51
|
Gaber M, Medhat W, Hany M, Saher N, Fang JY, Elzoghby A. Protein-lipid nanohybrids as emerging platforms for drug and gene delivery: Challenges and outcomes. J Control Release 2017; 254:75-91. [PMID: 28365294 DOI: 10.1016/j.jconrel.2017.03.392] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/24/2022]
Abstract
Nanoparticulate drug delivery systems have been long used to deliver a vast range of drugs and bioactives owing to their ability to demonstrate novel physical, chemical, and/or biological properties. An exponential growth has spurred in research and development of these nanocarriers which led to the evolution of a great number of diverse nanosystems including liposomes, nanoemulsions, solid lipid nanoparticles (SLNs), micelles, dendrimers, polymeric nanoparticles (NPs), metallic NPs, and carbon nanotubes. Among them, lipid-based nanocarriers have made the largest progress whether commercially or under development. Despite this progress, these lipid-based nanocarriers suffer from several limitations that led to the development of many protein-coated lipid nanocarriers. To less extent, protein-based nanocarriers suffer from limitations that led to the fabrication of some lipid bilayer enveloping protein nanocarriers. This review discusses in-depth some limitations associated with the lipid-based or protein-based nanocarriers and the fruitful outcomes brought by protein-lipid hybridization. Also discussed are the various hybridization techniques utilized to formulate these protein-lipid nanohybrids and the mechanisms involved in the drug loading process.
Collapse
Affiliation(s)
- Mohamed Gaber
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Waseem Medhat
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mark Hany
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Nourhan Saher
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan 333, Taiwan.
| | - Ahmed Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| |
Collapse
|
52
|
Xiaoli F, Junrong W, Xuan L, Yanli Z, Limin W, Jia L, Longquan S. Prenatal exposure to nanosized zinc oxide in rats: neurotoxicity and postnatal impaired learning and memory ability. Nanomedicine (Lond) 2017; 12:777-795. [PMID: 28322126 DOI: 10.2217/nnm-2016-0397] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM To examine the neurotoxicity of prenatal exposure to ZnO nanoparticles on rat offspring. MATERIALS & METHODS Pregnant Sprague-Dawley rats were exposed to ZnO nanoparticles (NPs) by gavage. Toxicity was assessed including zinc biodistribution, cerebral histopathology, antioxidant status and learning and memory capability. RESULTS A significantly elevated concentration of zinc was detected in offspring brains. Transmission electron microscope observations showed abnormal neuron ultrastructures. Histopathologic changes such as decreased proliferation and higher apoptotic death were observed. An obvious imbalanced antioxidant status occurred in brains. Adult experimental offspring exhibited impaired learning and memory behavior in the Morris water maze test compared with control groups. CONCLUSION These adverse effects on offspring brain may cause impaired learning and memory capabilities in adulthood, particularly in female rats.
Collapse
Affiliation(s)
- Feng Xiaoli
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wu Junrong
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lai Xuan
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhang Yanli
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wei Limin
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Liu Jia
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shao Longquan
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
53
|
Fiandra L, Capetti A, Sorrentino L, Corsi F. Nanoformulated Antiretrovirals for Penetration of the Central Nervous System: State of the Art. J Neuroimmune Pharmacol 2017; 12:17-30. [PMID: 27832401 DOI: 10.1007/s11481-016-9716-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 10/28/2016] [Indexed: 12/25/2022]
Abstract
The central nervous system is a very challenging HIV-1 sanctuary. But, despite complete suppression of plasmatic viral replication with current antiretroviral therapy, signs of HIV-1 replication can still be found in the cerebrospinal fluid in some patients. The main limitation to achieving HIV-1 eradication from the brain is related to the suboptimal concentrations of antiretrovirals within this site, due to their low permeation across the blood-brain barrier. In recent years, a number of reliable nanotechnological strategies have been developed with the aim of enhancing antiretroviral drug penetration across the blood-brain barrier. The aim of this review is to provide an overview of the different nanoformulated antiretrovirals, used in both clinical and preclinical studies, that are designed to improve their delivery into the brain by active or passive permeation mechanisms through the barrier. Different nanotechnological approaches have proven successful for optimizing antiretrovirals delivery to the central nervous system, with a likely benefit for HIV-associated neurocognitive disorders and a more debated contribution to the complete eradication of the HIV-1 infection.
Collapse
Affiliation(s)
- Luisa Fiandra
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, via G. B. Grassi 74, 20157, Milan, Italy
| | - Amedeo Capetti
- Division of Infectious Diseases, ASST Fatebenefratelli Sacco - "Luigi Sacco" University Hospital, via G. B. Grassi 74, 20157, Milan, Italy
| | - Luca Sorrentino
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, via G. B. Grassi 74, 20157, Milan, Italy
| | - Fabio Corsi
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, via G. B. Grassi 74, 20157, Milan, Italy.
- Surgery Department, Breast Unit, ICS Maugeri S.p.A. SB, via S. Maugeri 10, 27100, Pavia, Italy.
| |
Collapse
|
54
|
Fang F, Zou D, Wang W, Yin Y, Yin T, Hao S, Wang B, Wang G, Wang Y. Non-invasive approaches for drug delivery to the brain based on the receptor mediated transport. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:1316-1327. [PMID: 28482500 DOI: 10.1016/j.msec.2017.02.056] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/13/2016] [Accepted: 02/14/2017] [Indexed: 10/20/2022]
Abstract
The blood brain barrier (BBB) is a physical and biochemical barrier that prevents entry of toxic compounds into brain for preserving homeostasis. However, the BBB also strictly limits influx of most therapeutic agents into the brain. One promising method for overcoming this problem to deliver drugs is receptor mediated transport (RMT) system, which employs the vesicular trafficking machinery to transport substrates across the BBB endothelium in a noninvasive manner. The conjugates of drug or drug-loaded vector linked with appropriate ligands specifically binds to the endogenous targeting receptor on the surface of the endothelial cells. Then drugs could enter the cell body by means of transcytosis and eventual releasing into the brain parenchyma. Over the past 20years, there have been significant developments of RMT targeting strategies. Here, we will review the recent advance of various promising RMT systems and discuss the capability of these approaches for drug delivery to the brain.
Collapse
Affiliation(s)
- Fei Fang
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China
| | - Dan Zou
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China
| | - Wei Wang
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China
| | - Ying Yin
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China
| | - Tieying Yin
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China
| | - Shilei Hao
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China
| | - Bochu Wang
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China
| | - Guixue Wang
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China
| | - Yazhou Wang
- Key Laboratory of Bio-rheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Shapingba Street 174, Chongqing 404100, China.
| |
Collapse
|
55
|
|
56
|
Sauvage F, Messaoudi S, Fattal E, Barratt G, Vergnaud-Gauduchon J. Heat shock proteins and cancer: How can nanomedicine be harnessed? J Control Release 2017; 248:133-143. [PMID: 28088573 DOI: 10.1016/j.jconrel.2017.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 01/08/2017] [Indexed: 12/18/2022]
Abstract
Heat shock protein (hsp90) is an interesting target for cancer therapy because it is involved in the folding and stabilization of numerous proteins, including many that contribute to the development of cancer. It is part of the chaperone machinery that includes other heat shock proteins (hsp70, hsp27, hsp40) and is mainly localized in the cytosol, although many analogues or isoforms can be found in mitochondrion, endoplasmic reticulum and the cell membrane. Many potential inhibitors of hsp90 have been tested for cancer therapy but their usefulness is limited by their poor solubility in water and their ability to reach the target cells and the correct intracellular compartment. Nanomedicine, the incorporation of active molecules into an appropriate delivery system, could provide a solution to these drawbacks. In this review, we explain the rationale for using nanomedicine for this sort of cancer therapy, considering the properties of the chaperone machinery and of the different hsp90 analogues. We present some results that have already been obtained and put forward some strategies for delivery of hsp90 analogues to specific organelles.
Collapse
Affiliation(s)
- Félix Sauvage
- Institut Galien Paris-Sud, CNRS, UMR 8612, LabEx LERMIT, Univ. Paris-Sud/Univ. Paris-Saclay, 5 rue J.-B. Clément, Châtenay-Malabry, 92296, France
| | - Samir Messaoudi
- BioCIS-UMR 8076, Univ. Paris-Sud, CNRS, University Paris-Saclay, Châtenay-Malabry, 92296, France
| | - Elias Fattal
- Institut Galien Paris-Sud, CNRS, UMR 8612, LabEx LERMIT, Univ. Paris-Sud/Univ. Paris-Saclay, 5 rue J.-B. Clément, Châtenay-Malabry, 92296, France
| | - Gillian Barratt
- Institut Galien Paris-Sud, CNRS, UMR 8612, LabEx LERMIT, Univ. Paris-Sud/Univ. Paris-Saclay, 5 rue J.-B. Clément, Châtenay-Malabry, 92296, France
| | - Juliette Vergnaud-Gauduchon
- Institut Galien Paris-Sud, CNRS, UMR 8612, LabEx LERMIT, Univ. Paris-Sud/Univ. Paris-Saclay, 5 rue J.-B. Clément, Châtenay-Malabry, 92296, France.
| |
Collapse
|
57
|
Tomitaka A, Arami H, Raymond A, Yndart A, Kaushik A, Jayant RD, Takemura Y, Cai Y, Toborek M, Nair M. Development of magneto-plasmonic nanoparticles for multimodal image-guided therapy to the brain. NANOSCALE 2017; 9:764-773. [PMID: 27976764 PMCID: PMC5325696 DOI: 10.1039/c6nr07520g] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Magneto-plasmonic nanoparticles are one of the emerging multi-functional materials in the field of nanomedicine. Their potential for targeting and multi-modal imaging is highly attractive. In this study, magnetic core/gold shell (MNP@Au) magneto-plasmonic nanoparticles were synthesized by citrate reduction of Au ions on magnetic nanoparticle seeds. Hydrodynamic size and optical properties of magneto-plasmonic nanoparticles synthesized with the variation of Au ions and reducing agent concentrations were evaluated. The synthesized magneto-plasmonic nanoparticles exhibited superparamagnetic properties, and their magnetic properties contributed to the concentration-dependent contrast in magnetic resonance imaging (MRI). The imaging contrast from the gold shell part of the magneto-plasmonic nanoparticles was also confirmed by X-ray computed tomography (CT). The transmigration study of the magneto-plasmonic nanoparticles using an in vitro blood-brain barrier (BBB) model proved enhanced transmigration efficiency without disrupting the integrity of the BBB, and showed potential to be used for brain diseases and neurological disorders.
Collapse
Affiliation(s)
- Asahi Tomitaka
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA.
| | - Hamed Arami
- Molecular Imaging Program at Stanford (MIPS), The James H Clark Center, Stanford University, Stanford, California 94305, USA and Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Andrea Raymond
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA.
| | - Adriana Yndart
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA.
| | - Ajeet Kaushik
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA.
| | - Rahul Dev Jayant
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA.
| | - Yasushi Takemura
- Department of Electrical and Computer Engineering, Yokohama National University, Yokohama 240-8501, Japan
| | - Yong Cai
- Department of Chemistry & Biochemistry, Southeast Environmental Research Center, Florida International University, Miami, Florida 33199, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, Florida 33136, USA
| | - Madhavan Nair
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA. and Department of Immunology, Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| |
Collapse
|
58
|
Wang N, Jin X, Zhu X. Construction of biomimetic long-circulation delivery platform encapsulated by zwitterionic polymers for enhanced penetration of blood–brain barrier. RSC Adv 2017. [DOI: 10.1039/c7ra01532a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A core–shell protein-based long circulation delivery platform has been constructed for enhanced penetration of the blood–brain barrier.
Collapse
Affiliation(s)
- Nan Wang
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Xin Jin
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| |
Collapse
|
59
|
Tam VH, Sosa C, Liu R, Yao N, Priestley RD. Nanomedicine as a non-invasive strategy for drug delivery across the blood brain barrier. Int J Pharm 2016; 515:331-342. [DOI: 10.1016/j.ijpharm.2016.10.031] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/02/2016] [Accepted: 10/15/2016] [Indexed: 12/13/2022]
|
60
|
Vieira DB, Gamarra LF. Getting into the brain: liposome-based strategies for effective drug delivery across the blood-brain barrier. Int J Nanomedicine 2016; 11:5381-5414. [PMID: 27799765 PMCID: PMC5077137 DOI: 10.2147/ijn.s117210] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review summarizes articles that have been reported in literature on liposome-based strategies for effective drug delivery across the blood–brain barrier. Due to their unique physicochemical characteristics, liposomes have been widely investigated for their application in drug delivery and in vivo bioimaging for the treatment and/or diagnosis of neurological diseases, such as Alzheimer’s, Parkinson’s, stroke, and glioma. Several strategies have been used to deliver drug and/or imaging agents to the brain. Covalent ligation of such macromolecules as peptides, antibodies, and RNA aptamers is an effective method for receptor-targeting liposomes, which allows their blood–brain barrier penetration and/or the delivery of their therapeutic molecule specifically to the disease site. Additionally, methods have been employed for the development of liposomes that can respond to external stimuli. It can be concluded that the development of liposomes for brain delivery is still in its infancy, although these systems have the potential to revolutionize the ways in which medicine is administered.
Collapse
Affiliation(s)
| | - Lionel F Gamarra
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| |
Collapse
|
61
|
Atluri VSR, Jayant RD, Pilakka-Kanthikeel S, Garcia G, Samikkannu T, Yndart A, Kaushik A, Nair M. Development of TIMP1 magnetic nanoformulation for regulation of synaptic plasticity in HIV-1 infection. Int J Nanomedicine 2016; 11:4287-98. [PMID: 27621622 PMCID: PMC5012635 DOI: 10.2147/ijn.s108329] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Although the introduction of antiretroviral therapy has reduced the prevalence of severe forms of neurocognitive disorders, human immunodeficiency virus (HIV)-1-associated neurocognitive disorders were observed in 50% of HIV-infected patients globally. The blood–brain barrier is known to be impermeable to most of antiretroviral drugs. Successful delivery of antiretroviral drugs into the brain may induce an inflammatory response, which may further induce neurotoxicity. Therefore, alternate options to antiretroviral drugs for decreasing the HIV infection and neurotoxicity may help in reducing neurocognitive impairments observed in HIV-infected patients. In this study, we explored the role of magnetic nanoparticle (MNP)-bound tissue inhibitor of metalloproteinase-1 (TIMP1) protein in reducing HIV infection levels, oxidative stress, and recovering spine density in HIV-infected SK-N-MC neuroblastoma cells. We did not observe any neuronal cytotoxicity with either the free TIMP1 or MNP-bound TIMP1 used in our study. We observed significantly reduced HIV infection in both solution phase and in MNP-bound TIMP1-exposed neuronal cells. Furthermore, we also observed significantly reduced reactive oxygen species production in both the test groups compared to the neuronal cells infected with HIV alone. To observe the effect of both soluble-phase TIMP1 and MNP-bound TIMP1 on spine density in HIV-infected neuronal cells, confocal microscopy was used. We observed significant recovery of spine density in both the test groups when compared to the cells infected with HIV alone, indicting the neuroprotective effect of TIMP1. Therefore, our results suggest that the MNP-bound TIMP1 delivery method across the blood–brain barrier can be used for reducing HIV infectivity in brain tissue and neuronal toxicity in HIV-infected patients.
Collapse
Affiliation(s)
- Venkata Subba Rao Atluri
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Rahul Dev Jayant
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Sudheesh Pilakka-Kanthikeel
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Gabriella Garcia
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Thangavel Samikkannu
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Adriana Yndart
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Ajeet Kaushik
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Madhavan Nair
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
62
|
Nair M, Jayant RD, Kaushik A, Sagar V. Getting into the brain: Potential of nanotechnology in the management of NeuroAIDS. Adv Drug Deliv Rev 2016; 103:202-217. [PMID: 26944096 PMCID: PMC4935582 DOI: 10.1016/j.addr.2016.02.008] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/18/2022]
Abstract
In spite of significant advances in antiretroviral (ARV) therapy, the elimination of human immunodeficiency virus (HIV) reservoirs from the periphery and the central nervous system (CNS) remains a formidable task. The incapability of ARV to go across the blood-brain barrier (BBB) after systemic administration makes the brain one of the dominant HIV reservoirs. Thus, screening, monitoring, and elimination of HIV reservoirs from the brain remain a clinically daunting and key task. The practice and investigation of nanomedicine possesses potentials for therapeutics against neuroAIDS. This review highlights the advancements in nanoscience and nanotechnology to design and develop specific size therapeutic cargo for efficient navigation across BBB so as to recognize and eradicate HIV brain reservoirs. Different navigation and drug release strategies, their biocompatibility and efficacy with related challenges and future prospects are also discussed. This review would be an excellent platform to understand nano-enable multidisciplinary research to formulate efficient nanomedicine for the management of neuroAIDS.
Collapse
Key Words
- Anti-retroviral (ARV) therapy
- Blood–brain barrier (BBB)
- Bradykinin (PubChem CID: 439,201)
- CNS drug delivery
- Enfuvirtide (PubChem CID: 16,130,199), Lamivudine & Zidovudine (PubChem CID: 160,352)
- Ferrous oxide or iron (II) oxide (PubChem CID: 14,945)
- Foscarnet sodium (PubChem CID: 44,561)
- HIV monitoring
- HIV-1
- Magnetic nanoparticle
- Mannitol (PubChem CID: 6251)
- Nanotechnology
- Neopterin (PubChem CID: 4455)
- NeuroAIDS
- Pluronic-P85 (PubChem CID: 24,751)
- Saquinavir mesylate (PubChem CID: 60,934)
- Tenofovir disoproxil fumarate (PubChem CID: 6,398,764)
Collapse
Affiliation(s)
- Madhavan Nair
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Rahul Dev Jayant
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Ajeet Kaushik
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Vidya Sagar
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
63
|
Sagar V, Atluri VSR, Tomitaka A, Shah P, Nagasetti A, Pilakka-Kanthikeel S, El-Hage N, McGoron A, Takemura Y, Nair M. Coupling of transient near infrared photonic with magnetic nanoparticle for potential dissipation-free biomedical application in brain. Sci Rep 2016; 6:29792. [PMID: 27465276 PMCID: PMC4964614 DOI: 10.1038/srep29792] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/21/2016] [Indexed: 01/05/2023] Open
Abstract
Combined treatment strategies based on magnetic nanoparticles (MNPs) with near infrared ray (NIR) biophotonic possess tremendous potential for non-invasive therapeutic approach. Nonetheless, investigations in this direction have been limited to peripheral body region and little is known about the potential biomedical application of this approach for brain. Here we report that transient NIR exposure is dissipation-free and has no adverse effect on the viability and plasticity of major brain cells in the presence or absence superparamagnetic nanoparticles. The 808 nm NIR laser module with thermocouple was employed for functional studies upon NIR exposure to brain cells. Magnetic nanoparticles were characterized using transmission electron microscopy (TEM), X-ray diffraction (XRD), dynamic laser scattering (DLS), and vibrating sample magnetometer (VSM). Brain cells viability and plasticity were analyzed using electric cell-substrate impedance sensing system, cytotoxicity evaluation, and confocal microscopy. When efficacious non-invasive photobiomodulation and neuro-therapeutical targeting and monitoring to brain remain a formidable task, the discovery of this dissipation-free, transient NIR photonic approach for brain cells possesses remarkable potential to add new dimension.
Collapse
Affiliation(s)
- Vidya Sagar
- Center for Personalized Nanomedicine/Institute of Neuroimmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - V. S. R. Atluri
- Center for Personalized Nanomedicine/Institute of Neuroimmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - A. Tomitaka
- Center for Personalized Nanomedicine/Institute of Neuroimmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - P. Shah
- Department of Biomedical engineering, College of Engineering and Computing, Florida International University, Miami, 33174 Florida, USA
| | - A. Nagasetti
- Department of Biomedical engineering, College of Engineering and Computing, Florida International University, Miami, 33174 Florida, USA
| | - S. Pilakka-Kanthikeel
- Center for Personalized Nanomedicine/Institute of Neuroimmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - N. El-Hage
- Center for Personalized Nanomedicine/Institute of Neuroimmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - A. McGoron
- Department of Biomedical engineering, College of Engineering and Computing, Florida International University, Miami, 33174 Florida, USA
| | - Y. Takemura
- Department of Electrical and Computer Engineering, Yokohama National University, Yokohama 240-8501, Japan
| | - M. Nair
- Center for Personalized Nanomedicine/Institute of Neuroimmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| |
Collapse
|
64
|
Aparicio-Blanco J, Martín-Sabroso C, Torres-Suárez AI. In vitro screening of nanomedicines through the blood brain barrier: A critical review. Biomaterials 2016; 103:229-255. [PMID: 27392291 DOI: 10.1016/j.biomaterials.2016.06.051] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier accounts for the high attrition rate of the treatments of most brain disorders, which therefore remain one of the greatest health-care challenges of the twenty first century. Against this background of hindrance to brain delivery, nanomedicine takes advantage of the assembly at the nanoscale of available biomaterials to provide a delivery platform with potential to raising brain levels of either imaging or therapeutic agents. Nevertheless, to prevent later failure due to ineffective drug levels at the target site, researchers have been endeavoring to develop a battery of in vitro screening procedures that can predict earlier in the drug discovery process the ability of these cutting-edge drug delivery platforms to cross the blood-brain barrier for biomedical purposes. This review provides an in-depth analysis of the currently available in vitro blood-brain barrier models (both cell-based and non-cell-based) with the focus on their suitability for understanding the biological brain distribution of forthcoming nanomedicines. The relationship between experimental factors and underlying physiological assumptions that would ultimately lead to a more predictive capacity of their in vivo performance, and those methods already assayed for the evaluation of the brain distribution of nanomedicines are comprehensively discussed.
Collapse
Affiliation(s)
- Juan Aparicio-Blanco
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain
| | - Cristina Martín-Sabroso
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain
| | - Ana-Isabel Torres-Suárez
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain; University Institute of Industrial Pharmacy, Complutense University, 28040, Madrid, Spain.
| |
Collapse
|
65
|
Sagar V, Atluri VSR, Pilakka-Kanthikeel S, Nair M. Magnetic nanotherapeutics for dysregulated synaptic plasticity during neuroAIDS and drug abuse. Mol Brain 2016; 9:57. [PMID: 27216740 PMCID: PMC4878083 DOI: 10.1186/s13041-016-0236-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 05/06/2016] [Indexed: 01/02/2023] Open
Abstract
The human immunodeficiency virus (HIV) is a neurotropic virus. It induces neurotoxicity and subsequent brain pathologies in different brain cells. Addiction to recreational drugs remarkably affects the initiation of HIV infections and expedites the progression of acquired immunodeficiency syndrome (AIDS) associated neuropathogenesis. Symptoms of HIV-associated neurocognitive disorders (HAND) are noticed in many AIDS patients. At least 50 % of HIV diagnosed cases show one or other kind of neuropathological signs or symptoms during different stages of disease progression. In the same line, mild to severe neurological alterations are seen in at least 80 % autopsies of AIDS patients. Neurological illnesses weaken the connections between neurons causing significant altercations in synaptic plasticity. Synaptic plasticity alterations during HIV infection and recreational drug abuse are mediated by complex cellular phenomena involving changes in gene expression and subsequent loss of dendritic and spine morphology and physiology. New treatment strategies with ability to deliver drugs across blood-brain barrier (BBB) are being intensively investigated. In this context, magnetic nanoparticles (MNPs) based nanoformulations have shown significant potential for target specificity, drug delivery, drug release, and bioavailability of desired amount of drugs in non-invasive brain targeting. MNPs-based potential therapies to promote neuronal plasticity during HIV infection and recreational drug abuse are being developed.
Collapse
Affiliation(s)
- Vidya Sagar
- Department of Immunology, Center for Personalized Nanomedicine/Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Venkata Subba Rao Atluri
- Department of Immunology, Center for Personalized Nanomedicine/Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Sudheesh Pilakka-Kanthikeel
- Department of Immunology, Center for Personalized Nanomedicine/Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Madhavan Nair
- Department of Immunology, Center for Personalized Nanomedicine/Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA.
| |
Collapse
|
66
|
Bertrand L, Nair M, Toborek M. Solving the Blood-Brain Barrier Challenge for the Effective Treatment of HIV Replication in the Central Nervous System. Curr Pharm Des 2016; 22:5477-5486. [PMID: 27464720 PMCID: PMC7219022 DOI: 10.2174/1381612822666160726113001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
Abstract
Recent decades mark a great progress in the treatment of HIV infection. What was once a deadly disease is now a chronic infection. However, HIV-infected patients are prone to develop comorbidities, which severely affect their daily functions. For example, a large population of patients develop a variety of neurological and cognitive complications, called HIV associated neurological disorders (HAND). Despite efficient repression of viral replication in the periphery, evidence shows that the virus can remain active in the central nervous system (CNS). This low level of replication is believed to result in a progression of neurocognitive dysfunction in infected individuals. Insufficient viral inhibition in the brain results from the inability of several treatment drugs in crossing the blood-brain barrier (BBB) and reaching therapeutic concentrations in the CNS. The current manuscript discusses several strategies that are being developed to enable therapeutics to cross the BBB, including bypassing BBB, inhibition of efflux transporters, the use of active transporters present at the BBB, and nanotechnology. The increased concentration of therapeutics in the CNS is desirable to prevent viral replication; however, potential side effects of anti-retroviral drugs need also to be taken into consideration.
Collapse
Affiliation(s)
| | | | - Michal Toborek
- University of Miami. Miller School of Medicine, Department of Biochemistry and Molecular Biology, 1011 NW 15th Street, Miami, FL 33136, USA.
| |
Collapse
|
67
|
Busquets MA, Espargaró A, Sabaté R, Estelrich J. Magnetic Nanoparticles Cross the Blood-Brain Barrier: When Physics Rises to a Challenge. NANOMATERIALS (BASEL, SWITZERLAND) 2015; 5:2231-2248. [PMID: 28347118 PMCID: PMC5304810 DOI: 10.3390/nano5042231] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 11/25/2015] [Accepted: 12/08/2015] [Indexed: 12/21/2022]
Abstract
The blood-brain barrier is a physical and physiological barrier that protects the brain from toxic substances within the bloodstream and helps maintain brain homeostasis. It also represents the main obstacle in the treatment of many diseases of the central nervous system. Among the different approaches employed to overcome this barrier, the use of nanoparticles as a tool to enhance delivery of therapeutic molecules to the brain is particularly promising. There is special interest in the use of magnetic nanoparticles, as their physical characteristics endow them with additional potentially useful properties. Following systemic administration, a magnetic field applied externally can mediate the capacity of magnetic nanoparticles to permeate the blood-brain barrier. Meanwhile, thermal energy released by magnetic nanoparticles under the influence of radiofrequency radiation can modulate blood-brain barrier integrity, increasing its permeability. In this review, we present the strategies that use magnetic nanoparticles, specifically iron oxide nanoparticles, to enhance drug delivery to the brain.
Collapse
Affiliation(s)
- Maria Antònia Busquets
- Department of Physical Chemistry, Faculty of Pharmacy, University of Barcelona and Institute of Nanoscience and Nanotechnology (IN2UB), Avda. Joan XXIII, 08028 Barcelona, Spain.
| | - Alba Espargaró
- Department of Physical Chemistry, Faculty of Pharmacy, University of Barcelona and Institute of Nanoscience and Nanotechnology (IN2UB), Avda. Joan XXIII, 08028 Barcelona, Spain.
| | - Raimon Sabaté
- Department of Physical Chemistry, Faculty of Pharmacy, University of Barcelona and Institute of Nanoscience and Nanotechnology (IN2UB), Avda. Joan XXIII, 08028 Barcelona, Spain.
| | - Joan Estelrich
- Department of Physical Chemistry, Faculty of Pharmacy, University of Barcelona and Institute of Nanoscience and Nanotechnology (IN2UB), Avda. Joan XXIII, 08028 Barcelona, Spain.
| |
Collapse
|
68
|
Raymond AD, Diaz P, Chevelon S, Agudelo M, Yndart-Arias A, Ding H, Kaushik A, Jayant RD, Nikkhah-Moshaie R, Roy U, Pilakka-Kanthikeel S, Nair MP. Microglia-derived HIV Nef+ exosome impairment of the blood-brain barrier is treatable by nanomedicine-based delivery of Nef peptides. J Neurovirol 2015; 22:129-39. [PMID: 26631079 DOI: 10.1007/s13365-015-0397-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 10/13/2015] [Accepted: 10/14/2015] [Indexed: 10/22/2022]
Abstract
The negative factor (Nef) of human immunodeficiency virus (HIV) is an accessory protein that is thought to be integral to HIV-associated immune- and neuroimmune pathogenesis. Here, we show that nef-transfected microglia-released Nef+ exosome (exNef) disrupts the apical blood-brain barrier (BBB) and that only nef-transfected microglia release Nef in exosomes. nef-gfp-transduced neurons and astrocytes release exosomes but did not release exNef in the extracellular space. Apical administration of exNef derived from nef-transfected 293T cells reduced transendothelial electrical resistance (TEER) and increased permeability of the BBB. Microglia-derived exNef applied to either the apical/basal BBB significantly reduced expression of the tight junction protein, ZO-1, suggesting a mechanism of exNef-mediated neuropathogenesis. Microglia exposed to exNef release elevated levels of Toll-like receptor-induced cytokines and chemokines IL-12, IL-8, IL-6, RANTES, and IL-17A. Magnetic nanoparticle delivery of Nef peptides containing the Nef myrisolation site across an in vitro BBB ultimately reduced nef-transfected microglia release of Nef exosomes and prevented the loss of BBB integrity and permeability as measured by TEER and dextran-FITC transport studies, respectively. Overall, we show that exNef is released from nef-gfp-transfected microglia; exNef disrupts integrity and permeability, and tight junctions of the BBB, and induces microglial cytokine/chemokine secretion. These exNef-mediated effects were significantly restricted by Nef peptides. Taken together, this study provides preliminary evidence of the role of exNef in HIV neuroimmune pathogenesis and the feasibility of a nanomedicine-based therapeutics targeting exNef to treat HIV-associated neuropathogenesis.
Collapse
Affiliation(s)
- A D Raymond
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA. .,Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - P Diaz
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - S Chevelon
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - M Agudelo
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - A Yndart-Arias
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.,Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - H Ding
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - A Kaushik
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - R Dev Jayant
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - R Nikkhah-Moshaie
- Department of Mechanical and Materials Engineering, College of Engineering and Computing, Florida International University, 10555 W. Flagler St., Miami, FL, 33174, USA
| | - U Roy
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - S Pilakka-Kanthikeel
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - M P Nair
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.,Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| |
Collapse
|
69
|
Sagar V, Pilakka-Kanthikeel S, Atluri VSR, Ding H, Arias AY, Jayant RD, Kaushik A, Nair M. Therapeutical Neurotargeting via Magnetic Nanocarrier: Implications to Opiate-Induced Neuropathogenesis and NeuroAIDS. J Biomed Nanotechnol 2015; 11:1722-33. [PMID: 26502636 DOI: 10.1166/jbn.2015.2108] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Magnetite (Fe3O4) is the most commonly and extensively explored magnetic nanoparticles (MNPs) for drug-targeting and imaging in the field of biomedicine. Nevertheless, its potential application as safe and effective drug-carrier for CNS (Central Nervous System) anomalies is very limited. Previous studies have shown an entangled epidemic of opioid use and HIV infection and increased neuropathogenesis. Opiate such as morphine, heroine, etc. are used frequently as recreational drugs. Existing treatments to alleviate the action of opioid are less effective at CNS level due to impermeability of therapeutic molecules across brain barriers. Thus, development of an advanced nanomedicine based approach may pave the way for better treatment strategies. We herein report magnetic nanoformulation of a highly selective and potent morphine antagonist, CTOP (D-Pen-Cys-Tyr-DTrp-Orn-Thr-Pen-Thr-NH2), which is impenetrable to the brain. MNPs, synthesized in size range from 25 to 40 nm, were characterized by Transmission electron microscopy and assembly of MNPs-CTOP nanoformulations were confirmed by FTIR spectroscopy and fluorescent detection. Flow-cytometry analysis showed that biological efficacy of this nanoformulation in prevention of morphine induced apoptosis in peripheral blood mononuclear cells remains equivalent to that of free CTOP. Similarly, confocal microscopy reveals comparable efficacy of free and MNPs bound CTOP in protecting modulation of neuronal dendrite and spine morphology during morphine exposure and morphine-treated HIV infection. Further, typical transmigration assay showed increased translocation of MNPs across in vitro blood-brain barrier upon exposure of external magnetic force where barrier integrity remains unaltered. Thus, the developed nanoformulation could be effective in targeting brain by application of external magnetic force to treat morphine addiction in HIV patients.
Collapse
|
70
|
Abstract
Brain capillary endothelial cells denote the blood-brain barrier (BBB), and conjugation of nanoparticles with antibodies that target molecules expressed by these endothelial cells may facilitate their uptake and transport into the brain. Magnetic nanoparticles can be encapsulated in liposomes and carry large molecules with therapeutic potential, for example, siRNA, cDNA and polypeptides. An additional approach to enhance the transport of magnetic nanoparticles across the BBB is the application of extracranially applied magnetic force. Stepwise targeting of magnetic nanoparticles to brain capillary endothelial cells followed by transport through the BBB using magnetic force may prove a novel mechanism for targeted therapy of macromolecules to the brain.
Collapse
|
71
|
Roy U, Ding H, Pilakka-Kanthikeel S, Raymond AD, Atluri V, Yndart A, Kaftanovskaya EM, Batrakova E, Agudelo M, Nair M. Preparation and characterization of anti-HIV nanodrug targeted to microfold cell of gut-associated lymphoid tissue. Int J Nanomedicine 2015; 10:5819-35. [PMID: 26425084 PMCID: PMC4583111 DOI: 10.2147/ijn.s68348] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The human immunodeficiency virus 1 (HIV-1) still remains one of the leading life-threatening diseases in the world. The introduction of highly active antiretroviral therapy has significantly reduced disease morbidity and mortality. However, most of the drugs have variable penetrance into viral reservoir sites, including gut-associated lymphoid tissue (GALT). Being the largest lymphoid organ, GALT plays a key role in early HIV infection and host–pathogen interaction. Many different treatment options have been proposed to eradicate the virus from GALT. However, it becomes difficult to deliver traditional drugs to the GALT because of its complex physiology. In this regard, we developed a polymer-based Pluronic nanocarrier containing anti-HIV drug called efavirenz (EFV) targeting Microfold cells (M-cells) in the GALT. M-cells are specialized epithelial cells that are predominantly present in the GALT. In this work, we have exploited this paracellular transport property of M-cells for targeted delivery of Pluronic nanocarrier tagged EFV, bioconjugated with anti-M-cell-specific antibodies to the GALT (nanodrug). Preliminary characterization showed that the nanodrug (EFV-F12-COOH) is of 140 nm size with 0.3 polydispersion index, and the zeta potential of the particles was −19.38±2.2 mV. Further, drug dissolution study has shown a significantly improved sustained release over free drugs. Binding potential of nanodrug with M-cell was also confirmed with fluorescence microscopy and in vitro uptake and release studies. The anti-HIV activity of the nanodrug was also significantly higher compared to that of free drug. This novel formulation was able to show sustained release of EFV and inhibit the HIV-1 infection in the GALT compared to the free drug. The present study has potential for our in vivo targeted nanodrug delivery system by combining traditional enteric-coated capsule technique via oral administration.
Collapse
Affiliation(s)
- Upal Roy
- Center for Personalized NanoMedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Florida International University, Miami, FL, USA
| | - Hong Ding
- Center for Personalized NanoMedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Florida International University, Miami, FL, USA
| | - Sudheesh Pilakka-Kanthikeel
- Center for Personalized NanoMedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Florida International University, Miami, FL, USA
| | - Andrea D Raymond
- Center for Personalized NanoMedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Florida International University, Miami, FL, USA
| | - Venkata Atluri
- Center for Personalized NanoMedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Florida International University, Miami, FL, USA
| | - Adriana Yndart
- Center for Personalized NanoMedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Florida International University, Miami, FL, USA
| | - Elena M Kaftanovskaya
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Elena Batrakova
- UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marisela Agudelo
- Center for Personalized NanoMedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Florida International University, Miami, FL, USA
| | - Madhavan Nair
- Center for Personalized NanoMedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Florida International University, Miami, FL, USA
| |
Collapse
|
72
|
Setyawati MI, Tay CY, Docter D, Stauber RH, Leong DT. Understanding and exploiting nanoparticles' intimacy with the blood vessel and blood. Chem Soc Rev 2015; 44:8174-99. [PMID: 26239875 DOI: 10.1039/c5cs00499c] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While the blood vessel is seldom the target tissue, almost all nanomedicine will interact with blood vessels and blood at some point of time along its life cycle in the human body regardless of their intended destination. Despite its importance, many bionanotechnologists do not feature endothelial cells (ECs), the blood vessel cells, or consider blood effects in their studies. Including blood vessel cells in the study can greatly increase our understanding of the behavior of any given nanomedicine at the tissue of interest or to understand side effects that may occur in vivo. In this review, we will first describe the diversity of EC types found in the human body and their unique behaviors and possibly how these important differences can implicate nanomedicine behavior. Subsequently, we will discuss about the protein corona derived from blood with foci on the physiochemical aspects of nanoparticles (NPs) that dictate the protein corona characteristics. We would also discuss about how NPs characteristics can affect uptake by the endothelium. Subsequently, mechanisms of how NPs could cross the endothelium to access the tissue of interest. Throughout the paper, we will share some novel nanomedicine related ideas and insights that were derived from the understanding of the NPs' interaction with the ECs. This review will inspire more exciting nanotechnologies that had accounted for the complexities of the real human body.
Collapse
Affiliation(s)
- Magdiel Inggrid Setyawati
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | | | | | | | | |
Collapse
|
73
|
Atluri VSR, Hidalgo M, Samikkannu T, Kurapati KRV, Jayant RD, Sagar V, Nair MPN. Effect of human immunodeficiency virus on blood-brain barrier integrity and function: an update. Front Cell Neurosci 2015; 9:212. [PMID: 26113810 PMCID: PMC4461820 DOI: 10.3389/fncel.2015.00212] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/17/2015] [Indexed: 02/02/2023] Open
Abstract
The blood-brain barrier (BBB) is a diffusion barrier that has an important role in maintaining a precisely regulated microenvironment protecting the neural tissue from infectious agents and toxins in the circulating system. Compromised BBB integrity plays a major role in the pathogenesis of retroviral associated neurological diseases. Human Immunodeficiency Virus (HIV) infection in the Central Nervous System (CNS) is an early event even before the serodiagnosis for HIV positivity or the initiation of antiretroviral therapy (ART), resulting in neurological complications in many of the infected patients. Macrophages, microglia and astrocytes (in low levels) are the most productively/latently infected cell types within the CNS. In this brief review, we have discussed about the effect of HIV infection and viral proteins on the integrity and function of BBB, which may contribute to the progression of HIV associated neurocognitive disorders.
Collapse
Affiliation(s)
- Venkata Subba Rao Atluri
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Melissa Hidalgo
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Thangavel Samikkannu
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Kesava Rao Venkata Kurapati
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Rahul Dev Jayant
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Vidya Sagar
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| | - Madhavan P N Nair
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University Miami, FL, USA
| |
Collapse
|
74
|
Nanoparticle-mediated growth factor delivery systems: A new way to treat Alzheimer's disease. J Control Release 2015; 206:187-205. [DOI: 10.1016/j.jconrel.2015.03.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 01/03/2023]
|
75
|
Jayant RD, Atluri VSR, Agudelo M, Sagar V, Kaushik A, Nair M. Sustained-release nanoART formulation for the treatment of neuroAIDS. Int J Nanomedicine 2015; 10:1077-93. [PMID: 25709433 PMCID: PMC4327567 DOI: 10.2147/ijn.s76517] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A novel approach was developed for the coencapsulation of an anti-HIV drug (tenofovir) and a latency-breaking agent (vorinostat), using magnetically guided layer-by-layer (LbL) assembled nanocarriers for the treatment of neuroAIDS. Ultrasmall iron oxide (Fe3O4) nanoparticles (10±3 nm) were synthesized and characterized. The LbL technique was used to achieve a sustained release profile, and application of 2 bilayers ([tenofovir+dextran sulphate]2+vorinostat) to magnetic nanoparticles resulted in a 2.8 times increase in drug (tenofovir) loading and also resulted in an increase in the drug release period by 30-fold, with 100% drug release in sustained manner over a period of 5 days with the simultaneous stimulation of latent HIV expression. Nanoformulation showed a good blood-brain barrier transmigration ability (37.95%±1.5%) with good in vitro antiviral efficacy (~33% reduction of p24 level) over a period of 5 days after HIV infection in primary human astrocytes, with good cell viability (>90%). Hence, LbL arrangements of drugs on magnetic nanoparticles provides sustained release and, therefore, may improve the patient's adherence to therapy and lead to better compliance.
Collapse
Affiliation(s)
- Rahul Dev Jayant
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Venkata S R Atluri
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Marisela Agudelo
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Vidya Sagar
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Ajeet Kaushik
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Madhavan Nair
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
76
|
Wang X, Yu X, Vaughan W, Liu M, Guan Y. Novel drug-delivery approaches to the blood-brain barrier. Neurosci Bull 2015; 31:257-64. [PMID: 25595370 DOI: 10.1007/s12264-014-1498-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 07/21/2014] [Indexed: 11/30/2022] Open
Abstract
The blood-brain barrier (BBB) maintains homeostasis by blocking toxic molecules from the circulation, but drugs are blocked at the same time. When the dose is increased to enhance the drug concentration in the central nervous system, there are side-effects on peripheral organs. In recent years, genetic therapeutic agents and small molecules have been used in various strategies to penetrate the BBB while minimizing the damage to systemic organs. In this review, we describe several representative methods to circumvent or cross the BBB, including chemical and physical strategies.
Collapse
Affiliation(s)
- Xiaoqing Wang
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | | | | | | | | |
Collapse
|
77
|
Rao PSS, Yallapu MM, Sari Y, Fisher PB, Kumar S. Designing Novel Nanoformulations Targeting Glutamate Transporter Excitatory Amino Acid Transporter 2: Implications in Treating Drug Addiction. JOURNAL OF PERSONALIZED NANOMEDICINE 2015; 1:3-9. [PMID: 26635971 PMCID: PMC4666545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Chronic drug abuse is associated with elevated extracellular glutamate concentration in the brain reward regions. Deficit of glutamate clearance has been identified as a contributing factor that leads to enhanced glutamate concentration following extended drug abuse. Importantly, normalization of glutamate level through induction of glutamate transporter 1 (GLT1)/ excitatory amino acid transporter 2 (EAAT2) expression has been described in several in vivo studies. GLT1 upregulators including ceftriaxone, a beta-lactam antibiotic, have been effective in attenuating drug-seeking and drug-consumption behavior in rodent models. However, potential obstacles toward clinical translation of GLT1 (EAAT2) upregulators as treatment for drug addiction might include poor gastrointestinal absorption, serious peripheral adverse effects, and/or suboptimal CNS concentrations. Given the growing success of nanotechnology in targeting CNS ailments, nanoformulating known GLT1 (EAAT2) upregulators for selective uptake across the blood brain barrier presents an ideal therapeutic approach for treating drug addiction. In this review, we summarize the results obtained with promising GLT1 (EAAT2) inducing compounds in animal models recapitulating drug addiction. Additionally, the various nanoformulations that can be employed for selectively increasing the CNS bioavailability of GLT1 (EAAT2) upregulators are discussed. Finally, the applicability of GLT1 (EAAT2) induction via central delivery of drug-loaded nanoformulations is described.
Collapse
Affiliation(s)
- PSS Rao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA,Corresponding authors: (P.S.S.R), Tel: 901-448-7146. (S.K), Tel: 901-448-7157
| | - Murali M. Yallapu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA
| | - Paul B. Fisher
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA,Corresponding authors: (P.S.S.R), Tel: 901-448-7146. (S.K), Tel: 901-448-7157
| |
Collapse
|
78
|
Wang X, Zhang H, Jing H, Cui L. Highly Efficient Labeling of Human Lung Cancer Cells Using Cationic Poly-l-lysine-Assisted Magnetic Iron Oxide Nanoparticles. NANO-MICRO LETTERS 2015; 7:374-384. [PMID: 30464985 PMCID: PMC6223914 DOI: 10.1007/s40820-015-0053-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/23/2015] [Indexed: 05/18/2023]
Abstract
Cell labeling with magnetic iron oxide nanoparticles (IONPs) is increasingly a routine approach in the cell-based cancer treatment. However, cell labeling with magnetic IONPs and their leading effects on the biological properties of human lung carcinoma cells remain scarcely reported. Therefore, in the present study the magnetic γ-Fe2O3 nanoparticles (MNPs) were firstly synthesized and surface-modified with cationic poly-l-lysine (PLL) to construct the PLL-MNPs, which were then used to magnetically label human A549 lung cancer cells. Cell viability and proliferation were evaluated with propidium iodide/fluorescein diacetate double staining and standard 3-(4,5-dimethylthiazol-2-diphenyl-tetrazolium) bromide assay, and the cytoskeleton was immunocytochemically stained. The cell cycle of the PLL-MNP-labeled A549 lung cancer cells was analyzed using flow cytometry. Apoptotic cells were fluorescently analyzed with nuclear-specific staining after the PLL-MNP labeling. The results showed that the constructed PLL-MNPs efficiently magnetically labeled A549 lung cancer cells and that, at low concentrations, labeling did not affect cellular viability, proliferation capability, cell cycle, and apoptosis. Furthermore, the cytoskeleton in the treated cells was detected intact in comparison with the untreated counterparts. However, the results also showed that at high concentration (400 µg mL-1), the PLL-MNPs would slightly impair cell viability, proliferation, cell cycle, and apoptosis and disrupt the cytoskeleton in the treated A549 lung cancer cells. Therefore, the present results indicated that the PLL-MNPs at adequate concentrations can be efficiently used for labeling A549 lung cancer cells and could be considered as a feasible approach for magnetic targeted anti-cancer drug/gene delivery, targeted diagnosis, and therapy in lung cancer treatment.
Collapse
Affiliation(s)
- Xueqin Wang
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001 Henan People’s Republic of China
| | - Huiru Zhang
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001 Henan People’s Republic of China
| | - Hongjuan Jing
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001 Henan People’s Republic of China
| | - Liuqing Cui
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001 Henan People’s Republic of China
| |
Collapse
|
79
|
Nanoparticles and the blood-brain barrier: advancing from in-vitro models towards therapeutic significance. Pharm Res 2014; 32:1161-85. [PMID: 25446769 DOI: 10.1007/s11095-014-1545-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/06/2014] [Indexed: 01/12/2023]
Abstract
The blood-brain barrier is a unique cell-based restrictive barrier that prevents the entry of many substances, including most therapeutics, into the central nervous system. A wide range of nanoparticulate delivery systems have been investigated with the aim of targeting therapeutics (drugs, nucleic acids, proteins) to the brain following administration by various routes. This review provides a comprehensive description of the design and formulation of these nanoparticles including the rationale behind individual approaches. In addition, the ability of currently available in-vitro BBB models to accurately predict the in-vivo performance of targeted nanoparticles is critically assessed.
Collapse
|
80
|
Wang X, Wang L, Tan X, Zhang H, Sun G. Construction of doxorubicin-loading magnetic nanocarriers for assaying apoptosis of glioblastoma cells. J Colloid Interface Sci 2014; 436:267-75. [DOI: 10.1016/j.jcis.2014.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 12/12/2022]
|
81
|
Husseini GA, Pitt WG, Martins AM. Ultrasonically triggered drug delivery: breaking the barrier. Colloids Surf B Biointerfaces 2014; 123:364-86. [PMID: 25454759 DOI: 10.1016/j.colsurfb.2014.07.051] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 12/21/2022]
Abstract
The adverse side-effects of chemotherapy can be minimized by delivering the therapeutics in time and space to only the desired target site. Ultrasound offers one fairly non-invasive method of accomplishing such precise delivery because its energy can disrupt nanosized containers that are designed to sequester the drug until the ultrasonic event. Such containers include micelles, liposomes and solid nanoparticles. Conventional micelles and liposomes are less acoustically sensitive to ultrasound because the strongest forces associated with ultrasound are generated by gas-liquid interfaces, which both of these conventional constructs lack. Acoustically activated carriers often incorporate a gas phase, either actively as preformed bubbles, or passively such as taking advantage of dissolved gasses that form bubbles upon insonation. Newer concepts include using liquids that form gas when insonated. This review focuses on the ultrasonically activated delivery of therapeutics from micelles, liposomes and solid particles. In vitro and in vivo results are summarized and discussed. Novel structural concepts from micelles and liposomes are presented. Mechanisms of ultrasonically activated release are discussed. The future of ultrasound in drug delivery is envisioned.
Collapse
Affiliation(s)
| | | | - Ana M Martins
- American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
82
|
Kaushik A, Jayant RD, Sagar V, Nair M. The potential of magneto-electric nanocarriers for drug delivery. Expert Opin Drug Deliv 2014; 11:1635-46. [PMID: 24986772 PMCID: PMC4556109 DOI: 10.1517/17425247.2014.933803] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION The development and design of personalized nanomedicine for better health quality is receiving great attention. In order to deliver and release a therapeutic concentration at the target site, novel nanocarriers (NCs) were designed, for example, magneto-electric (ME) which possess ideal properties of high drug loading, site-specificity and precise on-demand controlled drug delivery. AREAS COVERED This review explores the potential of ME-NCs for on-demand and site-specific drug delivery and release for personalized therapeutics. The main features including effect of magnetism, improvement in drug loading, drug transport across blood-brain barriers and on-demand controlled release are also discussed. The future directions and possible impacts on upcoming nanomedicine are highlighted. EXPERT OPINION Numerous reports suggest that there is an urgent need to explore novel NC formulations for safe and targeted drug delivery and release at specific disease sites. The challenges of formulation lie in the development of NCs that improve biocompatibility and surface modifications for optimum drug loading/preservation/transmigration and tailoring of electrical-magnetic properties for on-demand drug release. Thus, the development of novel NCs is anticipated to overcome the problems of targeted delivery of therapeutic agents with desired precision that may lead to better patient compliance.
Collapse
Affiliation(s)
- Ajeet Kaushik
- Post-doctoral Associate, Florida International University, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Center of Personalized Nanomedicine, Department of Immunology, Miami, FL-33199, USA
| | - Rahul Dev Jayant
- Post-doctoral Associate, Florida International University, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Center of Personalized Nanomedicine, Department of Immunology, Miami, FL-33199, USA
| | - Vidya Sagar
- Post-doctoral Associate, Florida International University, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Center of Personalized Nanomedicine, Department of Immunology, Miami, FL-33199, USA
| | - Madhavan Nair
- Post-doctoral Associate, Florida International University, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Center of Personalized Nanomedicine, Department of Immunology, Miami, FL-33199, USA
- Professor and Chair, Director, Associate Dean of Bio-Medical Research, Florida International University, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Department of Immunology, AHC-1, 418A, 11200 SW 8th St, University Park, Miami, FL 33199, USA
| |
Collapse
|
83
|
Upadhyay RK. Drug delivery systems, CNS protection, and the blood brain barrier. BIOMED RESEARCH INTERNATIONAL 2014; 2014:869269. [PMID: 25136634 PMCID: PMC4127280 DOI: 10.1155/2014/869269] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/31/2014] [Accepted: 06/05/2014] [Indexed: 12/12/2022]
Abstract
Present review highlights various drug delivery systems used for delivery of pharmaceutical agents mainly antibiotics, antineoplastic agents, neuropeptides, and other therapeutic substances through the endothelial capillaries (BBB) for CNS therapeutics. In addition, the use of ultrasound in delivery of therapeutic agents/biomolecules such as proline rich peptides, prodrugs, radiopharmaceuticals, proteins, immunoglobulins, and chimeric peptides to the target sites in deep tissue locations inside tumor sites of brain has been explained. In addition, therapeutic applications of various types of nanoparticles such as chitosan based nanomers, dendrimers, carbon nanotubes, niosomes, beta cyclodextrin carriers, cholesterol mediated cationic solid lipid nanoparticles, colloidal drug carriers, liposomes, and micelles have been discussed with their recent advancements. Emphasis has been given on the need of physiological and therapeutic optimization of existing drug delivery methods and their carriers to deliver therapeutic amount of drug into the brain for treatment of various neurological diseases and disorders. Further, strong recommendations are being made to develop nanosized drug carriers/vehicles and noninvasive therapeutic alternatives of conventional methods for better therapeutics of CNS related diseases. Hence, there is an urgent need to design nontoxic biocompatible drugs and develop noninvasive delivery methods to check posttreatment clinical fatalities in neuropatients which occur due to existing highly toxic invasive drugs and treatment methods.
Collapse
Affiliation(s)
- Ravi Kant Upadhyay
- Department of Zoology, DDU Gorakhpur University, Gorakhpur 273009, India
| |
Collapse
|
84
|
Kuo YC, Chou PR. Neuroprotection against degeneration of sk-N-mc cells using neuron growth factor-encapsulated liposomes with surface cereport and transferrin. J Pharm Sci 2014; 103:2484-97. [PMID: 25041794 DOI: 10.1002/jps.24081] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/14/2014] [Accepted: 06/16/2014] [Indexed: 11/06/2022]
Abstract
Liposomes with Cereport (RMP-7) and transferrin (Tf) (RMP-7/Tf/liposomes) were employed to target the blood-brain barrier (BBB) and to inhibit the degeneration of neurons insulted with fibrillar β-amyloid peptide 1-42 (Aβ1-42). Neuron growth factor (NGF)-encapsulated RMP-7/Tf/liposomes (RMP-7/Tf/NGF-liposomes) were used to permeate a monolayer of human brain-microvascular endothelial cells (HBMECs) regulated by human astrocytes (HAs) and to treat Aβ1-42 -attacked SK-N-MC cells. An increase in RMT-7 concentration increased the particle size, zeta potential, propidium iodide (PI) permeability, and NGF permeability, but decreased the cross-linking efficiency of RMT-7, viability of HBMECs and HAs, and transendothelial electrical resistance (TEER). In addition, an increase in Tf concentration enhanced the particle size, viability of HBMECs, HAs, and SK-N-MC cells, PI permeability, and NGF permeability, but reduced the zeta potential, cross-linking efficiency of RMT-7 and Tf, and TEER. RMP-7/Tf/NGF-liposomes can transport NGF across the BBB and improve the neuroprotection for Alzheimer's disease therapy in preclinical trials.
Collapse
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, 62102, Republic of China
| | | |
Collapse
|
85
|
|
86
|
Atluri VSR, Pilakka-Kanthikeel S, Samikkannu T, Sagar V, Kurapati KRV, Saxena SK, Yndart A, Raymond A, Ding H, Hernandez O, Nair MPN. Vorinostat positively regulates synaptic plasticity genes expression and spine density in HIV infected neurons: role of nicotine in progression of HIV-associated neurocognitive disorder. Mol Brain 2014; 7:37. [PMID: 24886748 PMCID: PMC4040511 DOI: 10.1186/1756-6606-7-37] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/28/2014] [Indexed: 11/24/2022] Open
Abstract
Background HIV-associated neurocognitive disorder (HAND) is characterized by development of cognitive, behavioral and motor abnormalities, and occurs in approximately 50% of HIV infected individuals. In the United States, the prevalence of cigarette smoking ranges from 35-70% in HIV-infected individuals compared to 20% in general population. Cognitive impairment in heavy cigarette smokers has been well reported. However, the synergistic effects of nicotine and HIV infection and the underlying mechanisms in the development of HAND are unknown. Results In this study, we explored the role of nicotine in the progression of HAND using SK-N-MC, a neuronal cell line. SK-N-MC cells were infected with HIV-1 in the presence or absence of nicotine for 7 days. We observed significant increase in HIV infectivity in SK-N-MC treated with nicotine compared to untreated HIV-infected neuronal cells. HIV and nicotine synergize to significantly dysregulate the expression of synaptic plasticity genes and spine density; with a concomitant increase of HDAC2 levels in SK-N-MC cells. In addition, inhibition of HDAC2 up-regulation with the use of vorinostat resulted in HIV latency breakdown and recovery of synaptic plasticity genes expression and spine density in nicotine/HIV alone and in co-treated SK-N-MC cells. Furthermore, increased eIF2 alpha phosphorylation, which negatively regulates eukaryotic translational process, was observed in HIV alone and in co-treatment with nicotine compared to untreated control and nicotine alone treated SK-N-MC cells. Conclusions These results suggest that nicotine and HIV synergize to negatively regulate the synaptic plasticity gene expression and spine density and this may contribute to the increased risk of HAND in HIV infected smokers. Apart from disrupting latency, vorinostat may be a useful therapeutic to inhibit the negative regulatory effects on synaptic plasticity in HIV infected nicotine abusers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Madhavan P N Nair
- Department of Immunology, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL -33199, USA.
| |
Collapse
|
87
|
Yang KM, Cho HI, Choi HJ, Piao Y. Synthesis of water well-dispersed PEGylated iron oxide nanoparticles for MR/optical lymph node imaging. J Mater Chem B 2014; 2:3355-3364. [PMID: 32261598 DOI: 10.1039/c4tb00084f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We reported the synthesis of highly water-stable iron oxide nanoparticles by a simple one-pot reaction. Non-toxic polyethylene glycol MW 600 (PEG) acted as a solvent, capping agent and reducing agent in the synthesis of iron oxide nanoparticles. As a result of the synthesis, PEGylated small-size (4.2 ± 0.39 nm average diameter and 7.2 ± 1.9 nm hydrodynamic sizes measuring by DLS) iron oxide nanoparticles (USPIO) were obtained, which show great colloidal stability in water and tolerate high salt concentration (0.75 M sodium chloride) and a wide pH range of 4 to 12. Oxidation of PEG was observed during the synthesis of iron oxide nanoparticles, which makes USPIO easy to functionalize with other molecules. Functionalization of the USPIO surface with fluorescein isothiocyanate (FITC) was conducted for investigating the possibility for multimodal imaging. Also the cytotoxicity test and lymph node imaging were performed by using the FITC labelled USPIO (FITC@USPIO). According to these results, the stable water dispersed USPIO and FITC@USPIO are expected to apply for multimodal in vivo imaging.
Collapse
Affiliation(s)
- Kyung Mo Yang
- Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 151-742, Republic of Korea.
| | | | | | | |
Collapse
|