1
|
Li XY, Zhou XD, Hu JM. Peptides in the detection of metal ions. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:6589-6598. [PMID: 39269217 DOI: 10.1039/d4ay01232a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
By means of their specific interactions with different metal ions, naturally occurring proteins control structures and functions of many biological processes and functions in organisms. In view of natural metallopeptides, scientists have proposed artificial peptides which coordinate with metal ions through their functional groups either for introducing a special reactivity or for constructing various sensors. However, the design of new peptide ligands requires a deep understanding of the structures, assembly properties, and dynamic behaviors of such peptides. This review briefly describes detection strategies of metal ions via coordination to the binding sites in peptides. The principles and functions of sensing systems are described as well. We also highlight some examples of a metal-induced peptide self-assembly with relevance to biotechnology applications.
Collapse
Affiliation(s)
- Xin-Yi Li
- Core Facility of Wuhan University, Wuhan University, Wuhan 430072, PR China
| | - Xiao-Dong Zhou
- Core Facility of Wuhan University, Wuhan University, Wuhan 430072, PR China
| | - Ji-Ming Hu
- Core Facility of Wuhan University, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
2
|
Wang J, Zhang S, Li Y, Xu Q, Kritzer JA. Investigating the Cytosolic Delivery of Proteins by Lipid Nanoparticles Using the Chloroalkane Penetration Assay. Biochemistry 2024. [PMID: 38334719 DOI: 10.1021/acs.biochem.3c00614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Protein therapeutics are an expanding area for research and drug development, and lipid nanoparticles (LNPs) are the most prominent nonviral vehicles for protein delivery. The most common methods for assessing protein delivery by LNPs include assays that measure the total amount of protein taken up by cells and assays that measure the phenotypic changes associated with protein delivery. However, assays for total cellular uptake include large amounts of protein that are trapped in endosomes or are otherwise nonfunctional. Assays for functional delivery are important, but the readouts are indirect and amplified, limiting the quantitative interpretation. Here, we apply an assay for cytosolic delivery, the chloroalkane penetration assay (CAPA), to measure the cytosolic delivery of a (-30) green fluorescent protein (GFP) fused to Cre recombinase (Cre(-30)GFP) fusion protein by LNPs. We compare these data to the data from total cellular uptake and functional delivery assays to provide a richer analysis of uptake and endosomal escape for LNP-mediated protein delivery. We also use CAPA for a screen of a small library of lipidoids, identifying those with a promising ability to deliver Cre(-30)GFP to the cytosol of mammalian cells. With careful controls and optimized conditions, we expect that CAPA will be a useful tool for investigating the rate, efficiency, and mechanisms of LNP-mediated delivery of therapeutic proteins.
Collapse
Affiliation(s)
- Jing Wang
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Shiying Zhang
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Joshua A Kritzer
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
3
|
Li Z, Dang Q, Wang P, Zhao F, Huang J, Wang C, Liu X, Min W. Food-Derived Peptides: Beneficial CNS Effects and Cross-BBB Transmission Strategies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20453-20478. [PMID: 38085598 DOI: 10.1021/acs.jafc.3c06518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Food-derived peptides, as dietary supplements, have significant effects on promoting brain health and relieving central nervous system (CNS) diseases. However, the blood-brain barrier (BBB) greatly limits their in-brain bioavailability. Thus, overcoming the BBB to target the CNS is a major challenge for bioactive peptides in the prevention and treatment of CNS diseases. This review discusses improvement in the neuroprotective function of food-derived active peptides in CNS diseases, as well as the source of BBB penetrating peptides (BBB-shuttles) and the mechanism of transmembrane transport. Notably, this review also discusses various peptide modification methods to overcome the low permeability and stability of the BBB. Lipification, glycosylation, introduction of disulfide bonds, and cyclization are effective strategies for improving the penetration efficiency of peptides through the BBB. This review provides a new prospective for improving their neuroprotective function and developing treatments to delay or even prevent CNS diseases.
Collapse
Affiliation(s)
- Zehui Li
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P.R. China
| | - Qiao Dang
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P.R. China
| | - Peng Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
| | - Fanrui Zhao
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Jianqin Huang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Chongchong Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Xingquan Liu
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
| | - Weihong Min
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| |
Collapse
|
4
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Zaghmi A, Drouin-Ouellet J, Brambilla D, Gauthier MA. Treating brain diseases using systemic parenterally-administered protein therapeutics: Dysfunction of the brain barriers and potential strategies. Biomaterials 2020; 269:120461. [PMID: 33218788 DOI: 10.1016/j.biomaterials.2020.120461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022]
Abstract
The parenteral administration of protein therapeutics is increasingly gaining importance for the treatment of human diseases. However, the presence of practically impermeable blood-brain barriers greatly restricts access of such pharmaceutics to the brain. Treating brain disorders with proteins thus remains a great challenge, and the slow clinical translation of these therapeutics may be largely ascribed to the lack of appropriate brain delivery system. Exploring new approaches to deliver proteins to the brain by circumventing physiological barriers is thus of great interest. Moreover, parallel advances in the molecular neurosciences are important for better characterizing blood-brain interfaces, particularly under different pathological conditions (e.g., stroke, multiple sclerosis, Parkinson's disease, and Alzheimer's disease). This review presents the current state of knowledge of the structure and the function of the main physiological barriers of the brain, the mechanisms of transport across these interfaces, as well as alterations to these concomitant with brain disorders. Further, the different strategies to promote protein delivery into the brain are presented, including the use of molecular Trojan horses, the formulation of nanosystems conjugated/loaded with proteins, protein-engineering technologies, the conjugation of proteins to polymers, and the modulation of intercellular junctions. Additionally, therapeutic approaches for brain diseases that do not involve targeting to the brain are presented (i.e., sink and scavenging mechanisms).
Collapse
Affiliation(s)
- A Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada
| | - J Drouin-Ouellet
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - D Brambilla
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - M A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada.
| |
Collapse
|
6
|
Khan DH, Bashir S, Khan MI, Figueiredo P, Santos HA, Peltonen L. Formulation optimization and in vitro characterization of rifampicin and ceftriaxone dual drug loaded niosomes with high energy probe sonication technique. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
7
|
Xi J, Liu H. Recent Advances in the Design of Self‐Delivery Amphiphilic Drugs and Vaccines. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jingchao Xi
- Department of Chemical Engineering and Materials Science Wayne State University Detroit MI 48202 USA
| | - Haipeng Liu
- Department of Chemical Engineering and Materials Science Wayne State University Detroit MI 48202 USA
- Department of Oncology Wayne State University Detroit MI 48201 United States
- Tumor Biology and Microenvironment Program Barbara Ann Karmanos Cancer Institute Detroit MI 48201 United States
| |
Collapse
|
8
|
Hiremath CG, Heggnnavar GB, Kariduraganavar MY, Hiremath MB. Co-delivery of paclitaxel and curcumin to foliate positive cancer cells using Pluronic-coated iron oxide nanoparticles. Prog Biomater 2019; 8:155-168. [PMID: 31197663 PMCID: PMC6825627 DOI: 10.1007/s40204-019-0118-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/06/2019] [Indexed: 12/28/2022] Open
Abstract
Active targeting of folic acid and passive targeting of magnetic nanoparticles to bring about co-delivery of hydrophobic chemotherapeutic agents were the focus of this work. Co-precipitation in alkaline environment was employed for synthesizing Fe3O4 nanoparticles and stabilized by oleic acid. Aqueous dispersibility of oleic acid coated nanoparticles was brought about by folic acid modified Pluronic F127 and Pluronic F127 mixture. Folic acid is used as a targeting agent which was joined to Pluronic F127 via diethylene glycol bis(3-aminopropyl) ether spacer. The nanocomposite was used to delivery hydrophobic anticancer drugs, paclitaxel, and curcumin. Successful modification at each step was confirmed by FTIR and NMR. Quantitative analysis of attached folic acid indicated a total of 84.34% amount of conjugation. Nanoparticles characterization revealed the hydrodynamic size of and nanocomposite to be 94.2 nm nanometres. Furthermore, transmission electron micrograph reveals the size of the nanoparticle to be 12.5 nm hence also shows the superparamagnetic activity. Drug encapsulation efficiency of 34.7% and 59.5% was noted for paclitaxel and curcumin, respectively. Cytotoxic property of drug-loaded nanocomposites was increased in case of folic acid functionalized nanoparticles and further increased in the presence of an external magnetic field. Cellular uptake increased in the folic acid conjugated sample. Further many folds in the presence of an external magnetic field.
Collapse
Affiliation(s)
- Chinmay G Hiremath
- Department of Biotechnology and Microbiology, Karnatak University, Pavate Nagar, Dharwad, Karnataka, 580003, India
| | - Geetha B Heggnnavar
- Department of Chemistry, Karnatak University, Dharwad, Karnataka, 580003, India
| | | | - Murigendra B Hiremath
- Department of Biotechnology and Microbiology, Karnatak University, Pavate Nagar, Dharwad, Karnataka, 580003, India.
| |
Collapse
|
9
|
Khan DH, Bashir S, Figueiredo P, Santos HA, Khan MI, Peltonen L. Process optimization of ecological probe sonication technique for production of rifampicin loaded niosomes. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
10
|
Saeedi M, Eslamifar M, Khezri K, Dizaj SM. Applications of nanotechnology in drug delivery to the central nervous system. Biomed Pharmacother 2019; 111:666-675. [PMID: 30611991 DOI: 10.1016/j.biopha.2018.12.133] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/21/2018] [Accepted: 12/30/2018] [Indexed: 12/31/2022] Open
Abstract
In recent years, the researchers and drug designers have given growing attention to new nanotechnology strategies to improve drug delivery to the central nervous system (CNS). Nanotechnology has a great potential to affect the treatment of neurological disorders, mainly Alzheimer's disease, Parkinson's disease, brain tumors, and stroke. With regard to neurodegeneration, several studies showed that nanomaterials have been successfully used for the treatments of CNS disorders. In this regard, nanocarriers have facilitated the targeted delivery of chemotherapeutics resulting in the efficient inhibition of disease progression in malignant brain tumors. Therefore, the most efficacious application of nanomaterials is the use of these substances in the treatment of CNS disease that enhances the overall effect of drug and highlights the importance of nano-therapeutics. This study was conducted to review the evidence on the applications of nanotechnology in designing drug delivery systems with the ability to cross through the blood-brain barrier (BBB) in order to transfer the therapeutic agents to the CNS.
Collapse
Affiliation(s)
- Majid Saeedi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Masoumeh Eslamifar
- Department of Environmental Health Engineering, Faculty of Health, Mazandaran University of Medical Science, Sari, Iran.
| | - Khadijeh Khezri
- Student Research Committee, Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Science, Sari, Iran..
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center and Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Zhang N, Qi R, Chen Y, Ji X, Han Y, Wang Y. Partition of Glutamic Acid-Based Single-Chain and Gemini Amphiphiles into Phospholipid Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:13652-13661. [PMID: 30350992 DOI: 10.1021/acs.langmuir.8b02627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Understanding the interactions of amphiphile molecules with biological membranes is very important to many practical applications. Amino acid amphiphiles are a kind of mild surfactants and have many unique performances. However, their interactions with phospholipid membranes have scarcely been studied. This work has studied the interactions of glutamic acid-based gemini amphiphile C12(Glu)2C12 and single-chain amphiphile C12Glu with the model biomembrane formed by the phospholipid 1,2-dioleoyl- sn-glycero-3-phosphocholine (DOPC). The partition coefficients of C12(Glu)2C12 and C12Glu into the DOPC vesicles were derived from the observed enthalpy curves obtained by isothermal titration calorimetry at temperatures of 25.0 and 37.0 °C, and pHs of 5.6 and 7.4, corresponding to the skin surface and human physiological conditions. The results from cryogenic transmission electron microscopy, dynamic light scattering, and zeta potential measurements show that the amphiphile molecules form different aggregates, which make the amphiphile molecules exhibit different partition abilities to the DOPC vesicles. For C12Glu, the molecules form shorter wormlike micelles with a lower surface charge at all the pHs and temperatures used, and the partition coefficient of C12Glu into the DOPC vesicles does not change with temperature and pH. Differently, the C12(Glu)2C12 molecules form fibers with a larger negative charge and belts with a smaller negative charge at pHs 7.4 and 5.6, respectively, no matter what temperature is used. As a result, the partitions of C12(Glu)2C12 into the DOPC vesicles are markedly different at these two pH values, and the belts at pH 7.4 exhibit a stronger partition ability than the fibrils at pH 5.6. Moreover, at any temperature and pH, C12(Glu)2C12 shows a stronger partition ability than C12Glu. This work can help to understand the relationship between the molecular structure and aggregate structure of amino acid amphiphiles and their partition abilities into the biomembranes.
Collapse
Affiliation(s)
- Na Zhang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Ruilian Qi
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Yao Chen
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Xiuling Ji
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Yuchun Han
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Yilin Wang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| |
Collapse
|
12
|
Rodriguez‐Otormin F, Duro‐Castano A, Conejos‐Sánchez I, Vicent MJ. Envisioning the future of polymer therapeutics for brain disorders. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1532. [DOI: 10.1002/wnan.1532] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 01/09/2023]
Affiliation(s)
| | - Aroa Duro‐Castano
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Valencia Spain
| | | | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Valencia Spain
| |
Collapse
|
13
|
Vinzant N, Scholl JL, Wu CM, Kindle T, Koodali R, Forster GL. Iron Oxide Nanoparticle Delivery of Peptides to the Brain: Reversal of Anxiety during Drug Withdrawal. Front Neurosci 2017; 11:608. [PMID: 29163012 PMCID: PMC5672019 DOI: 10.3389/fnins.2017.00608] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 10/18/2017] [Indexed: 01/19/2023] Open
Abstract
Targeting neuropeptide systems is important for future advancements in treatment of neurological and psychiatric illnesses. However, many of the peptides and their analogs do not cross the blood-brain barrier (BBB) efficiently. Nanoparticles such as iron oxide can cross the BBB, and here we describe a novel method for the conjugation of a peptide antisauvagine-30 (ASV-30) to iron oxide nanoparticles. Previous research has shown that direct infusion of ASV-30 into the brain reduces anxiety-like behavior in animal models via actions on corticotropin releasing factor type 2 (CRF2) receptors. Therefore, we tested whether iron oxide+ASV-30 complexes cross the BBB of rats and then determined whether iron oxide+ASV-30 nanoparticles are localized with CRF2-expressing neurons. Finally we tested the hypothesis that systemic infusion of iron oxide+ASV-30 can reduce anxiety-like behavior. First we describe the synthesis and demonstrate the stability of iron oxide-peptide nanoparticle complexes. Next, nanoparticles (87.7 μg/kg Fe2O3) with or without ASV-30 (200 μg/kg, ip) were injected into male rats 30 min prior to transcardial perfusion and brain fixation for immunohistochemical analysis, or before testing on the elevated plus maze (EPM) in an amphetamine withdrawal model of anxiety. Systemically administered iron oxide+ASV-30 particles were present in the brain and associated with neurons, including those that express CRF2 receptors, but did not localize with the iron storage protein ferritin. Furthermore, systemic administration of ironoxide+ASV-30 reduced amphetamine withdrawal-induced anxiety without affecting locomotion, suggesting that the anxiolytic effects of ASV-30 were preserved and the bioavailability of ASV-30 was sufficient. The findings demonstrate a novel approach to peptide delivery across the BBB and provide insight as to the neural distribution and efficacy of this nanotechnology.
Collapse
Affiliation(s)
- Nathan Vinzant
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Jamie L Scholl
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Chia-Ming Wu
- Department of Chemistry, University of South Dakota, Vermillion, SD, United States
| | - Trevor Kindle
- Department of Chemistry, University of South Dakota, Vermillion, SD, United States
| | - Ranjit Koodali
- Department of Chemistry, University of South Dakota, Vermillion, SD, United States
| | - Gina L Forster
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
14
|
Ji WH, Xiao ZB, Liu GY, Zhang X. Development and application of nano-flavor-drug carriers in neurodegenerative diseases. CHINESE CHEM LETT 2017. [DOI: 10.1016/j.cclet.2017.06.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
Peng J, Rao Y, Yang X, Jia J, Wu Y, Lu J, Tao Y, Tu W. Targeting neuronal nitric oxide synthase by a cell penetrating peptide Tat-LK15/siRNA bioconjugate. Neurosci Lett 2017; 650:153-160. [PMID: 28450191 DOI: 10.1016/j.neulet.2017.04.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/11/2017] [Accepted: 04/21/2017] [Indexed: 02/01/2023]
Abstract
We developed a cell penetrating peptide (CPP) Tat-LK15, as a siRNA carrier to target nNOS. The feasibility, stability, efficiency and selectivity of this peptide-siRNA complex were evaluated in rat neuronal cells. We also compared the new method with conventional siRNA carrier Lipofectamine™. It was found that the CPP Tat-LK15 effectively and specifically delivered nNOS-siRNA into Rat retinal ganglia (RGC-5) cells and silenced the expression of nNOS. The CPP Tat-LK15 can conjugate with siRNA to form stable complex at a ratio of 2:1 (peptide/siRNA, w/w), which maintained stable in serum for as long as 4h. The CPP Tat-LK15 was low-toxicity to cells, as the apoptosis rate of treat cells was not increased significantly when the used peptide lower than 10μg/mL. Moreover, the cellular uptake of nNOS siRNA by Rat Neurons-dorsal spinal cord (RNdsc) cells was also significantly more than naked siRNA by RNdsc cells. The CPP Tat-LK15 was an efficient and stable, and non-cytotoxic siRNA delivery to neurons and effectively silenced the nNOS expression. The CPP Tat-LK15 mediated siRNA delivery was a potential tool to treat neuropathic diseases involving NO or nNOS neurotoxic cascades.
Collapse
Affiliation(s)
- Jie Peng
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China
| | - Yun Rao
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China; Department of Anesthesiology, Jiangxi Maternal and Child Health Hospital, Nanchang, 330000, Jiangxi Province, China
| | - Xue Yang
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China; Department of Anesthesiology, Second Affiliated Hospital, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, China
| | - Ji Jia
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China
| | - Youping Wu
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China
| | - Jianhua Lu
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China.
| | - Yuanxiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07013, USA
| | - Weifeng Tu
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China
| |
Collapse
|
16
|
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: 5.3] [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
|
17
|
Xu X, Li J, Han S, Tao C, Fang L, Sun Y, Zhu J, Liang Z, Li F. A novel doxorubicin loaded folic acid conjugated PAMAM modified with borneol, a nature dual-functional product of reducing PAMAM toxicity and boosting BBB penetration. Eur J Pharm Sci 2016; 88:178-90. [DOI: 10.1016/j.ejps.2016.02.015] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/24/2016] [Accepted: 02/26/2016] [Indexed: 12/20/2022]
|
18
|
Recent progress in biomedical applications of Pluronic (PF127): Pharmaceutical perspectives. J Control Release 2015; 209:120-38. [PMID: 25921088 DOI: 10.1016/j.jconrel.2015.04.032] [Citation(s) in RCA: 234] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 04/23/2015] [Accepted: 04/24/2015] [Indexed: 10/23/2022]
Abstract
Most of the administered anti-cancer drugs are hydrophobic in nature and are known to have poor water solubility, short residence time, rapid clearance from the body and systemic side effects. Polymeric-based targeted particulate carrier system has shown to directly deliver the encapsulated anti-cancer drug to the desired site of action and prevent the interaction of encapsulated drug with the normal cells. Pluronic F127 (PF127) has been widely investigated for its broad-range of therodiagnostic applications in biomedical and pharmaceutical sciences, but rapid dissolution in the physiological fluids, short residence time, rapid clearance, and weak mechanical strength are the main shortcomings that are associated with PF127 and have recently been overcome by making various modifications in the structure of PF127 notably through preparation of PF127-based mixed polymeric micelles, PF127-conjugated nanoparticles and PF127-based hydrophobically modified thermogels. In this article, we have briefly discussed the recent studies that have been conducted on various anti-cancer drugs using PF127 as nano-carrier modified with other copolymers and/or conjugated with magnetic nanoparticles. The key findings of these studies demonstrated that the modified form of PF127 can significantly increase the stability of incorporated hydrophobic drugs with enhanced in vitro cytotoxicity and cellular uptake of anti-cancer drugs. Moreover, the modified form of PF127 has also shown its therapeutic potentials as therodiagnostics in various types of tumors and cancers. Hence, it can be concluded that the modified form of PF127 exhibits significant therodiagnostic effects with increased tumor-specific delivery of anti-cancer drugs having minimal toxic effects as compared to PF127 alone and/or other copolymers.
Collapse
|
19
|
Exosomes as drug delivery vehicles for Parkinson's disease therapy. J Control Release 2015; 207:18-30. [PMID: 25836593 DOI: 10.1016/j.jconrel.2015.03.033] [Citation(s) in RCA: 1299] [Impact Index Per Article: 144.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/24/2015] [Accepted: 03/28/2015] [Indexed: 01/12/2023]
Abstract
Exosomes are naturally occurring nanosized vesicles that have attracted considerable attention as drug delivery vehicles in the past few years. Exosomes are comprised of natural lipid bilayers with the abundance of adhesive proteins that readily interact with cellular membranes. We posit that exosomes secreted by monocytes and macrophages can provide an unprecedented opportunity to avoid entrapment in mononuclear phagocytes (as a part of the host immune system), and at the same time enhance delivery of incorporated drugs to target cells ultimately increasing drug therapeutic efficacy. In light of this, we developed a new exosomal-based delivery system for a potent antioxidant, catalase, to treat Parkinson's disease (PD). Catalase was loaded into exosomes ex vivo using different methods: the incubation at room temperature, permeabilization with saponin, freeze-thaw cycles, sonication, or extrusion. The size of the obtained catalase-loaded exosomes (exoCAT) was in the range of 100-200nm. A reformation of exosomes upon sonication and extrusion, or permeabilization with saponin resulted in high loading efficiency, sustained release, and catalase preservation against proteases degradation. Exosomes were readily taken up by neuronal cells in vitro. A considerable amount of exosomes was detected in PD mouse brain following intranasal administration. ExoCAT provided significant neuroprotective effects in in vitro and in vivo models of PD. Overall, exosome-based catalase formulations have a potential to be a versatile strategy to treat inflammatory and neurodegenerative disorders.
Collapse
|
20
|
Lalatsa A, Schätzlein A, Garrett N, Moger J, Briggs M, Godfrey L, Iannitelli A, Freeman J, Uchegbu I. Chitosan amphiphile coating of peptide nanofibres reduces liver uptake and delivers the peptide to the brain on intravenous administration. J Control Release 2015; 197:87-96. [DOI: 10.1016/j.jconrel.2014.10.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/06/2014] [Accepted: 10/27/2014] [Indexed: 01/05/2023]
|
21
|
Kyle S, Saha S. Nanotechnology for the detection and therapy of stroke. Adv Healthc Mater 2014; 3:1703-20. [PMID: 24692428 DOI: 10.1002/adhm.201400009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Indexed: 01/06/2023]
Abstract
Over the years, nanotechnology has greatly developed, moving from careful design strategies and synthesis of novel nanostructures to producing them for specific medical and biological applications. The use of nanotechnology in diagnostics, drug delivery, and tissue engineering holds great promise for the treatment of stroke in the future. Nanoparticles are employed to monitor grafted cells upon implantation, or to enhance the imagery of the tissue, which is coupled with a noninvasive imaging modality such as magnetic resonance imaging, computed axial tomography or positron emission tomography scan. Contrast imaging agents used can range from iron oxide, perfluorocarbon, cerium oxide or platinum nanoparticles to quantum dots. The use of nanomaterial scaffolds for neuroregeneration is another area of nanomedicine, which involves the creation of an extracellular matrix mimic that not only serves as a structural support but promotes neuronal growth, inhibits glial differentiation, and controls hemostasis. Promisingly, carbon nanotubes can act as scaffolds for stem cell therapy and functionalizing these scaffolds may enhance their therapeutic potential for treatment of stroke. This Progress Report highlights the recent developments in nanotechnology for the detection and therapy of stroke. Recent advances in the use of nanomaterials as tissue engineering scaffolds for neuroregeneration will also be discussed.
Collapse
Affiliation(s)
- Stuart Kyle
- School of Medicine; University of Leeds; Leeds LS2 9JT UK
| | - Sikha Saha
- Division of Cardiovascular and Diabetes Research; Leeds Institute of Genetics; Health and Therapeutics; University of Leeds; Leeds LS2 9JT UK
| |
Collapse
|
22
|
Farkhani SM, Valizadeh A, Karami H, Mohammadi S, Sohrabi N, Badrzadeh F. Cell penetrating peptides: efficient vectors for delivery of nanoparticles, nanocarriers, therapeutic and diagnostic molecules. Peptides 2014; 57:78-94. [PMID: 24795041 DOI: 10.1016/j.peptides.2014.04.015] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/19/2014] [Accepted: 04/19/2014] [Indexed: 01/24/2023]
Abstract
Efficient delivery of therapeutic and diagnostic molecules to the cells and tissues is a difficult challenge. The cellular membrane is very effective in its role as a selectively permeable barrier. While it is essential for cell survival and function, also presents a major barrier for intracellular delivery of cargo such as therapeutic and diagnostic agents. In recent years, cell-penetrating peptides (CPPs), that are relatively short cationic and/or amphipathic peptides, received great attention as efficient cellular delivery vectors due to their intrinsic ability to enter cells and mediate uptake of a wide range of macromolecular cargo such as plasmid DNA (pDNA), small interfering RNA (siRNAs), drugs, and nanoparticulate pharmaceutical carriers. This review discusses the various uptake mechanisms of these peptides. Furthermore, we discuss recent advances in the use of CPP for the efficient delivery of nanoparticles, nanocarriers, DNA, siRNA, and anticancer drugs to the cells. In addition, we have been highlighting new results for improving endosomal escape of CPP-cargo molecules. Finally, pH-responsive and activable CPPs for tumor-targeting therapy have been described.
Collapse
Affiliation(s)
- Samad Mussa Farkhani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alireza Valizadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran.
| | - Hadi Karami
- Department of Medical Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Sardasht, 38481 Arak, Iran.
| | - Samane Mohammadi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran.
| | - Nasrin Sohrabi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran.
| | - Fariba Badrzadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran.
| |
Collapse
|
23
|
Yi X, Kabanov AV. Brain delivery of proteins via their fatty acid and block copolymer modifications. J Drug Target 2014; 21:940-55. [PMID: 24160902 DOI: 10.3109/1061186x.2013.847098] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It is well known that hydrophobic small molecules penetrate cell membranes better than hydrophilic molecules. Amphiphilic molecules that dissolve both in lipid and aqueous phases are best suited for membrane transport. Transport of biomacromolecules across physiological barriers, e.g. the blood-brain barrier, is greatly complicated by the unique structure and function of such barriers. Two decades ago we adopted a simple philosophy that to increase protein delivery to the brain one needs to modify this protein with hydrophobic moieties. With this general idea we began modifying proteins (antibodies, enzymes, hormones, etc.) with either hydrophobic fatty acid residues or amphiphilic block copolymer moieties, such as poy(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) (pluronics or poloxamers) and more recently, poly(2-oxasolines). This simple approach has resulted in impressive successes in CNS drug delivery. We present a retrospective overview of these works initiated in the Soviet Union in 1980s, and then continued in the United States and other countries. Notably some of the early findings were later corroborated by brain pharmacokinetic data. Industrial development of several drug candidates employing these strategies has followed. Overall modification by hydrophobic fatty acids residues or amphiphilic block copolymers represents a promising and relatively safe strategy to deliver proteins to the brain.
Collapse
Affiliation(s)
- Xiang Yi
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA and
| | | |
Collapse
|
24
|
Agile delivery of protein therapeutics to CNS. J Control Release 2014; 190:637-63. [PMID: 24956489 DOI: 10.1016/j.jconrel.2014.06.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/10/2014] [Accepted: 06/13/2014] [Indexed: 12/11/2022]
Abstract
A variety of therapeutic proteins have shown potential to treat central nervous system (CNS) disorders. Challenge to deliver these protein molecules to the brain is well known. Proteins administered through parenteral routes are often excluded from the brain because of their poor bioavailability and the existence of the blood-brain barrier (BBB). Barriers also exist to proteins administered through non-parenteral routes that bypass the BBB. Several strategies have shown promise in delivering proteins to the brain. This review, first, describes the physiology and pathology of the BBB that underscore the rationale and needs of each strategy to be applied. Second, major classes of protein therapeutics along with some key factors that affect their delivery outcomes are presented. Third, different routes of protein administration (parenteral, central intracerebroventricular and intraparenchymal, intranasal and intrathecal) are discussed along with key barriers to CNS delivery associated with each route. Finally, current delivery strategies involving chemical modification of proteins and use of particle-based carriers are overviewed using examples from literature and our own work. Whereas most of these studies are in the early stage, some provide proof of mechanism of increased protein delivery to the brain in relevant models of CNS diseases, while in few cases proof of concept had been attained in clinical studies. This review will be useful to broad audience of students, academicians and industry professionals who consider critical issues of protein delivery to the brain and aim developing and studying effective brain delivery systems for protein therapeutics.
Collapse
|
25
|
Guo X, Li D, Yang G, Shi C, Tang Z, Wang J, Zhou S. Thermo-triggered drug release from actively targeting polymer micelles. ACS APPLIED MATERIALS & INTERFACES 2014; 6:8549-8559. [PMID: 24804870 DOI: 10.1021/am501422r] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
How to deliver the drug to the target area at the right time and at the right concentration is still a challenge in cancer therapy. In this study, we present a facile strategy to control drug release by precisely controlling the thermo-sensitivity of the nanocarriers to the variation of environmental temperature. One type of thermoresponsive Pluronic F127-poly(d,l-lactic acid) (F127-PLA, abbreviated as FP) copolymer micelles was developed and decorated with folate (FA) for active targeting. FP100 micelles assembled from FP with PLA segment having polymerization degree of 100 had a low critical solution temperature of 39.2 °C close to body temperature. At 37 °C, little amount of encapsulated anticancer drug DOX is released from the FP100 micelles, while at a slightly elevated temperature (40 °C), the shrinkage of thermoresponsive segments causes a rapid release of DOX and instantly increases the drug concentration locally. The cytocompatibility analysis and cellular uptake efficiency were characterized with the fibroblast cell line NIH 3T3 and human cervix adenocarcinoma cell line HeLa. The results demonstrate that this copolymer has excellent cytocompatibility, and FA-decorated FP100 micelles present much better efficiency of cellular uptake and higher cytotoxicity to folate receptor (FR)-overexpressed HeLa cells. In particular, under hyperthermia (40 °C) the cytotoxicity of DOX-loaded FA-FP100 micelles against HeLa cells was significantly more obvious than that upon normothermia (37 °C). Therefore, these temperature-responsive micelles have great potential as a drug vehicle for cancer therapy.
Collapse
Affiliation(s)
- Xing Guo
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Niu J, Wang A, Ke Z, Zheng Z. Glucose transporter and folic acid receptor-mediated Pluronic P105 polymeric micelles loaded with doxorubicin for brain tumor treating. J Drug Target 2014; 22:712-23. [PMID: 24806516 DOI: 10.3109/1061186x.2014.913052] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this study, glucose transporter and folic acid (FA) receptor-mediated Pluronic P105 polymeric micelles loaded with DOX (GF-DOX) were prepared for enhancing the blood-brain barrier (BBB) transportation and improving the drug accumulation in the glioma cells. The pH-triggered DOX release of GF-DOX indicating a comparatively fast drug release at weak acidic condition and stable state of the carrier at physiological environment. The transport of GF-DOX across the in vitro BBB model showed that GF-DOX exhibited higher BBB transportation ability with the transporting ratio of 21.47% in 4 h. The carrier was internalized into C6 glioma cells upon crossing the BBB model for the combined effect of the brain targeting by transportation of glucose transporter and active tumor cell targeting by FA receptor-mediated endocytosis. Moreover, minimized weight changes and high suppression ratio of tumor growth were observed after intravenous injection of GF-DOX. In conclusion, the glucose transporter and FA dual-targeting micelles would provide a safe and effective strategy for new modalities to treat brain tumor.
Collapse
Affiliation(s)
- Jiangxiu Niu
- Department of Chemistry and Chemical Engineering, Huangshan University , Huangshan , People's Republic of China
| | | | | | | |
Collapse
|
27
|
Lalatsa A, Schatzlein AG, Uchegbu IF. Strategies to deliver peptide drugs to the brain. Mol Pharm 2014; 11:1081-93. [PMID: 24601686 DOI: 10.1021/mp400680d] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neurological diseases such as neurodegeneration, pain, psychiatric disorders, stroke, and brain cancers would greatly benefit from the use of highly potent and specific peptide pharmaceuticals. Peptides are especially desirable because of their low inherent toxicity. The presence of the blood brain barrier (BBB), their short duration of action, and their need for parenteral administration limits their clinical use. However, over the past decade there have been significant advances in delivering peptides to the central nervous system. Angiopep peptides developed by Angiochem (Montreal, Canada), transferrin antibodies developed by ArmaGen (Santa Monica, USA), and cell penetrating peptides have all shown promise in delivering therapeutic peptides across the BBB after intravenous administration. Noninvasive methods of delivering peptides to the brain include the use of chitosan amphiphile nanoparticles for oral delivery and nose to brain strategies. The uptake of the chitosan amphiphile nanoparticles by the gastrointestinal epithelium is important for oral peptide delivery. Finally protecting peptides from plasma degradation is integral to the success of most of these peptide delivery strategies.
Collapse
Affiliation(s)
- Aikaterini Lalatsa
- Department of Pharmaceutics, School of Pharmacy and Biomedical Sciences, University of Portsmouth , St Michael's Building 5.05, White Swan Road, Portsmouth, PO1 2DT, U.K
| | | | | |
Collapse
|
28
|
Basavaraj S, Betageri GV. Can formulation and drug delivery reduce attrition during drug discovery and development-review of feasibility, benefits and challenges. Acta Pharm Sin B 2014; 4:3-17. [PMID: 26579359 PMCID: PMC4590717 DOI: 10.1016/j.apsb.2013.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 12/06/2013] [Accepted: 12/16/2013] [Indexed: 12/21/2022] Open
Abstract
Drug discovery and development has become longer and costlier process. The fear of failure and stringent regulatory review process is driving pharmaceutical companies towards “me too” drugs and improved generics (505(b) (2)) fillings. The discontinuance of molecules at late stage clinical trials is common these years. The molecules are withdrawn at various stages of discovery and development process for reasons such as poor ADME properties, lack of efficacy and safety reasons. Hence this review focuses on possible applications of formulation and drug delivery to salvage molecules and improve the drugability. The formulation and drug delivery technologies are suitable for addressing various issues contributing to attrition are discussed in detail.
Collapse
|
29
|
Klyachko NL, Haney MJ, Zhao Y, Manickam DS, Mahajan V, Suresh P, Hingtgen SD, Mosley RL, Gendelman HE, Kabanov AV, Batrakova EV. Macrophages offer a paradigm switch for CNS delivery of therapeutic proteins. Nanomedicine (Lond) 2013; 9:1403-22. [PMID: 24237263 DOI: 10.2217/nnm.13.115] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
AIMS Active targeted transport of the nanoformulated redox enzyme, catalase, in macrophages attenuates oxidative stress and as such increases survival of dopaminergic neurons in animal models of Parkinson's disease. Optimization of the drug formulation is crucial for the successful delivery in living cells. We demonstrated earlier that packaging of catalase into a polyion complex micelle ('nanozyme') with a synthetic polyelectrolyte block copolymer protected the enzyme against degradation in macrophages and improved therapeutic outcomes. We now report the manufacture of nanozymes with superior structure and therapeutic indices. METHODS Synthesis, characterization and therapeutic efficacy of optimal cell-based nanoformulations are evaluated. RESULTS A formulation design for drug carriers typically works to avoid entrapment in monocytes and macrophages focusing on small-sized nanoparticles with a polyethylene glycol corona (to provide a stealth effect). By contrast, the best nanozymes for delivery in macrophages reported in this study have a relatively large size (≈ 200 nm), which resulted in improved loading capacity and release from macrophages. Furthermore, the cross-linking of nanozymes with the excess of a nonbiodegradable linker ensured their low cytotoxicity, and efficient catalase protection in cell carriers. Finally, the 'alternatively activated' macrophage phenotype (M2) utilized in these studies did not promote further inflammation in the brain, resulting in a subtle but statistically significant effect on neuronal regeneration and repair in vivo. CONCLUSION The optimized cross-linked nanozyme loaded into macrophages reduced neuroinflammatory responses and increased neuronal survival in mice. Importantly, the approach for nanoformulation design for cell-mediated delivery is different from the common requirements for injectable formulations.
Collapse
Affiliation(s)
- Natalia L Klyachko
- Laboratory for Chemical Design of Bionanomaterials & the Department of Chemical Enzymology, Faculty of Chemistry, MV Lomonosov Moscow State University, Moscow, Russia and Department of Pharmaceutical Sciences, Center for Drug Delivery & Nanomedicine, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The peptide therapeutic market is one of the fastest growth areas of the pharmaceutical industry. Although few orally administered peptides are marketed and many are in different phases of clinical development, there is no marketed oral peptide therapeutic used for CNS disorders. The major challenges involved in orally delivering peptides to the brain relate to their enzymatic instability and inability to permeate across physiological barriers. The paucity of therapies for the treatment of brain diseases and the presence of the blood-brain barrier excluding 98% of therapeutic molecules necessitates parenteral administration. Various approaches have been applied to enhance oral peptide bioavailability, but only nanoparticulate strategies were able to deliver orally therapeutic peptides to the brain. Although industry may be reluctant to invest in developing oral peptide nanomedicines, the increasingly unmet clinical need and economic burden associated with brain diseases will fuel the development of the first marketed oral-to-brain peptide therapy.
Collapse
|
31
|
Perera RH, Patel R, Wu H, Gangolli M, Traughber B, Oleinick N, Exner AA. Preclinical evaluation of radiosensitizing activity of Pluronic block copolymers. Int J Radiat Biol 2013; 89:801-12. [PMID: 23631609 DOI: 10.3109/09553002.2013.800246] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Pluronic block copolymers are non-ionic surfactants with demonstrated sensitizing activity in chemotherapy and hyperthermia in various tumor cell lines. In the current study we investigated the potential activity of Pluronic as a radiosensitizing agent. MATERIALS AND METHODS As a possible mechanism, the effect of Pluronic on Hsp70 and Hsp90 was examined. Gli36 human glioma cells were treated with radiation alone as well as with a combination treatment of Pluronic and radiation. RESULTS Clonogenic cell survival assays show that Pluronic has an elevated effect on radiosensitization (50% high, p < 0.01), even with radiation doses as low as 2 Gy. The Hsp90 level was reduced 24 h after the combined treatment in both in vitro and in vivo. Similarly, Hsp70 levels were also decreased 24 h post treatment. When Gli36 cells were exposed to Pluronic before and during irradiation, DNA DSB: double-strand breaks repair was reduced, and elevated apoptosis was also seen in tumor xenografts. CONCLUSION Data suggest the potential use of L10 as a radiosensitizer. While the mechanism of sensitization requires additional investigation, the presented results indicate that the effect may be due, in part, to a decrease in Hsp90 and 70 levels and increased DNA damage.
Collapse
Affiliation(s)
- Reshani H Perera
- Department of Radiology, Case Western Reserve University , Cleveland, Ohio , USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Guo X, Shi C, Wang J, Di S, Zhou S. pH-triggered intracellular release from actively targeting polymer micelles. Biomaterials 2013; 34:4544-54. [DOI: 10.1016/j.biomaterials.2013.02.071] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 02/28/2013] [Indexed: 12/24/2022]
|
33
|
Haney MJ, Zhao Y, Harrison EB, Mahajan V, Ahmed S, He Z, Suresh P, Hingtgen SD, Klyachko NL, Mosley RL, Gendelman HE, Kabanov AV, Batrakova EV. Specific transfection of inflamed brain by macrophages: a new therapeutic strategy for neurodegenerative diseases. PLoS One 2013; 8:e61852. [PMID: 23620794 PMCID: PMC3631190 DOI: 10.1371/journal.pone.0061852] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 03/15/2013] [Indexed: 12/25/2022] Open
Abstract
The ability to precisely upregulate genes in inflamed brain holds great therapeutic promise. Here we report a novel class of vectors, genetically modified macrophages that carry reporter and therapeutic genes to neural cells. Systemic administration of macrophages transfected ex vivo with a plasmid DNA (pDNA) encoding a potent antioxidant enzyme, catalase, produced month-long expression levels of catalase in the brain resulting in three-fold reductions in inflammation and complete neuroprotection in mouse models of Parkinson's disease (PD). This resulted in significant improvements in motor functions in PD mice. Mechanistic studies revealed that transfected macrophages secreted extracellular vesicles, exosomes, packed with catalase genetic material, pDNA and mRNA, active catalase, and NF-κb, a transcription factor involved in the encoded gene expression. Exosomes efficiently transfer their contents to contiguous neurons resulting in de novo protein synthesis in target cells. Thus, genetically modified macrophages serve as a highly efficient system for reproduction, packaging, and targeted gene and drug delivery to treat inflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Matthew J. Haney
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Yuling Zhao
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Emily B. Harrison
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Vivek Mahajan
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shaheen Ahmed
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Zhijian He
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Poornima Suresh
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shawn D. Hingtgen
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Natalia L. Klyachko
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Chemical Enzymology, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Alexander V. Kabanov
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Chemical Enzymology, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Elena V. Batrakova
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
34
|
Rosenbaugh EG, Savalia KK, Manickam DS, Zimmerman MC. Antioxidant-based therapies for angiotensin II-associated cardiovascular diseases. Am J Physiol Regul Integr Comp Physiol 2013; 304:R917-28. [PMID: 23552499 DOI: 10.1152/ajpregu.00395.2012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases, including hypertension and heart failure, are associated with activation of the renin-angiotensin system (RAS) and increased circulating and tissue levels of ANG II, a primary effector peptide of the RAS. Through its actions on various cell types and organ systems, ANG II contributes to the pathogenesis of cardiovascular diseases by inducing cardiac and vascular hypertrophy, vasoconstriction, sodium and water reabsorption in kidneys, sympathoexcitation, and activation of the immune system. Cardiovascular research over the past 15-20 years has clearly implicated an important role for elevated levels of reactive oxygen species (ROS) in mediating these pathophysiological actions of ANG II. As such, the use of antioxidants, to reduce the elevated levels of ROS, as potential therapies for various ANG II-associated cardiovascular diseases has been intensely investigated. Although some antioxidant-based therapies have shown therapeutic impact in animal models of cardiovascular disease and in human patients, others have failed. In this review, we discuss the benefits and limitations of recent strategies, including gene therapy, dietary sources, low-molecular-weight free radical scavengers, polyethylene glycol conjugation, and nanomedicine-based technologies, which are designed to deliver antioxidants for the improved treatment of cardiovascular diseases. Although much work has been completed, additional research focusing on developing specific antioxidant molecules or proteins and identifying the ideal in vivo delivery system for such antioxidants is necessary before the use of antioxidant-based therapies for cardiovascular diseases become a clinical reality.
Collapse
Affiliation(s)
- Erin G Rosenbaugh
- Department of Cellular and Integrative Physiology, Nebraska Center for Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | |
Collapse
|
35
|
Mazza M, Notman R, Anwar J, Rodger A, Hicks M, Parkinson G, McCarthy D, Daviter T, Moger J, Garrett N, Mead T, Briggs M, Schätzlein AG, Uchegbu IF. Nanofiber-based delivery of therapeutic peptides to the brain. ACS NANO 2013; 7:1016-1026. [PMID: 23289352 DOI: 10.1021/nn305193d] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The delivery of therapeutic peptides and proteins to the central nervous system is the biggest challenge when developing effective neuropharmaceuticals. The central issue is that the blood-brain barrier is impermeable to most molecules. Here we demonstrate the concept of employing an amphiphilic derivative of a peptide to deliver the peptide into the brain. The key to success is that the amphiphilic peptide should by design self-assemble into nanofibers wherein the active peptide epitope is tightly wrapped around the nanofiber core. The nanofiber form appears to protect the amphiphilic peptide from degradation while in the plasma, and the amphiphilic nature of the peptide promotes its transport across the blood-brain barrier. Therapeutic brain levels of the amphiphilic peptide are achieved with this strategy, compared with the absence of detectable peptide in the brain and the consequent lack of a therapeutic response when the underivatized peptide is administered.
Collapse
Affiliation(s)
- Mariarosa Mazza
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Teow HM, Zhou Z, Najlah M, Yusof SR, Abbott NJ, D’Emanuele A. Delivery of paclitaxel across cellular barriers using a dendrimer-based nanocarrier. Int J Pharm 2013; 441:701-11. [DOI: 10.1016/j.ijpharm.2012.10.024] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 10/10/2012] [Accepted: 10/10/2012] [Indexed: 12/13/2022]
|
37
|
Tong J, Yi X, Luxenhofer R, Banks WA, Jordan R, Zimmerman MC, Kabanov AV. Conjugates of superoxide dismutase 1 with amphiphilic poly(2-oxazoline) block copolymers for enhanced brain delivery: synthesis, characterization and evaluation in vitro and in vivo. Mol Pharm 2012; 10:360-77. [PMID: 23163230 DOI: 10.1021/mp300496x] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Superoxide dismutase 1 (SOD1) efficiently catalyzes dismutation of superoxide, but its poor delivery to the target sites in the body, such as brain, hinders its use as a therapeutic agent for superoxide-associated disorders. Here to enhance the delivery of SOD1 across the blood-brain barrier (BBB) and in neurons the enzyme was conjugated with poly(2-oxazoline) (POx) block copolymers, P(MeOx-b-BuOx) or P(EtOx-b-BuOx), composed of (1) hydrophilic 2-methyl-2-oxazoline (MeOx) or 2-ethyl-2-oxazoline (EtOx) and (2) hydrophobic 2-butyl-2-oxazoline (BuOx) repeating units. The conjugates contained from 2 to 3 POx chains joining the protein amino groups via cleavable -(ss)- or noncleavable -(cc)- linkers at the BuOx block terminus. They retained 30% to 50% of initial SOD1 activity, were conformationally and thermally stable, and assembled in 8 or 20 nm aggregates in aqueous solution. They had little if any toxicity to CATH.a neurons and displayed enhanced uptake in these neurons as compared to native or PEGylated SOD1. Of the two conjugates, SOD1-(cc)-P(MeOx-b-BuOx) and SOD1-(cc)-P(EtOx-b-BuOx), compared, the latter was entering cells 4 to 7 times faster and at 6 h colocalized predominantly with endoplasmic reticulum (41 ± 3%) and mitochondria (21 ± 2%). Colocalization with endocytosis markers and pathway inhibition assays suggested that it was internalized through lipid raft/caveolae, also employed by the P(EtOx-b-BuOx) copolymer. The SOD activity in cell lysates and ability to attenuate angiotensin II (Ang II)-induced superoxide in live cells were increased for this conjugate compared to SOD1 and PEG-SOD1. Studies in mice showed that SOD1-POx had ca. 1.75 times longer half-life in blood than native SOD1 (28.4 vs 15.9 min) and after iv administration penetrated the BBB significantly faster than albumin to accumulate in brain parenchyma. The conjugate maintained high stability both in serum and in brain (77% vs 84% at 1 h postinjection). Its amount taken up by the brain reached a maximum value of 0.08% ID/g (percent of the injected dose taken up per gram of brain) 4 h postinjection. The entry of SOD1-(cc)-P(EtOx-b-BuOx) to the brain was mediated by a nonsaturable mechanism. Altogether, SOD1-POx conjugates are promising candidates as macromolecular antioxidant therapies for superoxide-associated diseases such as Ang II-induced neurocardiovascular diseases.
Collapse
Affiliation(s)
- Jing Tong
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | | | | | | | | | | | | |
Collapse
|
38
|
Adolph O, Köster S, Georgieff M, Georgieff EM, Moulig W, Föhr KJ. Promethazine inhibits NMDA-induced currents – New pharmacological aspects of an old drug. Neuropharmacology 2012; 63:280-91. [DOI: 10.1016/j.neuropharm.2012.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 02/27/2012] [Accepted: 03/11/2012] [Indexed: 12/24/2022]
|
39
|
Lalatsa A, Schätzlein AG, Uchegbu* IF. Nanostructures Overcoming the Blood-Brain Barrier: Physiological Considerations and Mechanistic Issues. NANOSTRUCTURED BIOMATERIALS FOR OVERCOMING BIOLOGICAL BARRIERS 2012. [DOI: 10.1039/9781849735292-00329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
40
|
Lalatsa A, Lee V, Malkinson JP, Zloh M, Schätzlein AG, Uchegbu IF. A Prodrug Nanoparticle Approach for the Oral Delivery of a Hydrophilic Peptide, Leucine5-enkephalin, to the Brain. Mol Pharm 2012; 9:1665-80. [DOI: 10.1021/mp300009u] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Aikaterini Lalatsa
- UCL School of Pharmacy, University
of London, 29-39
Brunswick Square, London, WC1N 1AX, U.K
| | - Vivian Lee
- UCL School of Pharmacy, University
of London, 29-39
Brunswick Square, London, WC1N 1AX, U.K
| | - John P. Malkinson
- UCL School of Pharmacy, University
of London, 29-39
Brunswick Square, London, WC1N 1AX, U.K
| | - Mire Zloh
- UCL School of Pharmacy, University
of London, 29-39
Brunswick Square, London, WC1N 1AX, U.K
| | - Andreas G. Schätzlein
- UCL School of Pharmacy, University
of London, 29-39
Brunswick Square, London, WC1N 1AX, U.K
| | - Ijeoma F. Uchegbu
- UCL School of Pharmacy, University
of London, 29-39
Brunswick Square, London, WC1N 1AX, U.K
| |
Collapse
|
41
|
Haney MJ, Suresh P, Zhao Y, Kanmogne GD, Kadiu I, Sokolsky-Papkov M, Klyachko NL, Mosley RL, Kabanov AV, Gendelman HE, Batrakova EV. Blood-borne macrophage-neural cell interactions hitchhike on endosome networks for cell-based nanozyme brain delivery. Nanomedicine (Lond) 2012; 7:815-33. [PMID: 22236307 DOI: 10.2217/nnm.11.156] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Macrophage-carried nanoformulated catalase ('nanozyme') attenuates neuroinflammation and protects nigrostriatal neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxication. This is facilitated by effective enzyme transfer from blood-borne macrophages to adjacent endothelial cells and neurons leading to the decomposition of reactive oxygen species. MATERIALS & METHODS We examined the intra- and inter-cellular trafficking mechanisms of nanozymes by confocal microscopy. Improved neuronal survival mediated by nanozyme-loaded macrophages was demonstrated by fluorescence-activated cell sorting. RESULTS In macrophages, nanozymes were internalized mainly by clathrin-mediated endocytosis then trafficked to recycling endosomes. The enzyme is subsequently released in exosomes facilitated by bridging conduits. Nanozyme transfer from macrophages to adjacent cells by endocytosis-independent mechanisms diffusing broadly throughout the recipient cells. In contrast, macrophage-free nanozymes were localized in lysosomes following endocytic entry. CONCLUSION Facilitated transfer of nanozyme from cell to cell can improve neuroprotection against oxidative stress commonly seen during neurodegenerative disease processes.
Collapse
|
42
|
Banks WA, Gertler A, Solomon G, Niv-Spector L, Shpilman M, Yi X, Batrakova E, Vinogradov S, Kabanov AV. Principles of strategic drug delivery to the brain (SDDB): development of anorectic and orexigenic analogs of leptin. Physiol Behav 2011; 105:145-9. [PMID: 21669216 DOI: 10.1016/j.physbeh.2011.05.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 05/19/2011] [Accepted: 05/23/2011] [Indexed: 11/19/2022]
Abstract
The blood-brain barrier (BBB) presents a tremendous challenge for the delivery of drugs to the central nervous system (CNS). This includes drugs that target brain receptors for the treatment of obesity and anorexia. Strategic drug delivery to brain (SDDB) is an approach that considers in depth the relations among the BBB, the candidate therapeutic, the CNS target, and the disease state to be treated. Here, we illustrate principles of SDDB with two different approaches to developing drugs based on leptin. In normal body weight humans and in non-obese rodents, leptin is readily transported across the BBB and into the CNS where it inhibits feeding and enhances thermogenesis. However, in obesity, the transport of leptin across the BBB is impaired, resulting in a resistance to leptin. As a result, it is difficult to treat obesity with leptin or its analogs that depend on the leptin transporter for access to the CNS. To treat obesity, we developed a leptin agonist modified by the addition of pluronic block copolymers (P85-leptin). P85-leptin retains biological activity and is capable of crossing the BBB by a mechanism that is not dependent on the leptin transporter. As such, P85-leptin is able to cross the BBB of obese mice at a rate similar to that of native leptin in lean mice. To treat anorexia, we developed a leptin antagonist modified by pegylation (PEG-MLA) that acts primarily by blocking the BBB transporter for endogenous, circulating leptin. This prevents blood-borne, endogenous leptin from entering the CNS, essentially mimicking the leptin resistance seen in obesity, and resulting in a significant increase in adiposity. These examples illustrate two strategies in which an understanding of the interactions among the BBB, CNS targets, and candidate therapeutics under physiologic and diseased conditions can be used to develop drugs effective for the treatment of brain disease.
Collapse
Affiliation(s)
- W A Banks
- Geriatrics Research Education and Clinical Center - Veterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington, Seattle, WA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhao Y, Haney MJ, Klyachko NL, Li S, Booth SL, Higginbotham SM, Jones J, Zimmerman MC, Mosley RL, Kabanov AV, Gendelman HE, Batrakova EV. Polyelectrolyte complex optimization for macrophage delivery of redox enzyme nanoparticles. Nanomedicine (Lond) 2011; 6:25-42. [PMID: 21182416 DOI: 10.2217/nnm.10.129] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND We posit that cell-mediated drug delivery can improve transport of therapeutic enzymes to the brain and decrease inflammation and neurodegeneration seen during Parkinson's disease. Our prior works demonstrated that macrophages loaded with nanoformulated catalase ('nanozyme') then parenterally injected protect the nigrostriatum in a murine model of Parkinson's disease. Packaging of catalase into block ionomer complex with a synthetic polyelectrolyte block copolymer precludes enzyme degradation in macrophages. METHODS We examined relationships between the composition and structure of block ionomer complexes with a range of block copolymers, their physicochemical characteristics, and loading, release and catalase enzymatic activity in bone marrow-derived macrophages. RESULTS Formation of block ionomer complexes resulted in improved aggregation stability. Block ionomer complexes with ε-polylysine and poly(L-glutamic acid)-poly(ethylene glycol) demonstrated the least cytotoxicity and high loading and release rates. However, these formulations did not efficiently protect catalase inside macrophages. CONCLUSION Nanozymes with polyethyleneimine- and poly(L-lysine)(10)-poly(ethylene glycol) provided the best protection of enzymatic activity for cell-mediated drug delivery.
Collapse
Affiliation(s)
- Yuling Zhao
- Center for Drug Delivery & Nanomedicine, 985830 Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Haney MJ, Zhao Y, Li S, Higginbotham SM, Booth SL, Han HY, Vetro JA, Mosley RL, Kabanov AV, Gendelman HE, Batrakova EV. Cell-mediated transfer of catalase nanoparticles from macrophages to brain endothelial, glial and neuronal cells. Nanomedicine (Lond) 2011; 6:1215-30. [PMID: 21449849 DOI: 10.2217/nnm.11.32] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Our laboratories forged the concept of macrophage delivery of protein antioxidants to attenuate neuroinflammation and nigrostriatal neurodegeneration in Parkinson's disease. Notably, the delivery of the redox enzyme, catalase, incorporated into a polyion complex micelle ('nanozyme') by bone marrow-derived macrophages protected nigrostriatum against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxication. Nonetheless, how macrophage delivery of nanozyme increases the efficacy of catalase remains unknown. METHODS In this study, we examined the transfer of nanozyme from macrophages to brain microvessel endothelial cells, neurons and astrocytes. RESULTS Facilitated transport of the nanozyme from macrophages to endothelial, neuronal and glial target cells occurred through endocytosis-independent mechanisms that involved fusion of cellular membranes, macrophage bridging conduits and nanozyme lipid coatings. Nanozyme transfer was operative across an artificial blood-brain barrier and showed efficient reactive oxygen species decomposition. CONCLUSION This is the first demonstration, to our knowledge, that drug-loaded macrophages discharge particles to contiguous target cells for therapeutic brain enzyme delivery. The data shown are of potential value for the treatment of neurodegenerative disorders and notably, Parkinson's disease.
Collapse
Affiliation(s)
- Matthew J Haney
- Center for Drug Delivery & Nanomedicine, University of Nebraska, Omaha, NE, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lai TC, Kataoka K, Kwon GS. Pluronic-based cationic block copolymer for forming pDNA polyplexes with enhanced cellular uptake and improved transfection efficiency. Biomaterials 2011; 32:4594-603. [PMID: 21453964 DOI: 10.1016/j.biomaterials.2011.02.065] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 02/24/2011] [Indexed: 01/11/2023]
Abstract
PEGylated cationic polymers have been extensively studied for substituting virus as gene delivery vehicles. These polymers can produce water-soluble polyionic complexes (polyplexes) with plasmid DNA (pDNA) and show enhanced stability compared to non-PEGylated polyplexes. However, PEGylation always diminishes the transfection efficiency of polyplexes probably due to poor cellular internalization of the particles and difficulty in releasing the pDNA cargo from the complexes intracellularly for gene expression. As non-ionic surfactants, Pluronic block copolymers have been shown to interact with plasma membrane and promote cellular uptake of various small molecules and biomacromolecules. To evaluate whether Pluronic could improve the transfection efficiency of polyplexes, Pluronic P85- and PEG-based cationomers comprising poly{N-[N-(2-aminoethyl)-2-aminoethyl] aspartamide (P[Asp(DET)]) cationic blocks were synthesized and tested for their transfection ability. In this study, it was demonstrated that although the stability of the PEG-based polyplexes was better than that of the P85-based polyplexes based cationic polymers, the P85-based polyplex could achieve significantly higher transfection than the PEG counterparts. The improvement of gene delivering ability was shown to be correlated with the enhanced cellular internalization of the P85-based polyplexes.
Collapse
Affiliation(s)
- Tsz Chung Lai
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705-2222, USA
| | | | | |
Collapse
|
46
|
Multifunctional Pluronic P123/F127 mixed polymeric micelles loaded with paclitaxel for the treatment of multidrug resistant tumors. Biomaterials 2011; 32:2894-906. [PMID: 21256584 DOI: 10.1016/j.biomaterials.2010.12.039] [Citation(s) in RCA: 293] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Accepted: 12/26/2010] [Indexed: 10/18/2022]
Abstract
The aim of this study was to exploit the possibility of combination of active targeting function of folic acid by folate receptor-mediated endocytosis and overcoming multidrug resistance (MDR) by Pluronic block copolymers to promote drug delivery to MDR tumor following intravenous administration with paclitaxel (PTX) as model drug. Folic acid functionalized Pluronic P123/F127 mixed micelles encapsulating PTX (FPF-PTX) was firstly developed and tested in vitro and in vivo, while PTX-loaded Pluronic P123/F127 mixed micelles (PF-PTX) and Taxol were used as control. FPF-PTX was about 20 nm in diameter with spherical shape and high encapsulation efficiency. Cellular uptake of FPF-PTX was found to be higher than that of PF-PTX due to the folate receptor-mediated endocytosis effect. In vitro cytotoxicity, cell apoptosis and cell cycle arrest studies also revealed that FPF-PTX was more potent than those of PF-PTX and Taxol. In vivo pharmacokinetic study in rats showed that the polymeric micelles significantly enhanced the bioavailability of PTX (∼3 fold) than Taxol. Moreover, in BALB/c mice bearing KBv MDR tumor xenografts, stronger antitumor efficacy was shown in FPF-PTX group, with good correlation between in vitro and in vivo. In conclusion, folate-conjugated Pluronic micelles could be a potential vehicle for delivering hydrophobic chemotherapeutic drugs to MDR tumors.
Collapse
|
47
|
Zhao Y, Haney MJ, Mahajan V, Reiner BC, Dunaevsky A, Mosley RL, Kabanov AV, Gendelman HE, Batrakova EV. Active Targeted Macrophage-mediated Delivery of Catalase to Affected Brain Regions in Models of Parkinson's Disease. ACTA ACUST UNITED AC 2011; S4. [PMID: 22288024 DOI: 10.4172/2157-7439.s4-003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We previously demonstrated that monocyte-macrophage based drug delivery can be applied to a spectrum of infectious, neoplastic, and degenerative disorders. In particular, bone marrow-derived macrophages (BMM) loaded with nano formulated catalase, "nanozyme", were shown to attenuate neuro inflammation and nigrostriatal degeneration in rodent models of Parkinson's disease (PD). Nonetheless, the pharmacokinetics and biodistribution of BMM-incorporated nanozyme has not been explored. To this end, we now demonstrate that BMM, serving as a "depot" for nanozyme, increased area under the curve(AUC), half-life, and mean residence time in blood circulation of the protein when compared to the nanozyme administered alone. Accordingly, bioavailability of the nanozyme for the brain, spleen, kidney, and liver was substantially increased. Importantly, nanozyme-loaded BMM targeted diseased sites and improved transport across the blood brain barrier. This was seen specifically in affected brain subregions in models of PD. Engaging natural immune cells such as monocyte-macrophages as drug carriers provides a new perspective for therapeutic delivery for PD and also likely a range of other inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Yuling Zhao
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tong J, Luxenhofer R, Yi X, Jordan R, Kabanov AV. Protein modification with amphiphilic block copoly(2-oxazoline)s as a new platform for enhanced cellular delivery. Mol Pharm 2010; 7:984-92. [PMID: 20550191 DOI: 10.1021/mp100102p] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Several homopolymers, random copolymers and block copolymers based on poly(2-oxazoline)s (POx) were synthesized and conjugated to horseradish peroxidase (HRP) using biodegradable and nonbiodegradable linkers. These conjugates were characterized by amino group titration, polyacrylamide gel electrophoresis (PAGE), isoelectric focusing, enzymatic activity assay and conformation analysis. The conjugates contained on average from about one to two polymer chains per enzyme. From 70% to 90% of enzymatic activity was retained in most cases. Circular dichroism (CD) analysis revealed that HRP modification affected the secondary structure of the apoprotein but did not affect the tertiary structure and heme environment. Enhanced cellular uptake was found in the conjugates of two block copolymers using both MDCK cells and Caco-2 cells, but not in the conjugates of random copolymer and homopolymer. Conjugation with a block copolymer of 2-methyl-2-oxazoline and 2-butyl-2-oxazoline led to the highest cellular uptake as compared to other conjugates. Our data indicates that modification with amphiphilic POx has the potential to modulate and enhance cellular delivery of proteins.
Collapse
Affiliation(s)
- Jing Tong
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198-5830, USA
| | | | | | | | | |
Collapse
|
49
|
Yi X, Zimmerman MC, Yang R, Tong J, Vinogradov S, Kabanov AV. Pluronic-modified superoxide dismutase 1 attenuates angiotensin II-induced increase in intracellular superoxide in neurons. Free Radic Biol Med 2010; 49:548-58. [PMID: 20493251 PMCID: PMC2998907 DOI: 10.1016/j.freeradbiomed.2010.04.039] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 04/29/2010] [Accepted: 04/30/2010] [Indexed: 11/21/2022]
Abstract
Overexpressing superoxide dismutase 1 (SOD1; also called Cu/ZnSOD), an intracellular superoxide (O(2)(*-))-scavenging enzyme, in central neurons inhibits angiotensin II (AngII) intraneuronal signaling and normalizes cardiovascular dysfunction in diseases associated with enhanced AngII signaling in the brain, including hypertension and heart failure. However, the blood-brain barrier and neuronal cell membranes impose a tremendous impediment for the delivery of SOD1 to central neurons, which hinders the potential therapeutic impact of SOD1 treatment on these diseases. To address this, we developed conjugates of SOD1 with poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) block copolymer (Pluronic) (SOD1-P85 and SOD1-L81), which retained significant SOD1 enzymatic activity. The modified SOD1 effectively scavenged xanthine oxidase/hypoxanthine-derived O(2)(*-), as determined by HPLC and the measurement of 2-hydroxyethidium. Using catecholaminergic neurons, we observed an increase in neuronal uptake of SOD1-Pluronic after 1, 6, or 24h, compared to neurons treated with pure SOD1 or PEG-SOD1. Importantly, without inducing neuronal toxicity, SOD1-Pluronic conjugates significantly inhibited AngII-induced increases in intraneuronal O(2)(*-) levels. These data indicate that SOD1-Pluronic conjugates penetrate neuronal cell membranes, which results in elevated intracellular levels of functional SOD1. Pluronic conjugation may be a new delivery system for SOD1 into central neurons and therapeutically beneficial for AngII-related cardiovascular diseases.
Collapse
Affiliation(s)
- Xiang Yi
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-5830
| | - Matthew C. Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5830
| | - Ruifang Yang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5830
| | - Jing Tong
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-5830
| | - Serguei Vinogradov
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-5830
| | - Alexander V. Kabanov
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-5830
- Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119899 Moscow, Russia
- Correspondence: Alexander V. Kabanov, Center for Drug Delivery and Nanomedicine, 985830 Nebraska Medical Center, Omaha, NE 68198-5830; Tel: (402) 559-9364; Fax (402) 559-9365,
| |
Collapse
|
50
|
Nanoparticle-mediated brain-specific drug delivery, imaging, and diagnosis. Pharm Res 2010; 27:1759-71. [PMID: 20593303 DOI: 10.1007/s11095-010-0141-7] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2009] [Accepted: 03/29/2010] [Indexed: 12/16/2022]
Abstract
Central nervous system (CNS) diseases represent the largest and fastest-growing area of unmet medical need. Nanotechnology plays a unique instrumental role in the revolutionary development of brain-specific drug delivery, imaging, and diagnosis. With the aid of nanoparticles of high specificity and multifunctionality, such as dendrimers and quantum dots, therapeutics, imaging agents, and diagnostic molecules can be delivered to the brain across the blood-brain barrier (BBB), enabling considerable progress in the understanding, diagnosis, and treatment of CNS diseases. Nanoparticles used in the CNS for drug delivery, imaging, and diagnosis are reviewed, as well as their administration routes, toxicity, and routes to cross the BBB. Future directions and major challenges are outlined.
Collapse
|