permeability and reduced the monolayer integrity; PBN treatment reversed these effects. An analysis of the BBB structural properties, by immunostaining junction proteins and cytoskeleton in HBMECs, indicated that METH treatment changed the cellular localization of the tight (ZO-1) and adherens junctions (VE-cadherin) from the membrane to cytoplasm. Furthermore, METH induced cytoskeletal reorganization via the formation of robust stress fibers. METH-induced junctional protein redistribution and cytoskeletal reorganization were attenuated by PBN treatment. Our results suggest that PBN can act as a therapeutic reagent for METH-induced BBB dysfunction by inhibiting excess ROS generation.
Collapse
Affiliation(s)
- Jong Su Hwang
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Eun Hye Cha
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Eunyoung Ha
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea.
| | - Ji Hae Seo
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
25
|
Venn AA, Bernardet C, Chabenat A, Tambutté E, Tambutté S. Paracellular transport to the coral calcifying medium: effects of environmental parameters. J Exp Biol 2020; 223:jeb227074. [PMID: 32675232 DOI: 10.1242/jeb.227074] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
Coral calcification relies on the transport of ions and molecules to the extracellular calcifying medium (ECM). Little is known about paracellular transport (via intercellular junctions) in corals and other marine calcifiers. Here, we investigated whether the permeability of the paracellular pathway varied in different environmental conditions in the coral Stylophora pistillata Using the fluorescent dye calcein, we characterised the dynamics of calcein influx from seawater to the ECM and showed that increases in paracellular permeability (leakiness) induced by hyperosmotic treatment could be detected by changes in calcein influx rates. We then used the calcein-imaging approach to investigate the effects of two environmental stressors on paracellular permeability: seawater acidification and temperature change. Under conditions of seawater acidification (pH 7.2) known to depress pH in the ECM and the calcifying cells of S. pistillata, we observed a decrease in half-times of calcein influx, indicating increased paracellular permeability. By contrast, high temperature (31°C) had no effect, whereas low temperature (20°C) caused decreases in paracellular permeability. Overall, our study establishes an approach to conduct further in vivo investigation of paracellular transport and suggests that changes in paracellular permeability could form an uncharacterised aspect of the physiological response of S. pistillata to seawater acidification.
Collapse
Affiliation(s)
- Alexander A Venn
- Marine Biology Department, Centre Scientifique de Monaco, 8 Quai Antoine 1er, 98000 Monaco
| | - Coralie Bernardet
- Marine Biology Department, Centre Scientifique de Monaco, 8 Quai Antoine 1er, 98000 Monaco
| | - Apolline Chabenat
- Marine Biology Department, Centre Scientifique de Monaco, 8 Quai Antoine 1er, 98000 Monaco
| | - Eric Tambutté
- Marine Biology Department, Centre Scientifique de Monaco, 8 Quai Antoine 1er, 98000 Monaco
| | - Sylvie Tambutté
- Marine Biology Department, Centre Scientifique de Monaco, 8 Quai Antoine 1er, 98000 Monaco
| |
Collapse
|
26
|
Norouzi M, Yathindranath V, Thliveris JA, Kopec BM, Siahaan TJ, Miller DW. Doxorubicin-loaded iron oxide nanoparticles for glioblastoma therapy: a combinational approach for enhanced delivery of nanoparticles. Sci Rep 2020; 10:11292. [PMID: 32647151 PMCID: PMC7347880 DOI: 10.1038/s41598-020-68017-y] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/16/2020] [Indexed: 01/05/2023] Open
Abstract
Although doxorubicin (DOX) is an effective anti-cancer drug with cytotoxicity in a variety of different tumors, its effectiveness in treating glioblastoma multiforme (GBM) is constrained by insufficient penetration across the blood–brain barrier (BBB). In this study, biocompatible magnetic iron oxide nanoparticles (IONPs) stabilized with trimethoxysilylpropyl-ethylenediamine triacetic acid (EDT) were developed as a carrier of DOX for GBM chemotherapy. The DOX-loaded EDT-IONPs (DOX-EDT-IONPs) released DOX within 4 days with the capability of an accelerated release in acidic microenvironments. The DOX-loaded EDT-IONPs (DOX-EDT-IONPs) demonstrated an efficient uptake in mouse brain-derived microvessel endothelial, bEnd.3, Madin–Darby canine kidney transfected with multi-drug resistant protein 1 (MDCK-MDR1), and human U251 GBM cells. The DOX-EDT-IONPs could augment DOX’s uptake in U251 cells by 2.8-fold and significantly inhibited U251 cell proliferation. Moreover, the DOX-EDT-IONPs were found to be effective in apoptotic-induced GBM cell death (over 90%) within 48 h of treatment. Gene expression studies revealed a significant downregulation of TOP II and Ku70, crucial enzymes for DNA repair and replication, as well as MiR-155 oncogene, concomitant with an upregulation of caspase 3 and tumor suppressors i.e., p53, MEG3 and GAS5, in U251 cells upon treatment with DOX-EDT-IONPs. An in vitro MDCK-MDR1-GBM co-culture model was used to assess the BBB permeability and anti-tumor activity of the DOX-EDT-IONPs and DOX treatments. While DOX-EDT-IONP showed improved permeability of DOX across MDCK-MDR1 monolayers compared to DOX alone, cytotoxicity in U251 cells was similar in both treatment groups. Using a cadherin binding peptide (ADTC5) to transiently open tight junctions, in combination with an external magnetic field, significantly enhanced both DOX-EDT-IONP permeability and cytotoxicity in the MDCK-MDR1-GBM co-culture model. Therefore, the combination of magnetic enhanced convective diffusion and the cadherin binding peptide for transiently opening the BBB tight junctions are expected to enhance the efficacy of GBM chemotherapy using the DOX-EDT-IONPs. In general, the developed approach enables the chemotherapeutic to overcome both BBB and multidrug resistance (MDR) glioma cells while providing site-specific magnetic targeting.
Collapse
Affiliation(s)
- Mohammad Norouzi
- Department of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba, A205 Chown Bldg., 753 McDermot Avenue, Winnipeg, MB, Canada
| | - Vinith Yathindranath
- Department of Pharmacology and Therapeutics, University of Manitoba, A205 Chown Bldg., 753 McDermot Avenue, Winnipeg, MB, Canada
| | - James A Thliveris
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Brian M Kopec
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Teruna J Siahaan
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Donald W Miller
- Department of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada. .,Department of Pharmacology and Therapeutics, University of Manitoba, A205 Chown Bldg., 753 McDermot Avenue, Winnipeg, MB, Canada.
| |
Collapse
|
27
|
UV irradiation of Type I collagen gels changed the morphology of the interconnected brain capillary endothelial cells on them. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 112:110907. [PMID: 32409061 DOI: 10.1016/j.msec.2020.110907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 03/07/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022]
Abstract
We cultured mouse brain capillary endothelial cell line bEnd.3 on the UV-irradiated Type I collagen gel. Morphology of bEnd.3 cells on the Type I collagen gel was drastically changed if the gel was crosslinked by UV irradiation. The interconnecting network of bEnd.3 cells which have cord-like morphology on the soft collagen gels was converted to the monolayer of the flat cells, tightly-bound each other covering the gel surface, in a confluent state. The collagen gels were mechanically stiffened by UV irradiation for 15 min with UV light at 254 nm showing approximately two times higher value of Young's modulus E (1.51 ± 0.58 kPa) than the control gel (3.17 ± 1.17 kPa). AFM images of the collagen fibrils were not severely changed after irradiation. Collagen subunit proteins were crosslinked and degraded simultaneously under UV irradiation proved by results of SDS-PAGE and separation by centrifugation. Expression of Integrin gene was measured by quantitative real-time PCR. Expression of the integrin α2 gene, tight junction protein 1 gene, and claudin 5 gene were down-regulated in cells on the UV irradiated collagen gel in comparison with the unirradiated one while expression of the integrin β1 gene and Integrin α1 gene did not significantly change. Thick actin filaments were more clearly observed in the cells on the UV-irradiated collagen gel than the unirradiated one by fluorescent microscopy. We conclude that UV irradiation made the collagen gel stiffened and changed the physiological state of bEnd.3 cells including their adhesion, extension, and proliferation.
Collapse
|
28
|
Suzuki T, Yasumoto M, Suzuki Y, Asai K, Imaizumi Y, Yamamura H. TMEM16A Ca 2+-Activated Cl - Channel Regulates the Proliferation and Migration of Brain Capillary Endothelial Cells. Mol Pharmacol 2020; 98:61-71. [PMID: 32358165 DOI: 10.1124/mol.119.118844] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 04/20/2020] [Indexed: 11/22/2022] Open
Abstract
The blood-brain barrier (BBB) is essential for the maintenance of homeostasis in the brain. Brain capillary endothelial cells (BCECs) comprise the BBB, and thus a delicate balance between their proliferation and death is required. Although the activity of ion channels in BCECs is involved in BBB functions, the underlying molecular mechanisms remain unclear. In the present study, the molecular components of Ca2+-activated Cl- (ClCa) channels and their physiological roles were examined using mouse BCECs (mBCECs) and a cell line derived from bovine BCECs, t-BBEC117. Expression analyses revealed that TMEM16A was strongly expressed in mBCECs and t-BBEC117 cells. In t-BBEC117 cells, whole-cell Cl- currents were sensitive to the ClCa channel blockers, 100 μM niflumic acid and 10 μM T16Ainh-A01, and were also reduced markedly by small-interfering RNA (siRNA) knockdown of TMEM16A. Importantly, block of ClCa currents with ClCa channel blockers or TMEM16A siRNA induced membrane hyperpolarization. Moreover, treatment with TMEM16A siRNA caused an increase in resting cytosolic Ca2+ concentration ([Ca2+]cyt). T16Ainh-A01 reduced cell viability in a concentration-dependent manner. Either ClCa channel blockers or TMEM16A siRNA also curtailed cell proliferation and migration. Furthermore, ClCa channel blockers attenuated the trans-endothelial permeability. In combination, these results strongly suggest that TMEM16A contributes to ClCa channel conductance and can regulate both the resting membrane potential and [Ca2+]cyt in BCECs. Our data also reveal how these BCECs may be involved in the maintenance of BBB functions, as both the proliferation and migration are altered following changes in channel activity. SIGNIFICANCE STATEMENT: In brain capillary endothelial cells (BCECs) of the blood-brain barrier (BBB), TMEM16A is responsible for Ca2+-activated Cl- channels and can regulate both the resting membrane potential and cytosolic Ca2+ concentration, contributing to the proliferation and migration of BCECs. The present study provides novel information on the molecular mechanisms underlying the physiological functions of BCECs in the BBB and a novel target for therapeutic drugs for disorders associated with dysfunctions in the BBB.
Collapse
Affiliation(s)
- Takahisa Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences (T.S., M.Y., Y.S., Y.I., H.Y.) and Department of Molecular Neurobiology, Graduate School of Medical Sciences (K.A.), Nagoya City University, Nagoya, Japan
| | - Miki Yasumoto
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences (T.S., M.Y., Y.S., Y.I., H.Y.) and Department of Molecular Neurobiology, Graduate School of Medical Sciences (K.A.), Nagoya City University, Nagoya, Japan
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences (T.S., M.Y., Y.S., Y.I., H.Y.) and Department of Molecular Neurobiology, Graduate School of Medical Sciences (K.A.), Nagoya City University, Nagoya, Japan
| | - Kiyofumi Asai
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences (T.S., M.Y., Y.S., Y.I., H.Y.) and Department of Molecular Neurobiology, Graduate School of Medical Sciences (K.A.), Nagoya City University, Nagoya, Japan
| | - Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences (T.S., M.Y., Y.S., Y.I., H.Y.) and Department of Molecular Neurobiology, Graduate School of Medical Sciences (K.A.), Nagoya City University, Nagoya, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences (T.S., M.Y., Y.S., Y.I., H.Y.) and Department of Molecular Neurobiology, Graduate School of Medical Sciences (K.A.), Nagoya City University, Nagoya, Japan
| |
Collapse
|
29
|
Aoki H, Yamashita M, Hashita T, Iwao T, Matsunaga T. Laminin 221 fragment is suitable for the differentiation of human induced pluripotent stem cells into brain microvascular endothelial-like cells with robust barrier integrity. Fluids Barriers CNS 2020; 17:25. [PMID: 32228708 PMCID: PMC7106710 DOI: 10.1186/s12987-020-00186-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/17/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND In vitro blood-brain barrier (BBB) models using human induced pluripotent stem (iPS) cell-derived brain microvascular endothelial-like cells (iBMELCs) have been developed to predict the BBB permeability of drug candidates. For the differentiation of iBMELCs, Matrigel, which is a gelatinous protein mixture, is often used as a coating substrate. However, the components of Matrigel can vary among lots, as it is obtained from mouse sarcoma cells with the use of special technics and also contains various basement membranes. Therefore, fully defined substrates as substitutes for Matrigel are needed for a stable supply of iBMELCs with less variation among lots. METHODS iBMELCs were differentiated from human iPS cells on several matrices. The barrier integrity of iBMELCs was evaluated based on transendothelial electrical resistance (TEER) values and permeability of fluorescein isothiocyanate-dextran 4 kDa (FD4) and Lucifer yellow (LY). Characterization of iBMELCs was conducted by RT-qPCR and immunofluorescence analysis. Functions of efflux transporters were defined by intracellular accumulation of the substrates in the wells of multiwell plates. RESULTS iBMELCs differentiated on laminin 221 fragment (LN221F-iBMELCs) had higher TEER values and lower permeability of LY and FD4 as compared with iBMELCs differentiated on Matrigel (Matrigel-iBMELCs). Besides, the gene and protein expression levels of brain microvascular endothelial cells (BMEC)-related markers were similar between LN221F-iBMELCs and Matrigel-iBMELCs. Moreover, both Matrigel- and LN221F-iBMELCs had functions of P-glycoprotein and breast cancer resistance protein, which are essential efflux transporters for barrier functions of the BBB. CONCLUSION The fully defined substrate LN221F presents as an optimal coating matrix for differentiation of iBMELCs. The LN221F-iBMELCs had more robust barrier function for a longer period than Matrigel-iBMELCs with characteristics of BMECs. This finding will contribute the establishment of an iBMELC supply system for pharmacokinetic and pathological models of the BBB.
Collapse
Affiliation(s)
- Hiromasa Aoki
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Misaki Yamashita
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Tadahiro Hashita
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Takahiro Iwao
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan.
| |
Collapse
|
30
|
Alamu O, Rado M, Ekpo O, Fisher D. Differential Sensitivity of Two Endothelial Cell Lines to Hydrogen Peroxide Toxicity: Relevance for In Vitro Studies of the Blood-Brain Barrier. Cells 2020; 9:cells9020403. [PMID: 32050666 PMCID: PMC7072657 DOI: 10.3390/cells9020403] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/20/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress (OS) has been linked to blood–brain barrier (BBB) dysfunction which in turn has been implicated in the initiation and propagation of some neurological diseases. In this study, we profiled, for the first time, two endothelioma cell lines of mouse brain origin, commonly used as in vitro models of the blood–brain barrier, for their resistance against oxidative stress using viability measures and glutathione contents as markers. OS was induced by exposing cultured cells to varying concentrations of hydrogen peroxide and fluorescence microscopy/spectrometry was used to detect and estimate cellular glutathione contents. A colorimetric viability assay was used to determine changes in the viability of OS-exposed cells. Both the b.End5 and bEnd.3 cell lines investigated showed demonstrable content of glutathione with a statistically insignificant difference in glutathione quantity per unit cell, but with a statistically significant higher capacity for the b.End5 cell line for de novo glutathione synthesis. Furthermore, the b.End5 cells demonstrated greater oxidant buffering capacity to higher concentrations of hydrogen peroxide than the bEnd.3 cells. We concluded that mouse brain endothelial cells, derived from different types of cell lines, differ enormously in their antioxidant characteristics. We hereby recommend caution in making comparisons across BBB models utilizing distinctly different cell lines and require further prerequisites to ensure that in vitro BBB models involving these cell lines are reliable and reproducible.
Collapse
Affiliation(s)
- Olufemi Alamu
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa; (O.A.); (M.R.); (O.E.)
- Anatomy Department, Ladoke Akintola University of Technology, Ogbomoso 210241, Nigeria
| | - Mariam Rado
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa; (O.A.); (M.R.); (O.E.)
| | - Okobi Ekpo
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa; (O.A.); (M.R.); (O.E.)
| | - David Fisher
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa; (O.A.); (M.R.); (O.E.)
- Correspondence: ; Tel.: +27-21-959-2185
| |
Collapse
|
31
|
Macdonald J, Denoyer D, Henri J, Jamieson A, Burvenich IJ, Pouliot N, Shigdar S. Bifunctional Aptamer-Doxorubicin Conjugate Crosses the Blood-Brain Barrier and Selectively Delivers Its Payload to EpCAM-Positive Tumor Cells. Nucleic Acid Ther 2020; 30:117-128. [PMID: 32027209 PMCID: PMC7133447 DOI: 10.1089/nat.2019.0807] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The prognosis for breast cancer patients diagnosed with brain metastases is poor, with survival time measured merely in months. This can largely be attributed to the limited treatment options capable of reaching the tumor as a result of the highly restrictive blood-brain barrier (BBB). While methods of overcoming this barrier have been developed and employed with current treatment options, the majority are highly invasive and nonspecific, leading to severe neurotoxic side effects. A novel approach to address these issues is the development of therapeutics targeting receptor-mediated transport mechanisms on the BBB endothelial cell membranes. Using this approach, we intercalated doxorubicin (DOX) into a bifunctional aptamer targeting the transferrin receptor on the BBB and epithelial cell adhesion molecule (EpCAM) on metastatic cancer cells. The ability of the DOX-loaded aptamer to transcytose the BBB and selectively deliver the payload to EpCAM-positive tumors was evaluated in an in vitro model and confirmed for the first time in vivo using the MDA-MB-231 breast cancer metastasis model (MDA-MB-231Br). We show that colocalized aptamer and DOX are clearly detectable within the brain lesions 75 min postadministration. Collectively, results from this study demonstrate that through intercalation of a cytotoxic drug into the bifunctional aptamer, a therapeutic delivery vehicle can be developed for specific targeting of EpCAM-positive brain metastases.
Collapse
Affiliation(s)
- Joanna Macdonald
- School of Medicine, Deakin University, Geelong, Australia
- Centre for Molecular and Medical Research, Deakin University, Geelong, Australia
| | - Delphine Denoyer
- Matrix Microenvironment and Metastasis Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | - Justin Henri
- School of Medicine, Deakin University, Geelong, Australia
| | | | - Ingrid J.G. Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Normand Pouliot
- Matrix Microenvironment and Metastasis Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- Department of Pathology, The University of Melbourne, Parkville, Australia
| | - Sarah Shigdar
- School of Medicine, Deakin University, Geelong, Australia
- Centre for Molecular and Medical Research, Deakin University, Geelong, Australia
- Address correspondence to: Sarah Shigdar, MSc, PhD, School of Medicine, Deakin University, Geelong, Victoria 3216, Australia
| |
Collapse
|
32
|
Li X, Yang Y, Zhao H, Zhu T, Yang Z, Xu H, Fu Y, Lin F, Pan X, Li L, Cui C, Hong M, Yang L, Wang KK, Tan W. Enhanced in Vivo Blood–Brain Barrier Penetration by Circular Tau–Transferrin Receptor Bifunctional Aptamer for Tauopathy Therapy. J Am Chem Soc 2020; 142:3862-3872. [DOI: 10.1021/jacs.9b11490] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Xiaowei Li
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Yu Yang
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Hengzhi Zhao
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Tian Zhu
- Department of Emergency Medicine, Department of Neuroscience, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Zhihui Yang
- Department of Emergency Medicine, Department of Neuroscience, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Haiyan Xu
- Department of Emergency Medicine, Department of Neuroscience, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Yueqiang Fu
- Department of Emergency Medicine, Department of Neuroscience, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Fan Lin
- Department of Emergency Medicine, Department of Neuroscience, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Xiaoshu Pan
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Long Li
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Cheng Cui
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Min Hong
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong 252059, China
| | - Lu Yang
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| | - Kevin K. Wang
- Department of Emergency Medicine, Department of Neuroscience, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
- Brain Rehabilitation Research Center (BRRC), Malcom Randall Veterans Affairs Medical Center, 1601 SW Archer Road, Gainesville Florida 32608, United States
| | - Weihong Tan
- Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences; The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
33
|
Lan P, Du M, Teng Y, Banwell MG, Nie H, Reaney MJT, Wang Y. Structural Modifications of a Flaxseed Lignan in Pursuit of Higher Liposolubility: Evaluation of the Antioxidant and Permeability Properties of the Resulting Derivatives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:14152-14159. [PMID: 31747278 DOI: 10.1021/acs.jafc.9b06264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
While lignans and their biogenetic precursors can have various health benefits, the poor liposolubilities of such phenolic systems have restricted their application as antioxidants in the food industry. The research reported here was aimed at addressing these matters through derivatizing certain forms of such compounds and then assessing their properties as potential nutraceuticals. In particular, crude flaxseed lignan was purified to afford secoisolariciresinol diglucoside (SDG, 1) that was then subjected to structural modification. By such means, the SDG long-chain fatty acid esters 4-9 and 11-13, the fully acetylated SDG 10, secoisolariciresinol (SECO, 2), and anhydrosecoisolariciresinol (ASECO, 14) were obtained. The antioxidant activities of these derivatives were determined while their permeability properties were evaluated. Such studies revealed that certain SDG derivatives possessing useful liposolubilities also retained their antioxidative properties, as well as being capable of permeating Caco-2 cell monolayers while being nontoxic to them. SDG fatty acid esters 4-9 and 11-13 could be developed into emulsifiers with enhanced health benefits, especially considering their improved antioxidative (ca. <11 000 μmol Trolox/g) and permeability properties. This study thus highlights strategies for the structural modification of SDG so as to generate derivatives with superior properties in terms of their utility in the food and pharmaceutical industries.
Collapse
Affiliation(s)
| | - Muxiang Du
- National R&D Center for Freshwater Fish Processing , Jiangxi Normal University , Nanchang , Jiangxi 330022 , China
| | | | - Martin G Banwell
- Research School of Chemistry, Institute of Advanced Studies , The Australian National University , Canberra , Australian Capital Territory 2601 , Australia
| | | | - Martin J T Reaney
- Department of Plant Sciences , University of Saskatchewan , 51 Campus Dr. , Saskatoon , Saskatchewan S7N 5A8 , Canada
| | | |
Collapse
|
34
|
Joshi U, Pearson A, Evans JE, Langlois H, Saltiel N, Ojo J, Klimas N, Sullivan K, Keegan AP, Oberlin S, Darcey T, Cseresznye A, Raya B, Paris D, Hammock B, Vasylieva N, Hongsibsong S, Stern LJ, Crawford F, Mullan M, Abdullah L. A permethrin metabolite is associated with adaptive immune responses in Gulf War Illness. Brain Behav Immun 2019; 81:545-559. [PMID: 31325531 PMCID: PMC7155744 DOI: 10.1016/j.bbi.2019.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/17/2019] [Accepted: 07/11/2019] [Indexed: 10/31/2022] Open
Abstract
Gulf War Illness (GWI), affecting 30% of veterans from the 1991 Gulf War (GW), is a multi-symptom illness with features similar to those of patients with autoimmune diseases. The objective of the current work is to determine if exposure to GW-related pesticides, such as permethrin (PER), activates peripheral and central nervous system (CNS) adaptive immune responses. In the current study, we focused on a PER metabolite, 3-phenoxybenzoic acid (3-PBA), as this is a common metabolite previously shown to form adducts with endogenous proteins. We observed the presence of 3-PBA and 3-PBA modified lysine of protein peptides in the brain, blood and liver of pyridostigmine bromide (PB) and PER (PB+PER) exposed mice at acute and chronic post-exposure timepoints. We tested whether 3-PBA-haptenated albumin (3-PBA-albumin) can activate immune cells since it is known that chemically haptenated proteins can stimulate immune responses. We detected autoantibodies against 3-PBA-albumin in plasma from PB + PER exposed mice and veterans with GWI at chronic post-exposure timepoints. We also observed that in vitro treatment of blood with 3-PBA-albumin resulted in the activation of B- and T-helper lymphocytes and that these immune cells were also increased in blood of PB + PER exposed mice and veterans with GWI. These immune changes corresponded with elevated levels of infiltrating monocytes in the brain and blood of PB + PER exposed mice which coincided with alterations in the markers of blood-brain barrier disruption, brain macrophages and neuroinflammation. These studies suggest that pesticide exposure associated with GWI may have resulted in the activation of the peripheral and CNS adaptive immune responses, possibly contributing to an autoimmune-type phenotype in veterans with GWI.
Collapse
Affiliation(s)
- Utsav Joshi
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Andrew Pearson
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - James E. Evans
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Heather Langlois
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Nicole Saltiel
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Joseph Ojo
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Nancy Klimas
- NOVA Southeastern University, Ft. Lauderdale, FL, USA,Miami VAMC, Miami, FL, USA
| | | | | | - Sarah Oberlin
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Teresa Darcey
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Adam Cseresznye
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Balaram Raya
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Daniel Paris
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Bruce Hammock
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Natalia Vasylieva
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Surat Hongsibsong
- Environment and Health Research Unit, Research Institute for Health Science, Chiang Mai University, Chiang, Thailand
| | - Lawrence J. Stern
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA,Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Fiona Crawford
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Michael Mullan
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Laila Abdullah
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA; Open University, Milton Keynes, UK; James A. Haley VA Hospital, Tampa, FL, USA.
| |
Collapse
|
35
|
Sitia L, Catelani T, Guarnieri D, Pompa PP. In Vitro Blood-Brain Barrier Models for Nanomedicine: Particle-Specific Effects and Methodological Drawbacks. ACS APPLIED BIO MATERIALS 2019; 2:3279-3289. [PMID: 35030770 DOI: 10.1021/acsabm.9b00305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Predicting the therapeutic efficacy of a nanocarrier, in a rapid and cost-effective way, is pivotal for the drug delivery to the central nervous system (CNS). In this context, in vitro testing platforms, like the transwell systems, offer numerous advantages to study the passage through the blood-brain barrier (BBB), such as overcoming ethical and methodological issues of in vivo models. However, the use of different transwell filters and nanocarriers with various physical-chemical features makes it difficult to assess the nanocarrier efficacy and achieve data reproducibility. In this work, we performed a systematic study to elucidate the role of the most widely used transwell filters in affecting the passage of nanocarriers, as a function of filter pore size and density. In particular, the transport of carboxyl- and amine-modified 100 nm polystyrene nanoparticles (NPs), chosen as model nanocarriers, was quantified and compared to the behavior of Lucifer yellow (LY), a molecular marker of paracellular transport. Results indicate that the filter type affects the growth and formation of the confluent endothelial barrier, as well as the transport of NPs. Interestingly, the in situ dispersion of NPs was found to play a key role in governing their passage through the filters, both in absence and in presence of the cellular barrier. By framing the underlying nanobiointeractions, we found that particle-specific effects modulated cellular uptake and barrier intracellular distribution, eventually governing transcytosis through their interplay with "size exclusion effects" by the porous filters. This study highlights the importance of a careful evaluation of the physical-chemical profile of the tested nanocarrier along with filter parameters for a correct methodological approach to test BBB permeability in nanomedicine.
Collapse
Affiliation(s)
- Leopoldo Sitia
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), via Morego 30, Genova 16163, Italy.,Department of Biomedical and Clinical Sciences "L. Sacco″, Università Degli Studi di Milano, via G. B. Grassi 74, Milano 20157, Italy
| | - Tiziano Catelani
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego 30, Genova 16163, Italy.,Piattaforma Interdipartimentale di Microscopia, Università Degli Studi di Milano-Bicocca, Piazza della Scienza 2, Milano 20126, Italy
| | - Daniela Guarnieri
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), via Morego 30, Genova 16163, Italy.,Dipartimento di Chimica e Biologia "A. Zambelli", Università di Salerno, via Giovanni Paolo II 132, Fisciano, Salerno I-84084, Italy
| | - Pier Paolo Pompa
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), via Morego 30, Genova 16163, Italy
| |
Collapse
|
36
|
Jiang L, Li S, Zheng J, Li Y, Huang H. Recent Progress in Microfluidic Models of the Blood-Brain Barrier. MICROMACHINES 2019; 10:mi10060375. [PMID: 31195652 PMCID: PMC6630552 DOI: 10.3390/mi10060375] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/29/2019] [Accepted: 06/02/2019] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier (BBB) is a critical physical and chemical barrier that maintains brain homeostasis. Researchers in academia and industry are highly motivated to develop experimental models that can accurately mimic the physiological characteristics of the BBB. Microfluidic systems, which manipulate fluids at the micrometer scale, are ideal tools for simulating the BBB microenvironment. In this review, we summarized the progress in the design and evaluation of microfluidic in vitro BBB models, including advances in chip materials, porous membranes, the use of endothelial cells, the importance of shear stress, the detection specific markers to monitor tight junction formation and integrity, measurements of TEER and permeability. We also pointed out several shortcomings of the current microfluidic models. The purpose of this paper is to let the readers understand the characteristics of different types of model design, and select appropriate design parameters according to the research needs, so as to obtain the best experimental results. We believe that the microfluidics BBB models will play an important role in neuroscience and pharmaceutical research.
Collapse
Affiliation(s)
- Lili Jiang
- Department of Clinical and Military Laboratory Medicine, Army Medical University, Chongqing 400038, China.
| | - Shu Li
- Department of Microbiology, Army Medical University, Chongqing 400038, China.
| | - Junsong Zheng
- Department of Clinical and Military Laboratory Medicine, Army Medical University, Chongqing 400038, China.
| | - Yan Li
- Department of Clinical and Military Laboratory Medicine, Army Medical University, Chongqing 400038, China.
| | - Hui Huang
- Department of Clinical and Military Laboratory Medicine, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
37
|
Liu H, Huang CX, He Q, Li D, Luo MH, Zhao F, Lu W. Proteomics analysis of HSV-1-induced alterations in mouse brain microvascular endothelial cells. J Neurovirol 2019; 25:525-539. [PMID: 31144288 DOI: 10.1007/s13365-019-00752-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/01/2019] [Accepted: 04/08/2019] [Indexed: 02/08/2023]
Abstract
Herpes simplex virus 1 (HSV-1) is a predominant cause of herpes simplex encephalitis (HSE), leading to a high mortality rate and severe neurological sequelae worldwide. HSE is typically accompanied by the blood-brain barrier (BBB) disruption, but the underlying mechanisms are unclear. To explore the disruption mechanisms of the BBB, quantitative analysis of the cellular proteome was carried out to investigate the proteomic changes that occur after infection. In this study, bEnd.3 cells were infected with HSV-1, followed by liquid chromatography-tandem mass spectrometry. A total of 6761 proteins were identified in three independent mass spectrometry analyses. Compared to the uninfected cells, 386 and 293 differentially expressed proteins were markedly upregulated or downregulated, respectively. Bioinformatic analysis showed that the activator protein-1 factor, including Fos, Jun, and ATF family proteins and cell adhesion molecules were significantly changed. Further validation of the changes observed for these proteins was carried out by western blotting and quantitative real-time PCR. Transendothelial electrical resistance (TEER) studies were performed to explore the effects of ATF3, Fra1, or JunB overexpression on the function of bEnd.3 cells. Characterization of the differential expression of these proteins in bEnd.3 cells will facilitate further exploration of BBB disruption upon HSV-1 infection.
Collapse
Affiliation(s)
- Hui Liu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chu-Xin Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Qiang He
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Dong Li
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430000, China
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430000, China
| | - Fei Zhao
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430000, China.
| | - Wei Lu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
38
|
Dubey SK, Ram MS, Krishna KV, Saha RN, Singhvi G, Agrawal M, Ajazuddin, Saraf S, Saraf S, Alexander A. Recent Expansions on Cellular Models to Uncover the Scientific Barriers Towards Drug Development for Alzheimer's Disease. Cell Mol Neurobiol 2019; 39:181-209. [PMID: 30671696 DOI: 10.1007/s10571-019-00653-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/12/2019] [Indexed: 12/17/2022]
Abstract
Globally, the central nervous system (CNS) disorders appear as the most critical pathological threat with no proper cure. Alzheimer's disease (AD) is one such condition frequently observed with the aged population and sometimes in youth too. Most of the research utilizes different animal models for in vivo study of AD pathophysiology and to investigate the potency of the newly developed therapy. These in vivo models undoubtably provide a powerful investigation tool to study human brain. Although, it sometime fails to mimic the exact environment and responses as the human brain owing to the distinctive genetic and anatomical features of human and rodent brain. In such condition, the in vitro cell model derived from patient specific cell or human cell lines can recapitulate the human brain environment. In addition, the frequent use of animals in research increases the cost of study and creates various ethical issues. Instead, the use of in vitro cellular models along with animal models can enhance the translational values of in vivo models and represent a better and effective mean to investigate the potency of therapeutics. This strategy also limits the excessive use of laboratory animal during the drug development process. Generally, the in vitro cell lines are cultured from AD rat brain endothelial cells, the rodent models, human astrocytes, human brain capillary endothelial cells, patient derived iPSCs (induced pluripotent stem cells) and also from the non-neuronal cells. During the literature review process, we observed that there are very few reviews available which describe the significance and characteristics of in vitro cell lines, for AD investigation. Thus, in the present review article, we have compiled the various in vitro cell lines used in AD investigation including HBMEC, BCECs, SHSY-5Y, hCMEC/D3, PC-2 cell line, bEND3 cells, HEK293, hNPCs, RBE4 cells, SK-N-MC, BMVECs, CALU-3, 7W CHO, iPSCs and cerebral organoids cell lines and different types of culture media such as SCM, EMEM, DMEM/F12, RPMI, EBM and 3D-cell culture.
Collapse
Affiliation(s)
- Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India.
| | - Munnangi Siva Ram
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Kowthavarapu Venkata Krishna
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Ranendra Narayan Saha
- Department of Biotechnology, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Dubai Campus, Dubai, United Arab Emirates
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Mukta Agrawal
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Kohka, Kurud Road, Bhilai, Chhattisgarh, 490024, India
| | - Ajazuddin
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Kohka, Kurud Road, Bhilai, Chhattisgarh, 490024, India
| | - Swarnlata Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492 010, Chhattisgarh, India
| | - Shailendra Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492 010, Chhattisgarh, India
- Hemchand Yadav University, Durg, Chhattisgarh, 491 001, India
| | - Amit Alexander
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Kohka, Kurud Road, Bhilai, Chhattisgarh, 490024, India.
| |
Collapse
|
39
|
Yang S, Jin H, Zhao ZG. Epidermal growth factor treatment has protective effects on the integrity of the blood-brain barrier against cerebral ischemia injury in bEnd3 cells. Exp Ther Med 2019; 17:2397-2402. [PMID: 30867725 DOI: 10.3892/etm.2019.7186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022] Open
Abstract
Tight junctions (TJs) serve an important role in maintaining the integrity of the blood-brain barrier (BBB), while neurological disorders, including ischemic stroke, induce TJ disruption and increase BBB permeability; results include edema formation and hemorrhage transformation. Cerebral endothelium protection presents a promising approach in ischemic stroke therapy. In the current study, protective effects of the epidermal growth factor (EGF) on ischemia-induced disruption of BBB integrity were examined using an oxygen-glucose deprivation (OGD) model in bEnd3 cells. Expression levels of claudin-5 and TJ protein-1 (ZO-1) were determined by reverse transcription-quantitative polymerase chain reaction and western blot analysis. Cell viability was evaluated by cell counting kit-8 assay and the endothelial permeability of Lucifer yellow (LY) was assessed using Transwell assays. The results revealed that post-ischemia administration of EGF (250 ng/ml) significantly attenuated the decrease in mRNA (P<0.05) and protein (P<0.01) expression levels of claudin-5 and ZO-1, and the increase in endothelial permeability of LY (P<0.05) induced by 4 h OGD exposure followed by 24 h reoxygenation. In addition, EGF did not significant affect cell viability. The current study suggested a potential of EGF to improve BBB integrity against ischemic injury by upregulating the expression of TJ proteins and reducing endothelial permeability.
Collapse
Affiliation(s)
- Shu Yang
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Hong Jin
- Disinfection Evaluation Research Center, Institute of Disease Prevention and Control of PLA, Beijing 100071, P.R. China
| | - Zhi-Gang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
40
|
Battaglini M, Tapeinos C, Cavaliere I, Marino A, Ancona A, Garino N, Cauda V, Palazon F, Debellis D, Ciofani G. Design, Fabrication, and In Vitro Evaluation of Nanoceria-Loaded Nanostructured Lipid Carriers for the Treatment of Neurological Diseases. ACS Biomater Sci Eng 2019; 5:670-682. [PMID: 33405830 DOI: 10.1021/acsbiomaterials.8b01033] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases comprise a large group of disorders characterized by a dramatic synaptic connections loss, occurring as a result of neurodegeneration, which is closely related to the overproduction of reactive oxygen and nitrogen species. Currently, the treatment of neurodegenerative diseases has been limited mainly because of the inability of the synthesized delivery systems to cross the blood-brain barrier and to successfully deliver their therapeutic cargo to the diseased tissue. Taking into consideration the aforementioned limitations, we designed a lipid-based nanotherapeutic vector composed of biomimetic lipids and CeO2 nanoparticles (nanoceria, NC). NC have shown to be a promising tool for the treatment of several pathological conditions ranging from cancer to neurological diseases, mainly because of their antioxidant properties, while lipid-based structures have been shown to have an inherent ability to cross the blood-brain barrier. The lipid-based nanotherapeutics were successfully fabricated using a combination of ultrasonication and high-pressure homogenization techniques, and they were fully characterized morphologically and physicochemically. Their antioxidant ability was demonstrated using electron paramagnetic resonance spectroscopy and antioxidant assays. These innovative nanotherapeutics demonstrated a higher colloidal stability with respect to free NC, preserving at the same time their antioxidant properties. Finally, the ability of the lipid carriers to cross a model of the blood-brain barrier and to be internalized by neurons, acting both as neuroprotective and pro-neurogenic agents, was demonstrated using single- and triple-culture systems.
Collapse
Affiliation(s)
- Matteo Battaglini
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy.,Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Christos Tapeinos
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Ivana Cavaliere
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Andrea Ancona
- Politecnico di Torino, Department of Applied Science and Technology, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Nadia Garino
- Politecnico di Torino, Department of Applied Science and Technology, Corso Duca degli Abruzzi 24, 10129 Torino, Italy.,Istituto Italiano di Tecnologia, Center for Sustainable Future Technologies, Corso Trento 21, 10129 Torino, Italy
| | - Valentina Cauda
- Politecnico di Torino, Department of Applied Science and Technology, Corso Duca degli Abruzzi 24, 10129 Torino, Italy.,Istituto Italiano di Tecnologia, Center for Sustainable Future Technologies, Corso Trento 21, 10129 Torino, Italy
| | - Francisco Palazon
- Istituto Italiano di Tecnologia, Nanochemistry, Via Morego 30, 16163 Genova, Italy
| | - Doriana Debellis
- Istituto Italiano di Tecnologia, Electron Microscopy Facility, Via Morego 30, 16163 Genova, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy.,Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
41
|
Rady I, Bloch MB, Chamcheu RCN, Banang Mbeumi S, Anwar MR, Mohamed H, Babatunde AS, Kuiate JR, Noubissi FK, El Sayed KA, Whitfield GK, Chamcheu JC. Anticancer Properties of Graviola ( Annona muricata): A Comprehensive Mechanistic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1826170. [PMID: 30151067 PMCID: PMC6091294 DOI: 10.1155/2018/1826170] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/03/2018] [Indexed: 01/19/2023]
Abstract
Graviola (Annona muricata) is a small deciduous tropical evergreen fruit tree, belonging to the Annonaceae family, and is widely grown and distributed in tropical and subtropical regions around the world. The aerial parts of graviola have several functions: the fruits have been widely used as food confectionaries, while several preparations, especially decoctions of the bark, fruits, leaves, pericarp, seeds, and roots, have been extensively used in traditional medicine to treat multiple ailments including cancers by local communities in tropical Africa and South America. The reported therapeutic benefits of graviola against various human tumors and disease agents in in vitro culture and preclinical animal model systems are typically tested for their ability to specifically target the disease, while exerting little or no effect on normal cell viability. Over 212 phytochemical ingredients have been reported in graviola extracts prepared from different plant parts. The specific bioactive constituents responsible for the major anticancer, antioxidant, anti-inflammatory, antimicrobial, and other health benefits of graviola include different classes of annonaceous acetogenins (metabolites and products of the polyketide pathway), alkaloids, flavonoids, sterols, and others. This review summarizes the current understanding of the anticancer effects of A. muricata and its constituents on diverse cancer types and disease states, as well as efficacy and safety concerns. It also includes discussion of our current understanding of possible mechanisms of action, with the hope of further stimulating the development of improved and affordable therapies for a variety of ailments.
Collapse
Affiliation(s)
- Islam Rady
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, WI 53706, USA
| | - Melissa B. Bloch
- School of Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Roxane-Cherille N. Chamcheu
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, WI 53706, USA
- Madison West High School, 30 Ash St, Madison, WI 53726, USA
| | - Sergette Banang Mbeumi
- Division for Research and Innovation, POHOFI Inc., P.O. Box 44067, Madison, WI 53744, USA
| | - Md Rafi Anwar
- School of Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Hadir Mohamed
- Department of Biochemistry, Faculty of Science, University of Mansoura, Mansoura, Egypt
| | | | - Jules-Roger Kuiate
- Department of Biochemistry, Faculty of Sciences, University of Dschang, Dschang, Cameroon
- Section for Research and Innovation, POHOFCAM, P.O. Box 175, Kumba, Cameroon
| | - Felicite K. Noubissi
- Division for Research and Innovation, POHOFI Inc., P.O. Box 44067, Madison, WI 53744, USA
- Department of Biology/RCMI, Jackson State University, 1400 J R Lynch, 429 JAP, Jackson, MS 39217, USA
| | - Khalid A. El Sayed
- School of Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - G. Kerr Whitfield
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Jean Christopher Chamcheu
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, WI 53706, USA
- School of Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
42
|
Salaroglio IC, Gazzano E, Kopecka J, Chegaev K, Costamagna C, Fruttero R, Guglielmo S, Riganti C. New Tetrahydroisoquinoline Derivatives Overcome Pgp Activity in Brain-Blood Barrier and Glioblastoma Multiforme in Vitro. Molecules 2018; 23:molecules23061401. [PMID: 29890725 PMCID: PMC6099747 DOI: 10.3390/molecules23061401] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022] Open
Abstract
P-glycoprotein (Pgp) determines resistance to a broad spectrum of drugs used against glioblastoma multiforme (GB). Indeed, Pgp is highly expressed in GB stem cells and in the brain-blood barrier (BBB), the peculiar endothelium surrounding the brain. Inhibiting Pgp activity in the BBB and GB is still an open challenge. Here, we tested the efficacy of a small library of tetrahydroisoquinoline derivatives with an EC50 for Pgp ≤ 50 nM, in primary human BBB cells and in patient-derived GB samples, from which we isolated differentiated/adherent cells (AC, i.e., Pgp-negative/doxorubicin-sensitive cells) and stem cells (neurospheres, NS, i.e., Pgp-positive/doxorubicin-resistant cells). Three compounds used at 1 nM increased the delivery of doxorubicin, a typical substrate of Pgp, across BBB monolayer, without altering the expression and activity of other transporters. The compounds increased the drug accumulation within NS, restoring doxorubicin-induced necrosis and apoptosis, and reducing cell viability. In co-culture systems, the compounds added to the luminal face of BBB increased the delivery of doxorubicin to NS growing under BBB and rescued the drug’s cytotoxicity. Our work identified new ligands of Pgp active at low nanomolar concentrations. These compounds reduce Pgp activity in BBB and GB and improve in vitro chemotherapy efficacy in this tumor.
Collapse
Affiliation(s)
| | - Elena Gazzano
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Costanzo Costamagna
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Roberta Fruttero
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Stefano Guglielmo
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| |
Collapse
|
43
|
Yang S, Jin H, Zhao Z. Paracellular tightness and the functional expression of efflux transporters P-gp and BCRP in bEnd3 cells. Neurol Res 2018; 40:644-649. [PMID: 29683403 DOI: 10.1080/01616412.2018.1460701] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Objective The blood-brain barrier (BBB), regulating brain homeostasis and limiting the entry of most drugs, is characterized by intercellular tight junctions and the presence of transporters. In this study, the paracellular tightness and functional expression of efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) were evaluated in mouse brain immortalized cell line bEnd3 to prove it as a useful BBB-mimicking system for biological and pharmacological research. Methods The presence of P-gp, BCRP and tight junction proteins occludin, claudin-5 and ZO-1 were validated by RT-PCR and Western blot. The tightness of bEnd3 monolayers was evaluated by measuring the permeability of hydrophilic marker Lucifer yellow. The P-gp functionality was identified by intracellular uptake assay using Rhodamine 123 (R123) as P-gp substrate and verapamil as P-gp inhibitor. The BCRP functionality was identified by flow cytometric analysis of mitoxantrone accumulation and fluorescence microscopic analysis of Hoechst 33342 accumulation using Ko-143 as BCRP inhibitor. Results The bEnd3 cells demonstrated the expression of P-gp, BCRP and tight junction proteins occludin, claudin-5 and ZO-1 at mRNA and protein levels. The permeability coefficient of Lucifer yellow was 1.3 ± 0.13 × 10-3 cm/min, indicating the moderate paracellular tightness barrier formed by bEnd3 cells. The verapamil induced a higher cellular uptake of Rhodamine 123, and Ko-143 significantly elevated cellular accumulation of mitoxantrone and Hoechst 33342, suggesting the P-gp and BCRP functionality shown by bEnd3 cells. Conclusions The bEnd3 cell line represents a useful in vitro tool for studying BBB characteristics and drug transport mechanisms at the BBB.
Collapse
Affiliation(s)
- Shu Yang
- a Department of Pharmacy , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| | - Hong Jin
- b Disinfection Evaluation Research Center , Institute of Disease Prevention and Control of PLA , Beijing , China
| | - Zhigang Zhao
- a Department of Pharmacy , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| |
Collapse
|