1
|
Jakka SR, Mugesh G. Emerging Role of Noncovalent Interactions and Disulfide Bond Formation in the Cellular Uptake of Small Molecules and Proteins. Chem Asian J 2025:e202401734. [PMID: 39831847 DOI: 10.1002/asia.202401734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 01/22/2025]
Abstract
Intracellular delivery of proteins and small molecules is an important barrier in the development of strategies to deliver functional proteins and therapeutics into the cells to realize their full potential in biotechnology, biomedicine, cell-based therapies, and gene editing protein systems. Most of the intracellular protein delivery strategies involve the conjugation of cell penetrating peptides to enable the permeability of plasma membrane of mammalian cells to allow proteins to enter cytosol. The conjugations of small molecules such as (p-methylphenyl) glycine, pyrenebutyrate and cysteines are used for the same purpose. Molecular level interactions are governed mostly by ionic (cationic/anionic), covalent and noncovalent interactions with various molecular entities of glycocalyx matrix on plasma membrane lipid bilayer. Although the role of noncovalent interactions in cellular uptake is not fully understood, several recent advances have focused on the noncovalent interaction-based strategies of intracellular delivery of small molecules and proteins into mammalian cells. These are achieved by simple modification of protein surfaces with chemical moieties which can form noncovalent interactions other than hydrogen bonding. In this review, we describe the recent advances and the mechanistic aspects of intracellular delivery and role of noncovalent interactions in the cellular uptake of proteins and small molecules.
Collapse
Affiliation(s)
- Surendar R Jakka
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560 012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560 012, India
| |
Collapse
|
2
|
de Pellegars-Malhortie A, Picque Lasorsa L, Mazard T, Granier F, Prévostel C. Why Is Wnt/β-Catenin Not Yet Targeted in Routine Cancer Care? Pharmaceuticals (Basel) 2024; 17:949. [PMID: 39065798 PMCID: PMC11279613 DOI: 10.3390/ph17070949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Despite significant progress in cancer prevention, screening, and treatment, the still limited number of therapeutic options is an obstacle towards increasing the cancer cure rate. In recent years, many efforts were put forth to develop therapeutics that selectively target different components of the oncogenic Wnt/β-catenin signaling pathway. These include small molecule inhibitors, antibodies, and more recently, gene-based approaches. Although some of them showed promising outcomes in clinical trials, the Wnt/β-catenin pathway is still not targeted in routine clinical practice for cancer management. As for most anticancer treatments, a critical limitation to the use of Wnt/β-catenin inhibitors is their therapeutic index, i.e., the difficulty of combining effective anticancer activity with acceptable toxicity. Protecting healthy tissues from the effects of Wnt/β-catenin inhibitors is a major issue due to the vital role of the Wnt/β-catenin signaling pathway in adult tissue homeostasis and regeneration. In this review, we provide an up-to-date summary of clinical trials on Wnt/β-catenin pathway inhibitors, examine their anti-tumor activity and associated adverse events, and explore strategies under development to improve the benefit/risk profile of this therapeutic approach.
Collapse
Affiliation(s)
- Auriane de Pellegars-Malhortie
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| | - Laurence Picque Lasorsa
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| | - Thibault Mazard
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
- Medical Oncology Department, ICM, University of Montpellier, CEDEX 5, 34298 Montpellier, France
| | | | - Corinne Prévostel
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| |
Collapse
|
3
|
Graceffa V. Intracellular protein delivery: New insights into the therapeutic applications and emerging technologies. Biochimie 2023; 213:82-99. [PMID: 37209808 DOI: 10.1016/j.biochi.2023.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
The inability to cross the plasma membranes traditionally limited the therapeutic use of recombinant proteins. However, in the last two decades, novel technologies made delivering proteins inside the cells possible. This allowed researchers to unlock intracellular targets, once considered 'undruggable', bringing a new research area to emerge. Protein transfection systems display a large potential in a plethora of applications. However, their modality of action is often unclear, and cytotoxic effects are elevated, whereas experimental conditions to increase transfection efficacy and cell viability still need to be identified. Furthermore, technical complexity often limits in vivo experimentation, while challenging industrial and clinical translation. This review highlights the applications of protein transfection technologies, and then critically discuss the current methodologies and their limitations. Physical membrane perforation systems are compared to systems exploiting cellular endocytosis. Research evidence of the existence of either extracellular vesicles (EVs) or cell-penetrating peptides (CPPs)- based systems, that circumvent the endosomal systems is critically analysed. Commercial systems, novel solid-phase reverse protein transfection systems, and engineered living intracellular bacteria-based mechanisms are finally described. This review ultimately aims at finding new methodologies and possible applications of protein transfection systems, while helping the development of an evidence-based research approach.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Centre for Mathematical Modelling and Intelligent Systems for Health and Environment (MISHE), Atlantic Technological University (ATU), Sligo, Ireland.
| |
Collapse
|
4
|
Lalzawmliana V, Mukherjee P, Roy S, Roy M, Nandi SK. Ceramic Biomaterials in Advanced Biomedical Applications. FUNCTIONAL BIOMATERIALS 2022:371-408. [DOI: 10.1007/978-981-16-7152-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Zhang T, Gao Y, Cui W, Li Y, Xiao D, Zhou R. Nanomaterials-based Cell Osteogenic Differentiation and Bone Regeneration. Curr Stem Cell Res Ther 2020; 16:36-47. [PMID: 32436831 DOI: 10.2174/1574888x15666200521083834] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/07/2020] [Accepted: 01/16/2020] [Indexed: 02/08/2023]
Abstract
With the rapid development of nanotechnology, various nanomaterials have been applied to bone repair and regeneration. Due to the unique chemical, physical and mechanical properties, nanomaterials could promote stem cells osteogenic differentiation, which has great potentials in bone tissue engineering and exploiting nanomaterials-based bone regeneration strategies. In this review, we summarized current nanomaterials with osteo-induction ability, which could be potentially applied to bone tissue engineering. Meanwhile, the unique properties of these nanomaterials and their effects on stem cell osteogenic differentiation are also discussed. Furthermore, possible signaling pathways involved in the nanomaterials- induced cell osteogenic differentiation are also highlighted in this review.
Collapse
Affiliation(s)
- Tianxu Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yang Gao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Weitong Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yanjing Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Tamemoto N, Akishiba M, Sakamoto K, Kawano K, Noguchi H, Futaki S. Rational Design Principles of Attenuated Cationic Lytic Peptides for Intracellular Delivery of Biomacromolecules. Mol Pharm 2020; 17:2175-2185. [DOI: 10.1021/acs.molpharmaceut.0c00312] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Naoki Tamemoto
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- Institute for Solid State Physics, University of Tokyo, Kashiwa, Chiba 277-8581, Japan
| | - Misao Akishiba
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Kentarou Sakamoto
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Kenichi Kawano
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Hiroshi Noguchi
- Institute for Solid State Physics, University of Tokyo, Kashiwa, Chiba 277-8581, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| |
Collapse
|
7
|
Sasaki Y, Tsuchido Y, Yoshimura T, Akiyoshi K. Nanogelation and Thermal Stabilization of Enzyme by Vitamin B 6-Bearing Polysaccharide as Biocrosslinker. ACS Biomater Sci Eng 2019; 5:5752-5758. [PMID: 33405708 DOI: 10.1021/acsbiomaterials.9b00026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nanogels containing a protein (carbonic anhydrase, CA) were prepared by cross-linking CA and pyridoxal (vitamin B6)-bearing pullulan (PLPP) as a biocrosslinker via Schiff base formation. UV titration and high-performance liquid chromatography confirmed that CA was quantitatively complexed with PLPP in the presence of zinc ions. Dynamic light scattering and transmission electron microscopy showed that the nanogel diameter was about 20 nm. CA retained 90% of its native activity after complexation with PLPP. Moreover, the residual enzymatic activity of CA after heating and its long-term storage stability at room temperature were improved by complexation with PLPP. Enzyme nanogelation with PLPP is an efficient method for enzyme stabilization.
Collapse
Affiliation(s)
- Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 606-8501, Japan
| | - Yuji Tsuchido
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioicho, Chiyoda, Tokyo 102-8554, Japan
| | - Takahiro Yoshimura
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 606-8501, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 606-8501, Japan.,The Exploratory Research for Advanced Technology (ERATO), Japan Science and Technology Agency (JST), K's Goban-cho bldg., 7 Goban-cho, chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
8
|
Qin X, Yu C, Wei J, Li L, Zhang C, Wu Q, Liu J, Yao SQ, Huang W. Rational Design of Nanocarriers for Intracellular Protein Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902791. [PMID: 31496027 DOI: 10.1002/adma.201902791] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/04/2019] [Indexed: 06/10/2023]
Abstract
Protein/antibody therapeutics have exhibited the advantages of high specificity and activity even at an extremely low concentration compared to small molecule drugs. However, they are accompanied by unfavorable physicochemical properties such as fragile tertiary structure, large molecular size, and poor penetration of the membrane, and thus the clinical use of protein drugs is hindered by inefficient delivery of proteins into the host cells. To overcome the challenges associated with protein therapeutics and enhance their biopharmaceutical applications, various protein-loaded nanocarriers with desired functions, such as lipid nanocapsules, polymeric nanoparticles, inorganic nanoparticles, and peptides, are developed. In this review, the different strategies for intracellular delivery of proteins are comprehensively summarized. Their designed routes, mechanisms of action, and potential therapeutics in live cells or in vivo are discussed in detail. Furthermore, the perspective on the new generation of delivery systems toward the emerging area of protein-based therapeutics is presented as well.
Collapse
Affiliation(s)
- Xiaofei Qin
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Changmin Yu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Jing Wei
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Chengwu Zhang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Qiong Wu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Jinhua Liu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
- Shaanxi Institute of Flexible Electronics (SIFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, P. R. China
| |
Collapse
|
9
|
Lv J, Fan Q, Wang H, Cheng Y. Polymers for cytosolic protein delivery. Biomaterials 2019; 218:119358. [DOI: 10.1016/j.biomaterials.2019.119358] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/11/2019] [Accepted: 07/13/2019] [Indexed: 12/31/2022]
|
10
|
Abstract
Nanostructured devices are able to foster the technology for cell membrane poration. With the size smaller than a cell, nanostructures allow efficient poration on the cell membrane. Emerging nanostructures with various physical transduction have been demonstrated to accommodate effective intracellular delivery. Aside from improving poration and intracellular delivery performance, nanostructured devices also allow for the discovery of novel physiochemical phenomena and the biological response of the cell. This article provides a brief introduction to the principles of nanostructured devices for cell poration and outlines the intracellular delivery capability of the technology. In the future, we envision more exploration on new nanostructure designs and creative applications in biomedical fields.
Collapse
Affiliation(s)
- Apresio K Fajrial
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80309 United States of America
| | | |
Collapse
|
11
|
Yun CK, Hwang JW, Kwak TJ, Chang WJ, Ha S, Han K, Lee S, Choi YS. Nanoinjection system for precise direct delivery of biomolecules into single cells. LAB ON A CHIP 2019; 19:580-588. [PMID: 30623953 DOI: 10.1039/c8lc00709h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Intracellular delivery of functional molecules such as proteins, transcription factors and DNA is effective and promising in cell biology. However, existing transfection methods are often unsuitable to deliver big molecules into cells or require carriers such as viruses and peptides specific to the target molecules. In addition, the nature of bulk processing does not generally provide accurate dose control of individual cells. The concept of single-cell-based material injection based on electrokinetic pumping through nanocapillaries could overcome these problems, yet the fabrication and operation of nanoscale 3-dimensional structures have remained unsolved. In this research, a hybrid (PDMS/glass) microfluidic chip with a true 3-dimensional nanoinjection structure (called "nanoinjection system") is presented. The nanoinjection structure was fabricated by femtosecond-laser (fs-laser) ablation in a single solid glass, which showed very successful delivery of red fluorescent protein (RFP) and expression of plasmid DNA in several different types of cells. This system is promising in that the amount of molecules to be delivered is controllable and the processed cells are systematically separated into a harvesting chamber, which can radically improve the purity of the processed cells. In addition, it was confirmed that the cells were healthy even after the molecule injection for a few seconds, indicating that the injection time can be significantly elongated, further improving the delivery efficiency of biomolecules without affecting the cell viability. We envision that the nanoinjection system having the major features of being carrier-free and dose-controllable, having an unlimited injection period, and ease of harvesting will greatly contribute to the next-generation research studies in the fields of cell biology and cell therapeutics.
Collapse
Affiliation(s)
- Chang-Koo Yun
- Department of Biotechnology, CHA University, 335 Pankyoro, Bundang-gu, Seongnam, Gyeonggi-do 13488, Republic of Korea. and Department of Mechanical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Jung Wook Hwang
- Department of Biotechnology, CHA University, 335 Pankyoro, Bundang-gu, Seongnam, Gyeonggi-do 13488, Republic of Korea.
| | - Tae Joon Kwak
- Department of Mechanical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Woo-Jin Chang
- Department of Mechanical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Sungjae Ha
- Femtobiomed Inc., Seongnam, 13487, Republic of Korea.
| | - Kyuboem Han
- Paean Biotechnology Inc., Daejeon, 34028, Republic of Korea
| | - Sanghyun Lee
- Femtobiomed Inc., Seongnam, 13487, Republic of Korea.
| | - Yong-Soo Choi
- Department of Biotechnology, CHA University, 335 Pankyoro, Bundang-gu, Seongnam, Gyeonggi-do 13488, Republic of Korea.
| |
Collapse
|
12
|
Garcia-Hernandez C, Freese AK, Rodriguez-Mendez ML, Wanekaya AK. In situ synthesis, stabilization and activity of protein-modified gold nanoparticles for biological applications. Biomater Sci 2019; 7:2511-2519. [DOI: 10.1039/c9bm00129h] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We demonstrate an in situ synthesis, stabilization and activity of a nanoparticle-based protein carrier platform via the Layer-by-Layer (LbL) technology.
Collapse
Affiliation(s)
- Celia Garcia-Hernandez
- Chemistry Department
- Missouri State University
- 901 S. National Ave
- USA
- Group UVASENS. Chemistry Department
| | | | | | - Adam K. Wanekaya
- Chemistry Department
- Missouri State University
- 901 S. National Ave
- USA
| |
Collapse
|
13
|
Park M, Xu X, Min W, Sugiman-Marangos SN, Beilhartz GL, Adams JJ, Sidhu SS, Grunebaum E, Melnyk RA. Intracellular Delivery of Human Purine Nucleoside Phosphorylase by Engineered Diphtheria Toxin Rescues Function in Target Cells. Mol Pharm 2018; 15:5217-5226. [DOI: 10.1021/acs.molpharmaceut.8b00735] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
| | | | | | | | | | - Jarret J. Adams
- Banting and Best Department of Medical Research, Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Sachdev S. Sidhu
- Banting and Best Department of Medical Research, Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Eyal Grunebaum
- Division of Immunology and Allergy, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | | |
Collapse
|
14
|
Liu XY, Zhou CB, Fang C. Nanomaterial-involved neural stem cell research: Disease treatment, cell labeling, and growth regulation. Biomed Pharmacother 2018; 107:583-597. [PMID: 30114642 DOI: 10.1016/j.biopha.2018.08.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) have been widely investigated for their potential in the treatment of various diseases and transplantation therapy. However, NSC growth regulation, labeling, and its application to disease diagnosis and treatment are outstanding challenges. Recently, nanomaterials have shown promise for various applications including genetic modification, imaging, and controlled drug release. Here we summarize the recent progress in the use of nanomaterials in combination with NSCs for disease treatment and diagnosis, cell labeling, and NSC growth regulation. The toxicity of nanomaterials to NSCs is also discussed.
Collapse
Affiliation(s)
- Xiang-Yu Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Cheng-Bin Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240 China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
15
|
Rudramurthy GR, Swamy MK. Potential applications of engineered nanoparticles in medicine and biology: an update. J Biol Inorg Chem 2018; 23:1185-1204. [PMID: 30097748 DOI: 10.1007/s00775-018-1600-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/26/2018] [Indexed: 12/22/2022]
Abstract
Nanotechnology advancements have led to the development of its allied fields, such as nanoparticle synthesis and their applications in the field of biomedicine. Nanotechnology driven innovations have given a hope to the patients as well as physicians in solving the complex medical problems. Nanoparticles with a size ranging from 0.2 to 100 nm are associated with an increased surface to volume ratio. Moreover, the physico-chemical and biological properties of nanoparticles can be modified depending on the applications. Different nanoparticles have been documented with a wide range of applications in various fields of medicine and biology including cancer therapy, drug delivery, tissue engineering, regenerative medicine, biomolecules detection, and also as antimicrobial agents. However, the development of stable and effective nanoparticles requires a profound knowledge on both physico-chemical features of nanomaterials and their intended applications. Further, the health risks associated with the use of engineered nanoparticles needs a serious attention.
Collapse
Affiliation(s)
| | - Mallappa Kumara Swamy
- Department of Crop Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
16
|
Zhang W, Wang W, Yu DX, Xiao Z, He Z. Application of nanodiagnostics and nanotherapy to CNS diseases. Nanomedicine (Lond) 2018; 13:2341-2371. [PMID: 30088440 DOI: 10.2217/nnm-2018-0163] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease, Parkinson's disease and stroke are the most common CNS diseases, all characterized by progressive cellular dysfunction and death in specific areas of the nervous system. Therapeutic development for these diseases has lagged behind other disease areas due to difficulties in early diagnosis, long disease courses and drug delivery challenges, not least due to the blood-brain barrier. Over recent decades, nanotechnology has been explored as a potential tool for the diagnosis, treatment and monitoring of CNS diseases. In this review, we describe the application of nanotechnology to common CNS diseases, highlighting disease pathogenesis and the underlying mechanisms and promising functional outcomes that make nanomaterials ideal candidates for early diagnosis and therapy. Moreover, we discuss the limitations of nanotechnology, and possible solutions.
Collapse
Affiliation(s)
- Weiyuan Zhang
- Yunnan Key Laboratory of Stem Cell & Regenerative Medicine, Institute of Molecular & Clinical Medicine, Kunming Medical University, Kunming 650500, PR China
| | - Wenyue Wang
- Department of Anatomy & Developmental Biology, Monash University, Clayton, 3800 Clayton, Melbourne 3800, Australia
| | - David X Yu
- Department of Anatomy & Developmental Biology, Monash University, Clayton, 3800 Clayton, Melbourne 3800, Australia
| | - Zhicheng Xiao
- Department of Anatomy & Developmental Biology, Monash University, Clayton, 3800 Clayton, Melbourne 3800, Australia
| | - Zhiyong He
- Yunnan Key Laboratory of Stem Cell & Regenerative Medicine, Institute of Molecular & Clinical Medicine, Kunming Medical University, Kunming 650500, PR China.,Department of Anatomy & Developmental Biology, Monash University, Clayton, 3800 Clayton, Melbourne 3800, Australia
| |
Collapse
|
17
|
Zhang B, Yan W, Zhu Y, Yang W, Le W, Chen B, Zhu R, Cheng L. Nanomaterials in Neural-Stem-Cell-Mediated Regenerative Medicine: Imaging and Treatment of Neurological Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705694. [PMID: 29543350 DOI: 10.1002/adma.201705694] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/17/2017] [Indexed: 05/24/2023]
Abstract
Patients are increasingly being diagnosed with neuropathic diseases, but are rarely cured because of the loss of neurons in damaged tissues. This situation creates an urgent clinical need to develop alternative treatment strategies for effective repair and regeneration of injured or diseased tissues. Neural stem cells (NSCs), highly pluripotent cells with the ability of self-renewal and potential for multidirectional differentiation, provide a promising solution to meet this demand. However, some serious challenges remaining to be addressed are the regulation of implanted NSCs, tracking their fate, monitoring their interaction with and responsiveness to the tissue environment, and evaluating their treatment efficacy. Nanomaterials have been envisioned as innovative components to further empower the field of NSC-based regenerative medicine, because their unique physicochemical characteristics provide unparalleled solutions to the imaging and treatment of diseases. By building on the advantages of nanomaterials, tremendous efforts have been devoted to facilitate research into the clinical translation of NSC-based therapy. Here, recent work on emerging nanomaterials is highlighted and their performance in the imaging and treatment of neurological diseases is evaluated, comparing the strengths and weaknesses of various imaging modalities currently used. The underlying mechanisms of therapeutic efficacy are discussed, and future research directions are suggested.
Collapse
Affiliation(s)
- Bingbo Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
- Department of Spine Surgery, Tongji Hospital, Institute of Spine and Spinal Cord Injury, Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Wei Yan
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory of Green Preparation and Application for Functional Materials, Ministry of Education, School of Materials Science & Engineering, Hubei University, Wuhan, 430062, China
| | - Yanjing Zhu
- Department of Spine Surgery, Tongji Hospital, Institute of Spine and Spinal Cord Injury, Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Weitao Yang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
| | - Wenjun Le
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
| | - Bingdi Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
| | - Rongrong Zhu
- Department of Spine Surgery, Tongji Hospital, Institute of Spine and Spinal Cord Injury, Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Liming Cheng
- Department of Spine Surgery, Tongji Hospital, Institute of Spine and Spinal Cord Injury, Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
18
|
Sun D, Gong L, Xie J, He X, Chen S, A L, Li Q, Gu Z, Xu H. Evaluating the toxicity of silicon dioxide nanoparticles on neural stem cells using RNA-Seq. RSC Adv 2017. [DOI: 10.1039/c7ra09512k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neural stem cells are characterized by self-renewal and multipotency, and a capacity to regenerate in response to brain injury or neurodegenerative disease.
Collapse
Affiliation(s)
- Dayu Sun
- Southwest Hospital/Southwest Eye Hospital
- Third Military Medical University
- Chongqing 400038
- China
- Department of Physiology
| | - Linji Gong
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
- Institute of High Energy Physics
- Chinese Academy of Sciences
- Beijing 100049
- China
| | - Jing Xie
- Southwest Hospital/Southwest Eye Hospital
- Third Military Medical University
- Chongqing 400038
- China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing
| | - Xiao He
- Southwest Hospital/Southwest Eye Hospital
- Third Military Medical University
- Chongqing 400038
- China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing
| | - Siyu Chen
- Southwest Hospital/Southwest Eye Hospital
- Third Military Medical University
- Chongqing 400038
- China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing
| | - Luodan A
- Southwest Hospital/Southwest Eye Hospital
- Third Military Medical University
- Chongqing 400038
- China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing
| | - Qiyou Li
- Southwest Hospital/Southwest Eye Hospital
- Third Military Medical University
- Chongqing 400038
- China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing
| | - Zhanjun Gu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
- Institute of High Energy Physics
- Chinese Academy of Sciences
- Beijing 100049
- China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital
- Third Military Medical University
- Chongqing 400038
- China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing
| |
Collapse
|
19
|
Brodin JD, Sprangers AJ, McMillan JR, Mirkin CA. DNA-Mediated Cellular Delivery of Functional Enzymes. J Am Chem Soc 2015; 137:14838-41. [PMID: 26587747 DOI: 10.1021/jacs.5b09711] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We report a strategy for creating a new class of protein transfection materials composed of a functional protein core chemically modified with a dense shell of oligonucleotides. These materials retain the native structure and catalytic ability of the hydrolytic enzyme β-galactosidase, which serves as the protein core, despite the functionalization of its surface with ∼25 DNA strands. The covalent attachment of a shell of oligonucleotides to the surface of β-galactosidase enhances its cellular uptake of by up to ∼280-fold and allows for the use of working concentrations as low as 100 pM enzyme. DNA-functionalized β-galactosidase retains its ability to catalyze the hydrolysis of β-glycosidic linkages once endocytosed, whereas equal concentrations of protein show little to no intracellular catalytic activity.
Collapse
Affiliation(s)
- Jeffrey D Brodin
- International Institute of Nanotechnology, ‡Department of Chemistry, and §Department of Biomedical Engineering, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Anthony J Sprangers
- International Institute of Nanotechnology, ‡Department of Chemistry, and §Department of Biomedical Engineering, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Janet R McMillan
- International Institute of Nanotechnology, ‡Department of Chemistry, and §Department of Biomedical Engineering, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- International Institute of Nanotechnology, ‡Department of Chemistry, and §Department of Biomedical Engineering, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
20
|
Walmsley GG, McArdle A, Tevlin R, Momeni A, Atashroo D, Hu MS, Feroze AH, Wong VW, Lorenz PH, Longaker MT, Wan DC. Nanotechnology in bone tissue engineering. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2015; 11:1253-63. [PMID: 25791811 PMCID: PMC4476906 DOI: 10.1016/j.nano.2015.02.013] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 11/23/2014] [Accepted: 02/21/2015] [Indexed: 02/04/2023]
Abstract
Nanotechnology represents a major frontier with potential to significantly advance the field of bone tissue engineering. Current limitations in regenerative strategies include impaired cellular proliferation and differentiation, insufficient mechanical strength of scaffolds, and inadequate production of extrinsic factors necessary for efficient osteogenesis. Here we review several major areas of research in nanotechnology with potential implications in bone regeneration: 1) nanoparticle-based methods for delivery of bioactive molecules, growth factors, and genetic material, 2) nanoparticle-mediated cell labeling and targeting, and 3) nano-based scaffold construction and modification to enhance physicochemical interactions, biocompatibility, mechanical stability, and cellular attachment/survival. As these technologies continue to evolve, ultimate translation to the clinical environment may allow for improved therapeutic outcomes in patients with large bone deficits and osteodegenerative diseases. FROM THE CLINICAL EDITOR Traditionally, the reconstruction of bony defects has relied on the use of bone grafts. With advances in nanotechnology, there has been significant development of synthetic biomaterials. In this article, the authors provided a comprehensive review on current research in nanoparticle-based therapies for bone tissue engineering, which should be useful reading for clinicians as well as researchers in this field.
Collapse
Affiliation(s)
- Graham G Walmsley
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Adrian McArdle
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ruth Tevlin
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Arash Momeni
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - David Atashroo
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael S Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Abdullah H Feroze
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Victor W Wong
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter H Lorenz
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
21
|
Jabbari E. Nanoparticles for Stem‐Cell Engineering. STEM‐CELL NANOENGINEERING 2015:143-169. [DOI: 10.1002/9781118540640.ch9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
22
|
Ashjari M. Delivery of Molecules and Genes/Small Interfering RNA into Stem Cells by Nanoengineering. STEM‐CELL NANOENGINEERING 2015:223-242. [DOI: 10.1002/9781118540640.ch13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
23
|
Wang Y, Santos A, Evdokiou A, Losic D. An overview of nanotoxicity and nanomedicine research: principles, progress and implications for cancer therapy. J Mater Chem B 2015; 3:7153-7172. [DOI: 10.1039/c5tb00956a] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The studies of nanomaterial-based drug delivery and nanotoxicity are closely interconnected.
Collapse
Affiliation(s)
- Ye Wang
- School of Chemical Engineering
- The University of Adelaide
- 5005 Adelaide
- Australia
- School of Medicine
| | - Abel Santos
- School of Chemical Engineering
- The University of Adelaide
- 5005 Adelaide
- Australia
| | - Andreas Evdokiou
- School of Medicine
- Discipline of Surgery
- The University of Adelaide
- Australia
| | - Dusan Losic
- School of Chemical Engineering
- The University of Adelaide
- 5005 Adelaide
- Australia
| |
Collapse
|
24
|
Fu A, Tang R, Hardie J, Farkas M, Rotello VM. Promises and pitfalls of intracellular delivery of proteins. Bioconjug Chem 2014; 25:1602-8. [PMID: 25133522 PMCID: PMC4166028 DOI: 10.1021/bc500320j] [Citation(s) in RCA: 241] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 08/10/2014] [Indexed: 02/07/2023]
Abstract
The direct delivery of functional proteins into the cell cytosol is a key issue for protein therapy, with many current strategies resulting in endosomal entrapment. Protein delivery to the cytosol is challenging due to the high molecular weight and the polarity of therapeutic proteins. Here we review strategies for the delivery of proteins into cells, including cell-penetrating peptides, virus-like particles, supercharged proteins, nanocarriers, polymers, and nanoparticle-stabilized nanocapsules. The advantages and disadvantages of these approaches including cytosolar delivery are compared and contrasted, with promising pathways forward identified.
Collapse
Affiliation(s)
- Ailing Fu
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- School
of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Rui Tang
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Joseph Hardie
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Michelle
E. Farkas
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Vincent M. Rotello
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
25
|
Hsu BB, Jamieson KS, Hagerman SR, Holler E, Ljubimova JY, Hammond PT. Ordered and kinetically discrete sequential protein release from biodegradable thin films. Angew Chem Int Ed Engl 2014; 53:8093-8. [PMID: 24938739 PMCID: PMC4387866 DOI: 10.1002/anie.201403702] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Indexed: 11/09/2022]
Abstract
Multidrug regimens can sometimes treat recalcitrant diseases when single-drug therapies fail. Recapitulating complex multidrug administration from controlled release films for localized delivery remains challenging because their release kinetics are frequently intertwined, and an initial burst release of each drug is usually uncontrollable. Kinetic control over protein release is demonstrated by cross-linking layer-by-layer films during the assembly process. We used biodegradable and naturally derived components and relied on copper-free click chemistry for bioorthogonal covalent cross-links throughout the film that entrap but do not modify the embedded protein. We found that this strategy restricted the interdiffusion of protein while maintaining its activity. By depositing a barrier layer and a second protein-containing layer atop this construct, we generated well-defined sequential protein release with minimal overlap that follows their spatial distribution within the film.
Collapse
Affiliation(s)
- Bryan B. Hsu
- Koch Institute for Integrative Cancer Research and the Institute for Soldier Nanotechnologies, Massachusetts Institute for Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA, Department of Chemistry, Massachusetts Institute for Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
| | - Kelsey S. Jamieson
- Department of Chemical Engineering, Massachusetts Institute for Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
| | - Samantha R. Hagerman
- Department of Chemical Engineering, Massachusetts Institute for Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
| | - Eggehard Holler
- Nanomedicine Research Center; Department of Neurosurgery, Cedars Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA
| | - Julia Y. Ljubimova
- Nanomedicine Research Center; Department of Neurosurgery, Cedars Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 USA
| | - Paula T. Hammond
- Koch Institute for Integrative Cancer Research and the Institute for Soldier Nanotechnologies, Massachusetts Institute for Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA, Department of Chemical Engineering, Massachusetts Institute for Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
| |
Collapse
|
26
|
A novel strategy for the treatment of chronic wounds based on the topical administration of rhEGF-loaded lipid nanoparticles: In vitro bioactivity and in vivo effectiveness in healing-impaired db/db mice. J Control Release 2014; 185:51-61. [DOI: 10.1016/j.jconrel.2014.04.032] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/15/2014] [Accepted: 04/18/2014] [Indexed: 12/29/2022]
|
27
|
Ordered and Kinetically Discrete Sequential Protein Release from Biodegradable Thin Films. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201403702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
28
|
Yu X, Khalil A, Dang PN, Alsberg E, Murphy WL. Multilayered Inorganic Microparticles for Tunable Dual Growth Factor Delivery. ADVANCED FUNCTIONAL MATERIALS 2014; 24:3082-3093. [PMID: 25342948 PMCID: PMC4204399 DOI: 10.1002/adfm.201302859] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
There is an increasing need to control the type, quantity, and timing of growth factors released during tissue healing. Sophisticated delivery systems offering the ability to deliver multiple growth factors with independently tunable kinetics are highly desirable. Here, a multilayered, mineral coated micro-particle (MCMs) platform that can serve as an adaptable dual growth factor delivery system is developed. Bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) are bound to the mineral coatings with high binding efficiencies of up to 80%. BMP-2 is firstly bound onto a 1st mineral coating layer; then VEGF is bound onto a 2nd mineral coating layer. The release of BMP-2 is sustained over a period of 50 days while the release of VEGF is a typical two-phase release with rapid release in the first 14 days and more sustained release for the following 36 days. Notably, the release behaviors of both growth factors can be independently tailored by changing the intrinsic properties of the mineral coatings. Furthermore, the release of BMP-2 can be tuned by changing the thickness of the 2nd layer. This injectable microparticle based delivery platform with tunable growth factor release has immense potential for applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Xiaohua Yu
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Andrew Khalil
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Phuong Ngoc Dang
- Department of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- AO Foundation Collaborative Research Center, Clavadelerstrasse 8, Davos, 7270, Switzerland
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
- AO Foundation Collaborative Research Center, Clavadelerstrasse 8, Davos, 7270, Switzerland
- Department of Orthopedics and Rehabilitation, 1300 University Ave, University of Wisconsin, Madison, WI, 53705, USA
| |
Collapse
|
29
|
Tonga GY, Saha K, Rotello VM. 25th anniversary article: interfacing nanoparticles and biology: new strategies for biomedicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2014; 26:359-70. [PMID: 24105763 PMCID: PMC4067239 DOI: 10.1002/adma.201303001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/02/2013] [Indexed: 05/09/2023]
Abstract
The exterior surface of nanoparticles (NPs) dictates the behavior of these systems with the outside world. Understanding the interactions of the NP surface functionality with biosystems enables the design and fabrication of effective platforms for therapeutics, diagnostics, and imaging agents. In this review, we highlight the role of chemistry in the engineering of nanomaterials, focusing on the fundamental role played by surface chemistry in controlling the interaction of NPs with proteins and cells.
Collapse
|
30
|
Vrana NE, Erdemli O, Francius G, Fahs A, Rabineau M, Debry C, Tezcaner A, Keskin D, Lavalle P. Double entrapment of growth factors by nanoparticles loaded into polyelectrolyte multilayer films. J Mater Chem B 2014; 2:999-1008. [DOI: 10.1039/c3tb21304h] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
31
|
Huang Y, Luo Q, Zha G, Zhang J, Li X, Zhao S, Li X. Biomimetic ECM coatings for controlled release of rhBMP-2: construction and biological evaluation. Biomater Sci 2014; 2:980-989. [DOI: 10.1039/c3bm60254k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
32
|
Costa RR, Mano JF. Polyelectrolyte multilayered assemblies in biomedical technologies. Chem Soc Rev 2014; 43:3453-79. [DOI: 10.1039/c3cs60393h] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
33
|
Choi DH, Subbiah R, Kim IH, Han DK, Park K. Dual growth factor delivery using biocompatible core-shell microcapsules for angiogenesis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:3468-3476. [PMID: 23585380 DOI: 10.1002/smll.201300427] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Indexed: 06/02/2023]
Abstract
An optimized electrodropping system produces homogeneous core-shell microcapsules (C-S MCs) by using poly(L-lactic-co-glycolic acid) (PLGA) and alginate. Fluorescence imaging clearly shows the C-S domain in the MC. For release control, the use of high-molecular-weight PLGA (HMW 270 000) restrains the initial burst release of protein compared to that of low-MW PLGA (LMW 40 000). Layer-by-layer (LBL) assembly of chitosan and alginate on MCs is also useful in controlling the release profile of biomolecules. LBL (7-layer) treatment is effective in suppressing the initial burst release of protein compared to no LBL (0-layer). The difference of cumulative albumin release between HMW (7-layer LBL) and LMW (0-layer LBL) PLGA is determined to be more than 40% on day 5. When dual angiogenic growth factors (GFs), such as platelet-derived GF (PDGF) and vascular endothelial GF (VEGF), are encapsulated separately in the core and shell domains, respectively, the VEGF release rate is much greater than that of PDGF, and the difference of the cumulative release percentage between the two GFs is about 30% on day 7 with LMW core PLGA and more than 45% with HMW core PLGA. As for the angiogenic potential of MC GFs with human umbilical vein endothelial cells (HUVECs), the fluorescence signal of CD31+ suggests that the angiogenic sprout of ECs is more active in MC-mediated GF delivery than conventional GF delivery, and this difference is significant, based on the number of capillary branches in the unit area. This study demonstrates that the fabrication of biocompatible C-S MCs is possible, and that the release control of biomolecules is adjustable. Furthermore, MC-mediated GFs remain in an active form and can upregulate the angiogenic activity of ECs.
Collapse
Affiliation(s)
- Dong Hoon Choi
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea; Department of Biological Science, Korea University, Seoul 136-701, Republic of Korea
| | | | | | | | | |
Collapse
|
34
|
Sato M, Sawahata R, Sakuma C, Takenouchi T, Kitani H. Single domain intrabodies against WASP inhibit TCR-induced immune responses in transgenic mice T cells. Sci Rep 2013; 3:3003. [PMID: 24141565 PMCID: PMC3801110 DOI: 10.1038/srep03003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/04/2013] [Indexed: 11/26/2022] Open
Abstract
Intrabody technology provides a novel approach to decipher the molecular mechanisms of protein function in cells. Single domains composed of only the variable regions (VH or VL) of antibodies are the smallest recombinant antibody fragments to be constructed thus far. In this study, we developed transgenic (Tg) mice expressing the VH or VL single domains derived from a monoclonal antibody raised against the N-terminal domain of Wiskott–Aldrich syndrome protein (WASP), which is an adaptor molecule in immune cells. In T cells from anti-WASP VH and VL single domain Tg mice, interleukin-2 production induced by T cell receptor (TCR) stimulation were impaired, and specific interaction between the WASP N-terminal domain and the Fyn SH3 domain was strongly inhibited by masking the binding sites in WASP. These results strongly suggest that the VH/VL single domain intrabodies are sufficient to knockdown the domain function of target proteins in the cytosol.
Collapse
Affiliation(s)
- Mitsuru Sato
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, 1-2 Ohwashi, Tsukuba, Ibaraki 305-8634, Japan
| | | | | | | | | |
Collapse
|
35
|
Tang R, Kim CS, Solfiell DJ, Rana S, Mout R, Velázquez-Delgado EM, Chompoosor A, Jeong Y, Yan B, Zhu ZJ, Kim C, Hardy JA, Rotello VM. Direct delivery of functional proteins and enzymes to the cytosol using nanoparticle-stabilized nanocapsules. ACS NANO 2013; 7:6667-6673. [PMID: 23815280 PMCID: PMC3757120 DOI: 10.1021/nn402753y] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Intracellular protein delivery is an important tool for both therapeutic and fundamental applications. Effective protein delivery faces two major challenges: efficient cellular uptake and avoiding endosomal sequestration. We report here a general strategy for direct delivery of functional proteins to the cytosol using nanoparticle-stabilized capsules (NPSCs). These NPSCs are formed and stabilized through supramolecular interactions between the nanoparticle, the protein cargo, and the fatty acid capsule interior. The NPSCs are ~130 nm in diameter and feature low toxicity and excellent stability in serum. The effectiveness of these NPSCs as therapeutic protein carriers was demonstrated through the delivery of fully functional caspase-3 to HeLa cells with concomitant apoptosis. Analogous delivery of green fluorescent protein (GFP) confirmed cytosolic delivery as well as intracellular targeting of the delivered protein, demonstrating the utility of the system for both therapeutic and imaging applications.
Collapse
Affiliation(s)
- Rui Tang
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Chang Soo Kim
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - David J. Solfiell
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Subinoy Rana
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Rubul Mout
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Elih M. Velázquez-Delgado
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Apiwat Chompoosor
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Youngdo Jeong
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Bo Yan
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Zheng-Jiang Zhu
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Chaekyu Kim
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Jeanne A. Hardy
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
36
|
Christ GJ, Saul JM, Furth ME, Andersson KE. The pharmacology of regenerative medicine. Pharmacol Rev 2013; 65:1091-133. [PMID: 23818131 DOI: 10.1124/pr.112.007393] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Regenerative medicine is a rapidly evolving multidisciplinary, translational research enterprise whose explicit purpose is to advance technologies for the repair and replacement of damaged cells, tissues, and organs. Scientific progress in the field has been steady and expectations for its robust clinical application continue to rise. The major thesis of this review is that the pharmacological sciences will contribute critically to the accelerated translational progress and clinical utility of regenerative medicine technologies. In 2007, we coined the phrase "regenerative pharmacology" to describe the enormous possibilities that could occur at the interface between pharmacology, regenerative medicine, and tissue engineering. The operational definition of regenerative pharmacology is "the application of pharmacological sciences to accelerate, optimize, and characterize (either in vitro or in vivo) the development, maturation, and function of bioengineered and regenerating tissues." As such, regenerative pharmacology seeks to cure disease through restoration of tissue/organ function. This strategy is distinct from standard pharmacotherapy, which is often limited to the amelioration of symptoms. Our goal here is to get pharmacologists more involved in this field of research by exposing them to the tools, opportunities, challenges, and interdisciplinary expertise that will be required to ensure awareness and galvanize involvement. To this end, we illustrate ways in which the pharmacological sciences can drive future innovations in regenerative medicine and tissue engineering and thus help to revolutionize the discovery of curative therapeutics. Hopefully, the broad foundational knowledge provided herein will spark sustained conversations among experts in diverse fields of scientific research to the benefit of all.
Collapse
Affiliation(s)
- George J Christ
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | | | | | | |
Collapse
|
37
|
Kant V, Gupta V, Gupta AR. Synthesis, Characterization and Biomedical Applications of Nanoparticles. ACTA ACUST UNITED AC 2013. [DOI: 10.5567/sciintl.2013.167.174] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
38
|
Aranda A, Sequedo L, Tolosa L, Quintas G, Burello E, Castell J, Gombau L. Dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay: A quantitative method for oxidative stress assessment of nanoparticle-treated cells. Toxicol In Vitro 2013; 27:954-63. [DOI: 10.1016/j.tiv.2013.01.016] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 10/24/2012] [Accepted: 01/11/2013] [Indexed: 10/27/2022]
|
39
|
Tautzenberger A, Kovtun A, Ignatius A. Nanoparticles and their potential for application in bone. Int J Nanomedicine 2012; 7:4545-57. [PMID: 22923992 PMCID: PMC3423651 DOI: 10.2147/ijn.s34127] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biomaterials are commonly applied in regenerative therapy and tissue engineering in bone, and have been substantially refined in recent years. Thereby, research approaches focus more and more on nanoparticles, which have great potential for a variety of applications. Generally, nanoparticles interact distinctively with bone cells and tissue, depending on their composition, size, and shape. Therefore, detailed analyses of nanoparticle effects on cellular functions have been performed to select the most suitable candidates for supporting bone regeneration. This review will highlight potential nanoparticle applications in bone, focusing on cell labeling as well as drug and gene delivery. Labeling, eg, of mesenchymal stem cells, which display exceptional regenerative potential, makes monitoring and evaluation of cell therapy approaches possible. By including bioactive molecules in nanoparticles, locally and temporally controlled support of tissue regeneration is feasible, eg, to directly influence osteoblast differentiation or excessive osteoclast behavior. In addition, the delivery of genetic material with nanoparticulate carriers offers the possibility of overcoming certain disadvantages of standard protein delivery approaches, such as aggregation in the bloodstream during systemic therapy. Moreover, nanoparticles are already clinically applied in cancer treatment. Thus, corresponding efforts could lead to new therapeutic strategies to improve bone regeneration or to treat bone disorders.
Collapse
Affiliation(s)
- Andrea Tautzenberger
- Institute of Orthopedic Research and Biomechanics, Centre of Musculoskeletal Research, Ulm University, Ulm, Germany.
| | | | | |
Collapse
|
40
|
Balmert SC, Little SR. Biomimetic delivery with micro- and nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2012; 24:3757-78. [PMID: 22528985 PMCID: PMC3627374 DOI: 10.1002/adma.201200224] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Indexed: 05/16/2023]
Abstract
The nascent field of biomimetic delivery with micro- and nanoparticles (MNP) has advanced considerably in recent years. Drawing inspiration from the ways that cells communicate in the body, several different modes of "delivery" (i.e., temporospatial presentation of biological signals) have been investigated in a number of therapeutic contexts. In particular, this review focuses on (1) controlled release formulations that deliver natural soluble factors with physiologically relevant temporal context, (2) presentation of surface-bound ligands to cells, with spatial organization of ligands ranging from isotropic to dynamically anisotropic, and (3) physical properties of particles, including size, shape and mechanical stiffness, which mimic those of natural cells. Importantly, the context provided by multimodal, or multifactor delivery represents a key element of most biomimetic MNP systems, a concept illustrated by an analogy to human interpersonal communication. Regulatory implications of increasingly sophisticated and "cell-like" biomimetic MNP systems are also discussed.
Collapse
Affiliation(s)
- Stephen C Balmert
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | | |
Collapse
|
41
|
Zhai W, He C, Wu L, Zhou Y, Chen H, Chang J, Zhang H. Degradation of hollow mesoporous silica nanoparticles in human umbilical vein endothelial cells. J Biomed Mater Res B Appl Biomater 2012; 100:1397-403. [DOI: 10.1002/jbm.b.32711] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 02/20/2012] [Accepted: 03/11/2012] [Indexed: 12/14/2022]
|
42
|
Shah NJ, Hong J, Hyder MN, Hammond PT. Osteophilic multilayer coatings for accelerated bone tissue growth. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2012; 24:1445-1450. [PMID: 22311551 PMCID: PMC3870474 DOI: 10.1002/adma.201104475] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 12/30/2011] [Indexed: 05/27/2023]
Abstract
Osteophilic modular nanostructured multilayers containing hydroxyapatite nanoparticles complexed with a natural polymer chitosan create an osteoconductive surface for mesenchymal stem cells (MSCs). Coupled with the sustained release of physiological amounts of osteoinductive bone morphogenetic protein over several days from degradable poly(β-amino ester) based multilayers, this single coating results in a synergistic accelerated and upregulated differentiation of MSCs into osteoblasts laying down new bone tissue on orthopedic implants.
Collapse
|
43
|
Santos T, Maia J, Agasse F, Xapelli S, Ferreira L, Bernardino L. Nanomedicine boosts neurogenesis: new strategies for brain repair. Integr Biol (Camb) 2012; 4:973-81. [DOI: 10.1039/c2ib20129a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
44
|
Varna M, Ratajczak P, Ferreira I, Leboeuf C, Bousquet G, Janin A. <i>In vivo</i> Distribution of Inorganic Nanoparticles in Preclinical Models. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/jbnb.2012.322033] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Paulo CSO, Pires das Neves R, Ferreira LS. Nanoparticles for intracellular-targeted drug delivery. NANOTECHNOLOGY 2011; 22:494002. [PMID: 22101232 DOI: 10.1088/0957-4484/22/49/494002] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Nanoparticles (NPs) are very promising for the intracellular delivery of anticancer and immunomodulatory drugs, stem cell differentiation biomolecules and cell activity modulators. Although initial studies in the area of intracellular drug delivery have been performed in the delivery of DNA, there is an increasing interest in the use of other molecules to modulate cell activity. Herein, we review the latest advances in the intracellular-targeted delivery of short interference RNA, proteins and small molecules using NPs. In most cases, the drugs act at different cellular organelles and therefore the drug-containing NPs should be directed to precise locations within the cell. This will lead to the desired magnitude and duration of the drug effects. The spatial control in the intracellular delivery might open new avenues to modulate cell activity while avoiding side-effects.
Collapse
Affiliation(s)
- Cristiana S O Paulo
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, Portugal
| | | | | |
Collapse
|