501
|
Rabiei M, Kashanian S, Samavati SS, Jamasb S, McInnes SJ. Active Targeting Towards and Inside the Brain based on Nanoparticles: A Review. Curr Pharm Biotechnol 2020; 21:374-383. [DOI: 10.2174/1389201020666191203094057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/08/2019] [Accepted: 11/22/2019] [Indexed: 11/22/2022]
Abstract
Background:
Treatment of neurological diseases using systemic and non-surgical techniques
presents a significant challenge in medicine. This challenge is chiefly associated with the condensation
and coherence of the brain tissue.
Methods:
The coherence structure of the brain is due to the presence of the blood-brain barrier (BBB),
which consists of a continuous layer of capillary endothelial cells. The BBB prevents most drugs from
entering the brain tissue and is highly selective, permitting only metabolic substances and nutrients to
pass through.
Results:
Although this challenge has caused difficulties for the treatment of neurological diseases, it
has opened up a broad research area in the field of drug delivery. Through the utilization of nanoparticles
(NPs), nanotechnology can provide the ideal condition for passing through the BBB.
Conclusion:
NPs with suitable dimensions and optimum hydrophobicity and charge, as well as appropriate
functionalization, can accumulate in the brain. Furthermore, NPs can facilitate the targeted delivery
of therapeutics into the brain areas involved in Alzheimer’s disease, Parkinson’s disease, stroke,
glioma, migraine, and other neurological disorders. This review describes these methods of actively
targeting specific areas of the brain.
Collapse
Affiliation(s)
- Morteza Rabiei
- Department of Nanobiotechnology, Razi University, Kermanshah, Iran
| | | | | | - Shahriar Jamasb
- Department of Biomedical Engineering, Hamedan University of Technology, Hamedan, 65169-13733, Iran
| | - Steven J.P. McInnes
- University of South Australia, Division of Information Technology, Engineering and the Environment, Mawson Lakes, Mawson Lakes 5095, Australia
| |
Collapse
|
502
|
Kirstein A, Schmid TE, Combs SE. The Role of miRNA for the Treatment of MGMT Unmethylated Glioblastoma Multiforme. Cancers (Basel) 2020; 12:cancers12051099. [PMID: 32354046 PMCID: PMC7281574 DOI: 10.3390/cancers12051099] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common high-grade intracranial tumor in adults. It is characterized by uncontrolled proliferation, diffuse infiltration due to high invasive and migratory capacities, as well as intense resistance to chemo- and radiotherapy. With a five-year survival of less than 3% and an average survival rate of 12 months after diagnosis, GBM has become a focus of current research to urgently develop new therapeutic approaches in order to prolong survival of GBM patients. The methylation status of the promoter region of the O6-methylguanine–DNA methyltransferase (MGMT) is nowadays routinely analyzed since a methylated promoter region is beneficial for an effective response to temozolomide-based chemotherapy. Furthermore, several miRNAs were identified regulating MGMT expression, apart from promoter methylation, by degrading MGMT mRNA before protein translation. These miRNAs could be a promising innovative treatment approach to enhance Temozolomide (TMZ) sensitivity in MGMT unmethylated patients and to increase progression-free survival as well as long-term survival. In this review, the relevant miRNAs are systematically reviewed.
Collapse
Affiliation(s)
- Anna Kirstein
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Thomas E. Schmid
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Stephanie E. Combs
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-4501
| |
Collapse
|
503
|
Adhikary RR, Koppaka O, Banerjee R. Development of color changing polydiacetylene-based biomimetic nanovesicle platforms for quick detection of membrane permeability across the blood brain barrier. NANOSCALE 2020; 12:8898-8908. [PMID: 32266882 DOI: 10.1039/c9nr07845b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Membrane permeability through passive diffusion is one of the important pathways for passage of drugs across the blood brain barrier (BBB). The present study describes the development of biomimetic unilamellar lipopolymeric nanovesicles of size 268 ± 37 nm, consisting of polar brain lipids in conjunction with polydiacetylene and validation of their application for an abbreviated yet accurate membrane permeability assay with high-throughput and rapid identification of BBB permeability of drugs. The nanovesicle suspension was tested with drugs of known permeability across the BBB to validate the detection of changes in hue, absorbance and fluorescence in response to permeation across the nanovesicles. A simple device was developed based on the nanovesicle sensors along with a mobile application which allowed for the determination of hue corresponding to qualitative identification of whether a drug is BBB permeable (BBB+) or not (BBB-). With respect to determination of a suitable endpoint in this assay, a hue cut off of 275°, reduction in %blueness by less than 59% and a fluorescence intensity of ≥0.22 a.u. at 560 nm accurately differentiated between drugs which are permeable and impermeable across the BBB within 5 minutes. Further quantification of BBB permeability can be done through the concentration at which the above end-points are achieved. For the quantification of the permeability, absorbance and fluorescence measurements were performed. The device thus developed allows the rapid determination of BBB permeability of various agents in drug discovery especially in smaller set-ups with minimal equipment through changes in color, absorbance and fluorescence.
Collapse
Affiliation(s)
- Rishi Rajat Adhikary
- Nanomedicine Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India.
| | | | | |
Collapse
|
504
|
González-Nieto D, Fernández-Serra R, Pérez-Rigueiro J, Panetsos F, Martinez-Murillo R, Guinea GV. Biomaterials to Neuroprotect the Stroke Brain: A Large Opportunity for Narrow Time Windows. Cells 2020; 9:E1074. [PMID: 32357544 PMCID: PMC7291200 DOI: 10.3390/cells9051074] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke represents one of the most prevalent pathologies in humans and is a leading cause of death and disability. Anti-thrombolytic therapy with tissue plasminogen activator (t-PA) and surgical thrombectomy are the primary treatments to recanalize occluded vessels and normalize the blood flow in ischemic and peri-ischemic regions. A large majority of stroke patients are refractory to treatment or are not eligible due to the narrow time window of therapeutic efficacy. In recent decades, we have significantly increased our knowledge of the molecular and cellular mechanisms that inexorably lead to progressive damage in infarcted and peri-lesional brain areas. As a result, promising neuroprotective targets have been identified and exploited in several stroke models. However, these considerable advances have been unsuccessful in clinical contexts. This lack of clinical translatability and the emerging use of biomaterials in different biomedical disciplines have contributed to developing a new class of biomaterial-based systems for the better control of drug delivery in cerebral disorders. These systems are based on specific polymer formulations structured in nanoparticles and hydrogels that can be administered through different routes and, in general, bring the concentrations of drugs to therapeutic levels for prolonged times. In this review, we first provide the general context of the molecular and cellular mechanisms impaired by cerebral ischemia, highlighting the role of excitotoxicity, inflammation, oxidative stress, and depolarization waves as the main pathways and targets to promote neuroprotection avoiding neuronal dysfunction. In the second part, we discuss the versatile role played by distinct biomaterials and formats to support the sustained administration of particular compounds to neuroprotect the cerebral tissue at risk of damage.
Collapse
Affiliation(s)
- Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Rocío Fernández-Serra
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- Brain Plasticity Group, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | | | - Gustavo V. Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
505
|
Kumar A, Naik PK, Pradhan D, Ghosh G, Rath G. Mucoadhesive formulations: innovations, merits, drawbacks, and future outlook. Pharm Dev Technol 2020; 25:797-814. [DOI: 10.1080/10837450.2020.1753771] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Amresh Kumar
- Department of Pharmaceutics, I.S.F. College of Pharmacy, Moga, Punjab, India
| | | | - Deepak Pradhan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Goutam Ghosh
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| |
Collapse
|
506
|
Lu X, Dong J, Zheng D, Li X, Ding D, Xu H. Reperfusion combined with intraarterial administration of resveratrol-loaded nanoparticles improved cerebral ischemia-reperfusion injury in rats. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102208. [PMID: 32334100 DOI: 10.1016/j.nano.2020.102208] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/13/2020] [Accepted: 03/28/2020] [Indexed: 01/28/2023]
Abstract
Endovascular thrombectomy (EVT) has been recommended as the first line therapy for large artery occlusion (LAO) stroke. However, abrupt recovery of blood flow induces oxidative stress which breaks down the blood-brain barrier (BBB), activates apoptosis and inhibits neurogenesis. Supplement of exogenous antioxidants to relieve the injuries related to oxidative stress is a rational treatment combined to EVT for acute LAO therapy. Resveratrol (RES), an antioxidant, was encapsulated into polymeric nanoparticles (RES-NPs). In transient middle cerebral artery occlusion (tMCAO) rats, intraarterial administration of RES-NPs demonstrated significant protection against cerebral ischemia/reperfusion (I/R) injuries. RES-NPs attenuated the oxidative stress induced by I/R, prevented brain edema, protected neurons from undergoing apoptosis, and contributed to neurogenesis through enhanced expression of brain-derived neurotrophic factor (BDNF). These results suggested that intra-arterial infusion of RES-NPs in conjunction with EVT could be a potential strategy for the LAO stroke therapy.
Collapse
Affiliation(s)
- Xiaowei Lu
- Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Jingde Dong
- Department of Geriatric Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Donghui Zheng
- Department of Nephrology, the Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an City, Huai'an, PR China
| | - Xiaolin Li
- Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, China
| | - Huae Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
507
|
Gil-Martins E, Barbosa DJ, Silva V, Remião F, Silva R. Dysfunction of ABC transporters at the blood-brain barrier: Role in neurological disorders. Pharmacol Ther 2020; 213:107554. [PMID: 32320731 DOI: 10.1016/j.pharmthera.2020.107554] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
ABC (ATP-binding cassette) transporters represent one of the largest and most diverse superfamily of proteins in living species, playing an important role in many biological processes such as cell homeostasis, cell signaling, drug metabolism and nutrient uptake. Moreover, using the energy generated from ATP hydrolysis, they mediate the efflux of endogenous and exogenous substrates from inside the cells, thereby reducing their intracellular accumulation. At present, 48 ABC transporters have been identified in humans, which were classified into 7 different subfamilies (A to G) according to their phylogenetic analysis. Nevertheless, the most studied members with importance in drug therapeutic efficacy and toxicity include P-glycoprotein (P-gp), a member of the ABCB subfamily, the multidrug-associated proteins (MPRs), members of the ABCC subfamily, and breast cancer resistance protein (BCRP), a member of the ABCG subfamily. They exhibit ubiquitous expression throughout the human body, with a special relevance in barrier tissues like the blood-brain barrier (BBB). At this level, they play a physiological function in tissue protection by reducing or limiting the brain accumulation of neurotoxins. Furthermore, dysfunction of ABC transporters, at expression and/or activity level, has been associated with many neurological diseases, including epilepsy, multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis. Additionally, these transporters are strikingly associated with the pharmacoresistance to central nervous system (CNS) acting drugs, because they contribute to the decrease in drug bioavailability. This article reviews the signaling pathways that regulate the expression and activity of P-gp, BCRP and MRPs subfamilies of transporters, with particular attention at the BBB level, and their mis-regulation in neurological disorders.
Collapse
Affiliation(s)
- Eva Gil-Martins
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.
| | - Vera Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
508
|
Gorter RP, Baron W. Matrix metalloproteinases shape the oligodendrocyte (niche) during development and upon demyelination. Neurosci Lett 2020; 729:134980. [PMID: 32315713 DOI: 10.1016/j.neulet.2020.134980] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 04/04/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
The oligodendrocyte lineage cell is crucial to proper brain function. During central nervous system development, oligodendrocyte progenitor cells (OPCs) migrate and proliferate to populate the entire brain and spinal cord, and subsequently differentiate into mature oligodendrocytes that wrap neuronal axons in an insulating myelin layer. When damage occurs to the myelin sheath, OPCs are activated and recruited to the demyelinated site, where they differentiate into oligodendrocytes that remyelinate the denuded axons. The process of OPC attraction and differentiation is influenced by a multitude of factors from the cell's niche. Matrix metalloproteinases (MMPs) are powerful and versatile enzymes that do not only degrade extracellular matrix proteins, but also cleave cell surface receptors, growth factors, signaling molecules, proteases and other precursor proteins, leading to their activation or degradation. MMPs are markedly upregulated during brain development and upon demyelinating injury, where their broad functions influence the behavior of neural progenitor cells (NPCs), OPCs and oligodendrocytes. In this review, we focus on the role of MMPs in (re)myelination. We will start out in the developing brain with describing the effects of MMPs on NPCs, OPCs and eventually oligodendrocytes. Then, we will outline their functions in oligodendrocyte process extension and developmental myelination. Finally, we will review their potential role in demyelination, describe their significance in remyelination and discuss the evidence for a role of MMPs in remyelination failure, focusing on multiple sclerosis. In conclusion, MMPs shape the oligodendrocyte (niche) both during development and upon demyelination, and thus are important players in directing the fate and behavior of oligodendrocyte lineage cells throughout their life cycle.
Collapse
Affiliation(s)
- Rianne P Gorter
- University of Groningen, University Medical Center Groningen, Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Wia Baron
- University of Groningen, University Medical Center Groningen, Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands.
| |
Collapse
|
509
|
Wu H, Lu H, Xiao W, Yang J, Du H, Shen Y, Qu H, Jia B, Manna SK, Ramachandran M, Xue X, Ma Z, Xu X, Wang Z, He Y, Lam KS, Zawadzki RJ, Li Y, Lin TY. Sequential Targeting in Crosslinking Nanotheranostics for Tackling the Multibarriers of Brain Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903759. [PMID: 32078198 PMCID: PMC7148201 DOI: 10.1002/adma.201903759] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 01/06/2020] [Indexed: 05/20/2023]
Abstract
The efficacy of therapeutics for brain tumors is seriously hampered by multiple barriers to drug delivery, including severe destabilizing effects in the blood circulation, the blood-brain barrier/blood-brain tumor barrier (BBB/BBTB), and limited tumor uptake. Here, a sequential targeting in crosslinking (STICK) nanodelivery strategy is presented to circumvent these important physiological barriers to improve drug delivery to brain tumors. STICK nanoparticles (STICK-NPs) can sequentially target BBB/BBTB and brain tumor cells with surface maltobionic acid (MA) and 4-carboxyphenylboronic acid (CBA), respectively, and simultaneously enhance nanoparticle stability with pH-responsive crosslinkages formed by MA and CBA in situ. STICK-NPs exhibit prolonged circulation time (17-fold higher area under curve) than the free agent, allowing increased opportunities to transpass the BBB/BBTB via glucose-transporter-mediated transcytosis by MA. The tumor acidic environment then triggers the transformation of the STICK-NPs into smaller nanoparticles and reveals a secondary CBA targeting moiety for deep tumor penetration and enhanced uptake in tumor cells. STICK-NPs significantly inhibit tumor growth and prolong the survival time with limited toxicity in mice with aggressive and chemoresistant diffuse intrinsic pontine glioma. This formulation tackles multiple physiological barriers on-demand with a simple and smart STICK design. Therefore, these features allow STICK-NPs to unleash the potential of brain tumor therapeutics to improve their treatment efficacy.
Collapse
Affiliation(s)
- Hao Wu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Hongwei Lu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Jinfan Yang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Hongxu Du
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Yingbin Shen
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Haijing Qu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Bei Jia
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Suman K Manna
- UC Davis RISE Eye-Pod Laboratory, Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, 95616, USA
| | - Mythili Ramachandran
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Xiangdong Xue
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Zhao Ma
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Xiaobao Xu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Zhongling Wang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Yixuan He
- Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Robert J Zawadzki
- UC Davis RISE Eye-Pod Laboratory, Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, 95616, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Tzu-Yin Lin
- Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
| |
Collapse
|
510
|
Chung YH, Lin CW, Huang HY, Chen SL, Huang HJ, Sun YC, Lee GC, Lee-Chen GJ, Chang YC, Hsieh-Li HM. Targeting Inflammation, PHA-767491 Shows a Broad Spectrum in Protein Aggregation Diseases. J Mol Neurosci 2020; 70:1140-1152. [PMID: 32170713 DOI: 10.1007/s12031-020-01521-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
Many protein aggregation diseases (PAD) affect the nervous system. Deposits of aggregated disease-specific proteins are found within or around the neuronal cells of neurodegenerative diseases. Although the main protein component is disease-specific, oligomeric aggregates are presumed to be the key agents causing the neurotoxicity. Evidence has shown that protein aggregates cause a chronic inflammatory reaction in the brain, resulting in neurodegeneration. Therefore, strategies targeting anti-inflammation could be beneficial to the therapeutics of PAD. PHA-767491 was originally identified as an inhibitor of CDC7/CDK9 and was found to reduce TDP-43 phosphorylation and prevent neurodegeneration in TDP-43 transgenic animals. We recently identified PHA-767491 as a GSK-3β inhibitor. In this study, we established mouse hippocampal primary culture with tau-hyperphosphorylation through the activation of GSK-3β using Wortmannin and GF109203X. We found that PHA-767491 significantly improved the neurite outgrowth of hippocampal primary neurons against the neurotoxicity induced by GSK-3β. We further showed that PHA-767491 had neuroprotective ability in hippocampal primary culture under oligomeric Aβ treatment. In addition, PHA-767491 attenuated the neuroinflammation in mouse cerebellar slice culture with human TBP-109Q agitation. Further study of SCA17 transgenic mice carrying human TBP-109Q showed that PHA-767491 ameliorated the gait ataxia and the inflammatory response both centrally and peripherally. Our findings suggest that PHA-767491 has a broad spectrum of activity in the treatment of different PAD and that this activity could be based on the anti-inflammation mechanism.
Collapse
Affiliation(s)
- Yu-Han Chung
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chia-Wei Lin
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsin-Yu Huang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Shu-Ling Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hei-Jen Huang
- Department of Nursing, Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Ying-Chieh Sun
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Guan-Chiun Lee
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ya-Ching Chang
- Department of Pharmacy, Taiwan Adventist Hospital, Taipei, Taiwan.
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan.
| |
Collapse
|
511
|
Gupta S, Kesarla R, Omri A. Approaches for CNS delivery of drugs - nose to brain targeting of antiretroviral agents as a potential attempt for complete elimination of major reservoir site of HIV to aid AIDS treatment. Expert Opin Drug Deliv 2020; 16:287-300. [PMID: 30779602 DOI: 10.1080/17425247.2019.1583206] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Human immune-deficiency virus (HIV) infection causing acquired immune-deficiency syndrome (AIDS) is one of the most life-threatening infections. The central nervous system (CNS) is reported to be the most important HIV reservoir site where the antiretroviral drugs are unable to reach. AREAS COVERED This article includes the review about HIV infections, its pathogenesis, HIV infections in CNS, its consequences, current therapies, challenges associated with the existing therapies, approaches to overcome them, CNS delivery of drugs - barriers, transport routes, approaches for transporting drugs across the blood-brain barrier, nasal route of drug delivery, and nose to brain targeting of antiretroviral agents as a potential approach for complete cure of AIDS. EXPERT OPINION Various approaches are exploited to enhance the drug delivery to the brain for various categories of drugs. However, very few have investigated on the delivery of antiretrovirals to the brain. Targeting antiretrovirals to CNS through oral/nasal routes along with oral/parenteral delivery of drug to the plasma can be a promising approach for an attempt to completely eradicate HIV reservoir and cure AIDS, after clinical trials. Further research is required to identify the exact location of the HIV reservoir in CNS and developing good animal models for evaluation of different newly developed formulations.
Collapse
Affiliation(s)
- Shweta Gupta
- a Department of Pharmaceutics, Ideal College of Pharmacy and Research , University of Mumbai , Mumbai, Maharashtra , India
| | - Rajesh Kesarla
- b Corporate Quality Assurance , Zydus Cadila , Ahmedabad , Gujarat , India
| | - Abdelwahab Omri
- c The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry , Laurentian University , Sudbury , ON , Canada
| |
Collapse
|
512
|
Wanat K. Biological barriers, and the influence of protein binding on the passage of drugs across them. Mol Biol Rep 2020; 47:3221-3231. [PMID: 32140957 DOI: 10.1007/s11033-020-05361-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/27/2020] [Indexed: 01/11/2023]
Abstract
Drug-protein binding plays a key role in determining the pharmacokinetics of a drug. The distribution and protein binding ability of a drug changes over a lifetime, and are important considerations during pregnancy and lactation. Although proteins are a significant fraction in plasma composition, they also exist beyond the bloodstream and bind with drugs in the skin, tissues or organs. Protein binding influences the bioavailability and distribution of active compounds, and is a limiting factor in the passage of drugs across biological membranes and barriers: drugs are often unable to cross membranes mainly due to the high molecular mass of the drug-protein complex, thus resulting in the accumulation of the active compounds and a significant reduction of their pharmacological activity. This review describes the consequences of drug-protein binding on drug transport across physiological barriers, whose role is to allow the passage of essential substances-such as nutrients or oxygen, but not of xenobiotics. The placental barrier regulates passage of xenobiotics into a fetus and protects the unborn organism. The blood-brain barrier is the most important barrier in the entire organism and the skin separates the human body from the environment.
Collapse
Affiliation(s)
- Karolina Wanat
- Department of Analytical Chemistry, Medical University of Lodz, Muszyńskiego 1, 90-151, Lodz, Poland.
| |
Collapse
|
513
|
Abstract
Drug formulations and suitable methods for their detection play a very crucial role in the development of therapeutics towards degenerative neurological diseases. For diseases such as Alzheimer’s disease, magnetic resonance imaging (MRI) is a non-invasive clinical technique suitable for early diagnosis. In this review, we will discuss the different experimental conditions which can push MRI as the technique of choice and the gold standard for early diagnosis of Alzheimer’s disease. Here, we describe and compare various techniques for administration of nanoparticles targeted to the brain and suitable formulations of nanoparticles for use as magnetically active therapeutic probes in drug delivery targeting the brain. We explore different physiological pathways involved in the transport of such nanoparticles for successful entry in the brain. In our lab, we have used different formulations of iron oxide nanoparticles (IONPs) and protein nanocages as contrast agents in anatomical MRI of an Alzheimer’s disease (AD) brain. We compare these coatings and their benefits to provide the best contrast in addition to biocompatibility properties to be used as sustainable drug-release systems. In the later sections, the contrast enhancement techniques in MRI studies are discussed. Examples of contrast-enhanced imaging using advanced pulse sequences are discussed with the main focus on important studies in the field of neurological diseases. In addition, T1 contrast agents such as gadolinium chelates are compared with the T2 contrast agents mainly made of superparamagnetic inorganic metal nanoparticles.
Collapse
|
514
|
Yamaguchi S, Ito S, Masuda T, Couraud PO, Ohtsuki S. Novel cyclic peptides facilitating transcellular blood-brain barrier transport of macromolecules in vitro and in vivo. J Control Release 2020; 321:744-755. [PMID: 32135226 DOI: 10.1016/j.jconrel.2020.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/26/2020] [Accepted: 03/02/2020] [Indexed: 12/15/2022]
Abstract
Brain delivery of nanoparticles and macromolecular drugs depends on blood-brain barrier (BBB)-permeable carriers. In this study, we searched for cyclic heptapeptides facilitating BBB permeation of M13 phages by phage library screening using a transcellular permeability assay with hCMEC/D3 cell monolayers, a human BBB model. The M13 phage, which is larger than macromolecular drugs and nanoparticles, served as a model macromolecule. The screen identified cyclic heptapeptide SLSHSPQ (SLS) as a human BBB-permeable peptide. The SLS-displaying phage (SLS-phage) exhibited improved permeation across the cell monolayer of monkey and rat BBB co-culture models. The SLS-phage internalized into hCMEC/D3 cells via macropinocytosis and externalized via the exosome excretion pathway. SLS-phage distribution into brain parenchyma was observed in mice after intravenous administration. Moreover, liposome permeated across the BBB as cyclic SLS peptide conjugates. In conclusion, the cyclic SLS heptapeptide is a novel carrier candidate for brain delivery of macromolecular drugs and nanoparticles.
Collapse
Affiliation(s)
- Shunsuke Yamaguchi
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Japan Society for the Promotion of Science, Research Fellowship for Young Scientists, Chiyoda-ku, Tokyo, Japan
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| |
Collapse
|
515
|
Nigam K, Kaur A, Tyagi A, Nematullah M, Khan F, Gabrani R, Dang S. Nose-to-brain delivery of lamotrigine-loaded PLGA nanoparticles. Drug Deliv Transl Res 2020; 9:879-890. [PMID: 30887226 DOI: 10.1007/s13346-019-00622-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Direct nose-to-brain delivery of drugs and faster onset of action have made intra-nasal route a much sought-after alternative to conventional routes of drug delivery to the brain. Lamotrigine is used for the treatment and management of neuropathic pain, and in the present work, lamotrigine (LTG)-PLGA nanoparticles were developed for intra-nasal delivery. The LTG-PLGA nanoparticles were prepared using modified nanoprecipitation method via high-speed homogenization and ultra-sonication techniques. Entrapment efficiency (EE%) of developed LTG-PLGA-NPs was found to be 84.87 ± 1.2% with drug loading of 10.21 ± 0.89%. The particle size of developed nanoparticles was found to be 184.6 nm with PDI value of 0.082 and zeta potential of - 18.8 mV. Dissolution profiles were studied in PBS (pH 7.4), simulated nasal fluid, and simulated cerebrospinal fluid where almost complete release was observed within 5 h in CSF. In vitro, cytotoxicity was analyzed using MTT assay where dose-dependent cytotoxicity was observed for developed LTG-PLGA-NPs. In vitro cytokine analysis showed positive effects of LTG-PLGA-NPs as pro-inflammatory cytokine suppressors. Further, in vivo studies were performed for radiolabeled formulation and drug (99mTc-LTG-PLGA-NPs and 99mTc-LTG-aqueous) using Sprague Dawley rats where with the help of gamma scintigraphy studies, various routes of administration viz. oral, intra-nasal, and intra-venous were compared. Various pharmacokinetic parameters were evaluated using biodistribution studies to estimate the drug levels in blood and brain. For 99mTc-LTG-PLGA-NPs via intra-nasal route, drug targeting efficiency (DTE%) was found to be 129.81% and drug target organ transport (DTP%) to be 22.81% in brain with Cmax of 3.82%/g within Tmax 1.5 h. Thus, the developed PLGA nanoparticles for intra-nasal delivery provide a possible alternative for existing available drug formulation for neuropathic pain management.
Collapse
Affiliation(s)
- Kuldeep Nigam
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, 201309, India
| | - Atinderpal Kaur
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, 201309, India
| | - Amit Tyagi
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Timarpur, Delhi, 110054, India
| | - Md Nematullah
- Department of Biochemistry, Faculty of Science, Jamia Hamdard, New Delhi, 110062, India
| | - Farah Khan
- Department of Biochemistry, Faculty of Science, Jamia Hamdard, New Delhi, 110062, India
| | - Reema Gabrani
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, 201309, India
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, 201309, India.
| |
Collapse
|
516
|
Spencer AP, Torrado M, Custódio B, Silva-Reis SC, Santos SD, Leiro V, Pêgo AP. Breaking Barriers: Bioinspired Strategies for Targeted Neuronal Delivery to the Central Nervous System. Pharmaceutics 2020; 12:E192. [PMID: 32102252 PMCID: PMC7076453 DOI: 10.3390/pharmaceutics12020192] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/01/2020] [Accepted: 02/19/2020] [Indexed: 12/23/2022] Open
Abstract
Central nervous system (CNS) disorders encompass a vast spectrum of pathological conditions and represent a growing concern worldwide. Despite the high social and clinical interest in trying to solve these pathologies, there are many challenges to bridge in order to achieve an effective therapy. One of the main obstacles to advancements in this field that has hampered many of the therapeutic strategies proposed to date is the presence of the CNS barriers that restrict the access to the brain. However, adequate brain biodistribution and neuronal cells specific accumulation in the targeted site also represent major hurdles to the attainment of a successful CNS treatment. Over the last few years, nanotechnology has taken a step forward towards the development of therapeutics in neurologic diseases and different approaches have been developed to surpass these obstacles. The versatility of the designed nanocarriers in terms of physical and chemical properties, and the possibility to functionalize them with specific moieties, have resulted in improved neurotargeted delivery profiles. With the concomitant progress in biology research, many of these strategies have been inspired by nature and have taken advantage of physiological processes to achieve brain delivery. Here, the different nanosystems and targeting moieties used to achieve a neuronal delivery reported in the open literature are comprehensively reviewed and critically discussed, with emphasis on the most recent bioinspired advances in the field. Finally, we express our view on the paramount challenges in targeted neuronal delivery that need to be overcome for these promising therapeutics to move from the bench to the bedside.
Collapse
Affiliation(s)
- Ana P. Spencer
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
| | - Marília Torrado
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Beatriz Custódio
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Sara C. Silva-Reis
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Sofia D. Santos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Victoria Leiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Ana P. Pêgo
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
517
|
Koo JH, Kim DH, Cha D, Kang MJ, Choi JM. LRR domain of NLRX1 protein delivery by dNP2 inhibits T cell functions and alleviates autoimmune encephalomyelitis. Theranostics 2020; 10:3138-3150. [PMID: 32194859 PMCID: PMC7053182 DOI: 10.7150/thno.43441] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/16/2020] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating inflammatory disease of the central nervous system (CNS), which is a chronic progressive disease and is caused by uncontrolled activation of myelin antigen specific T cells. It has high unmet medical needs due to the difficulty of efficient drug delivery into the CNS to control tissue inflammation. In this study, we demonstrate that a fusion protein of NOD-like receptor family member X1 (NLRX1) and blood brain barrier (BBB)-permeable peptide, dNP2 ameliorates experimental autoimmune encephalomyelitis (EAE). Methods: We purified recombinant LRR or NBD regions of NLRX1 protein conjugated with dNP2. To examine intracellular delivery efficiency of the recombinant protein, we incubated the proteins with Jurkat T cells or murine splenic T cells and their delivery efficiency was analyzed by flow cytometry. To investigate the therapeutic efficacy in an EAE model, we injected the recombinant protein into mice with 3 different treatment schemes e.g., prevention, semi-therapeutic, and therapeutic. To analyze their functional roles in T cells, we treated MACS-sorted naïve CD4 T cells with the proteins during their activation and differentiation into Th1, Th17, and Treg cells. Results: dNP2-LRR protein treatment showed significantly higher delivery efficiency than TAT-LRR or LRR alone in Jurkat T cells and mouse splenic T cells. In all three treatment schemes of EAE experiments, dNP2-LRR administration showed ameliorated tissue inflammation and disease severity with reduced number of infiltrating T cells producing inflammatory cytokines such as IFNγ. In addition, dNP2-LRR inhibited T cell activation, cytokine production, and Th1 differentiation. Conclusion: These results suggest that dNP2-LRR is a novel agent, which regulates effector T cell functions and could be a promising molecule for the treatment of CNS autoimmune diseases such as multiple sclerosis.
Collapse
|
518
|
Martinelli C, Pucci C, Battaglini M, Marino A, Ciofani G. Antioxidants and Nanotechnology: Promises and Limits of Potentially Disruptive Approaches in the Treatment of Central Nervous System Diseases. Adv Healthc Mater 2020; 9:e1901589. [PMID: 31854132 DOI: 10.1002/adhm.201901589] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/26/2019] [Indexed: 12/11/2022]
Abstract
Many central nervous system (CNS) diseases are still incurable and only symptomatic treatments are available. Oxidative stress is suggested to be a common hallmark, being able to cause and exacerbate the neuronal cell dysfunctions at the basis of these pathologies, such as mitochondrial impairments, accumulation of misfolded proteins, cell membrane damages, and apoptosis induction. Several antioxidant compounds are tested as potential countermeasures for CNS disorders, but their efficacy is often hindered by the loss of antioxidant properties due to enzymatic degradation, low bioavailability, poor water solubility, and insufficient blood-brain barrier crossing efficiency. To overcome the limitations of antioxidant molecules, exploitation of nanostructures, either for their delivery or with inherent antioxidant properties, is proposed. In this review, after a brief discussion concerning the role of the blood-brain barrier in the CNS and the involvement of oxidative stress in some neurodegenerative diseases, the most interesting research concerning the use of nano-antioxidants is introduced and discussed, focusing on the synthesis procedures, functionalization strategies, in vitro and in vivo tests, and on recent clinical trials.
Collapse
Affiliation(s)
- Chiara Martinelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Carlotta Pucci
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Matteo Battaglini
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
- Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| |
Collapse
|
519
|
Li S, Wang Y, Jiang D, Ni D, Kutyreff CJ, Barnhart TE, Engle JW, Cai W. Spatiotemporal Distribution of Agrin after Intrathecal Injection and Its Protective Role in Cerebral Ischemia/Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902600. [PMID: 32076591 PMCID: PMC7029627 DOI: 10.1002/advs.201902600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/18/2019] [Indexed: 05/30/2023]
Abstract
Intrathecal injection, drugs transporting along perivascular spaces, represents an important route for maintaining blood-brain barrier (BBB) integrity after cerebral ischemia/reperfusion (I/R) injury. However, after being directly injected into cerebrospinal fluid (CSF), the temporal and spatial changes in the distribution of therapeutic protein drugs have remained unknown. Here, with positron emission tomography (PET) imaging, the uptake of 89Zr-agrin is noninvasively and dynamically monitored. These data demonstrate the time-activity curve of drugs in the brain subregions and their spatial distribution in different organs after intrathecal administration. Furthermore, agrin treatment effectively inhibits BBB disruption by reducing the loss of tight-junctional proteins. Importantly, the infarct volume is reduced; the number of apoptotic neurons is decreased; and neurological function is improved in mouse I/R injury models. Thus, intrathecal injection of agrin provides the basis for a new strategy to research and develop protein drugs for reducing the aggravation of I/R injury.
Collapse
Affiliation(s)
- Shiyong Li
- Department of RehabilitationSecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Ye Wang
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
| | - Dawei Jiang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Dalong Ni
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Christopher J. Kutyreff
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Todd E. Barnhart
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Jonathan W. Engle
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Weibo Cai
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| |
Collapse
|
520
|
Teixeira MI, Lopes CM, Amaral MH, Costa PC. Current insights on lipid nanocarrier-assisted drug delivery in the treatment of neurodegenerative diseases. Eur J Pharm Biopharm 2020; 149:192-217. [PMID: 31982574 DOI: 10.1016/j.ejpb.2020.01.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/16/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022]
Abstract
The central nervous system (CNS) is vulnerable to pathologic processes that lead to the development of neurodegenerative disorders like Alzheimer's, Parkinson's and Huntington's diseases, Multiple sclerosis or Amyotrophic lateral sclerosis. These are chronic and progressive pathologies characterized by the loss of neurons and the formation of misfolded proteins. Additionally, neurodegenerative diseases are accompanied by a structural and functional dysfunction of the blood-brain barrier (BBB). Although serving as a protection for the CNS, the existence of physiological barriers, especially the BBB, limits the access of several therapeutic agents to the brain, constituting a major hindrance in neurotherapeutics advancement. In this regard, nanotechnology-based approaches have arisen as a promising strategy to not only improve drug targeting to the brain, but also to increase bioavailability. Lipid nanocarriers such as liposomes, solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), microemulsions and nanoemulsions, have already proven their potential for enhancing brain transport, crossing more easily into the CNS and allowing the administration of medicines that could benefit the treatment of neurological pathologies. Given the socioeconomic impact of such conditions and the advent of nanotechnology that inevitably leads to more effective and superior therapeutics for their management, it is imperative to constantly update on the current knowledge of these topics. Herein, we provide insight on the BBB and the pathophysiology of the main neurodegenerative disorders. Moreover, this review seeks to highlight the several approaches that can be used to improve the delivery of therapeutic agents to the CNS, while also offering an extensive overview of the latest efforts regarding the use of lipid-based nanocarriers in the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- M I Teixeira
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - C M Lopes
- FP-ENAS/CEBIMED, Fernando Pessoa Energy, Environment and Health Research Unit/Biomedical Research Centre, Faculty of Health Sciences, Fernando Pessoa University, Rua Carlos da Maia, 296, 4200-150 Porto, Portugal
| | - M H Amaral
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - P C Costa
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
521
|
Pantshwa JM, Kondiah PPD, Choonara YE, Marimuthu T, Pillay V. Nanodrug Delivery Systems for the Treatment of Ovarian Cancer. Cancers (Basel) 2020; 12:E213. [PMID: 31952210 PMCID: PMC7017423 DOI: 10.3390/cancers12010213] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Despite advances achieved in medicine, chemotherapeutics still has detrimental side effects with ovarian cancer (OC), accounting for numerous deaths among females. The provision of safe, early detection and active treatment of OC remains a challenge, in spite of improvements in new antineoplastic discovery. Nanosystems have shown remarkable progress with impact in diagnosis and chemotherapy of various cancers, due to their ideal size; improved drug encapsulation within its interior core; potential to minimize drug degradation; improve in vivo drug release kinetics; and prolong blood circulation times. However, nanodrug delivery systems have few limitations regarding its accuracy of tumour targeting and the ability to provide sustained drug release. Hence, a cogent and strategic approach has focused on nanosystem functionalization with antibody-based ligands to selectively enhance cellular uptake of antineoplastics. Antibody functionalized nanosystems are (advanced) synthetic candidates, with a broad range of efficiency in specific tumour targeting, whilst leaving normal cells unaffected. This article comprehensively reviews the present status of nanosystems, with particular emphasis on nanomicelles for molecular diagnosis and treatment of OC. In addition, biomarkers of nanosystems provide important prospects as chemotherapeutic strategies to upsurge the survival rate of patients with OC.
Collapse
Affiliation(s)
| | | | | | | | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa; (J.M.P.); (P.P.D.K.); (Y.E.C.); (T.M.)
| |
Collapse
|
522
|
Cavaco M, Gaspar D, ARB Castanho M, Neves V. Antibodies for the Treatment of Brain Metastases, a Dream or a Reality? Pharmaceutics 2020; 12:E62. [PMID: 31940974 PMCID: PMC7023012 DOI: 10.3390/pharmaceutics12010062] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/13/2019] [Accepted: 12/28/2019] [Indexed: 12/25/2022] Open
Abstract
The incidence of brain metastases (BM) in cancer patients is increasing. After diagnosis, overall survival (OS) is poor, elicited by the lack of an effective treatment. Monoclonal antibody (mAb)-based therapy has achieved remarkable success in treating both hematologic and non-central-nervous system (CNS) tumors due to their inherent targeting specificity. However, the use of mAbs in the treatment of CNS tumors is restricted by the blood-brain barrier (BBB) that hinders the delivery of either small-molecules drugs (sMDs) or therapeutic proteins (TPs). To overcome this limitation, active research is focused on the development of strategies to deliver TPs and increase their concentration in the brain. Yet, their molecular weight and hydrophilic nature turn this task into a challenge. The use of BBB peptide shuttles is an elegant strategy. They explore either receptor-mediated transcytosis (RMT) or adsorptive-mediated transcytosis (AMT) to cross the BBB. The latter is preferable since it avoids enzymatic degradation, receptor saturation, and competition with natural receptor substrates, which reduces adverse events. Therefore, the combination of mAbs properties (e.g., selectivity and long half-life) with BBB peptide shuttles (e.g., BBB translocation and delivery into the brain) turns the therapeutic conjugate in a valid approach to safely overcome the BBB and efficiently eliminate metastatic brain cells.
Collapse
Affiliation(s)
| | | | - Miguel ARB Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (M.C.); (D.G.)
| | - Vera Neves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (M.C.); (D.G.)
| |
Collapse
|
523
|
May JN, Golombek SK, Baues M, Dasgupta A, Drude N, Rix A, Rommel D, von Stillfried S, Appold L, Pola R, Pechar M, van Bloois L, Storm G, Kuehne AJC, Gremse F, Theek B, Kiessling F, Lammers T. Multimodal and multiscale optical imaging of nanomedicine delivery across the blood-brain barrier upon sonopermeation. Am J Cancer Res 2020; 10:1948-1959. [PMID: 32042346 PMCID: PMC6993230 DOI: 10.7150/thno.41161] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/20/2019] [Indexed: 01/15/2023] Open
Abstract
Rationale: The blood-brain barrier (BBB) is a major obstacle for drug delivery to the brain. Sonopermeation, which relies on the combination of ultrasound and microbubbles, has emerged as a powerful tool to permeate the BBB, enabling the extravasation of drugs and drug delivery systems (DDS) to and into the central nervous system (CNS). When aiming to improve the treatment of high medical need brain disorders, it is important to systematically study nanomedicine translocation across the sonopermeated BBB. To this end, we here employed multimodal and multiscale optical imaging to investigate the impact of DDS size on brain accumulation, extravasation and penetration upon sonopermeation. Methods: Two prototypic DDS, i.e. 10 nm-sized pHPMA polymers and 100 nm-sized PEGylated liposomes, were labeled with fluorophores and intravenously injected in healthy CD-1 nude mice. Upon sonopermeation, computed tomography-fluorescence molecular tomography, fluorescence reflectance imaging, fluorescence microscopy, confocal microscopy and stimulated emission depletion nanoscopy were used to study the effect of DDS size on their translocation across the BBB. Results: Sonopermeation treatment enabled safe and efficient opening of the BBB, which was confirmed by staining extravasated endogenous IgG. No micro-hemorrhages, edema and necrosis were detected in H&E stainings. Multimodal and multiscale optical imaging showed that sonopermeation promoted the accumulation of nanocarriers in mouse brains, and that 10 nm-sized polymeric DDS accumulated more strongly and penetrated deeper into the brain than 100 nm-sized liposomes. Conclusions: BBB opening via sonopermeation enables safe and efficient delivery of nanomedicine formulations to and into the brain. When looking at accumulation and penetration (and when neglecting issues such as drug loading capacity and therapeutic efficacy) smaller-sized DDS are found to be more suitable for drug delivery across the BBB than larger-sized DDS. These findings are valuable for better understanding and further developing nanomedicine-based strategies for the treatment of CNS disorders.
Collapse
|
524
|
Liang W, Xu W, Zhu J, Zhu Y, Gu Q, Li Y, Guo C, Huang Y, Yu J, Wang W, Hu Y, Zhao Y, Han B, Bei W, Guo J. Ginkgo biloba extract improves brain uptake of ginsenosides by increasing blood-brain barrier permeability via activating A1 adenosine receptor signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 246:112243. [PMID: 31541722 DOI: 10.1016/j.jep.2019.112243] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/04/2019] [Accepted: 09/15/2019] [Indexed: 12/24/2022]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginkgo biloba leaves and Panax ginseng are Chinese medicine commonly used in combination for cerebral disease. AIM OF THE STUDY To investigate the effect of standard extract of Ginkgo biloba leaves (EGb) on facilitating brain uptake of ginsenoside and its underlying mechanisms. MATERIALS AND METHODS The increasing uptake of ginsenosides in the brain of rats by EGb were detected by LC-MS/MS analysis. Evans blue and FITC-dextran leakage were determined to evaluate blood-brain barrier (BBB) permeability in vivo. Transendothelial electrical resistance (TEER) and Na-F penetration rate were measured with a co-culture of the human cerebral microvascular endothelial cell line (hCMEC/D3) and human normal glial cell line (HEB) in vitro BBB model. WB were used to analyzed the expression of BBB tight junctions (TJs) related protein (ZO-1, Occludin, Claudin-3, p-ERM, and p-MLC), ultrastructure of TJs was determined by transmission electron microscope. RESULTS LC-MS/MS analysis demonstrated that EGb could improve brain uptake of ginsenoside Rg1, Re, Rd and Rb1. In vivo study showed that, BBB permeability was significantly increased after EGb administration, evidenced by the markedly increased penetration of FITC-dextran and Evans Blue into the mice brain parenchyma. In the in vitro BBB model, reduced TEER and increased Na-F penetration rate was observed in EGb group, which was associated with alteration of TJs ultrastructure. Furthermore, the expression of p-ERM and p-MLC in hCMEC/D3 as well as mice brain microvessels were significantly upregulated, but no significant change on the expression of TJs proteins (ZO-1, Occludin and Claudin-3). Moreover, the effect of EGb on in vitro BBB permeability and ERM, MLC phosphorylation was counteracted by DPCPX, an A1 adenosine receptor (A1R) antagonist. CONCLUSIONS EGb might induce ERM/MLC phosphorylation and increase the cell-cell junction gaps to cause a reversible increase of the BBB permeability via A1R signaling pathway. Our results may contribute to better use of EGb in the treatment of brain diseases.
Collapse
Affiliation(s)
- Wenyi Liang
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Wei Xu
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Jing Zhu
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Yadong Zhu
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Quanlin Gu
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Yuping Li
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Caijuan Guo
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Yijian Huang
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Jiangfeng Yu
- The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Weixuan Wang
- Guangdong Province Research Centre for Chinese Integrative Medicine Against Metabolic Disease, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Guangdong TCM Key Laboratory for Metabolic Diseases, China; The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Yinming Hu
- Guangdong Province Research Centre for Chinese Integrative Medicine Against Metabolic Disease, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Guangdong TCM Key Laboratory for Metabolic Diseases, China; The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Yanqun Zhao
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,510515, China
| | - Bin Han
- College of Traditional Chinese Medicine (TCM), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Weijian Bei
- Guangdong Province Research Centre for Chinese Integrative Medicine Against Metabolic Disease, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Guangdong TCM Key Laboratory for Metabolic Diseases, China; The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China.
| | - Jiao Guo
- Guangdong Province Research Centre for Chinese Integrative Medicine Against Metabolic Disease, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Guangdong TCM Key Laboratory for Metabolic Diseases, China; The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China.
| |
Collapse
|
525
|
Branca JV, Pacini A, Carrino D. Targeting cannabidiol to specific areas of the brain: an ultrasound-based strategy. Neural Regen Res 2020; 15:2247-2248. [PMID: 32594040 PMCID: PMC7749472 DOI: 10.4103/1673-5374.284992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
526
|
Sharma A, Sharma R, Zhang Z, Liaw K, Kambhampati SP, Porterfield JE, Lin KC, DeRidder LB, Kannan S, Kannan RM. Dense hydroxyl polyethylene glycol dendrimer targets activated glia in multiple CNS disorders. SCIENCE ADVANCES 2020; 6:eaay8514. [PMID: 32010790 PMCID: PMC6976300 DOI: 10.1126/sciadv.aay8514] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/20/2019] [Indexed: 05/23/2023]
Abstract
Poor transport of neuropharmaceutics through central nervous system (CNS) barriers limits the development of effective treatments for CNS disorders. We present the facile synthesis of a novel neuroinflammation-targeting polyethylene glycol-based dendrimer (PEGOL-60) using an efficient click chemistry approach. PEGOL-60 reduces synthetic burden by achieving high hydroxyl surface density at low generation, which plays a key role in brain penetration and glia targeting of dendrimers in CNS disorders. Systemically administered PEGOL-60 crosses impaired CNS barriers and specifically targets activated microglia/macrophages at the injured site in diverse animal models for cerebral palsy, glioblastoma, and age-related macular degeneration, demonstrating its potential to overcome impaired blood-brain, blood-tumor-brain, and blood-retinal barriers and target key cells in the CNS. PEGOL-60 also exhibits powerful intrinsic anti-oxidant and anti-inflammatory effects in inflamed microglia in vitro. Therefore, PEGOL-60 is an effective vehicle to specifically deliver therapies to sites of CNS injury for enhanced therapeutic outcomes in a range of neuroinflammatory diseases.
Collapse
Affiliation(s)
- Anjali Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Rishi Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Zhi Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kevin Liaw
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore MD 21218, USA
| | - Siva P. Kambhampati
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Joshua E. Porterfield
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore MD 21218, USA
| | - Ku Chien Lin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore MD 21218, USA
| | - Louis B. DeRidder
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore MD 21218, USA
| | - Sujatha Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Hugo W. Moser Research Institute at Kennedy Krieger Inc., Baltimore, MD 21205, USA
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore MD 21218, USA
- Hugo W. Moser Research Institute at Kennedy Krieger Inc., Baltimore, MD 21205, USA
| |
Collapse
|
527
|
Khosa A, Krishna KV, Dubey SK, Saha RN. Lipid Nanocarriers for Enhanced Delivery of Temozolomide to the Brain. Methods Mol Biol 2020; 2059:285-298. [PMID: 31435928 DOI: 10.1007/978-1-4939-9798-5_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Brain disorders, a diverse range of conditions comprising of neurological and psychiatric conditions, are the leading cause of disability, severely affect the quality of life, and in many cases lead to mortality. The prime challenge in treatment of brain disorders is to deliver therapeutics by overcoming the blood-brain barrier (BBB), a unique anatomical and physiological barrier which restricts the passage of a number of molecules, proteins, and cells from the bloodstream. Lipid nanoparticles have emerged as promising drug delivery systems primarily because of biodegradability, low toxicity potential, and the ability to cross physiological barriers especially the BBB even without surface modifications.In this chapter we discuss the preparation and characterization of nanostructured lipid carriers of temozolomide, a chemotherapeutic drug. Evaluation of pharmacokinetics and biodistribution of the nanocarrier system in rats revealed improved delivery of the chemotherapeutic agent to the brain with the potential of lesser side effects.
Collapse
Affiliation(s)
- Archana Khosa
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani, India.
| | - Kowthavarapu V Krishna
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani, India
| | | |
Collapse
|
528
|
Pacheco C, Sousa F, Sarmento B. Chitosan-based nanomedicine for brain delivery: Where are we heading? REACT FUNCT POLYM 2020. [DOI: 10.1016/j.reactfunctpolym.2019.104430] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
529
|
Mauri E, Veglianese P, Papa S, Rossetti A, De Paola M, Mariani A, Posel Z, Posocco P, Sacchetti A, Rossi F. Effects of primary amine-based coatings on microglia internalization of nanogels. Colloids Surf B Biointerfaces 2020; 185:110574. [DOI: 10.1016/j.colsurfb.2019.110574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/20/2022]
|
530
|
Nishioka S, Wu PH, Yakabe T, Giaccia AJ, Le QT, Aoyama H, Shimizu S, Shirato H, Onodera Y, Nam JM. Rab27b contributes to radioresistance and exerts a paracrine effect via epiregulin in glioblastoma. Neurooncol Adv 2020; 2:vdaa091. [PMID: 33409495 PMCID: PMC7770522 DOI: 10.1093/noajnl/vdaa091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Radiotherapy is the standard treatment for glioblastoma (GBM). However, radioresistance of GBM cells leads to recurrence and poor patient prognosis. Recent studies suggest that secretion factors have important roles in radioresistance of tumor cells. This study aims to determine whether Rab27b, a small GTPase involved in secretory vesicle trafficking, plays a role in radioresistance of GBM. METHODS Microarray analysis, cell viability analysis, apoptosis assay, immunostaining, and in vivo experiments were performed to assess the effect of Rab27b on radioresistance of GBM. We further investigated paracrine effects mediated by Rab27b after X-ray irradiation using coculture systems of glioma cell lines. RESULTS Rab27b was specifically upregulated in irradiated U87MG cells. Furthermore, Rab27b knockdown decreased the proliferation of GBM cells after irradiation. Knockdown of Rab27b in U87MG cells combined with radiation treatment suppressed orthotopic tumor growth in the mouse brain and prolonged the survival of recipient mice. Interestingly, the co-upregulation of Rab27b and epiregulin (EREG), a member of the epidermal growth factor (EGF) family, correlated with radioresistance in glioma cell lines. Additionally, EREG, which was secreted from U87MG cells via Rab27b-mediated mechanism, activated EGF receptor and contributed to H4 cell proliferation in a paracrine manner. CONCLUSIONS Our results show that Rab27b mediates the radioresistance of highly malignant GBM cells. Rab27b promotes the proliferation of adjacent cells through EREG-mediated paracrine signaling after irradiation. Thus, the Rab27b-EREG pathway is a novel potential target to improve the efficacy of radiotherapy in GBM.
Collapse
Affiliation(s)
- Soichiro Nishioka
- Molecular and Cellular Dynamics Research, Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo, Japan
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Ping-Hsiu Wu
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Hidefumi Aoyama
- Department of Radiation Oncology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinichi Shimizu
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Radiation Medical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroki Shirato
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhito Onodera
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Molecular Biology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Jin-Min Nam
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
531
|
Overview of Current Drug Delivery Methods Across the Blood-Brain Barrier for the Treatment of Primary Brain Tumors. CNS Drugs 2020; 34:1121-1131. [PMID: 32965590 PMCID: PMC7658069 DOI: 10.1007/s40263-020-00766-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2020] [Indexed: 01/05/2023]
Abstract
Existing drug delivery methods have not led to a significant increase in survival for patients with malignant primary brain tumors. While the combination of conventional therapies consisting of surgery, radiotherapy, and chemotherapy has improved survival for some types of brain tumors (e.g., WNT medulloblastoma), other types of brain tumors (e.g., glioblastoma and diffuse midline glioma) still have a poor prognosis. The reason for the differences in response can be largely attributed to the blood-brain barrier (BBB), a specialized structure at the microvasculature level that regulates the transport of molecules across the blood vessels into the brain parenchyma. This structure hampers the delivery of most chemotherapeutic agents for the treatment of primary brain tumors. Several drug delivery methods such as nanoparticles, convection enhanced delivery, focused ultrasound, intranasal delivery, and intra-arterial delivery have been developed to overcome the BBB in primary brain tumors. However, prognosis of most primary brain tumors still remains poor. The heterogeneity of the BBB in primary brain tumors and the distinct vasculature of tumors make it difficult to design a drug delivery method that targets the entire tumor. Drug delivery methods that combine strategies such as focused ultrasound and nanoparticles might be a more successful approach. However, more research is needed to optimize and develop new drug delivery techniques to improve survival of patients with primary brain tumors.
Collapse
|
532
|
Zhao M, van Straten D, Broekman ML, Préat V, Schiffelers RM. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics 2020; 10:1355-1372. [PMID: 31938069 PMCID: PMC6956816 DOI: 10.7150/thno.38147] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022] Open
Abstract
The current achievements in treating glioblastoma (GBM) patients are not sufficient because many challenges exist, such as tumor heterogeneity, the blood brain barrier, glioma stem cells, drug efflux pumps and DNA damage repair mechanisms. Drug combination therapies have shown increasing benefits against those challenges. With the help of nanocarriers, enhancement of the efficacy and safety could be gained using synergistic combinations of different therapeutic agents. In this review, we will discuss the major issues for GBM treatment, the rationales of drug combinations with or without nanocarriers and the principle of enhanced permeability and retention effect involved in nanomedicine-based tumor targeting and promising nanodiagnostics or -therapeutics. We will also summarize the recent progress and discuss the clinical perspectives of nanocarrier-based combination therapies. The goal of this article was to provide better understanding and key considerations to develop new nanomedicine combinations and nanotheranostics options to fight against GBM.
Collapse
Affiliation(s)
- Mengnan Zhao
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1 73.12, 1200 Brussels, Belgium
| | - Demian van Straten
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Marike L.D. Broekman
- Department of Neurosurgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1 73.12, 1200 Brussels, Belgium
| | - Raymond M. Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| |
Collapse
|
533
|
Zhou Z, Ni K, Deng H, Chen X. Dancing with reactive oxygen species generation and elimination in nanotheranostics for disease treatment. Adv Drug Deliv Rev 2020; 158:73-90. [PMID: 32526453 DOI: 10.1016/j.addr.2020.06.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) play important roles in cell signaling and tissue homeostasis, in which the level of ROS is critical through the equilibrium between ROS generating and eliminating events. A disruption of the balance leads to disease development either by a surplus or a dearth of ROS, which requires ROS-modulating strategies to overturn the defect for disease treatment. Over the past decade, there have been tremendous advances in nanomedicine centering ROS generation and/or elimination as major mechanisms to treat a variety of diseases. In this review, we will discuss the research achievements on two opposite approaches of ROS-generating and ROS-eliminating strategies for treating cancer and other related diseases. Importantly, we will highlight the conceptual and strategic advances of ROS-mediated immunomodulation, including macrophage polarization, immunogenic cell death and T cell activation, which are currently rising as one of the mainstreams of cancer therapy. At the end, the future challenges and opportunities of mediating ROS-based mechanisms are envisioned. In light of the pleiotropic roles of ROS in different diseases, we hope this review is timely to deliver a clear logic of designing principles on ROS generation and elimination for different disease treatments.
Collapse
|
534
|
Charabati M, Rabanel JM, Ramassamy C, Prat A. Overcoming the Brain Barriers: From Immune Cells to Nanoparticles. Trends Pharmacol Sci 2019; 41:42-54. [PMID: 31839374 DOI: 10.1016/j.tips.2019.11.001] [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: 07/31/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 01/04/2023]
Abstract
Nanoparticulate carriers, often referred to as nanoparticles (NPs), represent an important pharmacological advance for drug protection and tissue-specific drug delivery. Accessing the central nervous system (CNS), however, is a complex process regulated by mainly three brain barriers. While some leukocyte (i.e., immune cell) subsets are equipped with the adequate molecular machinery to infiltrate the CNS in physiological and/or pathological contexts, the successful delivery of NPs into the CNS remains hindered by the tightness of the brain barriers. Here, we present an overview of the three major brain barriers and the mechanisms allowing leukocytes to migrate across each of them. We subsequently review different immune-inspired and -mediated strategies to deliver NPs into the CNS. Finally, we discuss the prospect of exploiting leukocyte trafficking mechanisms for further progress.
Collapse
Affiliation(s)
- Marc Charabati
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Neuroimmunology Unit, Centre de Recherche du CHUM (CRCHUM), Montréal, QC, Canada
| | - Jean-Michel Rabanel
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531, Boulevard des Prairies, Laval, QC, Canada
| | - Charles Ramassamy
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531, Boulevard des Prairies, Laval, QC, Canada.
| | - Alexandre Prat
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, and Neuroimmunology Unit, Centre de Recherche du CHUM (CRCHUM), Montréal, QC, Canada.
| |
Collapse
|
535
|
Mikkelsen RB, Frederiksen HRS, Gjerris M, Holst B, Hyttel P, Luo Y, Freude K, Sandøe P. Genetic Protection Modifications: Moving Beyond the Binary Distinction Between Therapy and Enhancement for Human Genome Editing. CRISPR J 2019; 2:362-369. [PMID: 31860350 DOI: 10.1089/crispr.2019.0024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The current debate and policy surrounding the use of genome editing in humans typically relies on a binary distinction between therapy and human enhancement. Here, we argue that this dichotomy fails to take into account perhaps the most significant potential uses of CRISPR-Cas9 genome editing in humans. We argue that genetic treatment of sporadic Alzheimer's disease, breast and ovarian cancer predisposing BRCA1/2 mutations, and the introduction of human immunodeficiency virus resistance in humans should be considered within a new category of genetic protection treatments. We suggest that if this category is not introduced, life-altering research might be unnecessarily limited by current or future policy. Otherwise ad hoc decisions might be made, which introduce a risk of unforeseen moral costs, and might overlook or fail to address some important opportunities.
Collapse
Affiliation(s)
| | | | - Mickey Gjerris
- Department of Food and Resource Economics, Aarhus University, Aarhus, Denmark
| | - Bjørn Holst
- Department of Bioneer A/S, Hørsholm, Denmark
| | - Poul Hyttel
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Yonglun Luo
- Department of Lars Bolund Institute of Regenerative Medicine, BGI-Shenzhen, Shenzhen, PR China
- Department of Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Peter Sandøe
- Department of Food and Resource Economics, Aarhus University, Aarhus, Denmark
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
536
|
Luo Y, Yang H, Zhou YF, Hu B. Dual and multi-targeted nanoparticles for site-specific brain drug delivery. J Control Release 2019; 317:195-215. [PMID: 31794799 DOI: 10.1016/j.jconrel.2019.11.037] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/26/2022]
Abstract
In recent years, nanomedicines have emerged as a promising method for central nervous system drug delivery, enabling the drugs to overcome the blood-brain barrier and accumulate preferentially in the brain. Despite the current success of brain-targeted nanomedicines, limitations still exist in terms of the targeting specificity. Based on the molecular mechanism, the exact cell populations and subcellular organelles where the injury occurs and the drugs take effect have been increasingly accepted as a more specific target for the next generation of nanomedicines. Dual and multi-targeted nanoparticles integrate different targeting functionalities and have provided a paradigm for precisely delivering the drug to the pathological site inside the brain. The targeting process often involves the sequential or synchronized navigation of the targeting moieties, which allows highly controlled drug delivery compared to conventional targeting strategies. Herein, we focus on the up-to-date design of pathological site-specific nanoparticles for brain drug delivery, highlighting the dual and multi-targeting strategies that were employed and their impact on improving targeting specificity and therapeutic effects. Furthermore, the background discussion of the basic properties of a brain-targeted nanoparticle and the common lesion features classified by neurological pathology are systematically summarized.
Collapse
Affiliation(s)
- Yan Luo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hang Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi-Fan Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
537
|
d'Angelo M, Castelli V, Benedetti E, Antonosante A, Catanesi M, Dominguez-Benot R, Pitari G, Ippoliti R, Cimini A. Theranostic Nanomedicine for Malignant Gliomas. Front Bioeng Biotechnol 2019; 7:325. [PMID: 31799246 PMCID: PMC6868071 DOI: 10.3389/fbioe.2019.00325] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
Brain tumors mainly originate from glial cells and are classified as gliomas. Malignant gliomas represent an incurable disease; indeed, after surgery and chemotherapy, recurrence appears within a few months, and mortality has remained high in the last decades. This is mainly due to the heterogeneity of malignant gliomas, indicating that a single therapy is not effective for all patients. In this regard, the advent of theranostic nanomedicine, a combination of imaging and therapeutic agents, represents a strategic tool for the management of malignant brain tumors, allowing for the detection of therapies that are specific to the single patient and avoiding overdosing the non-responders. Here, recent theranostic nanomedicine approaches for glioma therapy are described.
Collapse
Affiliation(s)
- Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Reyes Dominguez-Benot
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giuseppina Pitari
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
538
|
Shen Y, Lifante J, Ximendes E, Santos HDA, Ruiz D, Juárez BH, Zabala Gutiérrez I, Torres Vera V, Rubio Retama J, Martín Rodríguez E, Ortgies DH, Jaque D, Benayas A, Del Rosal B. Perspectives for Ag 2S NIR-II nanoparticles in biomedicine: from imaging to multifunctionality. NANOSCALE 2019; 11:19251-19264. [PMID: 31560003 DOI: 10.1039/c9nr05733a] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Research on near-infrared (NIR) bioimaging has progressed very quickly in the past few years, as fluorescence imaging is reaching a credible implementation as a preclinical technique. The applications of NIR bioimaging in theranostics have contributed to its increasing impact. This has brought about the development of novel technologies and, simultaneously, of new contrast agents capable of acting as efficient NIR optical probes. Among these probes, Ag2S nanoparticles (NPs) have attracted increasing attention due to their temperature-sensitive NIR-II emission, which can be exploited for deep-tissue imaging and thermometry, and their heat delivery capabilities. This multifunctionality makes Ag2S NPs ideal candidates for theranostics. This review presents a critical analysis of the synthesis routes, properties and optical features of Ag2S NPs. We also discuss the latest and most remarkable achievements enabled by these NPs in preclinical imaging and theranostics, together with a critical assessment of their potential to face forthcoming challenges in biomedicine.
Collapse
Affiliation(s)
- Yingli Shen
- Fluorescence Imaging Group, Departamento de Física de Materiales - Facultad de Ciencias, Universidad Autónoma de Madrid, C/Francisco Tomás y Valiente 7, Madrid 28049, Spain
| | - José Lifante
- Fluorescence Imaging Group, Departamento de Fisiología - Facultad de Medicina, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid 28029, Spain and Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain
| | - Erving Ximendes
- Fluorescence Imaging Group, Departamento de Física de Materiales - Facultad de Ciencias, Universidad Autónoma de Madrid, C/Francisco Tomás y Valiente 7, Madrid 28049, Spain and Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain
| | - Harrison D A Santos
- Grupo de Nano-Fotônica e Imagens, Instituto de Física, Universidade Federal de Alagoas, Maceió-AL 57072-900, Brazil
| | - Diego Ruiz
- IMDEA Nanoscience, Faraday 9, Campus de Cantoblanco, Madrid 28049, Spain
| | - Beatriz H Juárez
- IMDEA Nanoscience, Faraday 9, Campus de Cantoblanco, Madrid 28049, Spain and Department of Applied Physical Chemistry and Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Irene Zabala Gutiérrez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Plaza de Ramón y Cajal, s/n, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Vivian Torres Vera
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Plaza de Ramón y Cajal, s/n, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Jorge Rubio Retama
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain and Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Plaza de Ramón y Cajal, s/n, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Emma Martín Rodríguez
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain and Fluorescence Imaging Group, Departamento de Física Aplicada - Facultad de Ciencias, Universidad Autónoma de Madrid, C/Francisco Tomás y Valiente 7, Madrid 28049, Spain
| | - Dirk H Ortgies
- Fluorescence Imaging Group, Departamento de Física de Materiales - Facultad de Ciencias, Universidad Autónoma de Madrid, C/Francisco Tomás y Valiente 7, Madrid 28049, Spain and Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain
| | - Daniel Jaque
- Fluorescence Imaging Group, Departamento de Física de Materiales - Facultad de Ciencias, Universidad Autónoma de Madrid, C/Francisco Tomás y Valiente 7, Madrid 28049, Spain and Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain
| | - Antonio Benayas
- Department of Physics and CICECO-Aveiro Institute of Materials; University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Blanca Del Rosal
- Centre for Micro-Photonics, Faculty of Science Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia.
| |
Collapse
|
539
|
Roux GL, Jarray R, Guyot AC, Pavoni S, Costa N, Théodoro F, Nassor F, Pruvost A, Tournier N, Kiyan Y, Langer O, Yates F, Deslys JP, Mabondzo A. Proof-of-Concept Study of Drug Brain Permeability Between in Vivo Human Brain and an in Vitro iPSCs-Human Blood-Brain Barrier Model. Sci Rep 2019; 9:16310. [PMID: 31690750 PMCID: PMC6831611 DOI: 10.1038/s41598-019-52213-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/10/2019] [Indexed: 12/22/2022] Open
Abstract
The development of effective central nervous system (CNS) drugs has been hampered by the lack of robust strategies to mimic the blood-brain barrier (BBB) and cerebrovascular impairments in vitro. Recent technological advancements in BBB modeling using induced pluripotent stem cells (iPSCs) allowed to overcome some of these obstacles, nonetheless the pertinence for their use in drug permeation study remains to be established. This mandatory information requires a cross comparison of in vitro and in vivo pharmacokinetic data in the same species to avoid failure in late clinical drug development. Here, we measured the BBB permeabilities of 8 clinical positron emission tomography (PET) radioligands with known pharmacokinetic parameters in human brain in vivo with a newly developed in vitro iPSC-based human BBB (iPSC-hBBB) model. Our findings showed a good correlation between in vitro and in vivo drug brain permeability (R2 = 0.83; P = 0.008) which contrasted with the limited correlation between in vitro apparent permeability for a set of 18 CNS/non-CNS compounds using the in vitro iPSCs-hBBB model and drug physicochemical properties. Our data suggest that the iPSC-hBBB model can be integrated in a flow scheme of CNS drug screening and potentially used to study species differences in BBB permeation.
Collapse
Affiliation(s)
- Gwenaëlle Le Roux
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Rafika Jarray
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Anne-Cécile Guyot
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Serena Pavoni
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France
| | - Narciso Costa
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Frédéric Théodoro
- Service de Pharmacologie et d'Immunoanalyse (SPI), Plateforme Smart-MS, CEA, INRA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Ferid Nassor
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Alain Pruvost
- Service de Pharmacologie et d'Immunoanalyse (SPI), Plateforme Smart-MS, CEA, INRA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Nicolas Tournier
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Univ. Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Yulia Kiyan
- Medizinische Hochschule Hannover, DE-30625, Hannover, Germany
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria.,Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, A-2444, Seibersdorf, Austria
| | - Frank Yates
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Jean Philippe Deslys
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France
| | - Aloïse Mabondzo
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France.
| |
Collapse
|
540
|
Peyravian N, Dikici E, Deo S, Toborek M, Daunert S. Opioid antagonists as potential therapeutics for ischemic stroke. Prog Neurobiol 2019; 182:101679. [PMID: 31398359 PMCID: PMC6814577 DOI: 10.1016/j.pneurobio.2019.101679] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/31/2019] [Indexed: 01/09/2023]
Abstract
Chronic use of prescription opioids exacerbates risk and severity of ischemic stroke. Annually, 6 million people die from stroke worldwide and there are no neuroprotective or neurorestorative agents to improve stroke outcomes and promote recovery. Prescribed opioids such as morphine have been shown to alter tight junction protein expression, resulting in the disruption of the blood brain barrier (BBB), ultimately leading to stroke pathogenesis. Consequently, protection of the BBB has been proposed as a therapeutic strategy for ischemic stroke. This perspective addresses the deficiency in stroke pharmacological options and examines a novel application and repurposing of FDA-approved opioid antagonists as a prospective neuroprotective therapeutic strategy to minimize BBB damage, reduce stroke severity, and promote neural recovery. Future directions discuss potential drug design and delivery methods to enhance these novel therapeutic targets.
Collapse
Affiliation(s)
- Nadia Peyravian
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA
| | - Sapna Deo
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA.
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA; University of Miami Clinical and Translational Science Institute, USA.
| |
Collapse
|
541
|
Adam H, Gopinath SC, Arshad MM, Adam T, Hashim U. Perspectives of nanobiotechnology and biomacromolecules in parkinson’s disease. Process Biochem 2019. [DOI: 10.1016/j.procbio.2019.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
542
|
Morales-Cruz M, Delgado Y, Castillo B, Figueroa CM, Molina AM, Torres A, Milián M, Griebenow K. Smart Targeting To Improve Cancer Therapeutics. Drug Des Devel Ther 2019; 13:3753-3772. [PMID: 31802849 PMCID: PMC6826196 DOI: 10.2147/dddt.s219489] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/06/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second largest cause of death worldwide with the number of new cancer cases predicted to grow significantly in the next decades. Biotechnology and medicine can and should work hand-in-hand to improve cancer diagnosis and treatment efficacy. However, success has been frequently limited, in particular when treating late-stage solid tumors. There still is the need to develop smart and synergistic therapeutic approaches to achieve the synthesis of strong and effective drugs and delivery systems. Much interest has been paid to the development of smart drug delivery systems (drug-loaded particles) that utilize passive targeting, active targeting, and/or stimulus responsiveness strategies. This review will summarize some main ideas about the effect of each strategy and how the combination of some or all of them has shown to be effective. After a brief introduction of current cancer therapies and their limitations, we describe the biological barriers that nanoparticles need to overcome, followed by presenting different types of drug delivery systems to improve drug accumulation in tumors. Then, we describe cancer cell membrane targets that increase cellular drug uptake through active targeting mechanisms. Stimulus-responsive targeting is also discussed by looking at the intra- and extracellular conditions for specific drug release. We include a significant amount of information summarized in tables and figures on nanoparticle-based therapeutics, PEGylated drugs, different ligands for the design of active-targeted systems, and targeting of different organs. We also discuss some still prevailing fundamental limitations of these approaches, eg, by occlusion of targeting ligands.
Collapse
Affiliation(s)
- Moraima Morales-Cruz
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR, USA
| | - Yamixa Delgado
- Department of Biochemistry & Pharmacology, San Juan Bautista School of Medicine, Caguas, PR, USA
| | - Betzaida Castillo
- Department of Chemistry, University of Puerto Rico, Humacao Campus, Humacao, PR, USA
| | - Cindy M Figueroa
- Department of Math and Sciences, Polytechnic University of Puerto Rico, San Juan, PR, USA
| | - Anna M Molina
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR, USA
| | - Anamaris Torres
- Department of Biochemistry & Pharmacology, San Juan Bautista School of Medicine, Caguas, PR, USA
| | - Melissa Milián
- Department of Biochemistry & Pharmacology, San Juan Bautista School of Medicine, Caguas, PR, USA
| | - Kai Griebenow
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR, USA
| |
Collapse
|
543
|
Lakkadwala S, Dos Santos Rodrigues B, Sun C, Singh J. Biodistribution of TAT or QLPVM coupled to receptor targeted liposomes for delivery of anticancer therapeutics to brain in vitro and in vivo. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 23:102112. [PMID: 31669083 DOI: 10.1016/j.nano.2019.102112] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/26/2019] [Accepted: 10/03/2019] [Indexed: 01/31/2023]
Abstract
Combination therapy has emerged as an efficient way to deliver chemotherapeutics for treatment of glioblastoma. It provides collaborative approach of targeting cancer cells by acting via multiple mechanisms, thereby reducing drug resistance. However, the presence of impermeable blood brain barrier (BBB) restricts the delivery of chemotherapeutic drugs into the brain. To overcome this limitation, we designed a dual functionalized liposomes by modifying their surface with transferrin (Tf) and a cell penetrating peptide (CPP) for receptor and adsorptive mediated transcytosis, respectively. In this study, we used two different CPPs (based on physicochemical properties) and investigated the influence of insertion of CPP to Tf-liposomes on biocompatibility, cellular uptake, and transport across the BBB both in vitro and in vivo. The biodistribution profile of Tf-CPP liposomes showed more than 10 and 2.7 fold increase in doxorubicin and erlotinib accumulation in mice brain, respectively as compared to free drugs with no signs of toxicity.
Collapse
Affiliation(s)
- Sushant Lakkadwala
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Bruna Dos Santos Rodrigues
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Chengwen Sun
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
544
|
Jung B, Huh H, Lee EH, Han M, Park J. An advanced focused ultrasound protocol improves the blood-brain barrier permeability and doxorubicin delivery into the rat brain. J Control Release 2019; 315:55-64. [PMID: 31669208 DOI: 10.1016/j.jconrel.2019.10.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
Despite the recent development of a focused ultrasound (FUS) technique for disrupting the blood-brain barrier (BBB) and enabling the delivery of drugs into the targeted brain region, different sonication protocols have not been fully explored. In this study, we suggest a simple and cost-effective protocol that improves the BBB permeability and drug delivery without damaging the tissue. In this protocol, called "FUS+BBBD protocol", an additional FUS stimulation without microbubbles ("FUS protocol"; 0.5, 1.0, or 2.0MPa acoustic pressure, 10ms tone burst, 1Hz pulse repetition frequency, 120s total duration) is applied prior to the conventional BBB disruption with microbubbles ("BBBD protocol"; 0.6∼0.72MPa acoustic pressure, 10ms tone burst, 1Hz pulse repetition frequency, 120s total duration). With the "FUS+BBBD protocol", the magnetic resonance signal intensity and doxorubicin delivery at the targeted brain region were increased by 1.59 and 1.75 times at an FUS intensity of 1.0MPa, respectively, compared to the conventional BBBD. Other conditions also increase the drug delivery, but the increase was smaller than that at 1.0MPa (1.15 times for 0.5MPa and 1.60 times for 2.0MPa). The H&E histopathological analysis of the sonicated brain region using the proposed "FUS+BBBD protocol" showed no significant brain tissue damage at a FUS intensity of 0.5 and 1.0MPa. However, region cavities due to the damage were observed after an FUS intensity of 2.0MPa. These results suggest that the 1.0MPa "FUS+BBBD protocol" increases the BBB permeability and enhances the drug delivery efficiency without noticeable brain tissue damage, compared with the conventional BBBD. Although further studies are needed to determine the underlying mechanism of this effect, drugs that have been reported to be effective in the treatment of brain disease but had limited use due to severe systemic side effects will benefit from the enhanced drug delivery of "FUS+BBBD protocol". Furthermore, the suggested protocol may facilitate the development of new strategies in clinical trials to treat brain disorders with improved drug delivery and safety.
Collapse
Affiliation(s)
- Byeongjin Jung
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Hyungkyu Huh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Eun-Hee Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Mun Han
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Juyoung Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea.
| |
Collapse
|
545
|
Nose-to-brain co-delivery of repurposed simvastatin and BDNF synergistically attenuates LPS-induced neuroinflammation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 23:102107. [PMID: 31655202 DOI: 10.1016/j.nano.2019.102107] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 12/29/2022]
Abstract
A therapeutic strategy that can combat the multifaceted nature of neuroinflammation pathology was investigated. Thus, we fabricated PEG-PdLLA polymersomes and evaluated the efficacy in co-delivery of simvastatin (Sim, as a repurposed anti-inflammatory agent) with brain derived neurotrophic factor (BDNF, as an exogeneous trophic factor supplementation). Using LPS model of neuroinflammation, intranasal administration of combination drug-loaded polymersomes (containing both Sim and BDNF; Sim-BDNF-Ps) markedly down-regulated brain levels of cytokines compared to free drug and single-drug-loaded polymersomes. Further, Sim-BDNF-Ps effectively replenished brain level of BDNF that was depleted following neuroinflammation, resulting in a 2-fold BDNF increase versus untreated LPS control group. We found out that the efficiency of the combination drug-loaded polymersomes to suppress microglia activation in brain regions followed the order: frontal cortex > striatum > hippocampus. Our findings indicated that Sim-BDNF-Ps could effectively inhibit microglial-mediated inflammation as well as potentially resolve the neurotoxic microenvironment that is often associated with neuroinflammation.
Collapse
|
546
|
Sellers DL, Tan JKY, Pineda JMB, Peeler DJ, Porubsky VL, Olden BR, Salipante SJ, Pun SH. Targeting Ligands Deliver Model Drug Cargo into the Central Nervous System along Autonomic Neurons. ACS NANO 2019; 13:10961-10971. [PMID: 31589023 PMCID: PMC7651855 DOI: 10.1021/acsnano.9b01515] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
While biologic drugs such as proteins, peptides, or nucleic acids have shown promise in the treatment of neurodegenerative diseases, the blood-brain barrier (BBB) severely limits drug delivery to the central nervous system (CNS) after systemic administration. Consequently, drug delivery challenges preclude biological drug candidates from the clinical armamentarium. In order to target drug delivery and uptake into to the CNS, we used an in vivo phage display screen to identify peptides able to target drug-uptake by the vast array of neurons of the autonomic nervous system (ANS). Using next-generation sequencing, we identified 21 candidate targeted ANS-to-CNS uptake ligands (TACL) that enriched bacteriophage accumulation and delivered protein-cargo into the CNS after intraperitoneal (IP) administration. The series of TACL peptides were synthesized and tested for their ability to deliver a model enzyme (NeutrAvidin-horseradish peroxidase fusion) to the brain and spinal cord. Three TACL-peptides facilitated significant active enzyme delivery into the CNS, with limited accumulation in off-target organs. Peptide structure and serum stability is increased when internal cysteine residues are cyclized by perfluoroarylation with decafluorobiphenyl, which increased delivery to the CNS further. TACL-peptide was demonstrated to localize in parasympathetic ganglia neurons in addition to neuronal structures in the hindbrain and spinal cord. By targeting uptake into ANS neurons, we demonstrate the potential for TACL-peptides to bypass the blood-brain barrier and deliver a model drug into the brain and spinal cord.
Collapse
Affiliation(s)
- Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - James-Kevin Y. Tan
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Julio Marco B. Pineda
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - David J. Peeler
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Veronica L. Porubsky
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Brynn R. Olden
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington, 98195, USA
| |
Collapse
|
547
|
Weber-Adrian D, Kofoed RH, Chan JWY, Silburt J, Noroozian Z, Kügler S, Hynynen K, Aubert I. Strategy to enhance transgene expression in proximity of amyloid plaques in a mouse model of Alzheimer's disease. Theranostics 2019; 9:8127-8137. [PMID: 31754385 PMCID: PMC6857057 DOI: 10.7150/thno.36718] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/29/2019] [Indexed: 12/26/2022] Open
Abstract
Gene therapy can be designed to efficiently counter pathological features characteristic of neurodegenerative disorders. Here, we took advantage of the glial fibrillary acidic protein (GFAP) promoter to preferentially enhance transgene expression near plaques composed of amyloid-beta peptides (Aβ), a hallmark of Alzheimer's disease (AD), in the TgCRND8 mouse model of amyloidosis. Methods: The delivery of intravenously injected recombinant adeno-associated virus mosaic serotype 1/2 (rAAV1/2) to the cortex and hippocampus of TgCRND8 mice was facilitated using transcranial MRI-guided focused ultrasound in combination with microbubbles (MRIgFUS), which transiently and locally increases the permeability of the blood-brain barrier (BBB). rAAV1/2 expression of the reporter green fluorescent protein (GFP) under a GFAP promoter was compared to GFP expression driven by the constitutive human beta actin (HBA) promoter. Results: MRIgFUS targeting the cortex and hippocampus facilitated the entry of rAAV1/2 and GFP expression under the GFAP promoter was localized to GFAP-positive astrocytes. Adjacent to Aβ plaques where GFAP is upregulated, the volume, surface area, and fluorescence intensity of the transgene GFP were greater in rAAV1/2-GFAP-GFP compared to rAAV1/2-HBA-GFP treated animals. In peripheral organs, GFP expression was particularly strong in the liver, irrespective of the promoter. Conclusion: The GFAP promoter enhanced transgene expression in proximity of Aβ plaques in the brain of TgCRND8 mice, and it also resulted in significant expression in the liver. Future gene therapies for neurological disorders could benefit from using a GFAP promoter to regulate transgene expression in response to disease-induced astrocytic reactivity.
Collapse
Affiliation(s)
- Danielle Weber-Adrian
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - Rikke Hahn Kofoed
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - Josephine Wing Yee Chan
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Joseph Silburt
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - Zeinab Noroozian
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - Sebastian Kügler
- Department of Neurology, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Kullervo Hynynen
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Isabelle Aubert
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| |
Collapse
|
548
|
Vanden-Hehir S, Cairns SA, Lee M, Zoupi L, Shaver MP, Brunton VG, Williams A, Hulme AN. Alkyne-Tagged PLGA Allows Direct Visualization of Nanoparticles In Vitro and Ex Vivo by Stimulated Raman Scattering Microscopy. Biomacromolecules 2019; 20:4008-4014. [PMID: 31408325 PMCID: PMC6794644 DOI: 10.1021/acs.biomac.9b01092] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 12/19/2022]
Abstract
Polymeric nanoparticles (NPs) are attractive candidates for the controlled and targeted delivery of therapeutics in vitro and in vivo. However, detailed understanding of the uptake, location, and ultimate cellular fate of the NPs is necessary to satisfy safety concerns, which is difficult because of the nanoscale size of these carriers. In this work, we show how small chemical labels can be appended to poly(lactic acid-co-glycolic acid) (PLGA) to synthesize NPs that can then be imaged by stimulated Raman scattering microscopy, a vibrational imaging technique that can elucidate bond-specific information in biological environments, such as the identification of alkyne signatures in modified PLGA terpolymers. We show that both deuterium and alkyne labeled NPs can be imaged within primary rat microglia, and the alkyne NPs can also be imaged in ex vivo cortical mouse brain tissue. Immunohistochemical analysis confirms that the NPs localize in microglia in the mouse brain tissue, demonstrating that these NPs have the potential to deliver therapeutics selectively to microglia.
Collapse
Affiliation(s)
- Sally Vanden-Hehir
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| | - Stefan A. Cairns
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| | - Martin Lee
- Edinburgh
Cancer Research UK Centre, University of
Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, United Kingdom
| | - Lida Zoupi
- MRC
Centre for Regenerative Medicine, The University
of Edinburgh, Edinburgh
BioQuarter, 5, Little France Drive, Edinburgh, EH16 4UU, United Kingdom
| | - Michael P. Shaver
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| | - Valerie G. Brunton
- Edinburgh
Cancer Research UK Centre, University of
Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, United Kingdom
| | - Anna Williams
- MRC
Centre for Regenerative Medicine, The University
of Edinburgh, Edinburgh
BioQuarter, 5, Little France Drive, Edinburgh, EH16 4UU, United Kingdom
| | - Alison N. Hulme
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| |
Collapse
|
549
|
Concetta Scuto M, Mancuso C, Tomasello B, Laura Ontario M, Cavallaro A, Frasca F, Maiolino L, Trovato Salinaro A, Calabrese EJ, Calabrese V. Curcumin, Hormesis and the Nervous System. Nutrients 2019; 11:2417. [PMID: 31658697 PMCID: PMC6835324 DOI: 10.3390/nu11102417] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
Curcumin is a polyphenol compound extracted from the rhizome of Curcuma longa Linn (family Zingiberaceae) commonly used as a spice to color and flavor food. Several preclinical studies have suggested beneficial roles for curcumin as an adjuvant therapy in free radical-based diseases, mainly neurodegenerative disorders. Indeed, curcumin belongs to the family of hormetins and the enhancement of the cell stress response, mainly the heme oxygenase-1 system, is actually considered the common denominator for this dual response. However, evidence-based medicine has clearly demonstrated the lack of any therapeutic effect of curcumin to contrast the onset or progression of neurodegeneration and related diseases. Finally, the curcumin safety profile imposes a careful analysis of the risk/benefit balance prior to proposing chronic supplementation with curcumin.
Collapse
Affiliation(s)
- Maria Concetta Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Cesare Mancuso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
- Institute of Pharmacology, Catholic University of Sacred Heart, 00168 Roma, Italy.
| | - Barbara Tomasello
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Maria Laura Ontario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Andrea Cavallaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Francesco Frasca
- Department of Clinical and experimental Medicine, Division of Endocrinology, University of Catania, 95125 Catania, Italy.
| | - Luigi Maiolino
- Department of Medical and Surgery Sciences, University of Catania, 95125 Catania, Italy.
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Science, University of Massachusetts, Amherst, MA 01003, USA.
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| |
Collapse
|
550
|
Moscariello P, Raabe M, Liu W, Bernhardt S, Qi H, Kaiser U, Wu Y, Weil T, Luhmann HJ, Hedrich J. Unraveling In Vivo Brain Transport of Protein-Coated Fluorescent Nanodiamonds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1902992. [PMID: 31465151 DOI: 10.1002/smll.201902992] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Nanotheranostics, combining diagnostics and therapy, has the potential to revolutionize treatment of neurological disorders. But one of the major obstacles for treating central nervous system diseases is the blood-brain barrier (BBB) preventing systemic delivery of drugs and optical probes into the brain. To overcome these limitations, nanodiamonds (NDs) are investigated in this study as they are a powerful sensing and imaging platform for various biological applications and possess outstanding stable far-red fluorescence, do not photobleach, and are highly biocompatible. Herein, fluorescent NDs encapsulated by a customized human serum albumin-based biopolymer (polyethylene glycol) coating (dcHSA-PEG) are taken up by target brain cells. In vitro BBB models reveal transcytosis and an additional direct cell-cell transport via tunneling nanotubes. Systemic application of dcHSA-NDs confirms their ability to cross the BBB in a mouse model. Tracking of dcHSA-NDs is possible at the single cell level and reveals their uptake into neurons and astrocytes in vivo. This study shows for the first time systemic NDs brain delivery and suggests transport mechanisms across the BBB and direct cell-cell transport. Fluorescent NDs are envisioned as traceable transporters for in vivo brain imaging, sensing, and drug delivery.
Collapse
Affiliation(s)
- Pierpaolo Moscariello
- Institute of Physiology, University Medical Center of Johannes Gutenberg University Mainz, Duesbergweg 6, 55128, Mainz, Germany
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Marco Raabe
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Weina Liu
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Sandra Bernhardt
- Institute of Physiology, University Medical Center of Johannes Gutenberg University Mainz, Duesbergweg 6, 55128, Mainz, Germany
| | - Haoyuan Qi
- Central Facility of Electron Microscopy, Electron Microscopy Group of Materials Science, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Ute Kaiser
- Central Facility of Electron Microscopy, Electron Microscopy Group of Materials Science, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Yuzhou Wu
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of Johannes Gutenberg University Mainz, Duesbergweg 6, 55128, Mainz, Germany
| | - Jana Hedrich
- Institute of Physiology, University Medical Center of Johannes Gutenberg University Mainz, Duesbergweg 6, 55128, Mainz, Germany
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| |
Collapse
|