151
|
Thom G, Tian MM, Hatcher JP, Rodrigo N, Burrell M, Gurrell I, Vitalis TZ, Abraham T, Jefferies WA, Webster CI, Gabathuler R. A peptide derived from melanotransferrin delivers a protein-based interleukin 1 receptor antagonist across the BBB and ameliorates neuropathic pain in a preclinical model. J Cereb Blood Flow Metab 2019; 39:2074-2088. [PMID: 29845881 PMCID: PMC6775589 DOI: 10.1177/0271678x18772998] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Delivery of biologic drugs across the blood-brain barrier is becoming a reality. However, the solutions often involve the assembly of complex multi-specific antibody molecules. Here we utilize a simple 12 amino-acid peptide originating from the melanotransferrin (MTf) protein that has shown improved brain delivery properties. 3D confocal fluorescence microscopic analysis demonstrated brain parenchymal localisation of a fluorescently labelled antibody (NIP228) when chemically conjugated to either the MTf peptide or full-length MTf protein. Measurement of plasma kinetics demonstrated the MTf peptide fusions had very similar kinetics to an unmodified NIP228 control antibody, whereas the fusion to MTf protein had significantly reduced plasma exposure most likely due to a higher tissue distribution in the periphery. Brain exposure for the MTf peptide fusions was significantly increased for the duration of the study, exceeding that of the fusions to full length MTf protein. Using a neuropathic pain model, we have demonstrated that fusions to interleukin-1 receptor antagonist (IL-1RA) are able to induce significant and durable analgesia following peripheral administration. These data demonstrate that recombinant and chemically conjugated MTf-based brain delivery vectors can deliver therapeutic levels of drug to the central nervous system.
Collapse
Affiliation(s)
| | | | - Jon P Hatcher
- Neuroscience IMED Biotech Unit, AstraZeneca, AKB, Cambridge, UK
| | | | | | - Ian Gurrell
- Neuroscience IMED Biotech Unit, AstraZeneca, AKB, Cambridge, UK
| | | | - Thomas Abraham
- Neural and Behavioural Sciences & Microscopy Imaging Core Lab, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Wilfred A Jefferies
- The Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; The Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Centre for Blood Research and Departments of Microbiology and Immunology, Medical Genetics, and Zoology University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
152
|
Johnsen KB, Burkhart A, Thomsen LB, Andresen TL, Moos T. Targeting the transferrin receptor for brain drug delivery. Prog Neurobiol 2019; 181:101665. [DOI: 10.1016/j.pneurobio.2019.101665] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/10/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
|
153
|
Bruch GE, Fernandes LF, Bassi BL, Alves MTR, Pereira IO, Frézard F, Massensini AR. Liposomes for drug delivery in stroke. Brain Res Bull 2019; 152:246-256. [DOI: 10.1016/j.brainresbull.2019.07.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 06/26/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022]
|
154
|
Liu C, Zhao Z, Gao H, Rostami I, You Q, Jia X, Wang C, Zhu L, Yang Y. Enhanced blood-brain-barrier penetrability and tumor-targeting efficiency by peptide-functionalized poly(amidoamine) dendrimer for the therapy of gliomas. Nanotheranostics 2019; 3:311-330. [PMID: 31687320 PMCID: PMC6821994 DOI: 10.7150/ntno.38954] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 09/14/2019] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma is one of the most common primary tumor types of central nervous system (CNS) with high malignance and lethality. Although many treatment options are currently available, the therapy of brain cancers remains challenging because of blood-brain-barrier (BBB) which prevents most of the chemotherapeutics into the CNS. In this work, a poly(amidoamine) dendrimer-based carrier was fabricated and modified with angiopep-2 (Ang2) peptide that has been demonstrated to bind to low density lipoprotein receptor-relative protein-1 (LRP1) on the endothelial cells of BBB and could therefore induce BBB penetration of the carrier. To improve tumor-targeting effect towards the glioma sites, the dendrimer was simultaneously functionalized with an epidermal growth factor receptor (EGFR)-targeting peptide (EP-1) which was screened from a "one-bead one-compound" (OBOC) combinatorial library. EP-1 peptide was demonstrated to have high affinity and specificity to EGFR at both the molecular and cellular levels. The dual-targeting dendrimer exhibited outstanding BBB penetrability and glioma targeting efficiency both in vitro and in vivo, which strikingly enhanced the anti-gliomas effect of the drugs and prolonged the survival of gliomas-bearing mice. These results show the potential of the dual-targeting dendrimer-based carrier in the therapy of gliomas through enhancing BBB penetrability and tumor targeting.
Collapse
Affiliation(s)
- Changliang Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zijian Zhao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Houqian Gao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Iman Rostami
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinru Jia
- Department of Chemistry, Peking University, Beijing 100871, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
155
|
You ZQ, Wu Q, Zhou XM, Zhang XS, Yuan B, Wen LL, Xu WD, Cui S, Tang XL, Zhang X. Receptor-Mediated Delivery of Astaxanthin-Loaded Nanoparticles to Neurons: An Enhanced Potential for Subarachnoid Hemorrhage Treatment. Front Neurosci 2019; 13:989. [PMID: 31619957 PMCID: PMC6759683 DOI: 10.3389/fnins.2019.00989] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
Astaxanthin (ATX) is a carotenoid that exerts strong anti-oxidant and anti-inflammatory property deriving from its highly unsaturated molecular structures. However, the low stability and solubility of ATX results in poor bioavailability, which markedly hampers its application as therapeutic agent in clinic advancement. This study investigated a promising way of transferrin conjugated to poly (ethylene glycol) (PEG)-encapsulated ATX nanoparticles (ATX-NPs) on targeted delivery and evaluated the possible mechanism underlying neuroprotection capability. As a result, the ATX integrated into nanocarrier presented both well water-dispersible and biocompatible, primely conquering its limitations. More than that, the transferrin-containing ATX-NPs exhibited enhanced cellular uptake efficiency than that of ATX-NPs without transferrin conjugated in primary cortical neurons. Additionally, compared to free ATX, transferrin-containing ATX-NPs with lower ATX concentration showed powerful neuroprotective effects on OxyHb-induced neuronal damage. Taken together, the improved bioavailability and enhanced neuroprotective effects enabled ATX-NPs as favorable candidates for targeted delivery and absorption of ATX. We believe that these in vitro findings will provide insights for advancement of subarachnoid hemorrhage therapy.
Collapse
Affiliation(s)
- Zong-Qi You
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Jiangsu University, Zhenjiang, China
| | - Qi Wu
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
| | - Xiao-Ming Zhou
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Second Military Medical University, Shanghai, China
| | - Xiang-Sheng Zhang
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
| | - Bin Yuan
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Li-Li Wen
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Wei-Dong Xu
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Southern Medical University (Guangzhou), Nanjing, China
| | - Sheng Cui
- College of Material Sciences and Engineering, Nanjing Tech University, Nanjing, China
| | - Xiang-Long Tang
- College of Material Sciences and Engineering, Nanjing Tech University, Nanjing, China
| | - Xin Zhang
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Jiangsu University, Zhenjiang, China.,Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China.,Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
156
|
Markova V, Holm C, Pinborg AB, Thomsen LL, Moos T. Impairment of the Developing Human Brain in Iron Deficiency: Correlations to Findings in Experimental Animals and Prospects for Early Intervention Therapy. Pharmaceuticals (Basel) 2019; 12:ph12030120. [PMID: 31416268 PMCID: PMC6789712 DOI: 10.3390/ph12030120] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
Due to the necessity of iron for a variety of cellular functions, the developing mammalian organism is vulnerable to iron deficiency, hence causing structural abnormalities and physiological malfunctioning in organs, which are particularly dependent on adequate iron stores, such as the brain. In early embryonic life, iron is already needed for proper development of the brain with the proliferation, migration, and differentiation of neuro-progenitor cells. This is underpinned by the widespread expression of transferrin receptors in the developing brain, which, in later life, is restricted to cells of the blood–brain and blood–cerebrospinal fluid barriers and neuronal cells, hence ensuring a sustained iron supply to the brain, even in the fully developed brain. In embryonic human life, iron deficiency is thought to result in a lower brain weight, with the impaired formation of myelin. Studies of fully developed infants that have experienced iron deficiency during development reveal the chronic and irreversible impairment of cognitive, memory, and motor skills, indicating widespread effects on the human brain. This review highlights the major findings of recent decades on the effects of gestational and lactational iron deficiency on the developing human brain. The findings are correlated to findings of experimental animals ranging from rodents to domestic pigs and non-human primates. The results point towards significant effects of iron deficiency on the developing brain. Evidence would be stronger with more studies addressing the human brain in real-time and the development of blood biomarkers of cerebral disturbance in iron deficiency. Cerebral iron deficiency is expected to be curable with iron substitution therapy, as the brain, privileged by the cerebral vascular transferrin receptor expression, is expected to facilitate iron extraction from the circulation and enable transport further into the brain.
Collapse
Affiliation(s)
- Veronika Markova
- Department of Obstetrics and Gynaecology, Hvidovre Hospital, Copenhagen University Hospital, 2650 Hvidovre, Denmark
- Pharmacosmos A/S, 4300 Holbæk, Denmark
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark
| | - Charlotte Holm
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark
| | - Anja Bisgaard Pinborg
- Fertility Clinic, Juliane Marie Centre, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Lars Lykke Thomsen
- Pharmacosmos A/S, 4300 Holbæk, Denmark
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark
| | - Torben Moos
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark.
| |
Collapse
|
157
|
Engineered antibodies: new possibilities for brain PET? Eur J Nucl Med Mol Imaging 2019; 46:2848-2858. [PMID: 31342134 PMCID: PMC6879437 DOI: 10.1007/s00259-019-04426-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022]
Abstract
Almost 50 million people worldwide are affected by Alzheimer’s disease (AD), the most common neurodegenerative disorder. Development of disease-modifying therapies would benefit from reliable, non-invasive positron emission tomography (PET) biomarkers for early diagnosis, monitoring of disease progression, and assessment of therapeutic effects. Traditionally, PET ligands have been based on small molecules that, with the right properties, can penetrate the blood–brain barrier (BBB) and visualize targets in the brain. Recently a new class of PET ligands based on antibodies have emerged, mainly in applications related to cancer. While antibodies have advantages such as high specificity and affinity, their passage across the BBB is limited. Thus, to be used as brain PET ligands, antibodies need to be modified for active transport into the brain. Here, we review the development of radioligands based on antibodies for visualization of intrabrain targets. We focus on antibodies modified into a bispecific format, with the capacity to undergo transferrin receptor 1 (TfR1)-mediated transcytosis to enter the brain and access pathological proteins, e.g. amyloid-beta. A number of such antibody ligands have been developed, displaying differences in brain uptake, pharmacokinetics, and ability to bind and visualize the target in the brain of transgenic mice. Potential pathological changes related to neurodegeneration, e.g. misfolded proteins and neuroinflammation, are suggested as future targets for this novel type of radioligand. Challenges are also discussed, such as the temporal match of radionuclide half-life with the ligand’s pharmacokinetic profile and translation to human use. In conclusion, brain PET imaging using bispecific antibodies, modified for receptor-mediated transcytosis across the BBB, is a promising method for specifically visualizing molecules in the brain that are difficult to target with traditional small molecule ligands.
Collapse
|
158
|
Abstract
The vertebrate vasculature displays high organotypic specialization, with the structure and function of blood vessels catering to the specific needs of each tissue. A unique feature of the central nervous system (CNS) vasculature is the blood-brain barrier (BBB). The BBB regulates substance influx and efflux to maintain a homeostatic environment for proper brain function. Here, we review the development and cell biology of the BBB, focusing on the cellular and molecular regulation of barrier formation and the maintenance of the BBB through adulthood. We summarize unique features of CNS endothelial cells and highlight recent progress in and general principles of barrier regulation. Finally, we illustrate why a mechanistic understanding of the development and maintenance of the BBB could provide novel therapeutic opportunities for CNS drug delivery.
Collapse
Affiliation(s)
- Urs H Langen
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Swathi Ayloo
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
159
|
Lakkadwala S, Dos Santos Rodrigues B, Sun C, Singh J. Dual functionalized liposomes for efficient co-delivery of anti-cancer chemotherapeutics for the treatment of glioblastoma. J Control Release 2019; 307:247-260. [PMID: 31252036 DOI: 10.1016/j.jconrel.2019.06.033] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/04/2019] [Accepted: 06/24/2019] [Indexed: 11/18/2022]
Abstract
Glioblastoma is a hostile brain tumor associated with high infiltration leading to poor prognosis. Anti-cancer chemotherapeutic agents have limited access into the brain due to the presence of the blood brain barrier (BBB). In this study, we designed a dual functionalized liposomal delivery system, surface modified with transferrin (Tf) for receptor mediated transcytosis and a cell penetrating peptide-penetratin (Pen) for enhanced cell penetration. We loaded doxorubicin and erlotinib into liposomes to enhance their translocation across the BBB to glioblastoma tumor. In vitro cytotoxicity and hemocompatibility studies demonstrated excellent biocompatibility for in vivo administration. Co-delivery of doxorubicin and erlotinib loaded Tf-Pen liposomes revealed significantly (p < 0.05) higher translocation (~15%) across the co-culture endothelial barrier resulting in regression of tumor in the in vitro brain tumor model. The biodistribution of Tf-Pen liposomes demonstrated ~12 and 3.3 fold increase in doxorubicin and erlotinib accumulation in mice brain, respectively compared to free drugs. In addition, Tf-Pen liposomes showed excellent antitumor efficacy by regressing ~90% of tumor in mice brain with significant increase in the median survival time (36 days) along with no toxicity. Thus, we believe that this study would have high impact for treating patients with glioblastoma.
Collapse
Affiliation(s)
- Sushant Lakkadwala
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Bruna Dos Santos Rodrigues
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Chengwen Sun
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA.
| |
Collapse
|
160
|
Fu M, He Q, Guo Z, Zhou X, Li H, Zhao L, Tang H, Zhou X, Zhu H, Shen G, He Y, Lei P. Therapeutic Bispecific T-Cell Engager Antibody Targeting the Transferrin Receptor. Front Immunol 2019; 10:1396. [PMID: 31293575 PMCID: PMC6598450 DOI: 10.3389/fimmu.2019.01396] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022] Open
Abstract
Bispecific T-cell engager antibodies (BiTE) have been explored as a means to recruit cytolytic T cells to kill tumor cells. The transferrin receptor (TfR) is highly expressed on the surface of rapidly proliferating tumor cells. Therefore, it holds great potential in T cell redirecting therapies. In this research, we developed a BiTE targeting TfR and CD3 (TfR-BiTE) and studied its therapeutic impact on TfR-positive cancer. TfR-BiTE had the ability to induce the selective lysis of various TfR-positive cancer cells through the activation of T cells, the release of cytokines, and then the coming proliferation of T cells, whereas TfR-negative cells were not affected. In a subcutaneous HepG2 xenograft model, low concentrations of TfR-BiTE inhibited tumor growth. Overall, these results reveal that TfR-BiTE can selectively deplete TfR-positive HepG2 cells; hence, it represents a novel immunotherapeutic approach for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mingpeng Fu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi He
- Department of Transfusion Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zilong Guo
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoran Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heli Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Zhao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongling Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huifen Zhu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong He
- Department of Nuclear Medicine and PET Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
161
|
Qian ZM, Ke Y. Brain iron transport. Biol Rev Camb Philos Soc 2019; 94:1672-1684. [PMID: 31190441 DOI: 10.1111/brv.12521] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 12/19/2022]
Abstract
Brain iron is a crucial participant and regulator of normal physiological activity. However, excess iron is involved in the formation of free radicals, and has been associated with oxidative damage to neuronal and other brain cells. Abnormally high brain iron levels have been observed in various neurodegenerative diseases, including neurodegeneration with brain iron accumulation, Alzheimer's disease, Parkinson's disease and Huntington's disease. However, the key question of why iron levels increase in the relevant regions of the brain remains to be answered. A full understanding of the homeostatic mechanisms involved in brain iron transport and metabolism is therefore critical not only for elucidating the pathophysiological mechanisms responsible for excess iron accumulation in the brain but also for developing pharmacological interventions to disrupt the chain of pathological events occurring in these neurodegenerative diseases. Numerous studies have been conducted, but to date no effort to synthesize these studies and ideas into a systematic and coherent summary has been made, especially concerning iron transport across the luminal (apical) membrane of the capillary endothelium and the membranes of different brain cell types. Herein, we review key findings on brain iron transport, highlighting the mechanisms involved in iron transport across the luminal (apical) as well as the abluminal (basal) membrane of the blood-brain barrier, the blood-cerebrospinal fluid barrier, and iron uptake and release in neurons, oligodendrocytes, astrocytes and microglia within the brain. We offer suggestions for addressing the many important gaps in our understanding of this important topic, and provide new insights into the potential causes of abnormally increased iron levels in regions of the brain in neurodegenerative disorders.
Collapse
Affiliation(s)
- Zhong-Ming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, 226019, China.,Laboratory of Neuropharmacology, School of Pharmacy, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
162
|
Sato K, Tachikawa M, Watanabe M, Uchida Y, Terasaki T. Selective Protein Expression Changes of Leukocyte-Migration-Associated Cluster of Differentiation Antigens at the Blood–Brain Barrier in a Lipopolysaccharide-Induced Systemic Inflammation Mouse Model without Alteration of Transporters, Receptors or Tight Junction-Related Protein. Biol Pharm Bull 2019; 42:944-953. [DOI: 10.1248/bpb.b18-00939] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kazuki Sato
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Michitoshi Watanabe
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
163
|
Durcanova B, Appleton J, Gurijala N, Belov V, Giffenig P, Moeller E, Hogan M, Lee F, Papisov M. The Configuration of the Perivascular System Transporting Macromolecules in the CNS. Front Neurosci 2019; 13:511. [PMID: 31191221 PMCID: PMC6547014 DOI: 10.3389/fnins.2019.00511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/03/2019] [Indexed: 12/26/2022] Open
Abstract
Large blood vessels entering the CNS are surrounded by perivascular spaces that communicate with the cerebrospinal fluid and, at their termini, with the interstitial space. Solutes and particles can translocate along these perivascular conduits, reportedly in both directions. Recently, this prompted a renewed interest in the intrathecal therapy delivery route for CNS-targeted therapeutics. However, the extent of the CNS coverage by the perivascular system is unknown, making the outcome of drug administration to the CSF uncertain. We traced the translocation of model macromolecules from the CSF into the CNS of rats and non-human primates. Conduits transporting macromolecules were found to extend throughout the parenchyma from both external and internal (fissures) CNS boundaries, excluding ventricles, in large numbers, on average ca. 40 channels per mm2 in rats and non-human primates. The high density and depth of extension of the perivascular channels suggest that the perivascular route can be suitable for delivery of therapeutics to parenchymal targets throughout the CNS.
Collapse
Affiliation(s)
| | | | | | - Vasily Belov
- Massachusetts General Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Shriners Hospitals for Children - Boston, Boston, MA, United States
| | - Pilar Giffenig
- Massachusetts General Hospital, Boston, MA, United States
| | | | - Matthew Hogan
- Massachusetts General Hospital, Boston, MA, United States
| | - Fredella Lee
- Massachusetts General Hospital, Boston, MA, United States
| | - Mikhail Papisov
- Massachusetts General Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Shriners Hospitals for Children - Boston, Boston, MA, United States
| |
Collapse
|
164
|
Neely CLC, Lippi SLP, Lanzirotti A, Flinn JM. Localization of Free and Bound Metal Species through X-Ray Synchrotron Fluorescence Microscopy in the Rodent Brain and Their Relation to Behavior. Brain Sci 2019; 9:E74. [PMID: 30925761 PMCID: PMC6523809 DOI: 10.3390/brainsci9040074] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/23/2019] [Accepted: 03/26/2019] [Indexed: 12/27/2022] Open
Abstract
Biometals in the brain, such as zinc, copper, and iron, are often discussed in cases of neurological disorders; however, these metals also have important regulatory functions and mediate cell signaling and plasticity. With the use of synchrotron X-ray fluorescence, our lab localized total, both bound and free, levels of zinc, copper, and iron in a cross section of one hemisphere of a rat brain, which also showed differing metal distributions in different regions within the hippocampus, the site in the brain known to be crucial for certain types of memory. This review discusses the several roles of these metals in brain regions with an emphasis on hippocampal cell signaling, based on spatial mapping obtained from X-ray fluorescence microscopy. We also discuss the localization of these metals and emphasize different cell types and receptors in regions with metal accumulation, as well as the potential relationship between this physiology and behavior.
Collapse
Affiliation(s)
- Caroline L C Neely
- Department of Psychology, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA.
| | - Stephen L P Lippi
- Department of Psychology & Sociology, Angelo State University, 2601 W. Avenue N, ASU Station #10907, San Angelo, TX 76909, USA.
| | - Antonio Lanzirotti
- Center for Advanced Radiation Sources, University of Chicago, 9700 South Cass Avenue, Argonne, IL 60439, USA.
| | - Jane M Flinn
- Department of Psychology, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA.
| |
Collapse
|
165
|
Arranz-Gibert P, Guixer B, Prades R, Ciudad S, Giralt E, Teixidó M. A MALDI-TOF-based Method for Studying the Transport of BBB Shuttles-Enhancing Sensitivity and Versatility of Cell-Based In Vitro Transport Models. Sci Rep 2019; 9:4875. [PMID: 30890722 PMCID: PMC6424956 DOI: 10.1038/s41598-019-40973-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 02/13/2019] [Indexed: 12/27/2022] Open
Abstract
In recent decades, peptide blood-brain barrier shuttles have emerged as a promising solution for brain drugs that are not able to enter this organ. The research and development of these compounds involve the use of in vitro cell-based models of the BBB. Nevertheless, peptide transport quantification implies the use of large amounts of peptide (upper micromolar range for RP-HPLC-PDA) or of derivatives (e.g. fluorophore or quantum-dot attachment, radiolabeling) in the donor compartment in order to enhance the detection of these molecules in the acceptor well, although their structure is highly modified. Therefore, these methodologies either hamper the use of low peptide concentrations, thus hindering mechanistic studies, or do not allow the use of the unmodified peptide. Here we successfully applied a MALDI-TOF MS methodology for transport quantification in an in vitro BBB cell-based model. A light version of the acetylated peptide was evaluated, and the transport was subsequently quantified using a heavy internal standard (isotopically acetylated). We propose that this MALDI-TOF MS approach could also be applied to study the transport across other biological barriers using the appropriate in vitro transport models (e.g. Caco-2, PAMPA).
Collapse
Affiliation(s)
- Pol Arranz-Gibert
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain
| | - Bernat Guixer
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain
| | - Roger Prades
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain
| | - Sonia Ciudad
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain. .,Department of Inorganic and Organic Chemistry, University of Barcelona, Martí i Franquès 1-11, Barcelona, E-08028, Spain.
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain.
| |
Collapse
|
166
|
Abstract
PURPOSE OF REVIEW To discuss the mechanisms of iron regulation in the brain and the pathophysiological role of deregulation of iron homeostasis following a stroke, and to review existing evidence supporting the potential role of iron chelators in the treatment of ischemic and hemorrhagic stroke. RECENT FINDINGS In recent years, accumulating evidence has highlighted the role of neuroinflammation in neurological injury after ischemic and hemorrhagic stroke, and that free iron is central to this process. Via the Fenton reaction, free iron catalyzes the conversion of superoxide ion and hydrogen peroxide into hydroxyl radicals, which promote oxidative stress. Advances in our understanding of changes in brain iron metabolism and its relationship to neuronal injury in stroke could provide new therapeutic strategies to improve the outcome of stroke patients. Pharmacological agents targeting brain iron regulation hold promise as potentially effective treatments in both ischemic and hemorrhagic stroke.
Collapse
Affiliation(s)
- Khalid A Hanafy
- Beth Israel Deaconess Medical Center, Department of Neurology, Division of Stroke & Cerebrovascular Disease, Harvard Medical School, 330 Brookline Avenue - Palmer 127, Boston, MA, 02215, USA
| | - Joao A Gomes
- Cerebrovascular Center, Cleveland Clinic, Cleveland, OH, USA
| | - Magdy Selim
- Beth Israel Deaconess Medical Center, Department of Neurology, Division of Stroke & Cerebrovascular Disease, Harvard Medical School, 330 Brookline Avenue - Palmer 127, Boston, MA, 02215, USA.
| |
Collapse
|
167
|
Moura RP, Martins C, Pinto S, Sousa F, Sarmento B. Blood-brain barrier receptors and transporters: an insight on their function and how to exploit them through nanotechnology. Expert Opin Drug Deliv 2019; 16:271-285. [PMID: 30767695 DOI: 10.1080/17425247.2019.1583205] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The blood-brain barrier (BBB) is a highly limiting barrier that prevents the brain from contacting with several circulating molecules, including harmful agents. However, certain systemic nutrients and macromolecules are able to cross the BBB and reach the brain parenchyma, involving the interaction with multiple receptors and/or transporters at the BBB surface. Nanotechnology allows the creation of drug vehicles, functionalized with targeting ligands for binding specific BBB receptors and/or transporters, hence triggering the transport through this biobarrier. AREAS COVERED This review focuses the BBB receptors/transporters to be exploited in regard to their overall structure and biologic function, as well as their role in the development of strategies envisaging drug delivery to the brain. Then, the interplay between the targeting of these BBB receptors/transporters and nanotechnology is explored, as they can increase by several-fold the effectiveness of brain-targeted therapies. EXPERT OPINION Nanomedicine may be particularly useful in brain drug delivery, mainly due to the possibility of functionalizing nanoparticles to target specific receptors/transporters. Since the BBB is endowed with numerous receptors and transporters responsible for regulating the proper metabolic activity of the brain, their targeting can be a promising bypass strategy to circumvent the hurdle that the BBB represents for brain drug delivery.
Collapse
Affiliation(s)
- Rui Pedro Moura
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal
| | - Cláudia Martins
- b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Soraia Pinto
- b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal
| | - Flávia Sousa
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal.,b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Bruno Sarmento
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal.,b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal
| |
Collapse
|
168
|
Zhang SH, Liu DX, Wang L, Li YH, Wang YH, Zhang H, Su ZK, Fang WG, Qin XX, Shang DS, Li B, Han XN, Zhao WD, Chen YH. A CASPR1-ATP1B3 protein interaction modulates plasma membrane localization of Na +/K +-ATPase in brain microvascular endothelial cells. J Biol Chem 2019; 294:6375-6386. [PMID: 30792309 DOI: 10.1074/jbc.ra118.006263] [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] [Received: 10/14/2018] [Revised: 02/13/2019] [Indexed: 01/01/2023] Open
Abstract
Contactin-associated protein 1 (CASPR1 or CNTNAP1) was recently reported to be expressed in brain microvascular endothelial cells (BMECs), the major component of the blood-brain barrier. To investigate CASPR1's physiological role in BMECs, here we used CASPR1 as a bait in a yeast two-hybrid screen to identify CASPR1-interacting proteins and identified the β3 subunit of Na+/K+-ATPase (ATP1B3) as a CASPR1-binding protein. Using recombinant and purified CASPR1, RNAi, GST-pulldown, immunofluorescence, immunoprecipitation, and Na+/K+-ATPase activity assays, we found that ATP1B3's core proteins, but not its glycosylated forms, interact with CASPR1, which was primarily located in the endoplasmic reticulum of BMECs. CASPR1 knockdown reduced ATP1B3 glycosylation and prevented its plasma membrane localization, phenotypes that were reversed by expression of full-length CASPR1. We also found that the CASPR1 knockdown reduces the plasma membrane distribution of the α1 subunit of Na+/K+-ATPase, which is the major component assembled with ATP1B3 in the complete Na+/K+-ATPase complex. The binding of CASPR1 with ATP1B3, but not the α1 subunit, indicated that CASPR1 binds with ATP1B3 to facilitate the assembly of Na+/K+-ATPase. Furthermore, the activity of Na+/K+-ATPase was reduced in CASPR1-silenced BMECs. Interestingly, shRNA-mediated CASPR1 silencing reduced glutamate efflux through the BMECs. These results demonstrate that CASPR1 binds with ATP1B3 and thereby contributes to the regulation of Na+/K+-ATPase maturation and trafficking to the plasma membrane in BMECs. We conclude that CASPR1-mediated regulation of Na+/K+-ATPase activity is important for glutamate transport across the blood-brain barrier.
Collapse
Affiliation(s)
- Shu-Hong Zhang
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and.,the Department of Cell Biology, School of Basic Medicine, Jiamusi University, 258 Xuefu Street, Jiamusi 154007, Heilongjiang Province, China
| | - Dong-Xin Liu
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Li Wang
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Yu-Hua Li
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Yan-Hua Wang
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Hu Zhang
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Zheng-Kang Su
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Wen-Gang Fang
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Xiao-Xue Qin
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - De-Shu Shang
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Bo Li
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Xiao-Ning Han
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Wei-Dong Zhao
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| | - Yu-Hua Chen
- From the Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China and
| |
Collapse
|
169
|
Kohata A, Hashim PK, Okuro K, Aida T. Transferrin-Appended Nanocaplet for Transcellular siRNA Delivery into Deep Tissues. J Am Chem Soc 2019; 141:2862-2866. [DOI: 10.1021/jacs.8b12501] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ai Kohata
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - P. K. Hashim
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kou Okuro
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Takuzo Aida
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Riken Center for Emergent Matter Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
170
|
Ayloo S, Gu C. Transcytosis at the blood-brain barrier. Curr Opin Neurobiol 2019; 57:32-38. [PMID: 30708291 DOI: 10.1016/j.conb.2018.12.014] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) is a functional interface separating the brain from the circulatory system and is essential for homeostasis of the central nervous system (CNS). The BBB regulates molecular flux to maintain an optimal environment for neuronal function and protects the brain from toxins and pathogens. Endothelial cells forming the walls of CNS blood vessels constitute the BBB. CNS endothelial cells exhibit two features that underlie the restrictive properties of the BBB: specialized tight junctions that prevent paracellular passage between the blood and the brain, and unusually low levels of vesicle trafficking that limit transcellular transport or transcytosis. While the prevailing view in the field was that specialized tight junctions contributed to CNS barrier properties, recent findings have revealed the importance of maintaining low rates of transcytosis at the BBB. It is now clear that suppression of transcytosis at the BBB is an active process and CNS-specific genetic programs inhibit this pathway to maintain a functional barrier.
Collapse
Affiliation(s)
- Swathi Ayloo
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA.
| |
Collapse
|
171
|
Sun J, Martin JM, Vanderpoel V, Sumbria RK. The Promises and Challenges of Erythropoietin for Treatment of Alzheimer's Disease. Neuromolecular Med 2019; 21:12-24. [PMID: 30656553 DOI: 10.1007/s12017-019-08524-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder in the world, and intracellular neurofibrillary tangles and extracellular amyloid-beta protein deposits represent the major pathological hallmarks of the disease. Currently available treatments provide some symptomatic relief but fail to modify primary pathological processes that underlie the disease. Erythropoietin (EPO), a hematopoietic growth factor, acts primarily to stimulate erythroid cell production, and is clinically used to treat anemia. EPO has evolved as a therapeutic agent for neurodegeneration and has improved neurological outcomes and AD pathology in rodents. However, penetration of the blood-brain barrier (BBB) and negative hematopoietic effects are the two major challenges for the therapeutic development of EPO for chronic neurodegenerative diseases like AD. The transferrin receptors at the BBB, which are responsible for transporting transferrin-bound iron from the blood into the brain parenchyma, can be used to shuttle therapeutic molecules across the BBB. In this review, we discuss the role of EPO as a potential neurotherapeutic for AD, challenges associated with EPO development for AD, and targeting the BBB transferrin receptor for EPO brain delivery.
Collapse
Affiliation(s)
- Jiahong Sun
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, 535 Watson Dr, Claremont, CA, 91711, USA
| | - Jan Michelle Martin
- College of Medicine, California Northstate University, Elk Grove, CA, 95757, USA
| | | | - Rachita K Sumbria
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, 535 Watson Dr, Claremont, CA, 91711, USA. .,Department of Neurology, University of California, Irvine, CA, 92868, USA.
| |
Collapse
|
172
|
Bourassa P, Alata W, Tremblay C, Paris-Robidas S, Calon F. Transferrin Receptor-Mediated Uptake at the Blood-Brain Barrier Is Not Impaired by Alzheimer's Disease Neuropathology. Mol Pharm 2019; 16:583-594. [PMID: 30609376 DOI: 10.1021/acs.molpharmaceut.8b00870] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The transferrin receptor (TfR) is highly expressed by brain capillary endothelial cells (BCECs) forming the blood-brain barrier (BBB) and is therefore considered as a potential target for brain drug delivery. Monoclonal antibodies binding to the TfR, such as clone Ri7, have been shown to internalize into BCECs in vivo. However, since Alzheimer's disease (AD) is accompanied by a BBB dysfunction, it raises concerns about whether TfR-mediated transport becomes inefficient during the progression of the disease. Measurements of TfR levels using Western blot analysis in whole homogenates from human post-mortem parietal cortex and hippocampus did not reveal any significant difference between individuals with or without a neuropathological diagnosis of AD (respectively, n = 19 and 22 for the parietal cortex and n = 12 and 14 for hippocampus). Similarly, TfR concentrations in isolated human brain microvessels from parietal cortex were similar between controls and AD cases. TfR levels in isolated murine brain microvessels were not significantly different between groups of 12- and 18-month-old NonTg and 3xTg-AD mice, the latter modeling Aβ and τ neuropathologies. In situ brain perfusion assays were then conducted to measure the brain uptake and internalization of fluorolabeled Ri7 in BCECs upon binding. Consistently, TfR-mediated uptake in BCECs was similar between 3xTg-AD mice and nontransgenic controls (∼0.3 μL·g-1·s-1) at 12, 18, and 22 months of age. Fluorescence microscopy analysis following intravenous administration of fluorolabeled Ri7 highlighted that the signal from the antibody was widely distributed throughout the cerebral vasculature but not in neurons or astrocytes. Overall, our data suggest that both TfR protein levels and TfR-dependent internalization mechanisms are preserved in the presence of Aβ and τ neuropathologies, supporting the potential of TfR as a vector target for drug delivery into BCECs in AD.
Collapse
Affiliation(s)
- Philippe Bourassa
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Wael Alata
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Cyntia Tremblay
- Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Sarah Paris-Robidas
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Frédéric Calon
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| |
Collapse
|
173
|
Wong KH, Riaz MK, Xie Y, Zhang X, Liu Q, Chen H, Bian Z, Chen X, Lu A, Yang Z. Review of Current Strategies for Delivering Alzheimer's Disease Drugs across the Blood-Brain Barrier. Int J Mol Sci 2019; 20:ijms20020381. [PMID: 30658419 PMCID: PMC6358942 DOI: 10.3390/ijms20020381] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/16/2018] [Accepted: 01/11/2019] [Indexed: 12/20/2022] Open
Abstract
Effective therapy for Alzheimer’s disease is a major challenge in the pharmaceutical sciences. There are six FDA approved drugs (e.g., donepezil, memantine) that show some effectiveness; however, they only relieve symptoms. Two factors hamper research. First, the cause of Alzheimer’s disease is not fully understood. Second, the blood-brain barrier restricts drug efficacy. This review summarized current knowledge relevant to both of these factors. First, we reviewed the pathophysiology of Alzheimer’s disease. Next, we reviewed the structural and biological properties of the blood-brain barrier. We then described the most promising drug delivery systems that have been developed in recent years; these include polymeric nanoparticles, liposomes, metallic nanoparticles and cyclodextrins. Overall, we aim to provide ideas and clues to design effective drug delivery systems for penetrating the blood-brain barrier to treat Alzheimer’s disease.
Collapse
Affiliation(s)
- Ka Hong Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | | | - Yuning Xie
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Xue Zhang
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China.
| | - Qiang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Huoji Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu 215500, China.
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu 215500, China.
| |
Collapse
|
174
|
Johnsen KB, Bak M, Melander F, Thomsen MS, Burkhart A, Kempen PJ, Andresen TL, Moos T. Modulating the antibody density changes the uptake and transport at the blood-brain barrier of both transferrin receptor-targeted gold nanoparticles and liposomal cargo. J Control Release 2019; 295:237-249. [PMID: 30633947 DOI: 10.1016/j.jconrel.2019.01.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/04/2019] [Accepted: 01/05/2019] [Indexed: 01/17/2023]
Abstract
Transport of the majority of therapeutic molecules to the brain is precluded by the presence of the blood-brain barrier (BBB) rendering efficient treatment of many neurological disorders impossible. This BBB, nonetheless, may be circumvented by targeting receptors and transport proteins expressed on the luminal surface of the brain capillary endothelial cells (BCECs). The transferrin receptor (TfR) has remained a popular target since its original description for this purpose, although clinical progression of TfR-targeted drug constructs or nanomedicines remains unsuccessful. One proposed issue pertaining to the use of TfR-targeting in nanomedicines is the efficient tuning of the ligand density on the nanoparticle surface. We studied the impact of TfR antibody density on the uptake and transport of nanoparticles into the brain, taking a parallel approach to investigate the impact on both antibody-functionalized gold nanoparticles (AuNPs) and cargo-loaded liposomes. We report that among three different low-range mean ligand densities (0.15, 0.3, and 0.6 ∗ 103 antibodies/μm2), the highest density yielded the highest ability towards both targeting of the BCECs and subsequent transport across the BBB in vivo, and in vitro using primary cultures of the murine BBB. We also find that TfR-targeting on liposomes in the mouse may induce severe adverse effects after intravenous administration.
Collapse
Affiliation(s)
- Kasper Bendix Johnsen
- Laboratory for Neurobiology, Biomedicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark; Center for Nanomedicine and Theranostics, Department of Health Technology, Technical University of Denmark, Denmark
| | - Martin Bak
- Center for Nanomedicine and Theranostics, Department of Health Technology, Technical University of Denmark, Denmark
| | - Fredrik Melander
- Center for Nanomedicine and Theranostics, Department of Health Technology, Technical University of Denmark, Denmark
| | - Maj Schneider Thomsen
- Laboratory for Neurobiology, Biomedicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Annette Burkhart
- Laboratory for Neurobiology, Biomedicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Paul Joseph Kempen
- Center for Nanomedicine and Theranostics, Department of Health Technology, Technical University of Denmark, Denmark
| | - Thomas Lars Andresen
- Center for Nanomedicine and Theranostics, Department of Health Technology, Technical University of Denmark, Denmark
| | - Torben Moos
- Laboratory for Neurobiology, Biomedicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
175
|
Li M, Du C, Guo N, Teng Y, Meng X, Sun H, Li S, Yu P, Galons H. Composition design and medical application of liposomes. Eur J Med Chem 2019; 164:640-653. [PMID: 30640028 DOI: 10.1016/j.ejmech.2019.01.007] [Citation(s) in RCA: 345] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/04/2019] [Accepted: 01/04/2019] [Indexed: 12/11/2022]
Abstract
Liposomes, which possess the properties of nano-scale, biofilm similar structure, excellent biocompatibility, become more and more useful in the drug development as the delivery system. Liposomes are relatively stable, their aqueous phase could contain the hydrophilic drugs and their phospholipid bilayer should localize the lipophilic drugs. Moreover, their surface-modifiable characteristics have really extended the liposomes' application to targeting and environmental sensitive delivery system. In order to make the common liposome more fit the human and animal body's complex environment, the structural variation strategy in the head, tail and bond of lipid molecules have been employed to develop the different functionalized liposomes-based drug delivery system for the localizable relieve and organ/tissue targeting relieve. In this paper, we would like to summarize the recent development on the design and optimization of liposomes, including Long-circulation liposomes, Specific active targeting liposomes, Environmental sensitive liposomes, Multifunctional liposomes, and so on. And the liposome content selection and current status of clinical application are systematically discussed.
Collapse
Affiliation(s)
- Mingyuan Li
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Chunyang Du
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Na Guo
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yuou Teng
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Xin Meng
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Hua Sun
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Shuangshuang Li
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Peng Yu
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Hervé Galons
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/ Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| |
Collapse
|
176
|
Sato K, Tachikawa M, Watanabe M, Miyauchi E, Uchida Y, Terasaki T. Identification of Blood-Brain Barrier-Permeable Proteins Derived from a Peripheral Organ: In Vivo and in Vitro Evidence of Blood-to-Brain Transport of Creatine Kinase. Mol Pharm 2019; 16:247-257. [PMID: 30495961 DOI: 10.1021/acs.molpharmaceut.8b00975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Certain proteins, such as inflammatory cytokines, that are released from injured or diseased organs are transported from the circulating blood through the blood-brain barrier (BBB) into the brain and contribute to the pathogenesis of related central nervous system dysfunctions. However, little is known about the protein transport mechanisms involved in the central nervous system dysfunctions. The aims of the present study were to identify BBB-permeable protein(s) derived from liver and to clarify their transport characteristics at the BBB. After administration of biotin-labeled liver cytosolic protein fraction to mice in vivo, we identified 9 biotin-labeled proteins in the brain. Among them, we focused here on creatine kinase (CK). In vitro uptake studies with human brain microvessel endothelial cells (hCMEC/D3 cells) showed preferential uptake of muscle-type CK (CK-MM) compared with brain-type CK (CK-BB) at the BBB. Integration plot analysis revealed that CK-MM readily penetrated into brain parenchyma from the circulating blood across the BBB. The uptake of CK-MM by hCMEC/D3 cells was decreased at 4 °C and in the presence of clathrin- and caveolin-dependent endocytosis inhibitors. These results indicate that entry of CK into the brain is mediated by a transport system(s) at the BBB.
Collapse
Affiliation(s)
- Kazuki Sato
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Michitoshi Watanabe
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Eisuke Miyauchi
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8577 , Japan
| |
Collapse
|
177
|
Tang J, Wang Q, Yu Q, Qiu Y, Mei L, Wan D, Wang X, Li M, He Q. A stabilized retro-inverso peptide ligand of transferrin receptor for enhanced liposome-based hepatocellular carcinoma-targeted drug delivery. Acta Biomater 2019; 83:379-389. [PMID: 30395963 DOI: 10.1016/j.actbio.2018.11.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 10/02/2018] [Accepted: 11/01/2018] [Indexed: 12/16/2022]
Abstract
The application of tumor targeting ligands to the treatment of cancer holds promise for improving efficacy and reducing toxicity. LT7 (L(HAIYPRH)) peptide, a phage display-selected peptide, exhibited high binding affinity to transferrin receptor (TfR) overexpressed on tumor cells. However, its in vivo tumor targeting efficiency was impaired due to enzymatic degradation in blood circulation. To improve the stability and targeting ability, a retro-inverso analogue of LT7 peptide, named DT7 peptide (D(HRPYIAH)), was designed for targeted therapy of hepatocellular carcinoma. The result of computer simulation predicted that DT7 bound to TfR protein more efficiently than LT7, and this prediction was confirmed experimentally by surface plasmon resonance (SPR). Ex vivo stability experiment demonstrated that DT7 possessed stronger ability against proteolysis than LT7 in fresh mouse serum. We further prepared DT7-, LT7-, and transferrin (Tf)-modified liposomes (DT7-LIP, LT7-LIP, and Tf-LIP, respectively). DT7-LIP showed a significantly stronger in vitro targeting ability than LT7-LIP and Tf-LIP under normal condition and simulated biological condition. In addition, the in vitro antitumor effect of DTX-loaded DT7-LIP was markedly enhanced in comparison to DTX-loaded LT7-LIP and DTX-loaded Tf-LIP. In vivo imaging indicated that DT7-LIP had better tumor accumulation than LT7-LIP and Tf-LIP. For in vivo antitumor studies, the tumor growth rate of mice treated with DTX-loaded DT7-LIP was significantly inhibited compared to that in mice treated with DTX-loaded LT7-LIP and DTX-loaded Tf-LIP. Overall, this study verified the potential of the stable DT7 peptide in improving the efficacy of docetaxel in the treatment of hepatocellular carcinoma. STATEMENT OF SIGNIFICANCE: A phage display library-selected LT7 (L(HAIYPRH)) peptide exhibited high affinity to transferrin receptor (TfR). However, its bioactivity was impaired in vivo as L-peptides are susceptible to degradation by proteolytic enzymes. Here, we designed a retro-inverso peptide DT7(D(HRPYIAH)) and demonstrated its increased serum stability and higher binding affinity to TfR. A stabilized targeted drug delivery system was further constructed by modified DT7 peptide on the surface of liposomes. The data indicated that DT7 peptide-modified liposomes exhibited higher targeting ability in vitro and in vivo. More importantly, DT7-modified liposomes demonstrated positive preclinical significance in enhancing the therapeutic effects against hepatocellular carcinoma.
Collapse
|
178
|
Bu XL, Xiang Y, Guo Y. The Role of Iron in Amyotrophic Lateral Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1173:145-152. [PMID: 31456209 DOI: 10.1007/978-981-13-9589-5_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the degeneration of motor neurons in the motor cortex, brainstem, and spinal cord. The etiology and pathogenesis of this devastating disease remain largely unknown. Increasing evidence suggests that iron accumulation is involved in the onset and progression of ALS. In this review, we discuss the regulation of iron homoeostasis in the brain, the misregulation of iron homeostasis in ALS, and its possible roles in the mechanism of the disease. Finally, we summarize the recent progress and problems with respect to iron chelator therapies on ALS, aiming to propose a new therapeutic strategy to ameliorate the progression of the disease.
Collapse
Affiliation(s)
- Xian-Le Bu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yang Xiang
- Department of Neurology, Chengdu Military General Hospital, Chengdu, China
| | - Yansu Guo
- Beijing Geriatric Healthcare Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
179
|
Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol Rev 2019; 99:21-78. [PMID: 30280653 PMCID: PMC6335099 DOI: 10.1152/physrev.00050.2017] [Citation(s) in RCA: 1412] [Impact Index Per Article: 235.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) prevents neurotoxic plasma components, blood cells, and pathogens from entering the brain. At the same time, the BBB regulates transport of molecules into and out of the central nervous system (CNS), which maintains tightly controlled chemical composition of the neuronal milieu that is required for proper neuronal functioning. In this review, we first examine molecular and cellular mechanisms underlying the establishment of the BBB. Then, we focus on BBB transport physiology, endothelial and pericyte transporters, and perivascular and paravascular transport. Next, we discuss rare human monogenic neurological disorders with the primary genetic defect in BBB-associated cells demonstrating the link between BBB breakdown and neurodegeneration. Then, we review the effects of genes underlying inheritance and/or increased susceptibility for Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and amyotrophic lateral sclerosis (ALS) on BBB in relation to other pathologies and neurological deficits. We next examine how BBB dysfunction relates to neurological deficits and other pathologies in the majority of sporadic AD, PD, and ALS cases, multiple sclerosis, other neurodegenerative disorders, and acute CNS disorders such as stroke, traumatic brain injury, spinal cord injury, and epilepsy. Lastly, we discuss BBB-based therapeutic opportunities. We conclude with lessons learned and future directions, with emphasis on technological advances to investigate the BBB functions in the living human brain, and at the molecular and cellular level, and address key unanswered questions.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
180
|
Kopec BM, Ulapane KR, Moral MEG, Siahaan TJ. Methods of Delivering Molecules Through the Blood-Brain Barrier for Brain Diagnostics and Therapeutics. BLOOD-BRAIN BARRIER 2019. [DOI: 10.1007/978-1-4939-8946-1_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
181
|
Arranz-Gibert P, Prades R, Guixer B, Guerrero S, Araya E, Ciudad S, Kogan MJ, Giralt E, Teixidó M. HAI Peptide and Backbone Analogs-Validation and Enhancement of Biostability and Bioactivity of BBB Shuttles. Sci Rep 2018; 8:17932. [PMID: 30560894 PMCID: PMC6298966 DOI: 10.1038/s41598-018-35938-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 11/09/2018] [Indexed: 01/03/2023] Open
Abstract
Low effectiveness and resistance to treatments are commonplace in disorders of the central nervous system (CNS). These issues concern mainly the blood-brain barrier (BBB), which preserves homeostasis in the brain and protects this organ from toxic molecules and biohazards by regulating transport through it. BBB shuttles—short peptides able to cross the BBB—are being developed to help therapeutics to cross this barrier. BBB shuttles can be discovered by massive exploration of chemical diversity (e.g. computational means, phage display) or rational design (e.g. derivatives from a known peptide/protein able to cross). Here we present the selection of a peptide shuttle (HAI) from several candidates and the subsequent in-depth in vitro and in vivo study of this molecule. In order to explore the chemical diversity of HAI and enhance its biostability, and thereby its bioactivity, we explored two new protease-resistant versions of HAI (i.e. the retro-D-version, and a version that was N-methylated at the most sensitive sites to enzymatic cleavage). Our results show that, while both versions of HAI are resistant to proteases, the retro-D-approach preserved better transport properties.
Collapse
Affiliation(s)
- Pol Arranz-Gibert
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain
| | - Roger Prades
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain
| | - Bernat Guixer
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain
| | - Simón Guerrero
- Department of Pharmacological and Toxicological Chemistry, Faculty of Pharmaceutical Sciences, University of Chile, Sergio Livingstone, 1007, Independencia, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone, 1007, Independencia, Santiago, Chile
| | - Eyleen Araya
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone, 1007, Independencia, Santiago, Chile.,Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Av. Republica 275, Santiago, Chile
| | - Sonia Ciudad
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain
| | - Marcelo J Kogan
- Department of Pharmacological and Toxicological Chemistry, Faculty of Pharmaceutical Sciences, University of Chile, Sergio Livingstone, 1007, Independencia, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone, 1007, Independencia, Santiago, Chile
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain. .,Department of Inorganic and Organic Chemistry, University of Barcelona, Martí i Franquès 1-11, Barcelona, E-08028, Spain.
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, Barcelona, E-08028, Spain.
| |
Collapse
|
182
|
Kobayashi H. Recent trends in mucopolysaccharidosis research. J Hum Genet 2018; 64:127-137. [PMID: 30451936 DOI: 10.1038/s10038-018-0534-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023]
Abstract
Mucopolysaccharidosis (MPS) is a group of inherited conditions involving metabolic dysfunction. Lysosomal enzyme deficiency leads to the accumulation of glycosaminoglycan (GAG) resulting in systemic symptoms, and is categorized into seven types caused by deficiency in one of eleven different enzymes. The pathophysiological mechanism of these diseases has been investigated, indicating impaired autophagy in neuronal damage initiation, association of activated microglia and astrocytes with the neuroinflammatory processes, and involvement of tauopathy. A new inherited error of metabolism resulting in a multisystem disorder with features of the MPS was also identified. Additionally, new therapeutic methods are being developed that could improve conventional therapies, such as new recombinant enzymes that can penetrate the blood brain barrier, hematopoietic stem cell transplantation with reduced intensity conditioning, gene therapy using a viral vector system or gene editing, and substrate reduction therapy. In this review, we discuss the recent developments in MPS research and provide a framework for developing strategies.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Division of Gene Therapy, Research Center for Medical Sciences, Department of Pediatrics, The Jikei University School of Medicine, Tokyo, 105-8461, Japan.
| |
Collapse
|
183
|
Furtado D, Björnmalm M, Ayton S, Bush AI, Kempe K, Caruso F. Overcoming the Blood-Brain Barrier: The Role of Nanomaterials in Treating Neurological Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801362. [PMID: 30066406 DOI: 10.1002/adma.201801362] [Citation(s) in RCA: 390] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/09/2018] [Indexed: 05/24/2023]
Abstract
Therapies directed toward the central nervous system remain difficult to translate into improved clinical outcomes. This is largely due to the blood-brain barrier (BBB), arguably the most tightly regulated interface in the human body, which routinely excludes most therapeutics. Advances in the engineering of nanomaterials and their application in biomedicine (i.e., nanomedicine) are enabling new strategies that have the potential to help improve our understanding and treatment of neurological diseases. Herein, the various mechanisms by which therapeutics can be delivered to the brain are examined and key challenges facing translation of this research from benchtop to bedside are highlighted. Following a contextual overview of the BBB anatomy and physiology in both healthy and diseased states, relevant therapeutic strategies for bypassing and crossing the BBB are discussed. The focus here is especially on nanomaterial-based drug delivery systems and the potential of these to overcome the biological challenges imposed by the BBB. Finally, disease-targeting strategies and clearance mechanisms are explored. The objective is to provide the diverse range of researchers active in the field (e.g., material scientists, chemists, engineers, neuroscientists, and clinicians) with an easily accessible guide to the key opportunities and challenges currently facing the nanomaterial-mediated treatment of neurological diseases.
Collapse
Affiliation(s)
- Denzil Furtado
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mattias Björnmalm
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Materials, Department of Bioengineering, and the Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
- Cooperative Research Center for Mental Health, Parkville, Victoria, 3052, Australia
| | - Kristian Kempe
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
184
|
Mizrahy S, Gutkin A, Decuzzi P, Peer D. Targeting central nervous system pathologies with nanomedicines. J Drug Target 2018; 27:542-554. [PMID: 30296187 DOI: 10.1080/1061186x.2018.1533556] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
One of the major challenges in drug development is the delivery of therapeutics to the central nervous system (CNS). The blood-brain barrier (BBB), which modulates the passage of molecules from the CNS, presents a formidable obstacle that limits brain uptake of therapeutics and, therefore, impedes the treatment of multiple neurological pathologies. Targeted nanocarriers present an excellent opportunity for drug delivery into the brain leveraging on endogenous receptors to transport therapeutics across the BBB endothelium. Receptor-mediated transport endows multiple benefits over other conventional delivery methods such as the transient permeabilization of the BBB or the direct depositioning of intracranial depots. Herein, different strategies for nanocarrier targeting to the CNS are discussed, highlighting the challenges and recent developments.
Collapse
Affiliation(s)
- Shoshy Mizrahy
- a Laboratory of Precision NanoMedicine, School of Molecular Cell Biology and Biotechnology , George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv , Israel.,b Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering , Tel Aviv University , Tel Aviv , Israel.,c Center for Nanoscience and Nanotechnology , Tel Aviv University , Tel Aviv , Israel.,d Cancer Biology Research Center , Tel Aviv University , Tel Aviv , Israel.,e Laboratory of Nanotechnology for Precision Medicine , Fondazione Istituto Italiano di Tecnologia , Genoa , Italy
| | - Anna Gutkin
- a Laboratory of Precision NanoMedicine, School of Molecular Cell Biology and Biotechnology , George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv , Israel.,b Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering , Tel Aviv University , Tel Aviv , Israel.,c Center for Nanoscience and Nanotechnology , Tel Aviv University , Tel Aviv , Israel.,d Cancer Biology Research Center , Tel Aviv University , Tel Aviv , Israel
| | - Paolo Decuzzi
- e Laboratory of Nanotechnology for Precision Medicine , Fondazione Istituto Italiano di Tecnologia , Genoa , Italy
| | - Dan Peer
- a Laboratory of Precision NanoMedicine, School of Molecular Cell Biology and Biotechnology , George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv , Israel.,b Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering , Tel Aviv University , Tel Aviv , Israel.,c Center for Nanoscience and Nanotechnology , Tel Aviv University , Tel Aviv , Israel.,d Cancer Biology Research Center , Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
185
|
Chang R, Al Maghribi A, Vanderpoel V, Vasilevko V, Cribbs DH, Boado R, Pardridge WM, Sumbria RK. Brain Penetrating Bifunctional Erythropoietin-Transferrin Receptor Antibody Fusion Protein for Alzheimer's Disease. Mol Pharm 2018; 15:4963-4973. [PMID: 30252487 DOI: 10.1021/acs.molpharmaceut.8b00594] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Erythropoietin (EPO), a glycoprotein cytokine essential to hematopoiesis, has neuroprotective effects in rodent models of Alzheimer's disease (AD). However, high therapeutic doses or invasive routes of administration of EPO are required to achieve effective brain concentrations due to low blood-brain barrier (BBB) penetrability, and high EPO doses result in hematopoietic side effects. These obstacles can be overcome by engineering a BBB-penetrable analog of EPO, which is rapidly cleared from the blood, by fusing EPO to a chimeric monoclonal antibody targeting the transferrin receptor (cTfRMAb), which acts as a molecular Trojan horse to ferry the EPO into the brain via the transvascular route. In the current study, we investigated the effects of the BBB-penetrable analog of EPO on AD pathology in a double transgenic mouse model of AD. Five and a half month old male APPswe/PSEN1dE9 (APP/PS1) transgenic mice were treated with saline ( n = 10) or the BBB-penetrable EPO ( n = 10) 3 days/week intraperitoneally for 8 weeks, compared to same-aged C57BL/6J wild-type mice treated with saline ( n = 8) with identical regiment. At 9 weeks following treatment initiation, exploration and spatial memory were assessed with the open-field and Y-maze test, mice were sacrificed, and brains were evaluated for Aβ peptide load, synaptic loss, BBB disruption, microglial activation, and microhemorrhages. APP/PS1 mice treated with the BBB-penetrable cTfRMAb-EPO fusion protein had significantly lower cortical and hippocampal Aβ peptide number ( p < 0.05) and immune-positive area ( p < 0.05), a decrease in hippocampal synaptic loss ( p < 0.05) and cortical microglial activation ( p < 0.001), and improved spatial memory ( p < 0.05) compared with APP/PS1 saline controls. BBB-penetrating EPO was not associated with microhemorrhage development. The cTfRMAb-EPO fusion protein offers therapeutic benefits by targeting multiple targets of AD pathogenesis and progression (Aβ load, synaptic loss, microglial activation) and improving spatial memory in the APP/PS1 mouse model of AD.
Collapse
Affiliation(s)
- Rudy Chang
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences , Keck Graduate Institute , Claremont , California 91711 , United States
| | - Abrar Al Maghribi
- Henry E. Riggs School of Applied Life Sciences , Keck Graduate Institute , Claremont , California 91711 , United States
| | - Victoria Vanderpoel
- Department of Neuroscience , Pomona College , Claremont , California 91711 , United States
| | - Vitaly Vasilevko
- Institute for Memory Impairments and Neurological Disorders , University of California , Irvine , California 92697 , United States
| | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders , University of California , Irvine , California 92697 , United States
| | - Ruben Boado
- ArmaGen, Inc. , Calabasas , California 91302 , United States
| | | | - Rachita K Sumbria
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences , Keck Graduate Institute , Claremont , California 91711 , United States.,Institute for Memory Impairments and Neurological Disorders , University of California , Irvine , California 92697 , United States
| |
Collapse
|
186
|
Dos Santos Rodrigues B, Oue H, Banerjee A, Kanekiyo T, Singh J. Dual functionalized liposome-mediated gene delivery across triple co-culture blood brain barrier model and specific in vivo neuronal transfection. J Control Release 2018; 286:264-278. [PMID: 30071253 PMCID: PMC6138570 DOI: 10.1016/j.jconrel.2018.07.043] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/12/2018] [Accepted: 07/27/2018] [Indexed: 12/19/2022]
Abstract
Gene therapy has become a promising approach for neurodegenerative disease treatment, however there is an urgent need to develop an efficient gene carrier to transport gene across the blood brain barrier (BBB). In this study, we strategically designed dual functionalized liposomes for efficient neuronal transfection by combining transferrin (Tf) receptor targeting and enhanced cell penetration utilizing penetratin (Pen). A triple cell co-culture model of BBB confirmed the ability of the liposomes to cross the barrier layer and transfect primary neuronal cells. In vivo quantification of PenTf-liposomes demonstrated expressive accumulation in the brain (12%), without any detectable cellular damage or morphological change. The efficacy of these nanoparticles containing plasmid β-galactosidase in modulating transfection was assessed by β-galactosidase expression in vivo. As a consequence of accumulation in the brain, PenTf-liposomes significantly induced gene expression in mice. Immunofluorescence studies of brain sections of mice after tail vein injection of liposomes encapsulating pDNA encoding GFP (pGFP) illustrate the superior ability of dual-functionalized liposomes to accumulate in the brain and transfect neurons. Taken together, the multifunctional liposomes provide an excellent gene delivery platform for neurodegenerative diseases.
Collapse
Affiliation(s)
- Bruna Dos Santos Rodrigues
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA
| | - Hiroshi Oue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Amrita Banerjee
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND 58105, USA.
| |
Collapse
|
187
|
Haqqani AS, Thom G, Burrell M, Delaney CE, Brunette E, Baumann E, Sodja C, Jezierski A, Webster C, Stanimirovic DB. Intracellular sorting and transcytosis of the rat transferrin receptor antibody OX26 across the blood-brain barrier in vitro is dependent on its binding affinity. J Neurochem 2018; 146:735-752. [PMID: 29877588 PMCID: PMC6175443 DOI: 10.1111/jnc.14482] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/03/2018] [Accepted: 05/22/2018] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) is a formidable obstacle to the delivery of therapeutics to the brain. Antibodies that bind transferrin receptor (TfR), which is enriched in brain endothelial cells, have been shown to cross the BBB and are being developed as fusion proteins to deliver therapeutic cargos to brain targets. Various antibodies have been developed for this purpose and their in vivo evaluation demonstrated that either low affinity or monovalent receptor binding re-directs their transcellular trafficking away from lysosomal degradation and toward improved exocytosis on the abluminal side of the BBB. However, these studies have been performed with antibodies that recognize different TfR epitopes and have different binding characteristics, preventing inter-study comparisons. In this study, the efficiency of transcytosis in vitro and intracellular trafficking in endosomal compartments were evaluated in an in vitro BBB model for affinity variants (Kd from 5 to174 nM) of the rat TfR-binding antibody, OX26. Distribution in subcellular fractions of the rat brain endothelial cells was determined using both targeted quantitative proteomics-selected reaction monitoring and fluorescent imaging with markers of early- and late endosomes. The OX26 variants with affinities of 76 and 108 nM showed improved trancytosis (Papp values) across the in vitro BBB model compared with a 5 nM OX26. Although ~40% of the 5 nM OX26 and ~35% of TfR co-localized with late-endosome/lysosome compartment, 76 and 108 nM affinity variants showed lower amounts in lysosomes and a predominant co-localization with early endosome markers. The study links bivalent TfR antibody affinity to mechanisms of sorting and trafficking away from late endosomes and lysosomes, resulting in improvement in their transcytosis efficiency. OPEN PRACTICES Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ Cover Image for this issue: doi: 10.1111/jnc.14193.
Collapse
Affiliation(s)
- Arsalan S. Haqqani
- National Research Council of CanadaHuman Health Therapeutics Research CentreOttawaONCanada
| | - George Thom
- Antibody Discovery and Protein EngineeringMedImmune, Milstein BuildingGranta ParkCambridgeUK
| | - Matthew Burrell
- Antibody Discovery and Protein EngineeringMedImmune, Milstein BuildingGranta ParkCambridgeUK
| | - Christie E. Delaney
- National Research Council of CanadaHuman Health Therapeutics Research CentreOttawaONCanada
| | - Eric Brunette
- National Research Council of CanadaHuman Health Therapeutics Research CentreOttawaONCanada
| | - Ewa Baumann
- National Research Council of CanadaHuman Health Therapeutics Research CentreOttawaONCanada
| | - Caroline Sodja
- National Research Council of CanadaHuman Health Therapeutics Research CentreOttawaONCanada
| | - Anna Jezierski
- National Research Council of CanadaHuman Health Therapeutics Research CentreOttawaONCanada
| | - Carl Webster
- Antibody Discovery and Protein EngineeringMedImmune, Milstein BuildingGranta ParkCambridgeUK
| | - Danica B. Stanimirovic
- National Research Council of CanadaHuman Health Therapeutics Research CentreOttawaONCanada
| |
Collapse
|
188
|
Olajide OJ, Fatoye JO, Idowu OF, Ilekoya D, Gbadamosi IT, Gbadamosi MT, Asogwa NT. Reversal of behavioral decline and neuropathology by a complex vitamin supplement involves modulation of key neurochemical stressors. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 62:120-131. [PMID: 30005307 DOI: 10.1016/j.etap.2018.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/03/2018] [Accepted: 07/05/2018] [Indexed: 06/08/2023]
Abstract
Metal ions are crucial for normal neurochemical signaling and perturbations in their homeostasis have been associated with neurodegenerative processes. Hypothesizing that in vivo modulation of key neurochemical processes including metal ion regulation (by transferrin receptor-1: TfR-1) in cells can improve disease outcome, we investigated the efficacy of a complex vitamin supplement (CVS) containing B-vitamins and ascorbic acid in preventing/reversing behavioral decline and neuropathology in rats. Wistar rats (eight weeks-old) were assigned into five groups (n = 8), including controls and those administered CVS (400 mg/kg/day) for two weeks before or after AlCl3 (100 mg/kg)-induced neurotoxicity. Following behavioral assessments, prefrontal cortex (PFC) and hippocampus were prepared for biochemical analyses, histology and histochemistry. CVS significantly reversed reduction of exploratory/working memory, frontal-dependent motor deficits, cognitive decline, memory dysfunction and anxiety. These correlated with CVS-dependent modulation of TfP-1 expression that were accompanied by significant reversal of neural oxidative stress in expressed superoxide dismutase, nitric oxide, catalase, glutathione peroxidase and malondialdehyde. Furthermore, CVS inhibited neural bioenergetics dysfunction, with increased labelling of glucokinase within PFC and hippocampus correlating with increased glucose-6-phosphate dehydrogenase and decreased lactate dehydrogenase expressions. These relates to inhibition of over-expressed acetylcholinesterase and increased total protein synthesis. Histological and Nissl staining of thin sections corroborated roles of CVS in reversing AlCl3-induced neuropathology. Summarily, we showed the role of CVS in normalizing important neurochemical molecules linking concurrent progression of oxidative stress, bioenergetics deficits, synaptic dysfunction and cellular hypertrophy during neurodegeneration.
Collapse
Affiliation(s)
- Olayemi Joseph Olajide
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Nigeria.
| | - John Oluwasegun Fatoye
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Nigeria
| | - Oluwakunmi Folashade Idowu
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Nigeria
| | - Damilola Ilekoya
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Nigeria
| | - Ismail Temitayo Gbadamosi
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Nigeria
| | | | - Nnaemeka Tobechukwu Asogwa
- Department of Biochemistry, Faculty of Life Sciences, University of Ilorin, Nigeria; Central Research Laboratories Ltd, 132b University Road, Ilorin, Nigeria
| |
Collapse
|
189
|
Wevers NR, Kasi DG, Gray T, Wilschut KJ, Smith B, van Vught R, Shimizu F, Sano Y, Kanda T, Marsh G, Trietsch SJ, Vulto P, Lanz HL, Obermeier B. A perfused human blood-brain barrier on-a-chip for high-throughput assessment of barrier function and antibody transport. Fluids Barriers CNS 2018; 15:23. [PMID: 30165870 PMCID: PMC6117964 DOI: 10.1186/s12987-018-0108-3] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/30/2018] [Indexed: 12/13/2022] Open
Abstract
Background Receptor-mediated transcytosis is one of the major routes for drug delivery of large molecules into the brain. The aim of this study was to develop a novel model of the human blood–brain barrier (BBB) in a high-throughput microfluidic device. This model can be used to assess passage of large biopharmaceuticals, such as therapeutic antibodies, across the BBB. Methods The model comprises human cell lines of brain endothelial cells, astrocytes, and pericytes in a two-lane or three-lane microfluidic platform that harbors 96 or 40 chips, respectively, in a 384-well plate format. In each chip, a perfused vessel of brain endothelial cells was grown against an extracellular matrix gel, which was patterned by means of surface tension techniques. Astrocytes and pericytes were added on the other side of the gel to complete the BBB on-a-chip model. Barrier function of the model was studied using fluorescent barrier integrity assays. To test antibody transcytosis, the lumen of the model’s endothelial vessel was perfused with an anti-transferrin receptor antibody or with a control antibody. The levels of antibody that penetrated to the basal compartment were quantified using a mesoscale discovery assay. Results The perfused BBB on-a-chip model shows presence of adherens and tight junctions and severely limits the passage of a 20 kDa FITC-dextran dye. Penetration of the antibody targeting the human transferrin receptor (MEM-189) was markedly higher than penetration of the control antibody (apparent permeability of 2.9 × 10−5 versus 1.6 × 10−5 cm/min, respectively). Conclusions We demonstrate successful integration of a human BBB microfluidic model in a high-throughput plate-based format that can be used for drug screening purposes. This in vitro model shows sufficient barrier function to study the passage of large molecules and is sensitive to differences in antibody penetration, which could support discovery and engineering of BBB-shuttle technologies. Electronic supplementary material The online version of this article (10.1186/s12987-018-0108-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nienke R Wevers
- Mimetas BV, J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands. .,Department of Cell and Chemical Biology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands.
| | - Dhanesh G Kasi
- Mimetas BV, J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Taylor Gray
- Biogen, 225 Binney Street, Cambridge, MA, 02142, USA
| | | | | | - Remko van Vught
- Mimetas BV, J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | - Fumitaka Shimizu
- Yamaguchi University Graduate School of Medicine, Minamikogushi, Ube, Yamaguchi, 7558505, Japan
| | - Yasuteru Sano
- Yamaguchi University Graduate School of Medicine, Minamikogushi, Ube, Yamaguchi, 7558505, Japan
| | - Takashi Kanda
- Yamaguchi University Graduate School of Medicine, Minamikogushi, Ube, Yamaguchi, 7558505, Japan
| | - Graham Marsh
- Biogen, 225 Binney Street, Cambridge, MA, 02142, USA
| | | | - Paul Vulto
- Mimetas BV, J.H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| | | | | |
Collapse
|
190
|
Zou Z, Shen Q, Pang Y, Li X, Chen Y, Wang X, Luo X, Wu Z, Bao Z, Zhang J, Liang J, Kong L, Yan L, Xiong L, Zhu T, Yuan S, Wang M, Cai K, Yao Y, Wu J, Jiang Y, Liu H, Liu J, Zhou Y, Dong Q, Wang W, Zhu K, Li L, Lou Y, Wang H, Li Y, Lin H. The synthesized transporter K16APoE enabled the therapeutic HAYED peptide to cross the blood-brain barrier and remove excess iron and radicals in the brain, thus easing Alzheimer’s disease. Drug Deliv Transl Res 2018; 9:394-403. [DOI: 10.1007/s13346-018-0579-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
191
|
Gothwal A, Nakhate KT, Alexander A, Ajazuddin, Gupta U. Boosted Memory and Improved Brain Bioavailability of Rivastigmine: Targeting Effort to the Brain Using Covalently Tethered Lower Generation PAMAM Dendrimers with Lactoferrin. Mol Pharm 2018; 15:4538-4549. [DOI: 10.1021/acs.molpharmaceut.8b00537] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Avinash Gothwal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Kartik T. Nakhate
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai, Chhattisgarh 490024, India
| | - Amit Alexander
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai, Chhattisgarh 490024, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai, Chhattisgarh 490024, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| |
Collapse
|
192
|
Chung JY, Kim HS, Song J. Iron metabolism in diabetes-induced Alzheimer's disease: a focus on insulin resistance in the brain. Biometals 2018; 31:705-714. [PMID: 30043289 PMCID: PMC6133192 DOI: 10.1007/s10534-018-0134-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/18/2018] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is characterized by an excessive accumulation of toxic amyloid beta (Aβ) plaques and memory dysfunction. The onset of AD is influenced by age, genetic background, and impaired glucose metabolism in the brain. Several studies have demonstrated that diabetes involving insulin resistance and glucose tolerance could lead to AD, ultimately resulting in cognitive dysfunction. Even though the relationship between diabetes and AD was indicated by significant evidences, the critical mechanisms and metabolic alterations in diabetes induced AD are not clear until now. Recently, iron metabolism has been shown to play multiple roles in the central nervous system (CNS). Iron deficiency and overload are associated with neurodegenerative diseases. Iron binds to Aβ and subsequently regulates Aβ toxicity in the CNS. In addition, previous studies have shown that iron is involved in the aggravation of insulin resistance. Considering these effects of iron metabolism in CNS, we expect that iron metabolism may play crucial roles in diabetic AD brain. Thus, we review the recent evidence regarding the relationship between diabetes-induced AD and iron metabolism.
Collapse
Affiliation(s)
- Ji Yeon Chung
- Department of Neurology, Chosun University School of Medicine and Hospital, Gwangju, 61452, South Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| |
Collapse
|
193
|
Stochastic simulations of nanoparticle internalization through transferrin receptor dependent clathrin-mediated endocytosis. Biochim Biophys Acta Gen Subj 2018; 1862:2104-2111. [PMID: 29959983 DOI: 10.1016/j.bbagen.2018.06.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/14/2018] [Accepted: 06/26/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Receptor dependent clathrin-mediated endocytosis (CME) is one of the most important endocytic pathways for the internalization of bioparticles into cells. During CME, the ligand-receptor interactions, development of clathrin-coated pit (CCP) and membrane evolution all act together to drive the internalization of bioparticles. In this work, we develop a stochastic computational model to investigate the CME based on the Metropolis Monte Carlo simulations. METHODS The model is based on the combination of a stochastic particle binding model with a membrane model. The energetic costs of membrane bending, CCP formation and ligand-receptor interactions are systematically linked together. RESULTS We implement our model to investigate the effects of particle size, ligand density and membrane stiffness on the overall process of CME from the drug delivery perspectives. Consistent with some experiments, our results show that the intermediate particle size and ligand density favor the particle internalization. Moreover, our results show that it is easier for a particle to enter a cell with softer membrane. CONCLUSIONS The model presented here is able to provide mechanistic insights into CME and can be readily modified to include other important factors, such as actins. The predictions from the model will aid in the therapeutic design of intracellular/transcellular drug delivery and antiviral interventions.
Collapse
|
194
|
Zeiadeh I, Najjar A, Karaman R. Strategies for Enhancing the Permeation of CNS-Active Drugs through the Blood-Brain Barrier: A Review. Molecules 2018; 23:molecules23061289. [PMID: 29843371 PMCID: PMC6100436 DOI: 10.3390/molecules23061289] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/22/2018] [Accepted: 05/24/2018] [Indexed: 01/03/2023] Open
Abstract
Background: The blood brain barrier (BBB) is a dynamic and functional structure which poses a vast challenge in the development of drugs acting on the central nervous system (CNS). While most substances are denied BBB crossing, selective penetration of substances mainly occurs through diffusion, carrier mediated transport, or receptor mediated transcytosis. Methods: Strategies in enhancing BBB penetration have been reviewed and summarized in accordance with their type of formulation. Highlights in monoclonal antibodies, peptide-vectors, nanoparticles, and simple prodrugs were included. Conclusion: Nanoparticles and simple prodrugs, for example, can be used for efficient BBB penetration through inhibition of efflux mechanisms, however, monoclonal antibodies are the most promising strategy in BBB penetration. Close follow-up of future development in this area should confirm our expectation.
Collapse
Affiliation(s)
- Isra' Zeiadeh
- Department of Bioorganic & Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Quds University, Jerusalem P.O. Box 20002, Palestine.
| | - Anas Najjar
- Department of Bioorganic & Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Quds University, Jerusalem P.O. Box 20002, Palestine.
| | - Rafik Karaman
- Department of Bioorganic & Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Quds University, Jerusalem P.O. Box 20002, Palestine.
| |
Collapse
|
195
|
Zhang X, He T, Chai Z, Samulski RJ, Li C. Blood-brain barrier shuttle peptides enhance AAV transduction in the brain after systemic administration. Biomaterials 2018; 176:71-83. [PMID: 29860139 DOI: 10.1016/j.biomaterials.2018.05.041] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022]
Abstract
The adeno-associated virus (AAV) vector has been used in preclinical and clinical trials of gene therapy for central nervous system (CNS) diseases. One of the biggest challenges of effectively delivering AAV to the brain is to surmount the blood-brain barrier (BBB). Herein, we identified several potential BBB shuttle peptides that significantly enhanced AAV8 transduction in the brain after a systemic administration, the best of which was the THR peptide. The enhancement of AAV8 brain transduction by THR is dose-dependent, and neurons are the primary THR targets. Mechanism studies revealed that THR directly bound to the AAV8 virion, increasing its ability to cross the endothelial cell barrier. Further experiments showed that binding of THR to the AAV virion did not interfere with AAV8 infection biology, and that THR competitively blocked transferrin from binding to AAV8. Taken together, our results demonstrate, for the first time, that BBB shuttle peptides are able to directly interact with AAV and increase the ability of the AAV vectors to cross the BBB for transduction enhancement in the brain. These results will shed important light on the potential applications of BBB shuttle peptides for enhancing brain transduction with systemic administration of AAV vectors.
Collapse
Affiliation(s)
- Xintao Zhang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ting He
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zheng Chai
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27510, USA.
| |
Collapse
|
196
|
Saxena M, Delgado Y, Sharma RK, Sharma S, Guzmán SLPDL, Tinoco AD, Griebenow K. Inducing cell death in vitro in cancer cells by targeted delivery of cytochrome c via a transferrin conjugate. PLoS One 2018; 13:e0195542. [PMID: 29649293 PMCID: PMC5896948 DOI: 10.1371/journal.pone.0195542] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 03/23/2018] [Indexed: 02/07/2023] Open
Abstract
One of the major drawbacks of many of the currently used cancer drugs are off-target effects. Targeted delivery is one method to minimize such unwanted and detrimental events. To actively target lung cancer cells, we have developed a conjugate of the apoptosis inducing protein cytochrome c with transferrin because the transferrin receptor is overexpressed by many rapidly dividing cancer cells. Cytochrome c and transferrin were cross-linked with a redox sensitive disulfide bond for the intra-cellular release of the protein upon endocytosis by the transferrin receptor. Confocal results demonstrated the cellular uptake of the cytochrome c-transferrin conjugate by transferrin receptor overexpressing A549 lung cancer cells. Localization studies further validated that this conjugate escaped the endosome. Additionally, an in vitro assay showed that the conjugate could induce apoptosis by activating caspase-3. The neo-conjugate not only maintained an IC50 value similar to the well known drug cisplatin (50 μM) in A549 cancer cells but also was nontoxic to the normal lung (MRC5) cells. Our neo-conjugate holds promise for future development to target cancers with enhanced transferrin receptor expression.
Collapse
Affiliation(s)
- Manoj Saxena
- Department of Environmental Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico, United States of America
| | - Yamixa Delgado
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico, United States of America
| | - Rohit Kumar Sharma
- Department of Environmental Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico, United States of America
| | - Shweta Sharma
- Department of Environmental Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico, United States of America
| | | | - Arthur D. Tinoco
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico, United States of America
| | - Kai Griebenow
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico, United States of America
| |
Collapse
|
197
|
Thom G, Burrell M, Haqqani AS, Yogi A, Lessard E, Brunette E, Delaney C, Baumann E, Callaghan D, Rodrigo N, Webster CI, Stanimirovic DB. Enhanced Delivery of Galanin Conjugates to the Brain through Bioengineering of the Anti-Transferrin Receptor Antibody OX26. Mol Pharm 2018; 15:1420-1431. [PMID: 29485883 DOI: 10.1021/acs.molpharmaceut.7b00937] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB) is a formidable obstacle for brain delivery of therapeutic antibodies. However, antibodies against the transferrin receptor (TfR), enriched in brain endothelial cells, have been developed as delivery carriers of therapeutic cargoes into the brain via a receptor-mediated transcytosis pathway. In vitro and in vivo studies demonstrated that either a low-affinity or monovalent binding of these antibodies to the TfR improves their release on the abluminal side of the BBB and target engagement in brain parenchyma. However, these studies have been performed with mouse-selective TfR antibodies that recognize different TfR epitopes and have varied binding characteristics. In this study, we evaluated serum pharmacokinetics and brain and CSF exposure of the rat TfR-binding antibody OX26 affinity variants, having KDs of 5 nM, 76 nM, 108 nM, and 174 nM, all binding the same epitope in bivalent format. Pharmacodynamic responses were tested in the Hargreaves chronic pain model after conjugation of OX26 affinity variants with the analgesic and antiepileptic peptide, galanin. OX26 variants with affinities of 76 nM and 108 nM showed enhanced brain and cerebrospinal fluid (CSF) exposure and higher potency in the Hargreaves model, compared to a 5 nM affinity variant; lowering affinity to 174 nM resulted in prolonged serum pharmacokinetics, but reduced brain and CSF exposure. The study demonstrates that binding affinity optimization of TfR-binding antibodies could improve their brain and CSF exposure even in the absence of monovalent TfR engagement.
Collapse
Affiliation(s)
- George Thom
- Antibody Discovery and Protein Engineering , MedImmune , Milstein Building, Granta Park, Cambridge CB21 6GH , U.K
| | - Matthew Burrell
- Antibody Discovery and Protein Engineering , MedImmune , Milstein Building, Granta Park, Cambridge CB21 6GH , U.K
| | - Arsalan S Haqqani
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Alvaro Yogi
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Etienne Lessard
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Eric Brunette
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Christie Delaney
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Ewa Baumann
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Deborah Callaghan
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Natalia Rodrigo
- Antibody Discovery and Protein Engineering , MedImmune , Milstein Building, Granta Park, Cambridge CB21 6GH , U.K
| | - Carl I Webster
- Antibody Discovery and Protein Engineering , MedImmune , Milstein Building, Granta Park, Cambridge CB21 6GH , U.K
| | - Danica B Stanimirovic
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| |
Collapse
|
198
|
Sonoda H, Morimoto H, Yoden E, Koshimura Y, Kinoshita M, Golovina G, Takagi H, Yamamoto R, Minami K, Mizoguchi A, Tachibana K, Hirato T, Takahashi K. A Blood-Brain-Barrier-Penetrating Anti-human Transferrin Receptor Antibody Fusion Protein for Neuronopathic Mucopolysaccharidosis II. Mol Ther 2018; 26:1366-1374. [PMID: 29606503 PMCID: PMC5993955 DOI: 10.1016/j.ymthe.2018.02.032] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/18/2018] [Accepted: 02/21/2018] [Indexed: 12/28/2022] Open
Abstract
Mucopolysaccharidosis II (MPS II) is an X-linked recessive lysosomal storage disease caused by mutations in the iduronate-2-sulfatase (IDS) gene. Since IDS catalyzes the degradation of glycosaminoglycans (GAGs), deficiency in this enzyme leads to accumulation of GAGs in most cells in all tissues and organs, resulting in severe somatic and neurological disorders. Although enzyme replacement therapy with human IDS (hIDS) has been used for the treatment of MPS II, this therapy is not effective for defects in the CNS mainly because the enzyme cannot cross the blood-brain barrier (BBB). Here, we developed a BBB-penetrating fusion protein, JR-141, which consists of an anti-human transferrin receptor (hTfR) antibody and intact hIDS. The TfR-mediated incorporation of JR-141 was confirmed by using human fibroblasts in vitro. When administrated intravenously to hTfR knockin mice or monkeys, JR-141, but not naked hIDS, was detected in the brain. In addition, the intravenous administration of JR-141 reduced the accumulation of GAGs both in the peripheral tissues and in the brain of hTfR knockin mice lacking Ids, an animal model of MPS II. These data provide a proof of concept for the translation of JR-141 to clinical study for the treatment of patients with MPS II with CNS disorders.
Collapse
Affiliation(s)
| | | | - Eiji Yoden
- Research Division, JCR Pharmaceuticals, Kobe, Japan
| | | | | | | | | | | | | | - Akira Mizoguchi
- Department of Neural Regeneration and Cell Communication, Mie University Graduate School of Medicine, Tsu, Japan
| | | | - Tohru Hirato
- Research Division, JCR Pharmaceuticals, Kobe, Japan
| | | |
Collapse
|
199
|
Assessing the role of hypothalamic microglia and blood vessel disruption in the development of angiotensin II-dependent hypertension in Cyp1a1-Ren2 rats. Pflugers Arch 2018; 470:883-895. [PMID: 29500668 DOI: 10.1007/s00424-018-2128-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/15/2018] [Accepted: 02/19/2018] [Indexed: 10/17/2022]
Abstract
Elevated plasma levels of the hormone vasopressin have been implicated in the pathogenesis of some forms of hypertension. Hypothalamic paraventricular and supraoptic nuclei neurons regulate vasopressin secretion into the circulation. Vasopressin neuron activity is elevated by day 7 in the development of angiotensin II-dependent hypertension in Cyp1a1-Ren2 rats. While microglial activation and blood-brain barrier (BBB) breakdown contribute to the maintenance of well-established hypertension, it is not known whether these mechanisms contribute to the early onset of hypertension. Hence, we aimed to determine whether microglia are activated and/or the BBB is compromised during the onset of hypertension. Here, we used the Cyp1a1-Ren2 rat model of hypertension and showed that ionised calcium-binding adapter molecule 1 staining of microglia does not change in the paraventricular and supraoptic nuclei on day 7 (early onset) and day 28 (well established) of hypertension, compared to the normotensive control. Endothelial transferrin receptor staining, which stains endothelia and reflects blood vessel density, was also unchanged at day 7, but was reduced at day 28, suggesting that breakdown of the BBB begins between day 7 and day 28 in the development of hypertension. Hence, this study does not support the idea that microglial activation or BBB disruption contribute to the onset of angiotensin II-dependent hypertension in Cyp1a1-Ren2 rats, although BBB disruption might contribute to the progression from the early onset to well-established hypertension.
Collapse
|
200
|
Khan AI, Liu J, Dutta P. Iron transport kinetics through blood-brain barrier endothelial cells. Biochim Biophys Acta Gen Subj 2018; 1862:1168-1179. [PMID: 29466707 DOI: 10.1016/j.bbagen.2018.02.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 02/14/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Transferrin and its receptors play an important role during the uptake and transcytosis of iron through blood-brain barrier (BBB) endothelial cells (ECs) to maintain iron homeostasis in BBB endothelium and brain. Any disruptions in the cell environment may change the distribution of transferrin receptors on the cell surface, which eventually alter the homeostasis and initiate neurodegenerative disorders. In this paper, we developed a comprehensive mathematical model that considers the necessary kinetics for holo-transferrin internalization and acidification, apo-transferrin recycling, and exocytosis of free iron and transferrin-bound iron through basolateral side of BBB ECs. METHODS Ordinary differential equations are formulated based on the first order reaction kinetics to model the iron transport considering their interactions with transferrin and transferrin receptors. Unknown kinetics rate constants are determined from experimental data by applying a non-linear optimization technique. RESULTS Using the estimated kinetic rate constants, the presented model can effectively reproduce the experimental data of iron transports through BBB ECs for many in-vitro studies. Model results also suggest that the BBB ECs can regulate the extent of the two possible iron transport pathways (free and transferrin-bound iron) by controlling the receptor expression, internalization of holo-transferrin-receptor complexes and acidification of holo-transferrin inside the cell endosomes. CONCLUSION The comprehensive mathematical model described here can predict the iron transport through BBB ECs considering various possible routes from blood side to brain side. The model can also predict the transferrin and iron transport behavior in iron-enriched and iron-depleted cells, which has not been addressed in previous work.
Collapse
Affiliation(s)
- Aminul Islam Khan
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920, United States
| | - Jin Liu
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920, United States
| | - Prashanta Dutta
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920, United States.
| |
Collapse
|