1
|
Ge X, Wekselblatt JB, Elmore S, Wang B, Wang T, Dai R, Zhang T, Dave H, Ghaderi M, Anilkumar AR, Wang B, Sirsi SR, Ahn JM, Shapiro MG, Oka Y, Lois C, Qin Z. In Vivo Cytosolic Delivery of Biomolecules into Neurons for Super-Resolution Imaging and Genome Modification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501033. [PMID: 40285608 DOI: 10.1002/advs.202501033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Efficient delivery of biomolecules into neurons has significant impacts on therapeutic applications in the central nervous system (CNS) and fundamental neuroscience research. Existing viral and non-viral delivery methods often suffer from inefficient intracellular access due to the endocytic pathway. Here, a neuron-targeting and direct cytosolic delivery platform is discovered by using a 15-amino-acid peptide, termed the N1 peptide, which enables neuron-specific targeting and cytosolic delivery of functional biomolecules. The N1 peptide initially binds hyaluronan in the extracellular matrix and subsequently passes the membrane of neurons without being trapped into endosome. This mechanism facilitates the efficient delivery of cell-impermeable and photo-stable fluorescent dye for super-resolution imaging of dendritic spines, and functional proteins, such as Cre recombinase, for site-specific genome modification. Importantly, the N1 peptide exhibits robust neuronal specificity across diverse species, including mice, rats, tree shrews, and zebra finches. Its targeting capability is further demonstrated through various administration routes, including intraparenchymal, intrathecal, and intravenous (i.v.) injections after blood-brain barrier (BBB) opening with focused ultrasound (FUS). These findings establish the N1 peptide as a versatile and functional platform with significant potential for bioimaging and advanced therapeutic applications.
Collapse
Affiliation(s)
- Xiaoqian Ge
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Joseph B Wekselblatt
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Department of Opthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Scott Elmore
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Bo Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Tongtong Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Renjinming Dai
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Tingting Zhang
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Harsh Dave
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Mohammadaref Ghaderi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Athul Raj Anilkumar
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Bill Wang
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Shashank R Sirsi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Jung-Mo Ahn
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Howard Hughes Medical Institute, Pasadena, CA, 91125, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Zhenpeng Qin
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| |
Collapse
|
2
|
Kamei N, Ikeda K, Ohmoto Y, Fujisaki S, Shirata R, Maki M, Miyata M, Miyauchi Y, Nishiyama N, Yamada M, Ohigashi Y, Takeda-Morishita M. Insulin-inspired hippocampal neuron-targeting technology for protein drug delivery. Proc Natl Acad Sci U S A 2024; 121:e2407936121. [PMID: 39348543 PMCID: PMC11474037 DOI: 10.1073/pnas.2407936121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 09/03/2024] [Indexed: 10/02/2024] Open
Abstract
Hippocampal neurons can be the first to be impaired with neurodegenerative disorders, including Alzheimer's disease (AD). Most drug candidates for causal therapy of AD cannot either enter the brain or accumulate around hippocampal neurons. Here, we genetically engineered insulin-fusion proteins, called hippocampal neuron-targeting (Ht) proteins, for targeting protein drugs to hippocampal neurons because insulin tends to accumulate in the neuronal cell layers of the hippocampus. In vitro examinations clarified that insulin and Ht proteins were internalized into the cultured hippocampal neurons through insulin receptor-mediated macropinocytosis. Cysteines were key determinants of the delivery of Ht proteins to hippocampal neurons, and insulin B chain mutant was most potent in delivering cargo proteins. In vivo accumulation of Ht proteins to hippocampal neuronal layers occurred after intracerebroventricular administration. Thus, hippocampal neuron-targeting technology can provide great help for developing protein drugs against neurodegenerative disorders.
Collapse
Affiliation(s)
- Noriyasu Kamei
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Kento Ikeda
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Yuka Ohmoto
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Seita Fujisaki
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Ryusei Shirata
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Maya Maki
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Mika Miyata
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Yuki Miyauchi
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Nanaka Nishiyama
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Mana Yamada
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Yuna Ohigashi
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| | - Mariko Takeda-Morishita
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Hyogo650-8586, Japan
| |
Collapse
|
3
|
Ma J, Ding L, Peng X, Jiang L, Liu G. Recent Advances of Engineered Cell Membrane-Based Nanotherapeutics to Combat Inflammatory Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308646. [PMID: 38334202 DOI: 10.1002/smll.202308646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/20/2024] [Indexed: 02/10/2024]
Abstract
An immune reaction known as inflammation serves as a shield from external danger signals, but an overactive immune system may additionally lead to tissue damage and even a variety of inflammatory disorders. By inheriting biological functionalities and serving as both a therapeutic medication and a drug carrier, cell membrane-based nanotherapeutics offer the potential to treat inflammatory disorders. To further strengthen the anti-inflammatory benefits of natural cell membranes, researchers alter and optimize the membranes using engineering methods. This review focuses on engineered cell membrane-based nanotherapeutics (ECMNs) and their application in treating inflammation-related diseases. Specifically, this article discusses the methods of engineering cell membranes for inflammatory diseases and examines the progress of ECMNs in inflammation-targeted therapy, inflammation-neutralizing therapy, and inflammation-immunomodulatory therapy. Additionally, the article looks into the perspectives and challenges of ECMNs in inflammatory treatment and offers suggestions as well as guidance to encourage further investigations and implementations in this area.
Collapse
Affiliation(s)
- Jiaxin Ma
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Linyu Ding
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xuqi Peng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Lai Jiang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Gang Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
4
|
Chen J, Xu Y, Zhou M, Xu S, Varley AJ, Golubovic A, Lu RXZ, Wang KC, Yeganeh M, Vosoughi D, Li B. Combinatorial design of ionizable lipid nanoparticles for muscle-selective mRNA delivery with minimized off-target effects. Proc Natl Acad Sci U S A 2023; 120:e2309472120. [PMID: 38060560 PMCID: PMC10723144 DOI: 10.1073/pnas.2309472120] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Ionizable lipid nanoparticles (LNPs) pivotal to the success of COVID-19 mRNA (messenger RNA) vaccines hold substantial promise for expanding the landscape of mRNA-based therapies. Nevertheless, the risk of mRNA delivery to off-target tissues highlights the necessity for LNPs with enhanced tissue selectivity. The intricate nature of biological systems and inadequate knowledge of lipid structure-activity relationships emphasize the significance of high-throughput methods to produce chemically diverse lipid libraries for mRNA delivery screening. Here, we introduce a streamlined approach for the rapid design and synthesis of combinatorial libraries of biodegradable ionizable lipids. This led to the identification of iso-A11B5C1, an ionizable lipid uniquely apt for muscle-specific mRNA delivery. It manifested high transfection efficiencies in muscle tissues, while significantly diminishing off-targeting in organs like the liver and spleen. Moreover, iso-A11B5C1 also exhibited reduced mRNA transfection potency in lymph nodes and antigen-presenting cells, prompting investigation into the influence of direct immune cell transfection via LNPs on mRNA vaccine effectiveness. In comparison with SM-102, while iso-A11B5C1's limited immune transfection attenuated its ability to elicit humoral immunity, it remained highly effective in triggering cellular immune responses after intramuscular administration, which is further corroborated by its strong therapeutic performance as cancer vaccine in a melanoma model. Collectively, our study not only enriches the high-throughput toolkit for generating tissue-specific ionizable lipids but also encourages a reassessment of prevailing paradigms in mRNA vaccine design. This study encourages rethinking of mRNA vaccine design principles, suggesting that achieving high immune cell transfection might not be the sole criterion for developing effective mRNA vaccines.
Collapse
Affiliation(s)
- Jingan Chen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ONM5S 3G9, Canada
| | - Yue Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Muye Zhou
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Shufen Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Andrew James Varley
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Alex Golubovic
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Rick Xing Ze Lu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Kevin Chang Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Mina Yeganeh
- Institute of Medical Science, University of Toronto, Toronto, ONM5G 1L7, Canada
| | - Daniel Vosoughi
- Institute of Medical Science, University of Toronto, Toronto, ONM5G 1L7, Canada
- Latner Thoracic Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ONM5G 2C4, Canada
| | - Bowen Li
- Institute of Biomedical Engineering, University of Toronto, Toronto, ONM5S 3G9, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G 2C1, Canada
| |
Collapse
|
5
|
Olshefsky A, Benasutti H, Sylvestre M, Butterfield GL, Rocklin GJ, Richardson C, Hicks DR, Lajoie MJ, Song K, Leaf E, Treichel C, Decarreau J, Ke S, Kher G, Carter L, Chamberlain JS, Baker D, King NP, Pun SH. In vivo selection of synthetic nucleocapsids for tissue targeting. Proc Natl Acad Sci U S A 2023; 120:e2306129120. [PMID: 37939083 PMCID: PMC10655225 DOI: 10.1073/pnas.2306129120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/21/2023] [Indexed: 11/10/2023] Open
Abstract
Controlling the biodistribution of protein- and nanoparticle-based therapeutic formulations remains challenging. In vivo library selection is an effective method for identifying constructs that exhibit desired distribution behavior; library variants can be selected based on their ability to localize to the tissue or compartment of interest despite complex physiological challenges. Here, we describe further development of an in vivo library selection platform based on self-assembling protein nanoparticles encapsulating their own mRNA genomes (synthetic nucleocapsids or synNCs). We tested two distinct libraries: a low-diversity library composed of synNC surface mutations (45 variants) and a high-diversity library composed of synNCs displaying miniproteins with binder-like properties (6.2 million variants). While we did not identify any variants from the low-diversity surface library that yielded therapeutically relevant changes in biodistribution, the high-diversity miniprotein display library yielded variants that shifted accumulation toward lungs or muscles in just two rounds of in vivo selection. Our approach should contribute to achieving specific tissue homing patterns and identifying targeting ligands for diseases of interest.
Collapse
Affiliation(s)
- Audrey Olshefsky
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Halli Benasutti
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Meilyn Sylvestre
- Department of Bioengineering, University of Washington, Seattle, WA98195
| | - Gabriel L. Butterfield
- Institute for Protein Design, University of Washington, Seattle, WA98195
- Department of Molecular and Cellular Biology, University of Washington, Seattle, WA98195
| | - Gabriel J. Rocklin
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Christian Richardson
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Derrick R. Hicks
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Marc J. Lajoie
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Kefan Song
- Department of Bioengineering, University of Washington, Seattle, WA98195
| | - Elizabeth Leaf
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Catherine Treichel
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Justin Decarreau
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Sharon Ke
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Gargi Kher
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Jeffrey S. Chamberlain
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Department of Neurology, University of Washington, Seattle, WA98195
| | - David Baker
- Institute for Protein Design, University of Washington, Seattle, WA98195
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA98195
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA98195
| |
Collapse
|
6
|
Wang YK, Zhao YP, Ye MZ, Wang L, Lan TS, Wang Y, Qi ZQ. Chimeric CNS-targeting-peptide engineered exosomes for experimental autoimmune encephalomyelitis therapy. Int Immunopharmacol 2023; 124:110835. [PMID: 37717320 DOI: 10.1016/j.intimp.2023.110835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/02/2023] [Accepted: 08/18/2023] [Indexed: 09/19/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) that causes demyelination, neuronal damage and white matter loss, but there is still no known cure. Exosomes are 30-200 nm-sized double-layered membrane vesicles that can easily cross the blood-brain barrier (BBB). Exosomes from umbilical cord mesenchymal stem cells(UMSCs) have been found to treat experimental autoimmune encephalomyelitis (EAE) through the action of anti-inflammatory and immunomodulatory, but its clinical translation has been hampered by their inefficacious accumulation in CNS. Therefore, we developed a TAxI-exos, also known as a TAxI-peptide-chimeric UMSC-exos, for CNS-specific accumulation and curative effect in EAE. We used the EAE model in vivo as well as active T cell and BV-2 cell models in vitro to explore the efficacy and mechanisms. Exosomes from UMSCs with TAxI or DiR labels were given to EAE mice in one dosage (150 g) prior to the peak at day 15. The mice were sacrificed on day 30 so that spinal cords, spleens, and blood could be taken for analysis of demyelination, inflammation, microglia, T-cell subset proportions, and inflammatory cytokine expression. In vitro, PBMCs and splenocytes isolated from healthy C57BL/6 mice were activated and incubated with 0.15 mg/mL of UMSC-exos or TAxI-exos for immune mechanism investigations. Activated BV-2 cells were used to investigate the targeting and controlling polarization ability and mechanism of UMSC-exos and TAxI-exos. As expected, TAxI-exos exhibited significantly greater therapeutic action in EAE mice than UMSC-exos due to their improved targeting-ability. The medication reduced T-cell subset proportions and inflammation, reduced active-microglia proportions and promoted M1 to M2 microglial cell polarization through TNF pathway, upregulated IL-4, IL-10, TGF-β, and IDO-1 expression, and downregulated IL-2, IL-6, IL-17A, IFN-γ, and TNF-α. The CNS-targeting properties of TAxI-exos and their capacity to inhibit degenerative processes in EAE mice have considerable potential therapeutic value for MS and other CNS illnesses.
Collapse
Affiliation(s)
- Ying-Kai Wang
- School of Medicine, Guangxi University, Nanning, Guangxi, China.
| | - Yun-Peng Zhao
- Department of Histology and Embryology, Naval Medical University, Shanghai, China; Shanghai Key Lab of Cell Engineering, China.
| | - Ming-Zhu Ye
- Department of Obstetrics and Gynecology, Zhong Shan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Ling Wang
- Shanghai Key Lab of Cell Engineering, China; Translational Medicine Research Center, Naval Medical University, Shanghai, China.
| | - Tian-Shu Lan
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian, China.
| | - Yue Wang
- Department of Histology and Embryology, Naval Medical University, Shanghai, China; Shanghai Key Lab of Cell Engineering, China; Translational Medicine Research Center, Naval Medical University, Shanghai, China.
| | - Zhong-Quan Qi
- School of Medicine, Guangxi University, Nanning, Guangxi, China.
| |
Collapse
|
7
|
Wang W, Hassan MM, Mao G. Colloidal Perspective on Targeted Drug Delivery to the Central Nervous System. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:3235-3245. [PMID: 36825490 DOI: 10.1021/acs.langmuir.2c02949] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
This article describes a new approach in targeted drug delivery to the central nervous system (CNS) in a significant departure from the predominant systematic drug administration attempting to penetrate the blood-brain barrier (BBB). Nanoparticles chemically conjugated to neural tract tracer proteins are capable of path-specific axonal retrograde transport, transneuronal transport, and anatomical tract flow to bypass the BBB. To celebrate the work by Dr. Bettye Washington Greene on the physical chemistry of colloidal particles, this article focuses on the physiochemical characteristics of the nanoparticles, various colloidal forces that impact the colloidal stability of nanoparticles in biological media, and surface chemistry strategies to avoid nanoparticle aggregation-induced poor therapeutic outcomes. The biological environment for the anatomical retrograde transport of neural tract tracers is examined to directly link factors impacting the colloidal stability of the new class of CNS-targeting nanoconjugates such as nanoconjugate size, shape, surface charge, surface chemistry, ionic strength, pH, and protein adsorption on the nanoparticle. We conclude with opportunities and challenges for future research.
Collapse
Affiliation(s)
- Wenqian Wang
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Md Musfizur Hassan
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Guangzhao Mao
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| |
Collapse
|
8
|
Sellers DL, Lee K, Murthy N, Pun SH. TAxI-peptide targeted Cas12a ribonuclease protein nanoformulations increase genome editing in hippocampal neurons. J Control Release 2023; 354:188-195. [PMID: 36596342 PMCID: PMC9975068 DOI: 10.1016/j.jconrel.2022.12.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023]
Abstract
Gene therapy approaches that utilize Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) ribonucleases have tremendous potential to treat human disease. However, CRISPR therapies delivered by integrating viral vectors are limited by potential off-target genome editing caused by constitutive activation of ribonuclease functions. Thus, biomaterial formulations are being used for the delivery of purified CRISPR components to increase the efficiency and safety of genome editing approaches. We previously demonstrated that a novel peptide identified by phage display, TAxI-peptide, mediates delivery of recombinant proteins into neurons. In this report we utilized NeutrAvidin protein to formulate neuron-targeted genome-editing nanoparticles. Cas12a ribonucleases was loaded with biotinylated guide RNA and biotinylated TAxI-peptide onto NeutrAvidin protein to coordinate the formation a targeted ribonuclease protein (RNP) complex. TAxI-RNP complexes are polydisperse with a 14.3 nm radius. The nanoparticles are stable after formulation and show good stability in the presence of normal mouse serum. TAxI-RNP nanoparticles increased neuronal delivery of Cas12a in reporter mice, resulting in induced tdTomato expression after direct injection into the dentate gyrus of the hippocampus. TAxI-RNP nanoparticles also increased genome editing efficacy in hippocampal neurons versus glia. These studies demonstrate the ability to assemble RNP nanoformulations with NeutrAvidin by binding biotinylated peptides and gRNA-loaded Cas12a ribonucleases into protein nanoparticles that target CRISPR delivery to specific cell-types in vivo. The potential to deliver CRISPR nanoparticles to specific cell-types and control off-target delivery to further reduce deleterious genome editing is essential for the creation of viable therapies to treat nervous system disease.
Collapse
Affiliation(s)
- Drew L Sellers
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, United States.
| | - Kunwoo Lee
- GenEdit Inc., Berkeley, CA, United States
| | - Niren Murthy
- Department of Bioengineering, University of California, Berkeley, CA, United States
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
9
|
Padmakumar S, D'Souza A, Parayath NN, Bleier BS, Amiji MM. Nucleic acid therapies for CNS diseases: Pathophysiology, targets, barriers, and delivery strategies. J Control Release 2022; 352:121-145. [PMID: 36252748 DOI: 10.1016/j.jconrel.2022.10.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/10/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
Nucleic acid therapeutics have emerged as one of the very advanced and efficacious treatment approaches for debilitating health conditions, including those diseases affecting the central nervous system (CNS). Precise targeting with an optimal control over gene regulation confers long-lasting benefits through the administration of nucleic acid payloads via viral, non-viral, and engineered vectors. The current review majorly focuses on the development and clinical translational potential of non-viral vectors for treating CNS diseases with a focus on their specific design and targeting approaches. These carriers must be able to surmount the various intracellular and extracellular barriers, to ensure successful neuronal transfection and ultimately attain higher therapeutic efficacies. Additionally, the specific challenges associated with CNS administration also include the presence of blood-brain barrier (BBB), the complex pathophysiological and biochemical changes associated with different disease conditions and the existence of non-dividing cells. The advantages offered by lipid-based or polymeric systems, engineered proteins, particle-based systems coupled with various approaches of neuronal targeting have been discussed in the context of a variety of CNS diseases. The possibilities of rapid yet highly efficient gene modifications rendered by the breakthrough methodologies for gene editing and gene manipulation have also opened vast avenues of research in neuroscience and CNS disease therapy. The current review also underscores the extensive scientific efforts to optimize specialized, efficacious yet non-invasive and safer administration approaches to overcome the therapeutic delivery challenges specifically posed by the CNS transport barriers and the overall obstacles to clinical translation.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| | - Anisha D'Souza
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 20115, USA
| | - Neha N Parayath
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| | - Benjamin S Bleier
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 20115, USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Wang D, Min H, Wang Z, Wang X, Li H, Cao M, Wang J. Core-Shell Structures from the Coassembly of Lipoprotein-like Nanoparticles and Plasmid DNA for Gene Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:12198-12206. [PMID: 36170670 DOI: 10.1021/acs.langmuir.2c01829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
We reported self-assembled core-shell nanoparticles (NPs) based on lipoprotein-like NPs and plasmid DNA (pDNA). Lipoprotein-like NPs were prepared using cholic acid (CA)-modified lipopeptides. We designed six different lipopeptides with different peptide segments to construct a series of NPs. It was proven that these NPs have different positive surface charges. These NPs could bind pDNA through electrostatic interaction to form core-shell complexes. The interactions between NPs and pDNA were systematically investigated. The number of NP charges determines the strength of the interaction between NPs and pDNA. Thus, various types of core-shell structures, such as loose and dense core-shell NPs, were found in this system. Cytotoxicity test confirmed that the carriers had no toxicity. We also proved that the core-shell structures have a good cell transfection effect. This study would expand the application of lipopeptide assemblies in the gene delivery field, which may lead to the development of peptide-based gene vectors for therapeutic application.
Collapse
Affiliation(s)
- Dong Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, Shandong, China
| | - Haofeng Min
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, Shandong, China
| | - Zhaoyu Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, Shandong, China
| | - Xinhao Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, Shandong, China
| | - Hui Li
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, Shandong, China
| | - Meiwen Cao
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, Shandong, China
| | - Jiqian Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, Shandong, China
| |
Collapse
|
11
|
Tsedev U, Lin CW, Hess GT, Sarkaria JN, Lam FC, Belcher AM. Phage Particles of Controlled Length and Genome for In Vivo Targeted Glioblastoma Imaging and Therapeutic Delivery. ACS NANO 2022; 16:11676-11691. [PMID: 35830573 DOI: 10.1021/acsnano.1c08720] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
M13 bacteriophage (phage) are versatile, genetically tunable nanocarriers that have been recently adapted for use as diagnostic and therapeutic platforms. Applying p3 capsid chlorotoxin fusion with the "inho" circular single-stranded DNA (cssDNA) gene packaging system, we produced miniature chlorotoxin inho (CTX-inho) phage particles with a minimum length of 50 nm that can target intracranial orthotopic patient-derived GBM22 glioblastoma tumors in the brains of mice. Systemically administered indocyanine green conjugated CTX-inho phage accumulated in brain tumors, facilitating shortwave infrared detection. Furthermore, we show that our inho phage can carry cssDNA that are transcriptionally active when delivered to GBM22 glioma cells in vitro. The ability to modulate the capsid display, surface loading, phage length, and cssDNA gene content makes the recombinant M13 phage particle an ideal delivery platform.
Collapse
Affiliation(s)
- Uyanga Tsedev
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ching-Wei Lin
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 10617, Taiwan
| | - Gaelen T Hess
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705, Unites States
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Fred C Lam
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division of Neurosurgery, Saint Elizabeth's Medical Center, Brighton, Massachusetts 02135, United States
| | - Angela M Belcher
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
12
|
Fukui T, Tateno H, Nakamura T, Yamada Y, Sato Y, Iwasaki N, Harashima H, Kadoya K. Retrograde Axonal Transport of Liposomes from Peripheral Tissue to Spinal Cord and DRGs by Optimized Phospholipid and CTB Modification. Int J Mol Sci 2022; 23:6661. [PMID: 35743104 PMCID: PMC9223829 DOI: 10.3390/ijms23126661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
Despite recent advancements in therapeutic options for disorders of the central nervous system (CNS), the lack of an efficient drug-delivery system (DDS) hampers their clinical application. We hypothesized that liposomes could be optimized for retrograde transport in axons as a DDS from peripheral tissues to the spinal cord and dorsal root ganglia (DRGs). Three types of liposomes consisting of DSPC, DSPC/POPC, or POPC in combination with cholesterol (Chol) and polyethylene glycol (PEG) lipid were administered to sciatic nerves or the tibialis anterior muscle of mature rats. Liposomes in cell bodies were detected with infrared fluorescence of DiD conjugated to liposomes. Three days later, all nerve-administered liposomes were retrogradely transported to the spinal cord and DRGs, whereas only muscle-administered liposomes consisting of DSPC reached the spinal cord and DRGs. Modification with Cholera toxin B subunit improved the transport efficiency of liposomes to the spinal cord and DRGs from 4.5% to 17.3% and from 3.9% to 14.3% via nerve administration, and from 2.6% to 4.8% and from 2.3% to 4.1% via muscle administration, respectively. Modification with octa-arginine (R8) improved the transport efficiency via nerve administration but abolished the transport capability via muscle administration. These findings provide the initial data for the development of a novel DDS targeting the spinal cord and DRGs via peripheral administration.
Collapse
Affiliation(s)
- Takafumi Fukui
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Sapporo 060-8638, Japan; (T.F.); (N.I.)
| | - Hironao Tateno
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; (H.T.); (Y.Y.); (Y.S.); (H.H.)
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; (H.T.); (Y.Y.); (Y.S.); (H.H.)
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; (H.T.); (Y.Y.); (Y.S.); (H.H.)
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; (H.T.); (Y.Y.); (Y.S.); (H.H.)
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Sapporo 060-8638, Japan; (T.F.); (N.I.)
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan; (H.T.); (Y.Y.); (Y.S.); (H.H.)
| | - Ken Kadoya
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Sapporo 060-8638, Japan; (T.F.); (N.I.)
| |
Collapse
|
13
|
Nance E, Pun SH, Saigal R, Sellers DL. Drug delivery to the central nervous system. NATURE REVIEWS. MATERIALS 2022; 7:314-331. [PMID: 38464996 PMCID: PMC10923597 DOI: 10.1038/s41578-021-00394-w] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 03/12/2024]
Abstract
Despite the rising global incidence of central nervous system (CNS) disorders, CNS drug development remains challenging, with high costs, long pathways to clinical use and high failure rates. The CNS is highly protected by physiological barriers, in particular, the blood-brain barrier and the blood-cerebrospinal fluid barrier, which limit access of most drugs. Biomaterials can be designed to bypass or traverse these barriers, enabling the controlled delivery of drugs into the CNS. In this Review, we first examine the effects of normal and diseased CNS physiology on drug delivery to the brain and spinal cord. We then discuss CNS drug delivery designs and materials that are administered systemically, directly to the CNS, intranasally or peripherally through intramuscular injections. Finally, we highlight important challenges and opportunities for materials design for drug delivery to the CNS and the anticipated clinical impact of CNS drug delivery.
Collapse
Affiliation(s)
- Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Rajiv Saigal
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| |
Collapse
|
14
|
Hou M, Wei Y, Zhao Z, Han W, Zhou R, Zhou Y, Zheng Y, Yin L. Immuno-Engineered Nanodecoys for the Multi-Target Anti-Inflammatory Treatment of Autoimmune Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108817. [PMID: 35044010 DOI: 10.1002/adma.202108817] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/04/2022] [Indexed: 05/24/2023]
Abstract
Overactivated T cells and overproduced pro-inflammatory cytokines form a self-amplified signaling loop to continuously exacerbate the dysregulated inflammatory response and propel the progression of autoimmune diseases (AIDs). Herein, immuno-engineered nanodecoys (NDs) based on poly(lactic-co-glycolic acid) nanoparticles coated with programmed death-ligand 1 (PD-L1)-expressing macrophage membrane (PRM) are developed to mediate multi-target interruption of the self-promoted inflammatory cascade in AIDs. The PRM collected from IFN-γ-treated RAW 264.7 cells possesses elevated surface levels of adhesion molecule receptors and pro-inflammatory cytokine receptors, and, thus, systemically administered PRM NDs afford higher accumulation level in inflamed tissues and stronger scavenging efficiency toward multiple pro-inflammatory cytokines. More importantly, IFN-γ treatment induces remarkable PD-L1 expression on PRM, thereby allowing PRM NDs to bind membrane-bound programmed death-1 (PD-1) on CD4+ T cell surfaces or neutralize free soluble PD-1, which reconstructs the PD-1/PD-L1 inhibitory axis to suppress CD4+ T cell activation and restore immune tolerance. As such, PRM NDs provoke potent and cooperative anti-inflammatory and immune-suppressive efficacies to alleviate autoimmune damages in Zymosan A-induced arthritis mice and dextran sulfate sodium-induced ulcerative colitis mice. This study provides an enlightened example for the immuno-engineering of cell-membrane-based NDs, rendering promising implications into the treatment of AIDs via multi-target immune-modulation.
Collapse
Affiliation(s)
- Mengying Hou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yuansong Wei
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Wenqing Han
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Renxiang Zhou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yang Zhou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yiran Zheng
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
15
|
Steinle H, Weber J, Stoppelkamp S, Große-Berkenbusch K, Golombek S, Weber M, Canak-Ipek T, Trenz SM, Schlensak C, Avci-Adali M. Delivery of synthetic mRNAs for tissue regeneration. Adv Drug Deliv Rev 2021; 179:114007. [PMID: 34710530 DOI: 10.1016/j.addr.2021.114007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/03/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023]
Abstract
In recent years, nucleic acid-based therapeutics have gained increasing importance as novel treatment options for disease prevention and treatment. Synthetic messenger RNAs (mRNAs) are promising nucleic acid-based drugs to transiently express desired proteins that are missing or defective. Recently, synthetic mRNA-based vaccines encoding viral proteins have been approved for emergency use against COVID-19. Various types of vehicles, such as lipid nanoparticles (LNPs) and liposomes, are being investigated to enable the efficient uptake of mRNA molecules into desired cells. In addition, the introduction of novel chemical modifications into mRNAs increased the stability, enabled the modulation of nucleic acid-based drugs, and increased the efficiency of mRNA-based therapeutic approaches. In this review, novel and innovative strategies for the delivery of synthetic mRNA-based therapeutics for tissue regeneration are discussed. Moreover, with this review, we aim to highlight the versatility of synthetic mRNA molecules for various applications in the field of regenerative medicine and also discuss translational challenges and required improvements for mRNA-based drugs.
Collapse
Affiliation(s)
- Heidrun Steinle
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Josefin Weber
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sandra Stoppelkamp
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Katharina Große-Berkenbusch
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sonia Golombek
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Marbod Weber
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Tuba Canak-Ipek
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sarah-Maria Trenz
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Christian Schlensak
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Meltem Avci-Adali
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany.
| |
Collapse
|
16
|
Kulubya ES, Clark K, Hao D, Lazar S, Ghaffari-Rafi A, Karnati T, Ebinu JO, Zwienenberg M, Farmer DL, Wang A. The Unique Properties of Placental Mesenchymal Stromal Cells: A Novel Source of Therapy for Congenital and Acquired Spinal Cord Injury. Cells 2021; 10:2837. [PMID: 34831060 PMCID: PMC8616037 DOI: 10.3390/cells10112837] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/28/2022] Open
Abstract
Spinal cord injury (SCI) is a devasting condition with no reliable treatment. Spina bifida is the most common cause of congenital SCI. Cell-based therapies using mesenchymal stem/stromal cells (MSCS) have been largely utilized in SCI. Several clinical trials for acquired SCI use adult tissue-derived MSC sources, including bone-marrow, adipose, and umbilical cord tissues. The first stem/stromal cell clinical trial for spina bifida is currently underway (NCT04652908). The trial uses early gestational placental-derived mesenchymal stem/stromal cells (PMSCs) during the fetal repair of myelomeningocele. PMSCs have been shown to exhibit unique neuroprotective, angiogenic, and antioxidant properties, all which are promising applications for SCI. This review will summarize the unique properties and current applications of PMSCs and discuss their therapeutic role for acquired SCI.
Collapse
Affiliation(s)
- Edwin S Kulubya
- Department of Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (E.S.K.); (K.C.); (D.H.); (S.L.); (D.L.F.)
- Department of Neurological Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (A.G.-R.); (T.K.); (J.O.E.); (M.Z.)
| | - Kaitlin Clark
- Department of Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (E.S.K.); (K.C.); (D.H.); (S.L.); (D.L.F.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Dake Hao
- Department of Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (E.S.K.); (K.C.); (D.H.); (S.L.); (D.L.F.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Sabrina Lazar
- Department of Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (E.S.K.); (K.C.); (D.H.); (S.L.); (D.L.F.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Arash Ghaffari-Rafi
- Department of Neurological Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (A.G.-R.); (T.K.); (J.O.E.); (M.Z.)
| | - Tejas Karnati
- Department of Neurological Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (A.G.-R.); (T.K.); (J.O.E.); (M.Z.)
| | - Julius Okudu Ebinu
- Department of Neurological Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (A.G.-R.); (T.K.); (J.O.E.); (M.Z.)
| | - Marike Zwienenberg
- Department of Neurological Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (A.G.-R.); (T.K.); (J.O.E.); (M.Z.)
| | - Diana L Farmer
- Department of Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (E.S.K.); (K.C.); (D.H.); (S.L.); (D.L.F.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Aijun Wang
- Department of Surgery, Davis School of Medicine, University of California, Sacramento, CA 95817, USA; (E.S.K.); (K.C.); (D.H.); (S.L.); (D.L.F.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, UC Davis, Davis, CA 95616, USA
| |
Collapse
|
17
|
Downing K, Prisby R, Varanasi V, Zhou J, Pan Z, Brotto M. Old and new biomarkers for volumetric muscle loss. Curr Opin Pharmacol 2021; 59:61-69. [PMID: 34146835 DOI: 10.1016/j.coph.2021.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022]
Abstract
Volumetric muscle loss (VML) impacts skeletal muscles and causes damage to associated tissues such as blood vessels and other structural tissues. Despite progress in the VML field, current preclinical approaches are often ineffective at restoring muscle volume. Additional research is paramount to develop strategies that improve muscle mass and function, while restoring supporting tissues. We highlight mechanisms that govern normal muscle function that are also key players for VML, including intracellular calcium signaling/homeostasis, mitochondria signaling (calcium, reactiove oxidative species (ROS)/oxidative stress), and angiogenesis. We propose an integration of these processes within the context of emerging biomaterials that provide structural support for muscle regeneration. We posit that new biomarkers (i.e. myokines and lipid signaling mediators) may serve as sentinels of early muscle injury and regeneration. We conclude that as new ideas, approaches, and models come together, new treatments will emerge to allow the full rebuilding of skeletal muscles and functional recovery of skeletal muscles after VML.
Collapse
Affiliation(s)
- Kerrie Downing
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Rhonda Prisby
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Venu Varanasi
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Jingsong Zhou
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Zui Pan
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA.
| | - Marco Brotto
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA.
| |
Collapse
|
18
|
Li J, Røise JJ, He M, Das R, Murthy N. Non-viral strategies for delivering genome editing enzymes. Adv Drug Deliv Rev 2021; 168:99-117. [PMID: 32931860 DOI: 10.1016/j.addr.2020.09.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 08/02/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022]
Abstract
Genome-editing tools such as Cre recombinase (Cre), zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and most recently the clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein system have revolutionized biomedical research, agriculture, microbial engineering, and therapeutic development. Direct delivery of genome editing enzymes, as opposed to their corresponding DNA and mRNA precursors, is advantageous since they do not require transcription and/or translation. In addition, prolonged overexpression is a problem when delivering viral vector or plasmid DNA which is bypassed when delivering whole proteins. This lowers the risk of insertional mutagenesis and makes for relatively easier manufacturing. However, a major limitation of utilizing genome editing proteins in vivo is their low delivery efficiency, and currently the most successful strategy involves using potentially immunogenic viral vectors. This lack of safe and effective non-viral delivery systems is still a big hurdle for the clinical translation of such enzymes. This review discusses the challenges of non-viral delivery strategies of widely used genome editing enzymes, including Cre recombinase, ZFNs and TALENs, CRISPR/Cas9, and Cas12a (Cpf1) in their protein format and highlights recent innovations of non-viral delivery strategies which have the potential to overcome current delivery limitations and advance the clinical translation of genome editing.
Collapse
|
19
|
Zhong Y, Meng F, Zhang W, Li B, van Hest JC, Zhong Z. CD44-targeted vesicles encapsulating granzyme B as artificial killer cells for potent inhibition of human multiple myeloma in mice. J Control Release 2020; 320:421-430. [DOI: 10.1016/j.jconrel.2020.02.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/26/2020] [Accepted: 02/02/2020] [Indexed: 12/11/2022]
|
20
|
Wang G, Rayner S, Chung R, Shi B, Liang X. Advances in nanotechnology-based strategies for the treatments of amyotrophic lateral sclerosis. Mater Today Bio 2020; 6:100055. [PMID: 32529183 PMCID: PMC7280770 DOI: 10.1016/j.mtbio.2020.100055] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/09/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease (MND), is a progressive neurodegenerative disease that affects both upper and lower motor neurons, which results in loss of muscle control and eventual paralysis [1]. Currently, there are as yet unresolved challenges regarding efficient drug delivery into the central nervous system (CNS). These challenges can be attributed to multiple factors including the presence of the blood-brain barrier (BBB), blood-spinal cord barrier (BSCB), as well as the inherent characteristics of the drugs themselves (e.g. low solubility, insufficient bioavailability/bio-stability, 'off-target' effects) etc. As a result, conventional drug delivery systems may not facilitate adequate dosage of the required drugs for functional recovery in ALS patients. Nanotechnology-based strategies, however, employ engineered nanostructures that show great potential in delivering single or combined therapeutic agents to overcome the biological barriers, enhance interaction with targeted sites, improve drug bioavailability/bio-stability and achieve real-time tracking while minimizing the systemic side-effects. This review provides a concise discussion of recent advances in nanotechnology-based strategies in relation to combating specific pathophysiology relevant to ALS disease progression and investigates the future scope of using nanotechnology to develop innovative treatments for ALS patients.
Collapse
Affiliation(s)
- G.Y. Wang
- Huaihe Hospital, Henan University, Kaifeng, Henan, 475004, China
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - S.L. Rayner
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - R. Chung
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - B.Y. Shi
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - X.J. Liang
- Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
21
|
Spencer AP, Torrado M, Custódio B, Silva-Reis SC, Santos SD, Leiro V, Pêgo AP. Breaking Barriers: Bioinspired Strategies for Targeted Neuronal Delivery to the Central Nervous System. Pharmaceutics 2020; 12:E192. [PMID: 32102252 PMCID: PMC7076453 DOI: 10.3390/pharmaceutics12020192] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/01/2020] [Accepted: 02/19/2020] [Indexed: 12/23/2022] Open
Abstract
Central nervous system (CNS) disorders encompass a vast spectrum of pathological conditions and represent a growing concern worldwide. Despite the high social and clinical interest in trying to solve these pathologies, there are many challenges to bridge in order to achieve an effective therapy. One of the main obstacles to advancements in this field that has hampered many of the therapeutic strategies proposed to date is the presence of the CNS barriers that restrict the access to the brain. However, adequate brain biodistribution and neuronal cells specific accumulation in the targeted site also represent major hurdles to the attainment of a successful CNS treatment. Over the last few years, nanotechnology has taken a step forward towards the development of therapeutics in neurologic diseases and different approaches have been developed to surpass these obstacles. The versatility of the designed nanocarriers in terms of physical and chemical properties, and the possibility to functionalize them with specific moieties, have resulted in improved neurotargeted delivery profiles. With the concomitant progress in biology research, many of these strategies have been inspired by nature and have taken advantage of physiological processes to achieve brain delivery. Here, the different nanosystems and targeting moieties used to achieve a neuronal delivery reported in the open literature are comprehensively reviewed and critically discussed, with emphasis on the most recent bioinspired advances in the field. Finally, we express our view on the paramount challenges in targeted neuronal delivery that need to be overcome for these promising therapeutics to move from the bench to the bedside.
Collapse
Affiliation(s)
- Ana P. Spencer
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
| | - Marília Torrado
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Beatriz Custódio
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Sara C. Silva-Reis
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Sofia D. Santos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Victoria Leiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Ana P. Pêgo
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.S.); (M.T.); (B.C.); (S.C.S.-R.); (S.D.S.); (V.L.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
22
|
Järvinen TA, Pemmari T. Systemically Administered, Target-Specific, Multi-Functional Therapeutic Recombinant Proteins in Regenerative Medicine. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E226. [PMID: 32013041 PMCID: PMC7075297 DOI: 10.3390/nano10020226] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 12/25/2022]
Abstract
Growth factors, chemokines and cytokines guide tissue regeneration after injuries. However, their applications as recombinant proteins are almost non-existent due to the difficulty of maintaining their bioactivity in the protease-rich milieu of injured tissues in humans. Safety concerns have ruled out their systemic administration. The vascular system provides a natural platform for circumvent the limitations of the local delivery of protein-based therapeutics. Tissue selectivity in drug accumulation can be obtained as organ-specific molecular signatures exist in the blood vessels in each tissue, essentially forming a postal code system ("vascular zip codes") within the vasculature. These target-specific "vascular zip codes" can be exploited in regenerative medicine as the angiogenic blood vessels in the regenerating tissues have a unique molecular signature. The identification of vascular homing peptides capable of finding these unique "vascular zip codes" after their systemic administration provides an appealing opportunity for the target-specific delivery of therapeutics to tissue injuries. Therapeutic proteins can be "packaged" together with homing peptides by expressing them as multi-functional recombinant proteins. These multi-functional recombinant proteins provide an example how molecular engineering gives to a compound an ability to home to regenerating tissue and enhance its therapeutic potential. Regenerative medicine has been dominated by the locally applied therapeutic approaches despite these therapies are not moving to clinical medicine with success. There might be a time to change the paradigm towards systemically administered, target organ-specific therapeutic molecules in future drug discovery and development for regenerative medicine.
Collapse
Affiliation(s)
- Tero A.H. Järvinen
- Faculty of Medicine & Health Technology, Tampere University, FI-33014 Tampere, Finland & Tampere University Hospital, 33520 Tampere, Finland
| | | |
Collapse
|
23
|
Sellers DL, Tan JKY, Pineda JMB, Peeler DJ, Porubsky VL, Olden BR, Salipante SJ, Pun SH. Targeting Ligands Deliver Model Drug Cargo into the Central Nervous System along Autonomic Neurons. ACS NANO 2019; 13:10961-10971. [PMID: 31589023 PMCID: PMC7651855 DOI: 10.1021/acsnano.9b01515] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
While biologic drugs such as proteins, peptides, or nucleic acids have shown promise in the treatment of neurodegenerative diseases, the blood-brain barrier (BBB) severely limits drug delivery to the central nervous system (CNS) after systemic administration. Consequently, drug delivery challenges preclude biological drug candidates from the clinical armamentarium. In order to target drug delivery and uptake into to the CNS, we used an in vivo phage display screen to identify peptides able to target drug-uptake by the vast array of neurons of the autonomic nervous system (ANS). Using next-generation sequencing, we identified 21 candidate targeted ANS-to-CNS uptake ligands (TACL) that enriched bacteriophage accumulation and delivered protein-cargo into the CNS after intraperitoneal (IP) administration. The series of TACL peptides were synthesized and tested for their ability to deliver a model enzyme (NeutrAvidin-horseradish peroxidase fusion) to the brain and spinal cord. Three TACL-peptides facilitated significant active enzyme delivery into the CNS, with limited accumulation in off-target organs. Peptide structure and serum stability is increased when internal cysteine residues are cyclized by perfluoroarylation with decafluorobiphenyl, which increased delivery to the CNS further. TACL-peptide was demonstrated to localize in parasympathetic ganglia neurons in addition to neuronal structures in the hindbrain and spinal cord. By targeting uptake into ANS neurons, we demonstrate the potential for TACL-peptides to bypass the blood-brain barrier and deliver a model drug into the brain and spinal cord.
Collapse
Affiliation(s)
- Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - James-Kevin Y. Tan
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Julio Marco B. Pineda
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - David J. Peeler
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Veronica L. Porubsky
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Brynn R. Olden
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington, 98195, USA
| |
Collapse
|
24
|
Hernandez‐Garcia A, Álvarez Z, Simkin D, Madhan A, Pariset E, Tantakitti F, de J. Vargas‐Dorantes O, Lee SS, Kiskinis E, Stupp SI. Peptide-siRNA Supramolecular Particles for Neural Cell Transfection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801458. [PMID: 30775231 PMCID: PMC6364495 DOI: 10.1002/advs.201801458] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/21/2018] [Indexed: 05/26/2023]
Abstract
Small interfering ribonucleic acid (siRNA)-based gene knockdown is an effective tool for gene screening and therapeutics. However, the use of nonviral methods has remained an enormous challenge in neural cells. A strategy is reported to design artificial noncationic modular peptides with amplified affinity for siRNA via supramolecular assembly that shows efficient protein knockdown in neural cells. By solid phase synthesis, a sequence that binds specifically double-stranded ribonucleic acid (dsRNA) with a self-assembling peptide for particle formation is integrated. These supramolecular particles can be further functionalized with bioactive sequences without affecting their biophysical properties. The peptide carrier is found to silence efficiently up to 83% of protein expression in primary astroglial and neuronal cell cultures without cytotoxicity. In the case of neurons, a reduction in electrical activity is observed once the presynaptic protein synaptophysin is downregulated by the siRNA-peptide particles. The results demonstrate that the supramolecular particles offer an siRNA delivery platform for efficient nonviral gene screening and discovery of novel therapies for neural cells.
Collapse
Affiliation(s)
- Armando Hernandez‐Garcia
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
- Department of Chemistry of BiomacromoleculesInstitute of ChemistryNational Autonomous University of MexicoCiudad UniversitariaMexico City04510Mexico
| | - Zaida Álvarez
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
| | - Dina Simkin
- Department of PharmacologyFeinberg School of MedicineNorthwestern UniversityChicagoIL60611USA
- The Ken & Ruth Davee Department of Neurology & Clinical Neurological SciencesDepartment of PhysiologyFeinberg School of MedicineNorthwestern UniversityChicagoIL60611USA
| | - Ashwin Madhan
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
| | - Eloise Pariset
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
| | | | - Oscar de J. Vargas‐Dorantes
- Department of Chemistry of BiomacromoleculesInstitute of ChemistryNational Autonomous University of MexicoCiudad UniversitariaMexico City04510Mexico
| | - Sungsoo S. Lee
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology & Clinical Neurological SciencesDepartment of PhysiologyFeinberg School of MedicineNorthwestern UniversityChicagoIL60611USA
| | - Samuel I. Stupp
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
- Department of ChemistryNorthwestern UniversityEvanstonIL60208USA
- Department of Materials Science and EngineeringNorthwestern UniversityEvanstonIL60208USA
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIL60208USA
- Department of MedicineNorthwestern UniversityChicagoIL60611USA
| |
Collapse
|
25
|
Current state of in vivo panning technologies: Designing specificity and affinity into the future of drug targeting. Adv Drug Deliv Rev 2018; 130:39-49. [PMID: 29964079 DOI: 10.1016/j.addr.2018.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/24/2018] [Accepted: 06/22/2018] [Indexed: 11/20/2022]
Abstract
Targeting ligands are used in drug delivery to improve drug distribution to desired cells or tissues and to facilitate cellular entry. In vivo biopanning, whereby billions of potential ligand sequences are screened in biologically-relevant and complex conditions, is a powerful method for identification of novel target ligands. This tool has impacted drug delivery technologies and expanded our arsenal of therapeutics and diagnostics. Within this review we will discuss current in vivo panning technologies and ways that these technologies can be improved to advance next-generation drug delivery strategies.
Collapse
|
26
|
Järvinen TAH, Rashid J, Valmari T, May U, Ahsan F. Systemically Administered, Target-Specific Therapeutic Recombinant Proteins and Nanoparticles for Regenerative Medicine. ACS Biomater Sci Eng 2017; 3:1273-1282. [DOI: 10.1021/acsbiomaterials.6b00746] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Tero A. H. Järvinen
- Faculty
of Medicine and Life Sciences, University of Tampere, Lääkärinkatu
1, 33014 Tampere, Finland
- Department of Orthopedics & Traumatology, Tampere University Hospital, Teiskontie 35, 33520 Tampere, Finland
| | - Jahidur Rashid
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 Coulter Street, Amarillo, Texas 79106, United States
| | - Toini Valmari
- Faculty
of Medicine and Life Sciences, University of Tampere, Lääkärinkatu
1, 33014 Tampere, Finland
| | - Ulrike May
- Faculty
of Medicine and Life Sciences, University of Tampere, Lääkärinkatu
1, 33014 Tampere, Finland
| | - Fakhrul Ahsan
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 Coulter Street, Amarillo, Texas 79106, United States
| |
Collapse
|
27
|
Tan JKY, Sellers DL, Pham B, Pun SH, Horner PJ. Non-Viral Nucleic Acid Delivery Strategies to the Central Nervous System. Front Mol Neurosci 2016; 9:108. [PMID: 27847462 PMCID: PMC5088201 DOI: 10.3389/fnmol.2016.00108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022] Open
Abstract
With an increased prevalence and understanding of central nervous system (CNS) injuries and neurological disorders, nucleic acid therapies are gaining promise as a way to regenerate lost neurons or halt disease progression. While more viral vectors have been used clinically as tools for gene delivery, non-viral vectors are gaining interest due to lower safety concerns and the ability to deliver all types of nucleic acids. Nevertheless, there are still a number of barriers to nucleic acid delivery. In this focused review, we explore the in vivo challenges hindering non-viral nucleic acid delivery to the CNS and the strategies and vehicles used to overcome them. Advantages and disadvantages of different routes of administration including: systemic injection, cerebrospinal fluid injection, intraparenchymal injection and peripheral administration are discussed. Non-viral vehicles and treatment strategies that have overcome delivery barriers and demonstrated in vivo gene transfer to the CNS are presented. These approaches can be used as guidelines in developing synthetic gene delivery vectors for CNS applications and will ultimately bring non-viral vectors closer to clinical application.
Collapse
Affiliation(s)
- James-Kevin Y Tan
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington Seattle, WA, USA
| | - Drew L Sellers
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington Seattle, WA, USA
| | - Binhan Pham
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington Seattle, WA, USA
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington Seattle, WA, USA
| | - Philip J Horner
- Center for Neuroregenerative Medicine, Houston Methodist Research Institute Houston, TX, USA
| |
Collapse
|