1
|
Tang Z, Li R, Guo X, Wang Z, Wu J. Regulation of blood-brain barrier integrity by brain microvascular endothelial cells in ischemic stroke: A therapeutic opportunity. Eur J Pharmacol 2025; 996:177553. [PMID: 40147580 DOI: 10.1016/j.ejphar.2025.177553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Stroke is the second leading cause of death from cardiovascular diseases. Brain microvascular endothelial cells (BMECs) are crucial in the treatment of cerebral ischemic stroke, as their functional status directly affects the integrity of the blood-brain barrier (BBB). This review systematically discusses the central role of BMECs in ischemia. The mitochondrial dysfunction and activation of apoptosis/necrosis pathways in BMECs directly disrupt the integrity of the BBB and the degradation of junctional complexes (such as TJs and AJs) further exacerbates its permeability. In the neurovascular unit (NVU), astrocytes, microglia, and pericytes regulate the function of BMECs by secreting cytokines (such as TGF-β and VEGF), showing dual effects of promoting repair and damage. The dynamic changes of transporters, including those from the ATP-binding cassette and solute carrier families, as well as ion channels and exchangers, such as potassium and calcium channels, offer novel insights for the development of targeted drug delivery systems.
Collapse
Affiliation(s)
- Ziqi Tang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| | - Ruoxi Li
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Xi Guo
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China
| | - Zhongyu Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China; Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China.
| |
Collapse
|
2
|
Ranjan R, Kumar D, Singh MR, Singh D. Novel drug delivery systems in cerebral vascular disorders, transient ischaemic attack, and stroke interventions. NOVEL DRUG DELIVERY SYSTEMS IN THE MANAGEMENT OF CNS DISORDERS 2025:295-311. [DOI: 10.1016/b978-0-443-13474-6.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Darban YM, Askari H, Ghasemi-Kasman M, Yavarpour-Bali H, Dehpanah A, Gholizade P, Nosratiyan N. The Role of Induced Pluripotent Stem Cells in the Treatment of Stroke. Curr Neuropharmacol 2024; 22:2368-2383. [PMID: 39403058 PMCID: PMC11451314 DOI: 10.2174/1570159x22666240603084558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 10/19/2024] Open
Abstract
Stroke is a neurological disorder with high disability and mortality rates. Almost 80% of stroke cases are ischemic stroke, and the remaining are hemorrhagic stroke. The only approved treatment for ischemic stroke is thrombolysis and/or thrombectomy. However, these treatments cannot sufficiently relieve the disease outcome, and many patients remain disabled even after effective thrombolysis. Therefore, rehabilitative therapies are necessary to induce remodeling in the brain. Currently, stem cell transplantation, especially via the use of induced pluripotent stem cells (iPSCs), is considered a promising alternative therapy for stimulating neurogenesis and brain remodeling. iPSCs are generated from somatic cells by specific transcription factors. The biological functions of iPSCs are similar to those of embryonic stem cells (ESCs), including immunomodulation, reduced cerebral blood flow, cerebral edema, and autophagy. Although iPSC therapy plays a promising role in both hemorrhagic and ischemic stroke, its application is associated with certain limitations. Tumor formation, immune rejection, stem cell survival, and migration are some concerns associated with stem cell therapy. Therefore, cell-free therapy as an alternative method can overcome these limitations. This study reviews the therapeutic application of iPSCs in stroke models and the underlying mechanisms and constraints of these cells. Moreover, cell-free therapy using exosomes, apoptotic bodies, and microvesicles as alternative treatments is discussed.
Collapse
Affiliation(s)
| | - Hamid Askari
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Amirabbas Dehpanah
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Parnia Gholizade
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Nasrin Nosratiyan
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
4
|
Ronaldson PT, Davis TP. Blood-brain barrier transporters: a translational consideration for CNS delivery of neurotherapeutics. Expert Opin Drug Deliv 2024; 21:71-89. [PMID: 38217410 PMCID: PMC10842757 DOI: 10.1080/17425247.2024.2306138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/12/2024] [Indexed: 01/15/2024]
Abstract
INTRODUCTION Successful neuropharmacology requires optimization of CNS drug delivery and, by extension, free drug concentrations at brain molecular targets. Detailed assessment of blood-brain barrier (BBB) physiological characteristics is necessary to achieve this goal. The 'next frontier' in CNS drug delivery is targeting BBB uptake transporters, an approach that requires evaluation of brain endothelial cell transport processes so that effective drug accumulation and improved therapeutic efficacy can occur. AREAS COVERED BBB permeability of drugs is governed by tight junction protein complexes (i.e., physical barrier) and transporters/enzymes (i.e., biochemical barrier). For most therapeutics, a component of blood-to-brain transport involves passive transcellular diffusion. Small molecule drugs that do not possess acceptable physicochemical characteristics for passive permeability may utilize putative membrane transporters for CNS uptake. While both uptake and efflux transport mechanisms are expressed at the brain microvascular endothelium, uptake transporters can be targeted for optimization of brain drug delivery and improved treatment of neurological disease states. EXPERT OPINION Uptake transporters represent a unique opportunity to optimize brain drug delivery by leveraging the endogenous biology of the BBB. A rigorous understanding of these transporters is required to improve translation from the bench to clinical trials and stimulate the development of new treatment paradigms for neurological diseases.
Collapse
Affiliation(s)
| | - Thomas P. Davis
- Department of Pharmacology, University of Arizona College of Medicine
| |
Collapse
|
5
|
Zhang Y, Sharma S, Jonnalagadda S, Kumari S, Queen A, Esfahani SH, Archie SR, Nozohouri S, Patel D, Trippier PC, Karamyan VT, Abbruscato TJ. Discovery of the Next Generation of Non-peptidomimetic Neurolysin Activators with High Blood-Brain Barrier Permeability: a Pharmacokinetics Study in Healthy and Stroke Animals. Pharm Res 2023; 40:2747-2758. [PMID: 37833570 DOI: 10.1007/s11095-023-03619-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
PURPOSE There is growing interest in seeking pharmacological activation of neurolysin (Nln) for stroke treatment. Discovery of central nervous system drugs remains challenging due to the protection of the blood-brain barrier (BBB). The previously reported peptidomimetic Nln activators display unsatisfactory BBB penetration. Herein, we investigate the next generation of non-peptidomimetic Nln activators with high BBB permeability. METHODS A BBB-mimicking model was used to evaluate their in vitro BBB permeability. Protein binding, metabolic stability, and efflux assays were performed to determine their unbound fraction, half-lives in plasma and brains, and dependence of BBB transporter P-glycoprotein (P-gp). The in vivo pharmacokinetic profiles were elucidated in healthy and stroke mice. RESULTS Compounds KS52 and KS73 out of this generation exhibit improved peptidase activity and BBB permeability compared to the endogenous activator and previous peptidomimetic activators. They show reasonable plasma and brain protein binding, improved metabolic stability, and independence of P-gp-mediated efflux. In healthy animals, they rapidly distribute into brains and reach peak levels of 18.69% and 12.10% injected dose (ID)/ml at 10 min. After 4 h, their total brain concentrations remain 7.78 and 12.34 times higher than their A50(minimal concentration required for enhancing 50% peptidase activity). Moreover, the ipsilateral hemispheres of stroke animals show comparable uptake to the corresponding contralateral hemispheres and healthy brains. CONCLUSIONS This study provides essential details about the pharmacokinetic properties of a new generation of potent non-peptidomimetic Nln activators with high BBB permeability and warrants the future development of these agents as potential neuroprotective pharmaceutics for stroke treatment.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Shirisha Jonnalagadda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
| | - Shikha Kumari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
| | - Aarfa Queen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
| | - Shiva Hadi Esfahani
- Department of Foundational Medical Studies, William Beaumont School of Medicine, Oakland University, Rochester, MI, 48309, USA
- Laboratory for Neurodegenerative Disease & Drug Discovery, William Beaumont School of Medicine, Oakland University, Rochester, MI, 48309, USA
| | - Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Dhavalkumar Patel
- Office of Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center (UNMC), Omaha, NE, 68106, USA
| | - Vardan T Karamyan
- Department of Foundational Medical Studies, William Beaumont School of Medicine, Oakland University, Rochester, MI, 48309, USA
- Laboratory for Neurodegenerative Disease & Drug Discovery, William Beaumont School of Medicine, Oakland University, Rochester, MI, 48309, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
- Center for Blood Brain Barrier Research, Jerry. H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| |
Collapse
|
6
|
Sharma S, Zhang Y, Akter KA, Nozohouri S, Archie SR, Patel D, Villalba H, Abbruscato T. Permeability of Metformin across an In Vitro Blood-Brain Barrier Model during Normoxia and Oxygen-Glucose Deprivation Conditions: Role of Organic Cation Transporters (Octs). Pharmaceutics 2023; 15:1357. [PMID: 37242599 PMCID: PMC10220878 DOI: 10.3390/pharmaceutics15051357] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/19/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Our lab previously established that metformin, a first-line type two diabetes treatment, activates the Nrf2 pathway and improves post-stroke recovery. Metformin's brain permeability value and potential interaction with blood-brain barrier (BBB) uptake and efflux transporters are currently unknown. Metformin has been shown to be a substrate of organic cationic transporters (Octs) in the liver and kidneys. Brain endothelial cells at the BBB have been shown to express Octs; thus, we hypothesize that metformin uses Octs for its transport across the BBB. We used a co-culture model of brain endothelial cells and primary astrocytes as an in vitro BBB model to conduct permeability studies during normoxia and hypoxia using oxygen-glucose deprivation (OGD) conditions. Metformin was quantified using a highly sensitive LC-MS/MS method. We further checked Octs protein expression using Western blot analysis. Lastly, we completed a plasma glycoprotein (P-GP) efflux assay. Our results showed that metformin is a highly permeable molecule, uses Oct1 for its transport, and does not interact with P-GP. During OGD, we found alterations in Oct1 expression and increased permeability for metformin. Additionally, we showed that selective transport is a key determinant of metformin's permeability during OGD, thus, providing a novel target for improving ischemic drug delivery.
Collapse
Affiliation(s)
- Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Yong Zhang
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Khondker Ayesha Akter
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Dhavalkumar Patel
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Heidi Villalba
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Thomas Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
7
|
Liu G, Bai X, Yang J, Duan Y, Zhu J, Xiangyang L. Relationship between blood-brain barrier changes and drug metabolism under high-altitude hypoxia: obstacle or opportunity for drug transport? Drug Metab Rev 2023; 55:107-125. [PMID: 36823775 DOI: 10.1080/03602532.2023.2180028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
The blood-brain barrier is essential for maintaining the stability of the central nervous system and is also crucial for regulating drug metabolism, changes of blood-brain barrier's structure and function can influence how drugs are delivered to the brain. In high-altitude hypoxia, the central nervous system's function is drastically altered, which can cause disease and modify the metabolism of drugs in vivo. Changes in the structure and function of the blood-brain barrier and the transport of the drug across the blood-brain barrier under high-altitude hypoxia, are regulated by changes in brain microvascular endothelial cells, astrocytes, and pericytes, either regulated by drug metabolism factors such as drug transporters and drug-metabolizing enzymes. This article aims to review the effects of high-altitude hypoxia on the structure and function of the blood-brain barrier as well as the effects of changes in the blood-brain barrier on drug metabolism. We also hypothesized and explore the regulation and potential mechanisms of the blood-brain barrier and associated pathways, such as transcription factors, inflammatory factors, and nuclear receptors, in regulating drug transport under high-altitude hypoxia.
Collapse
Affiliation(s)
- Guiqin Liu
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
| | - Xue Bai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
| | - Jianxin Yang
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
| | - Yabin Duan
- Affiliated Hospital of Qinghai University, Xining, China
| | - Junbo Zhu
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China
| | - Li Xiangyang
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining, China.,State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| |
Collapse
|
8
|
Mendez EF, Grimm SL, Stertz L, Gorski D, Movva SV, Najera K, Moriel K, Meyer TD, Fries GR, Coarfa C, Walss-Bass C. A human stem cell-derived neuronal model of morphine exposure reflects brain dysregulation in opioid use disorder: Transcriptomic and epigenetic characterization of postmortem-derived iPSC neurons. Front Psychiatry 2023; 14:1070556. [PMID: 36873219 PMCID: PMC9978009 DOI: 10.3389/fpsyt.2023.1070556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Human-derived induced pluripotent stem cell (iPSC) models of brain promise to advance our understanding of neurotoxic consequences of drug use. However, how well these models recapitulate the actual genomic landscape and cell function, as well as the drug-induced alterations, remains to be established. New in vitro models of drug exposure are needed to advance our understanding of how to protect or reverse molecular changes related to substance use disorders. Methods We engineered a novel induced pluripotent stem cell-derived model of neural progenitor cells and neurons from cultured postmortem human skin fibroblasts, and directly compared these to isogenic brain tissue from the donor source. We assessed the maturity of the cell models across differentiation from stem cells to neurons using RNA cell type and maturity deconvolution analyses as well as DNA methylation epigenetic clocks trained on adult and fetal human tissue. As proof-of-concept of this model's utility for substance use disorder studies, we compared morphine- and cocaine-treated neurons to gene expression signatures in postmortem Opioid Use Disorder (OUD) and Cocaine Use Disorder (CUD) brains, respectively. Results Within each human subject (N = 2, 2 clones each), brain frontal cortex epigenetic age parallels that of skin fibroblasts and closely approximates the donor's chronological age; stem cell induction from fibroblast cells effectively sets the epigenetic clock to an embryonic age; and differentiation of stem cells to neural progenitor cells and then to neurons progressively matures the cells via DNA methylation and RNA gene expression readouts. In neurons derived from an individual who died of opioid overdose, morphine treatment induced alterations in gene expression similar to those previously observed in OUD ex-vivo brain tissue, including differential expression of the immediate early gene EGR1, which is known to be dysregulated by opioid use. Discussion In summary, we introduce an iPSC model generated from human postmortem fibroblasts that can be directly compared to corresponding isogenic brain tissue and can be used to model perturbagen exposure such as that seen in opioid use disorder. Future studies with this and other postmortem-derived brain cellular models, including cerebral organoids, can be an invaluable tool for understanding mechanisms of drug-induced brain alterations.
Collapse
Affiliation(s)
- Emily F. Mendez
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sandra L. Grimm
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, United States
| | - Laura Stertz
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Damian Gorski
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas Medical School at Houston, Houston, TX, United States
| | - Sai V. Movva
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Katherine Najera
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karla Moriel
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Thomas D. Meyer
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Gabriel R. Fries
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Cristian Coarfa
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, United States
| | - Consuelo Walss-Bass
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
9
|
Zhang S, Gan L, Cao F, Wang H, Gong P, Ma C, Ren L, Lin Y, Lin X. The barrier and interface mechanisms of the brain barrier, and brain drug delivery. Brain Res Bull 2022; 190:69-83. [PMID: 36162603 DOI: 10.1016/j.brainresbull.2022.09.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 08/25/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022]
Abstract
Three different barriers are formed between the cerebrovascular and the brain parenchyma: the blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the cerebrospinal fluid-brain barrier (CBB). The BBB is the main regulator of blood and central nervous system (CNS) material exchange. The semipermeable nature of the BBB limits the passage of larger molecules and hydrophilic small molecules, Food and Drug Administration (FDA)-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Although the complexity of the BBB affects CNS drug delivery, understanding the composition and function of the BBB can provide a platform for the development of new methods for CNS drug delivery. This review summarizes the classification of the brain barrier, the composition and role of the basic structures of the BBB, and the transport, barrier, and destruction mechanisms of the BBB; discusses the advantages and disadvantages of different drug delivery methods and prospects for future drug delivery strategies.
Collapse
Affiliation(s)
- Shanshan Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang Province, China
| | - Lin Gan
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Fengye Cao
- Yiyang The First Hospital of Traditional Chinese Medicine, Yiyang, Hunan Province, 413000, China
| | - Hao Wang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Peng Gong
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Congcong Ma
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Li Ren
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Yubo Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Xianming Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China.
| |
Collapse
|
10
|
Ronaldson PT, Davis TP. Transport Mechanisms at the Blood-Brain Barrier and in Cellular Compartments of the Neurovascular Unit: Focus on CNS Delivery of Small Molecule Drugs. Pharmaceutics 2022; 14:1501. [PMID: 35890396 PMCID: PMC9324459 DOI: 10.3390/pharmaceutics14071501] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a primary origin of morbidity and mortality in the United States and around the world. Indeed, several research projects have attempted to discover new drugs or repurpose existing therapeutics to advance stroke pharmacotherapy. Many of these preclinical stroke studies have reported positive results for neuroprotective agents; however, only one compound (3K3A-activated protein C (3K3A-APC)) has advanced to Phase III clinical trial evaluation. One reason for these many failures is the lack of consideration of transport mechanisms at the blood-brain barrier (BBB) and neurovascular unit (NVU). These endogenous transport processes function as a "gateway" that is a primary determinant of efficacious brain concentrations for centrally acting drugs. Despite the knowledge that some neuroprotective agents (i.e., statins and memantine) are substrates for these endogenous BBB transporters, preclinical stroke studies have largely ignored the role of transporters in CNS drug disposition. Here, we review the current knowledge on specific BBB transporters that either limit drug uptake into the brain (i.e., ATP-binding cassette (ABC) transporters) or can be targeted for optimized drug delivery (i.e., solute carrier (SLC) transporters). Additionally, we highlight the current knowledge on transporter expression in astrocytes, microglia, pericytes, and neurons with an emphasis on transport mechanisms in these cell types that can influence drug distribution within the brain.
Collapse
Affiliation(s)
- Patrick T. Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724-5050, USA;
| | | |
Collapse
|
11
|
Betterton RD, Abdullahi W, Williams EI, Lochhead JJ, Brzica H, Stanton J, Reddell E, Ogbonnaya C, Davis TP, Ronaldson PT. Regulation of Blood-Brain Barrier Transporters by Transforming Growth Factor- β/Activin Receptor-Like Kinase 1 Signaling: Relevance to the Brain Disposition of 3-Hydroxy-3-Methylglutaryl Coenzyme A Reductase Inhibitors (i.e., Statins). Drug Metab Dispos 2022; 50:942-956. [PMID: 35504656 PMCID: PMC11022862 DOI: 10.1124/dmd.121.000781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/08/2022] [Indexed: 11/22/2022] Open
Abstract
Our laboratory has shown that activation of transforming growth factor- β (TGF- β )/activin receptor-like kinase 1 (ALK1) signaling can increase protein expression and transport activity of organic anion transporting polypeptide 1a4 (Oatp1a4) at the blood-brain barrier (BBB). These results are relevant to treatment of ischemic stroke because Oatp transport substrates such as 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (i.e., statins) improve functional neurologic outcomes in patients. Advancement of our work requires determination if TGF- β /ALK1 signaling alters Oatp1a4 functional expression differently across brain regions and if such disparities affect central nervous system (CNS) statin disposition. Therefore, we studied regulation of Oatp1a4 by the TGF- β /ALK1 pathway, in vivo, in rat brain microvessels isolated from cerebral cortex, hippocampus, and cerebellum using the ALK1 agonist bone morphogenetic protein-9 (BMP-9) and the ALK1 inhibitor 4-[6-[4-(1-piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline dihydrochloride 193189. We showed that Oatp1a4 protein expression and brain distribution of three currently marketed statin drugs (i.e., atorvastatin, pravastatin, and rosuvastatin) were increased in cortex relative to hippocampus and cerebellum. Additionally, BMP-9 treatment enhanced Oatp-mediated statin transport in cortical tissue but not in hippocampus or cerebellum. Although brain drug delivery is also dependent upon efflux transporters, such as P-glycoprotein and/or Breast Cancer Resistance Protein, our data showed that administration of BMP-9 did not alter the relative contribution of these transporters to CNS disposition of statins. Overall, this study provides evidence for differential regulation of Oatp1a4 by TGF- β /ALK1 signaling across brain regions, knowledge that is critical for development of therapeutic strategies to target Oatps at the BBB for CNS drug delivery. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (Oatps) represent transporter targets for brain drug delivery. We have shown that Oatp1a4 statin uptake is higher in cortex versus hippocampus and cerebellum. Additionally, we report that the transforming growth factor- β /activin receptor-like kinase 1 agonist bone morphogenetic protein-9 increases Oatp1a4 functional expression, but not efflux transporters P-glycoprotein and Breast Cancer Resistance Protein, in cortical brain microvessels. Overall, this study provides critical data that will advance treatment for neurological diseases where drug development has been challenging.
Collapse
Affiliation(s)
- Robert D Betterton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Wazir Abdullahi
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Erica I Williams
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Jeffrey J Lochhead
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Hrvoje Brzica
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Joshua Stanton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Elizabeth Reddell
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Chidinma Ogbonnaya
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
12
|
Blood-Brain Barrier Transporters: Opportunities for Therapeutic Development in Ischemic Stroke. Int J Mol Sci 2022; 23:ijms23031898. [PMID: 35163820 PMCID: PMC8836701 DOI: 10.3390/ijms23031898] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/20/2022] Open
Abstract
Globally, stroke is a leading cause of death and long-term disability. Over the past decades, several efforts have attempted to discover new drugs or repurpose existing therapeutics to promote post-stroke neurological recovery. Preclinical stroke studies have reported successes in identifying novel neuroprotective agents; however, none of these compounds have advanced beyond a phase III clinical trial. One reason for these failures is the lack of consideration of blood-brain barrier (BBB) transport mechanisms that can enable these drugs to achieve efficacious concentrations in ischemic brain tissue. Despite the knowledge that drugs with neuroprotective properties (i.e., statins, memantine, metformin) are substrates for endogenous BBB transporters, preclinical stroke research has not extensively studied the role of transporters in central nervous system (CNS) drug delivery. Here, we review current knowledge on specific BBB uptake transporters (i.e., organic anion transporting polypeptides (OATPs in humans; Oatps in rodents); organic cation transporters (OCTs in humans; Octs in rodents) that can be targeted for improved neuroprotective drug delivery. Additionally, we provide state-of-the-art perspectives on how transporter pharmacology can be integrated into preclinical stroke research. Specifically, we discuss the utility of in vivo stroke models to transporter studies and considerations (i.e., species selection, co-morbid conditions) that will optimize the translational success of stroke pharmacotherapeutic experiments.
Collapse
|
13
|
Pervaiz I, Al-Ahmad AJ. In Vitro Models of the Human Blood-Brain Barrier Utilising Human Induced Pluripotent Stem Cells: Opportunities and Challenges. Methods Mol Biol 2022; 2492:53-72. [PMID: 35733038 DOI: 10.1007/978-1-0716-2289-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The blood-brain barrier (BBB) is a component of the neurovascular unit formed by specialized brain microvascular endothelial cells surrounded by astrocytes end-feet processes, pericytes, and a basement membrane. The BBB plays an important role in the maintenance of brain homeostasis and has seen a growing involvement in the pathophysiology of various neurological diseases. On the other hand, the presence of such a barrier remains an important challenge for drug delivery to treat such illnesses.Since the pioneering work describing the isolation and cultivation of primary brain microvascular cells about 50 years ago until now, the development of an in vitro model of the BBB that is scalable, capable to form tight monolayers, and predictive of drug permeability in vivo remained extremely challenging.The recent description of the use of induced pluripotent stem cells (iPSCs) as a modeling tool for neurological diseases raised momentum into the use of such cells to develop new in vitro models of the BBB. This chapter will provide an exhaustive description of the use of iPSCs as a source of cells for modeling the BBB in vitro, describe the advantages and limitations of such model, as well as describe their prospective use for disease modeling and drug permeability screening platforms.
Collapse
Affiliation(s)
- Iqra Pervaiz
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Abraham J Al-Ahmad
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| |
Collapse
|
14
|
Peng Q, Bi R, Chen S, Chen J, Li Z, Li J, Jin H, Hu B. Predictive value of different bilirubin subtypes for clinical outcomes in patients with acute ischemic stroke receiving thrombolysis therapy. CNS Neurosci Ther 2021; 28:226-236. [PMID: 34779141 PMCID: PMC8739039 DOI: 10.1111/cns.13759] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/19/2022] Open
Abstract
Aims To explore the association of total bilirubin (TBIL), direct bilirubin (DBIL), and indirect bilirubin (IBIL) levels with, as well as the incremental predictive value of different bilirubin subtypes for, poor outcomes in acute ischemic stroke patients after thrombolysis. Methods We analyzed 588 individuals out of 718 AIS participants, and all patients were followed up at 3 months after thrombolysis. The primary outcome was 3‐month death and major disability (modified Rankin Scale (mRS) score of 3–6). The secondary outcomes were 3‐month mortality (mRS score of 6), moderate‐severe cerebral edema, and symptomatic intracranial hemorrhage (sICH), respectively. Results Elevated DBIL pre‐thrombolysis was associated with an increased risk of primary outcome (OR 3.228; 95% CI 1.595–6.535; p for trend = 0.014) after fully adjustment. Elevated TBIL pre‐thrombolysis showed the similar results (OR 2.185; 95% CI 1.111–4.298; p for trend = 0.047), while IBIL pre‐thrombolysis was not significantly associated with primary outcome (OR 1.895; 95% CI 0.974–3.687; p for trend = 0.090). Multivariable‐adjusted spline regression model showed a positive linear dose‐response relationship between DBIL pre‐thrombolysis and risk of primary outcome (p for linearity = 0.004). Adding DBIL pre‐thrombolysis into conventional model had greater incremental predictive value for primary outcome, with net reclassification improvement (NRI) 95% CI = 0.275 (0.084–0.466) and integrated discrimination improvement (IDI) 95% CI = 0.011 (0.001–0.024). Increased DBIL post‐thrombolysis had an association with primary outcome (OR 2.416; 95%CI 1.184–4.930; p for trend = 0.039), and it also elevated the incremental predictive value for primary outcome, with NRI (95% CI) = 0.259 (0.066–0.453) and IDI (95% CI) = 0.025 (0.008–0.043). Conclusion Increased DBIL pre‐thrombolysis had a stronger association with, as well as greater incremental predictive value for, poor outcomes than TBIL and IBIL did in AIS patients after thrombolysis, which should be understood in the context of retrospective design. The effect of DBIL on targeted populations should be investigated in further researches.
Collapse
Affiliation(s)
- Qiwei Peng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengcai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiefang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhifang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianzhuang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Redkiewicz P, Dyniewicz J, Misicka A. Biphalin-A Potent Opioid Agonist-As a Panacea for Opioid System-Dependent Pathophysiological Diseases? Int J Mol Sci 2021; 22:11347. [PMID: 34768778 PMCID: PMC8582929 DOI: 10.3390/ijms222111347] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Biphalin, one of the opioid agonists, is a dimeric analog of enkephalin with a high affinity for opioid receptors. Opioid receptors are widespread in the central nervous system and in peripheral neuronal and non-neuronal tissues. Hence, these receptors and their agonists, which play an important role in pain blocking, may also be involved in the regulation of other physiological functions. Biphalin was designed and synthesized in 1982 by Lipkowski as an analgesic peptide. Extensive further research in various laboratories on the antinociceptive effects of biphalin has shown its excellent properties. It has been demonstrated that biphalin exhibits an analgesic effect in acute, neuropathic, and chronic animal pain models, and is 1000 times more potent than morphine when administered intrathecally. In the course of the broad conducted research devoted primarily to the antinociceptive effect of this compound, it has been found that biphalin may also potentially participate in the regulation of other opioid system-dependent functions. Nearly 40 years of research on the properties of biphalin have shown that it may play a beneficial role as an antiviral, antiproliferative, anti-inflammatory, and neuroprotective agent, and may also affect many physiological functions. This integral review analyzes the literature on the multidirectional biological effects of biphalin and its potential in the treatment of many opioid system-dependent pathophysiological diseases.
Collapse
Affiliation(s)
- Patrycja Redkiewicz
- Department of Neuropeptides, Mossakowski Medical Research Institute Polish Academy of Sciences, 02106 Warsaw, Poland;
| | - Jolanta Dyniewicz
- Department of Neuropeptides, Mossakowski Medical Research Institute Polish Academy of Sciences, 02106 Warsaw, Poland;
| | - Aleksandra Misicka
- Department of Neuropeptides, Mossakowski Medical Research Institute Polish Academy of Sciences, 02106 Warsaw, Poland;
- Faculty of Chemistry, University of Warsaw, 02093 Warsaw, Poland
| |
Collapse
|
16
|
Kim EH, Kim ES, Shin D, Kim D, Choi S, Shin YJ, Kim KA, Noh D, Caglayan AB, Rajanikant G, Majid A, Bae ON. Carnosine Protects against Cerebral Ischemic Injury by Inhibiting Matrix-Metalloproteinases. Int J Mol Sci 2021; 22:7495. [PMID: 34299128 PMCID: PMC8306548 DOI: 10.3390/ijms22147495] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/02/2021] [Accepted: 07/11/2021] [Indexed: 12/11/2022] Open
Abstract
Stroke is one of the leading causes of death and disability worldwide. However, treatment options for ischemic stroke remain limited. Matrix-metalloproteinases (MMPs) contribute to brain damage during ischemic strokes by disrupting the blood-brain barrier (BBB) and causing brain edemas. Carnosine, an endogenous dipeptide, was found by us and others to be protective against ischemic brain injury. In this study, we investigated whether carnosine influences MMP activity. Brain MMP levels and activity were measured by gelatin zymography after permanent occlusion of the middle cerebral artery (pMCAO) in rats and in vitro enzyme assays. Carnosine significantly reduced infarct volume and edema. Gelatin zymography and in vitro enzyme assays showed that carnosine inhibited brain MMPs. We showed that carnosine inhibited both MMP-2 and MMP-9 activity by chelating zinc. Carnosine also reduced the ischemia-mediated degradation of the tight junction proteins that comprise the BBB. In summary, our findings show that carnosine inhibits MMP activity by chelating zinc, an essential MMP co-factor, resulting in the reduction of edema and brain injury. We believe that our findings shed new light on the neuroprotective mechanism of carnosine against ischemic brain damage.
Collapse
Affiliation(s)
- Eun-Hye Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| | - Eun-Sun Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| | - Donggeun Shin
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| | - Donghyun Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| | - Sungbin Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| | - Young-Jun Shin
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| | - Kyeong-A Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| | - Dabi Noh
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| | - Ahmet B. Caglayan
- Department of Physiology, Faculty of Medicine, Istanbul Medipol University, 34810 Istanbul, Turkey;
| | - G.K. Rajanikant
- School of Biotechnology, National Institute of Technology Calicut, Calicut 673601, India;
| | - Arshad Majid
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2TN, UK
| | - Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan 15588, Korea; (E.-H.K.); (E.-S.K.); (D.S.); (D.K.); (S.C.); (Y.-J.S.); (K.-A.K.); (D.N.)
| |
Collapse
|
17
|
Abstract
This paper is the forty-second consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2019 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
18
|
Nozohouri S, Vaidya B, Abbruscato TJ. Exosomes in Ischemic Stroke. Curr Pharm Des 2021; 26:5533-5545. [PMID: 32534564 DOI: 10.2174/1381612826666200614180253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Ischemic stroke, a leading cause of mortality, results in severe neurological outcomes in the patients. Effective stroke therapies may significantly decrease the extent of injury. For this purpose, novel and efficient drug delivery strategies need to be developed. Among a myriad of therapeutic and drug delivery techniques, exosomes have shown promising results in ischemic stroke either by their intrinsic therapeutic characteristics, which can result in angiogenesis and neurogenesis or by acting as competent, biocompatible drug delivery vehicles to transport neurotherapeutic agents into the brain. In this review, we have discussed different methods of exosome isolation and cargo loading techniques, advantages and disadvantages of using exosomes as a drug delivery carrier and the therapeutic applications of exosomes with a focus on ischemic stroke therapy.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| |
Collapse
|
19
|
Ronaldson PT, Brzica H, Abdullahi W, Reilly BG, Davis TP. Transport Properties of Statins by Organic Anion Transporting Polypeptide 1A2 and Regulation by Transforming Growth Factor- β Signaling in Human Endothelial Cells. J Pharmacol Exp Ther 2021; 376:148-160. [PMID: 33168642 PMCID: PMC7839073 DOI: 10.1124/jpet.120.000267] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
Our in vivo rodent studies have shown that organic anion transporting polypeptide (Oatp) 1a4 is critical for blood-to-brain transport of statins, drugs that are effective neuroprotectants. Additionally, transforming growth factor-β (TGF-β) signaling via the activin receptor-like kinase 1 (ALK1) receptor regulates Oatp1a4 functional expression. The human ortholog of Oatp1a4 is OATP1A2. Therefore, the translational significance of our work requires demonstration that OATP1A2 can transport statins and is regulated by TGF-β/ALK1 signaling. Cellular uptake and monolayer permeability of atorvastatin, pravastatin, and rosuvastatin were investigated in vitro using human umbilical vein endothelial cells (HUVECs). Regulation of OATP1A2 by the TGF-β/ALK1 pathway was evaluated using bone morphogenetic protein 9 (BMP-9), a selective ALK1 agonist, and LDN193189, an ALK1 antagonist. We showed that statin accumulation in HUVECs requires OATP1A2-mediated uptake but is also affected by efflux transporters (i.e., P-glycoprotein, breast cancer resistance protein). Absorptive flux (i.e., apical-to-basolateral) for all statins was higher than secretory flux (i.e., basolateral-to-apical) and was decreased by an OATP inhibitor (i.e., estrone-3-sulfate). OATP1A2 protein expression, statin uptake, and cellular monolayer permeability were increased by BMP-9 treatment. This effect was attenuated in the presence of LDN193189. Apical-to-basolateral statin transport across human endothelial cellular monolayers requires functional expression of OATP1A2, which can be controlled by therapeutically targeting TGF-β/ALK1 signaling. Taken together with our previous work, the present data show that OATP-mediated drug transport is a critical mechanism in facilitating neuroprotective drug disposition across endothelial barriers of the blood-brain barrier. SIGNIFICANCE STATEMENT: Transporter data derived from rodent models requires validation in human models. Using human umbilical vein endothelial cells, this study has shown that statin transport is mediated by OATP1A2. Additionally, we demonstrated that OATP1A2 is regulated by transforming growth factor-β/activin receptor-like kinase 1 signaling. This work emphasizes the need to consider endothelial transporter kinetics and regulation during preclinical drug development. Furthermore, our forward-thinking approach can identify effective therapeutics for diseases for which drug development has been challenging (i.e., neurological diseases).
Collapse
Affiliation(s)
- Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Hrvoje Brzica
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Wazir Abdullahi
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Bianca G Reilly
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
20
|
Al-Ahmad AJ, Pervaiz I, Karamyan VT. Neurolysin substrates bradykinin, neurotensin and substance P enhance brain microvascular permeability in a human in vitro model. J Neuroendocrinol 2021; 33:e12931. [PMID: 33506602 PMCID: PMC8166215 DOI: 10.1111/jne.12931] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/31/2022]
Abstract
Increased brain microvascular permeability and disruption of blood-brain barrier (BBB) function are among hallmarks of several acute neurodegenerative disorders, including stroke. Numerous studies suggest the involvement of bradykinin (BK), neurotensin (NT) and substance P (SP) in BBB impairment and oedema formation after stroke; however, there is paucity of data in regard to the direct effects of these peptides on the brain microvascular endothelial cells (BMECs) and BBB. The present study aimed to evaluate the direct effects of BK, NT and SP on the permeability of BBB in an in vitro model based on human induced pluripotent stem cell (iPSC)-derived BMECs. Our data indicate that all three peptides increase BBB permeability in a concentration-dependent manner in an in vitro model formed from two different iPSC lines (CTR90F and CTR65M) and widely used hCMEC/D3 human BMECs. The combination of BK, NT and SP at a sub-effective concentration also resulted in increased BBB permeability in the iPSC-derived model indicating potentiation of their action. Furthermore, we observed abrogation of BK, NT and SP effects with pretreatment of pharmacological blockers targeting their specific receptors. Additional mechanistic studies indicate that the short-term effects of these peptides are not mediated through alteration of tight-junction proteins claudin-5 and occludin, but likely involve redistribution of F-actin and secretion of vascular endothelial growth factor. This is the first experimental study to document the increased permeability of the BBB in response to direct action of NT in an in vitro model. In addition, our study confirms the expected but not well-documented, direct effect of SP on BBB permeability and adds to the well-recognised actions of BK on BBB. Lastly, we demonstrate that peptidase neurolysin can neutralise the effects of these peptides on BBB, suggesting potential therapeutic implications.
Collapse
Affiliation(s)
- Abraham J Al-Ahmad
- Department of Pharmaceutical Sciences and Center for Blood Brain Barrier Research, School of Pharmacy, TTUHSC, Amarillo, TX, USA
| | - Iqra Pervaiz
- Department of Pharmaceutical Sciences and Center for Blood Brain Barrier Research, School of Pharmacy, TTUHSC, Amarillo, TX, USA
| | - Vardan T Karamyan
- Department of Pharmaceutical Sciences and Center for Blood Brain Barrier Research, School of Pharmacy, TTUHSC, Amarillo, TX, USA
| |
Collapse
|
21
|
Nozohouri S, Noorani B, Al-Ahmad A, Abbruscato TJ. Estimating Brain Permeability Using In Vitro Blood-Brain Barrier Models. Methods Mol Biol 2021; 2367:47-72. [PMID: 32789777 DOI: 10.1007/7651_2020_311] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The blood-brain barrier (BBB) is a vital biological interface that regulates transfer of different molecules between blood and brain and, therefore, maintains the homeostatic environment of the CNS. In order to perform high-throughput screening of therapeutics in drug discovery, specific properties of the BBB are investigated within in vitro BBB platforms. In this chapter, we detail the process and steps for the iPSC to BMEC and astrocyte differentiation as well as TEER and permeability measurement in Transwell platform of in vitro BBB model. Also, advanced microfluidic iPSCs-derived BMECs on chip and permeability measurement within this model have been elucidated.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Center for Blood-Brain Barrier Research, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Center for Blood-Brain Barrier Research, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Center for Blood-Brain Barrier Research, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
- Center for Blood-Brain Barrier Research, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| |
Collapse
|
22
|
Andjelkovic AV, Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF. Modeling blood-brain barrier pathology in cerebrovascular disease in vitro: current and future paradigms. Fluids Barriers CNS 2020; 17:44. [PMID: 32677965 PMCID: PMC7367394 DOI: 10.1186/s12987-020-00202-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
The complexity of the blood-brain barrier (BBB) and neurovascular unit (NVU) was and still is a challenge to bridge. A highly selective, restrictive and dynamic barrier, formed at the interface of blood and brain, the BBB is a "gatekeeper" and guardian of brain homeostasis and it also acts as a "sensor" of pathological events in blood and brain. The majority of brain and cerebrovascular pathologies are associated with BBB dysfunction, where changes at the BBB can lead to or support disease development. Thus, an ultimate goal of BBB research is to develop competent and highly translational models to understand mechanisms of BBB/NVU pathology and enable discovery and development of therapeutic strategies to improve vascular health and for the efficient delivery of drugs. This review article focuses on the progress being made to model BBB injury in cerebrovascular diseases in vitro.
Collapse
Affiliation(s)
- Anuska V Andjelkovic
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.
| | - Svetlana M Stamatovic
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Chelsea M Phillips
- Graduate Program in Neuroscience, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriela Martinez-Revollar
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Li C, Wu X, Liu S, Shen D, Zhu J, Liu K. Role of Resolvins in the Inflammatory Resolution of Neurological Diseases. Front Pharmacol 2020; 11:612. [PMID: 32457616 PMCID: PMC7225325 DOI: 10.3389/fphar.2020.00612] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
The occurrence of neurological diseases including neurodegenerative disorders, neuroimmune diseases, and cerebrovascular disorders is closely related to neuroinflammation. Inflammation is a response against infection or injury. Genetic abnormalities, the aging process, or environmental factors can lead to dysregulation of the inflammatory response. Our immune system can cause massive damage when the inflammatory response becomes dysregulated. Inflammatory resolution is an effective process that terminates the inflammatory response to maintain health. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are omega-three polyunsaturated fatty acids that play a crucial regulatory role in the development of inflammation. Resolvins (Rvs) derived from EPA and DHA constitute the Rvs E and Rvs D series, respectively. Numerous studies on the effect of Rvs over inflammation using animal models reveal that they have both anti-inflammatory and pro-resolving capabilities. Here, we review the current knowledge on the classification, biosynthesis, receptors, mechanisms of action, and role of Rvs in neurological diseases.
Collapse
Affiliation(s)
- Chunrong Li
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiujuan Wu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Shan Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Donghui Shen
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Kangding Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
24
|
Keep RF, Jones HC, Drewes LR. This was the year that was: brain barriers and brain fluid research in 2019. Fluids Barriers CNS 2020; 17:20. [PMID: 32138786 PMCID: PMC7059280 DOI: 10.1186/s12987-020-00181-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This editorial highlights advances in brain barrier and brain fluid research published in 2019, as well as addressing current controversies and pressing needs. Topics include recent advances related to: the cerebral endothelium and the neurovascular unit; the choroid plexus, arachnoid membrane; cerebrospinal fluid and the glymphatic hypothesis; the impact of disease states on brain barriers and brain fluids; drug delivery to the brain; and translation of preclinical data to the clinic. This editorial also mourns the loss of two important figures in the field, Malcolm B. Segal and Edward G. Stopa.
Collapse
Affiliation(s)
- Richard F. Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200 USA
| | | | - Lester R. Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN 55812 USA
| |
Collapse
|
25
|
Williams EI, Betterton RD, Davis TP, Ronaldson PT. Transporter-Mediated Delivery of Small Molecule Drugs to the Brain: A Critical Mechanism That Can Advance Therapeutic Development for Ischemic Stroke. Pharmaceutics 2020; 12:pharmaceutics12020154. [PMID: 32075088 PMCID: PMC7076465 DOI: 10.3390/pharmaceutics12020154] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/28/2022] Open
Abstract
Ischemic stroke is the 5th leading cause of death in the United States. Despite significant improvements in reperfusion therapies, stroke patients still suffer from debilitating neurocognitive deficits. This indicates an essential need to develop novel stroke treatment paradigms. Endogenous uptake transporters expressed at the blood-brain barrier (BBB) provide an excellent opportunity to advance stroke therapy via optimization of small molecule neuroprotective drug delivery to the brain. Examples of such uptake transporters include organic anion transporting polypeptides (OATPs in humans; Oatps in rodents) and organic cation transporters (OCTs in humans; Octs in rodents). Of particular note, small molecule drugs that have neuroprotective properties are known substrates for these transporters and include 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (i.e., statins) for OATPs/Oatps and 1-amino-3,5-dimethyladamantane (i.e., memantine) for OCTs/Octs. Here, we review current knowledge on specific BBB transporters that can be targeted for improvement of ischemic stroke treatment and provide state-of-the-art perspectives on the rationale for considering BBB transport properties during discovery/development of stroke therapeutics.
Collapse
|
26
|
Nozohouri S, Sifat AE, Vaidya B, Abbruscato TJ. Novel approaches for the delivery of therapeutics in ischemic stroke. Drug Discov Today 2020; 25:535-551. [PMID: 31978522 DOI: 10.1016/j.drudis.2020.01.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
Here, we review novel approaches to deliver neuroprotective drugs to salvageable penumbral brain areas of stroke injury with the goals of offsetting ischemic brain injury and enhancing recovery.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
27
|
Abstract
Knowledge about the transport of active compounds across the blood-brain barrier is of essential importance for drug development. Systemically applied drugs for the central nervous system (CNS) must be able to cross the blood-brain barrier in order to reach their target sites, whereas drugs that are supposed to act in the periphery should not permeate the blood-brain barrier so that they do not trigger any adverse central adverse effects. A number of approaches have been pursued, and manifold in silico, in vitro, and in vivo animal models were developed in order to be able to make a better prediction for humans about the possible penetration of active substances into the CNS. In this particular case, however, in vitro models play a special role, since the data basis for in silico models is usually in need of improvement, and the predictive power of in vivo animal models has to be checked for possible species differences. The blood-brain barrier is a dynamic, highly selective barrier formed by brain capillary endothelial cells. One of its main tasks is the maintenance of homeostasis in the CNS. The function of the barrier is regulated by cells of the microenvironment and the shear stress mediated by the blood flow, which makes the model development most complex. In general, one could follow the credo "as easy as possible, as complex as necessary" for the usage of in vitro BBB models for drug development. In addition to the description of the classical cell culture models (transwell, hollow fiber) and guidance how to apply them, the latest developments (spheroids, microfluidic models) will be introduced in this chapter, as it is attempted to get more in vivo-like and to be applicable for high-throughput usage with these models. Moreover, details about the development of models based on stem cells derived from different sources with a special focus on human induced pluripotent stem cells are presented.
Collapse
Affiliation(s)
- Winfried Neuhaus
- Competence Unit Molecular Diagnostics, Center Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria.
| |
Collapse
|